Correlation between renal metabolism and cardiovascular disease | Metabolism and Translational Medicine

Correlation between renal metabolism and cardiovascular disease

Authors

  • Siying Deng China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
  • Wei Huang Department of Cardiology, General Hospital of Central Theater Command of PLA, Wuhan 430070, Hubei Province, China

DOI:

https://doi.org/10.54844/mtm.2023.0351

Keywords:

cardiovascular disease, atherosclerosis, heart failure, chronic kidney disease, renal metabolism

Abstract

Cardiovascular disease is the main medical burden in the world, and coronary heart disease and heart failure are the main reasons for the increase of all-cause mortality and cardiovascular mortality year by year. There is a synergistic effect between cardiovascular and kidney diseases. Under pathological conditions, there is a significant correlation between various cardiovascular diseases and kidney diseases. In this review, we discussed the effects of metabolite accumulation caused by kidney disease on multiple mechanisms of atherosclerosis from the whole process of the formation and development of atherosclerosis. These metabolites can be mainly divided into uremic toxins, intestinal metabolites, fibroblast growth factor 23, and advanced glycosylation end products. Then we summarized the effects of metabolites related to renal metabolism on heart failure according to the biological changes of patients with heart failure at three different levels.

Downloads

Published

2023-08-31

How to Cite

1.
Deng S, Huang W. Correlation between renal metabolism and cardiovascular disease. Metabol Transl Med. 2023;1. doi:10.54844/mtm.2023.0351

Issue

Section

Review Article

Downloads

Download data is not yet available.
REVIEW ARTICLE

Correlation between renal metabolism and cardiovascular disease


Siying Deng1, Wei Huang2,3,4,*

1China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China

2Department of Cardiology, General Hospital of Central Theater Command of PLA, Wuhan 430070, Hubei Province, China

3Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China

4Wuhan Clinical Medical Research Center for Hypertension, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China


*Corresponding Author:

Wei Huang, Department of Cardiology, General Hospital of Central Theater Command of PLA, No.627, Wuluo Road, Wuchang District, Wuhan 430070, Hubei Province, China. Email: huangwei0521@126.com; https://orcid.org/0000-0001-6136-6653


Received: 21 February 2023 Revised: 2 August 2023 Accepted: 25 August 2023 Published: 31 August 2023


ABSTRACT

Cardiovascular disease is the main medical burden in the world, and coronary heart disease and heart failure are the main reasons for the increase of all-cause mortality and cardiovascular mortality year by year. There is a synergistic effect between cardiovascular and kidney diseases. Under pathological conditions, there is a significant correlation between various cardiovascular diseases and kidney diseases. In this review, we discussed the effects of metabolite accumulation caused by kidney disease on multiple mechanisms of atherosclerosis from the whole process of the formation and development of atherosclerosis. These metabolites can be mainly divided into uremic toxins, intestinal metabolites, fibroblast growth factor 23, and advanced glycosylation end products. Then we summarized the effects of metabolites related to renal metabolism on heart failure according to the biological changes of patients with heart failure at three different levels.

Key words: cardiovascular disease, atherosclerosis, heart failure, chronic kidney disease, renal metabolism

INTRODUCTION

Cardiovascular diseases (CVDs) are the main cause of death and burden of disease globally.[1] Due to various types and multiple related risk factors, the research on CVDs has never been overlooked. A study evaluated the trend of the burden of CVDs around the world from 1990 to 2019.[2] This article introduced 13 CVDs such as ischemic heart disease (IHD), stroke, hypertensive heart disease and rheumatic heart disease, and analyzed the epidemiology and indicators of burden of disease regarding 9 risk factors including high systolic blood pressure, high low-density lipoprotein cholesterol (LDL-C), high body mass index (BMI), and renal function impairment. Compared with the past, due to the change of lifestyle and the aging of society, the diagnosis, treatment, and prevention of CVD, we are facing are still full of challenges even if the medical conditions continue to be optimized.

The heart is the organ with the highest requirement for metabolism in the human body.[3] As the basis of most CVDs,[4] the disorder of heart energy metabolism can directly lead to cardiac dysfunction and eventually form organic changes. Utilizing the most advanced metabolomics techniques allows us to measure thousands of metabolites in biological fluids or living tissues, providing us with an individual's metabolic fingerprint.[3] Under different physiological and pathological conditions, these metabolites can reflect changes in the metabolism of various organs and tissues of the body. Further analysis can reveal the connection between cardiac metabolism and metabolism of other organs and tissues.

Among various metabolites, some link cardiovascular events with impaired renal function. As early as 1836, Bright[5] proposed a link between chronic kidney disease (CKD) and cardiovascular abnormalities. Since then, many studies have confirmed and extended this association, and given different explanations. Patients with a glomerular filtration rate (GFR) less than 60 mL/min/1.73 m² have approximately double the risk of developing heart failure compared with those with preserved GFR.[6] There is also an increased risk of stroke,[7] peripheral artery disease,[8] coronary artery disease (CAD), and atrial fibrillation.[9]

On the one hand, the kidneys excrete metabolites and harmful substances in the body through the production of urine to keep the internal environment of the body stable. Therefore, when the kidney function gradually loses, it is accompanied by the retention of a large number of metabolites. Many of these metabolites have been shown to be biologically active, thereby affecting cellular functions and metabolic processes, leading to uremic syndrome.[10] On the other hand, the kidneys can secrete renin and prostaglandins to regulate blood pressure, secrete erythropoietin to act on the bone marrow hematopoietic system, and promote the activation of vitamin D to promote calcium and phosphorus absorption and bone growth. In summary, the excretory function and endocrine function of the kidney play a key role in regulating the metabolic state of the body. However, only a few national and international guidelines on cardiovascular risk management pay special attention to CKD as a significant risk factor,[11] and the link between renal metabolism and CVD deserves greater attention.

This review will start with several classic CVDs such as CAD and heart failure. Then, the review demonstrates how several important metabolites related to renal metabolism affect the occurrence, development, and outcome of CVDs. The review is intended to provide reference for the development and application of therapeutics.

EPIDEMIOLOGY DEMONSTRATES SIGNIFICANT ASSOCIATION BETWEEN CARDIOVASCULAR DISEASE AND RENAL INSUFFICIENCY

Previous epidemiological surveys have pointed out that patients with CKD have increased cardiovascular risk and mortality, and conversely, cardiovascular events are the most common cause of death in patients with CKD.[12] The nature and extent of CVD of individuals with CKD have been considered distinct from those of individuals without renal disease, including atherosclerosis, arteriosclerosis, calcified arterial and valvular disease, left ventricular remodeling and dysfunction, cardiac arrhythmias, and sudden cardiac death.[10] In 2004, Go et al.[13] evaluated a longitudinal GFR of 1,120,295 adults who measured serum creatinine between 1996 and 2000 and did not undergo dialysis or kidney transplantation in a large general health care system. Multivariate associations were found between estimated GFR and risk of death, cardiovascular events, and hospitalization. Kottgen et al.[6] pairs of 14,857 participants, from 1987 to 2002, followed up the events of heart failure hospitalization or death and found that middle-aged people with moderate or severe renal dysfunction are high-risk groups for heart failure. Reboldi et al.[14] analyzed 8794 participants with a mean age of 52 in 8 prospective studies. In a multivariate Cox model including age, sex, 24-hour mean blood pressure, smoking, diabetes, and cholesterol, and high estimated glomerular filtration rate (eGFR), the risk of cardiovascular events in participants with low eGFR was significantly higher than that in participants with normal eGFR, which further verified the conclusion that the curves describing the relationship between GFR and cardiovascular outcomes and GFR and all-cause mortality were u-shaped.[1517] It shows that the rise and decline of GFR increase CVD risk and total mortality to the same extent. Compared with long-term dialysis treatment, successful kidney transplantation significantly reduces the risk of death from cardiovascular events.[18]

CORONARY HEART DISEASE AND RENAL METABOLISM

CAD is an important component of morbidity and mortality around the world.[19] According to Global Burden of Diseases (GBD) data, the global prevalence of CAD in 2016 was 154 million, accounting for 32.7% of the global burden of CVD and 2.2% of the total global burden of disease.[20] In 2019, the number of patients with IHD reached 197 million and the number of deaths was 9.14 million, and the absolute number is still increasing.

Atherosclerosis, as one of the main causes of CAD,[21] refers to the damage of the arterial intima due to various reasons, with lipid deposition, foam cell formation, and smooth muscle cell and fiber component proliferation, gradually forming plaques in the arterial wall. In the plaque, tissue necrotizes and disintegrates, and then combines with deposited lipids to form atheroma.[22] Clinical studies point out that renal insufficiency is associated with the severity of coronary atherosclerosis, the incidence of coronary events, and the mortality rate after myocardial infarction. Nakano et al.[23] investigated the relationship between CKD and severity of coronary atherosclerosis in a population-based autopsy sample. According to the research, a progressive increase in the frequency of advanced atherosclerotic lesions and an increase in the frequency of coronary calcified lesions are noticed as eGFR decreases. Bae et al.[24] conducted a retrospective cohort study to eGFR in 12,636 patients with acute myocardial infarction (AMI) and concluded that eGFR was independently associated with mortality and complications after AMI. Interpretation of the epidemiology of CAD is complicated due to the frequent coexistence of CKD and diabetes, which is also a predisposing factor for cardiac events.[25] Nevertheless, even with modest impairment of eGFR, the risk of myocardial infarction is still increased in nondiabetic patients.[26] Epidemiological evidence suggests that the progression of atherosclerosis in CKD cannot be fully explained by traditional CVD risk factors such as hypertension, diabetes, and dyslipidemia, meaning that there are also nontraditional risk factors that contribute to atherosclerosis in CKD Sclerosis formation, including proteinuria, accumulation of uremic toxins, etc.[25]

Atherosclerosis is a dynamic process, and changes in the microenvironment at each time point can affect the occurrence of outcome events. Therefore, when we explore the relationship between renal metabolism and CAD under pathological conditions, we can find the key points more carefully and logically according to the occurrence process of atherosclerosis.

Renal insufficiency can induce dyslipidemia

One of the mechanisms of atherosclerosis is lipid infiltration. Many factors can cause endothelial damage and increase the permeability of low-density lipoprotein (LDL), which accumulates under the endothelium. The combination of excessively deposited LDL and proteoglycans makes it easier to be oxidized or chemically modified in other ways to facilitate macrophage recognition and phagocytosis, and eventually get removed. Monocytes are stimulated by oxidized LDL and other stimuli to migrate to the subendothelium to differentiate into macrophages, and further recognize and phagocytize the modified oxidized LDL particles through scavenger receptors. Finally, foam cells are formed. A large number of foam cells die to form lipid pools and develop into typical atheromatous plaque.[22]

In previous studies, it has been reported that patients with chronic renal failure and experimental animals often have elevated plasma triglycerides, impaired clearance of very low-density lipoprotein (VLDL) and chylomicrons, sustained reduction in plasma high-density lipoprotein (HDL) concentration, and impaired maturation of cholesterol-ester-rich cardioprotective HDL-2 from HDL-3 with low level of cholesterol ester.[2729] In addition to changes in the content of plasma protein itself, its composition has also changed. A decrease in cholesteryl ester content and a relative increase in triglyceride content in several lipoproteins suggests a redistribution of cholesterol from HDL to VLDL and LDL, as well as a defect in the removal of triglyceride from LDL and HDL particles. However, other factors that accompany CKD, such as diabetes, insulin resistance, metabolic syndrome, obesity, and overt proteinuria, may enhance dyslipidemia and high triglyceride lipoprotein cholesterol, making it difficult to determine a causal independent effect of CKD on lipid abnormalities.[30]

Studies have indicated that strategies to reduce traditional risk factors for cardiac adverse events in the general population, such as LDL-C, do not benefit patients with CKD.[31] According to the Study of Heart and Renal Protection (SHARP), compared with placebo, although simvastatin plus ezetimibe reduces the composite cardiovascular outcome in subjects with CKD by 17%, it does not reduce all-cause mortality or CAD mortality.[32] A sub-study of the Treating to New Targets (TNT) trial shows that lowering LDL cholesterol with atorvastatin in CKD reduces the relative risk of major cardiovascular events in CKD patients.[33] Triglyceride-lowering drugs such as gemfibrozil and fenofibrate have been shown to be cardioprotective in patients with non-dialysis CKD.[34,35] These results suggest that LDL plays a more important role than high-density lipoprotein cholesterol (HDL-C) in CVD in CKD. Besides, triglyceride may also play a role, but there is no evidence to prove it. Studies of cholesteryl ester transfer protein (CETP) inhibition, such as torcetrapib and dalcetrapib, fail to demonstrate improved cardiovascular outcomes, although they successfully increase HDL-C levels.[36,37] The recently completed Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events (HPS2-THRIVE) study is the largest prospective randomized trial testing niacin in patients at high risk of cardiovascular events. However, it failed to demonstrate a reduction in the risk of major vascular events.[38]

Renal insufficiency promotes the develop-ment of insulin resistance

Insulin resistance is a core characteristic of several common human diseases such as type 2 diabetes and primary hypertension, which are potential risk factors for coronary heart disease. Epidemiological studies have shown that up to 40% of patients with coronary heart disease in whites may have diseases related to insulin resistance, while in South Asia, nearly 60% of the risk of coronary heart disease may be associated with insulin resistance.[39] Dugani et al. explored the baseline risk status of female coronary heart disease development based on the age of onset and found that insulin resistance appears to be one of the strongest risk factors for early-onset coronary heart disease.[40] In different community populations, insulin resistance is associated with an increased risk of cardiovascular disease events, regardless of whether it is directly measured, estimated using fasting insulin concentration (such as in homeostatic model assessment), or calculated through dynamic tests (such as the oral glucose tolerance test).[41] In recent years, Derek Klarin's team conducted a genome-wide association study, testing the association between approximately 9 million DNA sequence variations and CAD, and identified a new gene locus CCDC92 that may affect CAD through the insulin resistance pathway.[42]

In physiological conditions, insulin plays a regulatory role in cellular metabolism by enhancing glucose utilization in insulin-sensitive tissues, and also regulates vascular relaxation of small blood supplying arteries by activating endothelial nitric oxide synthase (eNOS) through the phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathway.[43] Insulin resistance is defined as a decreased tissue response to insulin stimulation and is thus characterized by defects in glucose uptake and oxidation, decreased glycogen synthesis, the ability to inhibit lipid oxidation to a lesser extent, and vasoconstriction.[44] Insulin resistance is closely associated with mitochondrial dysfunction, reactive oxygen species (ROS) production, endothelial dysfunction, and other cardiovascular risk factors, although it is difficult to disentangle the causal relationships between these processes.[45]

Research has shown that patients with CKD begin to develop insulin resistance before the onset of a decrease in GFR,[46] but the specific cause is not yet determined. Renal function impairment may be promoted by residual uremic toxins, acidosis, and deficiency of active vitamin D, while insulin resistance can lead to renal function impairment by damaging glomerular endothelial and epithelial cells.[41,47] In one epidemiological study, insulin resistance was found to be an independent risk factor for the development of CKD and the rapid decline of eGFR in the Korean population.[48]

Metabolic disorders caused by renal insufficiency can lead to endothelial dysfunction and can aggravate inflammation and oxidative stress

The functions of the endothelium in a healthy state include dynamic maintenance of vascular tone, angiogenesis, hemostasis, and providing an antioxidant, anti-inflammatory, and antithrombotic interface. Vascular endothelial dysfunction is manifested by impaired endothelium-dependent vasodilation, enhanced oxidative stress, chronic inflammation, leukocyte adhesion and hyperpermeability, and endothelial cell senescence.[49] In a cross-sectional study of patients with peripheral arterial disease (PAD), endothelial function is assessed by flow-mediated vasodilation in the brachial artery. It is estimated that GFR was significantly correlated with endothelial function. Under such a background, renal function can be described as one of the determinants of endothelial dysfunction.[50]

Uric acid (UA)

UA is the final product of purine metabolism in higher animals. It is mainly synthesized in the liver, intestine, and vascular endothelium. The kidney plays a leading role in the excretion of UA. The kidney excretes about 70% of the daily UA produced, and the rest 30% of UA is excreted by the gut.[51] When UA production exceeds UA excretion, hyperuricemia, defined as a serum UA concentration > 7.0 mg/dL, occurs. Various transporters expressed in the renal proximal tubules mediate UA exchange. These molecules include glucose transporter 9 (GLUT9), uric acid transporter 1 (URAT1), the human adenosine triphosphate-binding cassette subfamily G 2 (ABCG2), organic anion transporter (OAT) 1, 3, and 4.[52] A meta-analysis[53] including 15 studies shows that the carotid intima-media thickness (CIMT) in the high UA group is significantly higher than that in the control group, and further studies indicate that high UA is significantly correlated with the presence of carotid atherosclerotic plaques.[52] Based on a meta-analysis of 29 prospective cohort studies, Li et al.[54] verified that hyperuricemia may increase the risk of CAD events. For every 1 mg/dL increase in UA level, the pooled multivariable relative risk of CAD mortality is 1.13 (95% confidence intervals [CI] 1.06–1.20). Dose-response analysis shows that the combined relative risk for CHD mortality is 1.02 (95% CI 0.84–1.24) for a 1 mg UA increase per liter. There is research incubating human umbilical vein endothelial cells (HUVECs) with UA, ROS scavenger polyethylene glycol superoxide dismutase (PEG-SOD), endoplasmic reticulum (ER) stress inhibitor 4-phenylbutyric acid (4-PBA), and the protein kinase C (PKC) inhibitor polymyxin B. The research assesses nitric oxide (NO) production, eNOS activity, intracellular ROS, ER stress levels, and the interaction between eNOS and calmodulin (CaM) and cytosolic calcium levels. It is demonstrated that UA triggers oxidative stress and ER stress through PKC/eNOS-mediated eNOS activity and NO production, thereby inducing HUVEC apoptosis and endothelial dysfunction.[55] It is reported that high concentrations of UA can induce endothelial dysfunction through the high mobility group protein B1/receptor for advanced glycosylation end-product (HMGB1/RAGE) signaling pathway.[56] In addition, hyperuricemia-induced endothelial dysfunction is also been validated in many clinical studies.[57,58]

Urea

Urea is a toxic end product produced when amino acids and other nitrogen-containing metabolites are broken down in the liver. It is normally excreted by the kidneys. However, patients with end-stage renal disease (ESRD) are unable to urinate and thus have continuously increased blood urea concentrations. This condition can only be treated with intermittent hemodialysis or kidney transplantation. Although hemodialysis can improve uremia in patients with ESRD, it can only replace about 10% of normal renal function. Therefore, these patients still suffer from chronic urea overload.

As kidney function declines, accumulated urea spontaneously decomposes to form cyanate, which undergoes an irreversible non-enzymatic reaction with proteins and free amino groups, called carbamylation.[59] There is evidence that protein carbamylation induced by chronically elevated blood urea is associated with the risk of CVD in patients.[60] Studies with tissue culture and animal models also show that urea and protein carbamylation may contribute to CVD.[59] Carbamylated albumin has pro-inflammatory properties and is more nephrotoxic than albumin and traditional albumin modifications.[61] Apostolov et al.[62] demonstrated that carbamylated low-density lipoprotein (cLDL) can promote atherosclerosis by causing dysfunction of human endothelial cells through the lectin-like, oxidized low-density lipoprotein receptor-1 (LOX-1) and partially CD36, scavenger receptor expressed by endothelial cells 1 (SREC1) and scavenger receptors A1 (SR-A1) receptors. The research team also demonstrate that intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) play important roles in cLDL-activated human vascular endothelial monocyte adhesion in vitro.[63] Schreier et al.[64] found that carbamylation completely abolishes the proteolytic and cytoprotective effects of threonine against hypochlorous acid (HOCl) challenge, thus carbamylation may contribute to oxidative stress. Urea itself is also shown to induce ROS in cell culture studies.[25]

Free amino acids are competitive inhibitors of protein carbamylation. Studies manifest that amino acid scavenger therapy can reverse protein carbamylation.[65] Intensification of hemodialysis treatment also reduces protein carbamylation and may partly explain the benefit seen when dialysis dose (i.e., frequency) is increased. The impact of reduced protein carbamylation on clinical outcomes awaits further study and is a promising target for intervention.

Indoxyl sulfate (IS)

IS is a uremic toxin metabolized by dietary tryptophan. In tubular epithelial cells, IS is excreted from blood into urine by OATs and organic anion-transporting polypeptides (OATPs).[66] Clinical evidence suggests that IS is an independent risk factor for cardiovascular morbidity and mortality in patients with CKD.[67] Itoh et al.[68] revealed elevated levels of 11 protein-bound uremic toxins in the serum of hemodialysis patients. IS, p-cresol sulfate, and 3-carboxy-4-methyl-5-propyl-2-furan propionic acid (CMPF) cannot be effectively removed by hemodialysis due to high protein binding, among which indoxyl phenyl sulfate has the strongest induction effect on ROS production in endothelial cells. Ito et al.[69] demonstrated that IS enhances leukocyte-endothelial cell interaction and promotes vascular inflammation by upregulating E-selectin, possibly through c-Jun N-terminal kinase (JNK)- and nuclear factor κB (NF-κB)-dependent pathways. Matsuo et al.[70] exposed THP-1 cell-differentiated macrophages to IS in vitro. The result shows that IS reduces the viability of THP-1-derived macrophages and promotes generation of inflammatory cytokines such as interleukin-1 beta (IL-1β), tumor necrosis factor (TNF)-α, and ROS. Nakano et al.[71] proved that human and mouse macrophages express OAT and OATP family transporters through in vivo and in vitro experiments. Additionally, this family of proteins mediates the IS-induced pro-inflammatory activation of macrophages in vitro and in vivo, and accelerates the development of cardiovascular metabolic disorders. The activation of this inflammatory response is achieved through the organic anion transporting polypeptide 2B1 (OATP2B1)-Delta-like 4 (Dll4)-Notch axis.

P-cresol sulfate (PCS)

PCS is a major uremic toxin derived from tyrosine and phenylalanine via hepatic metabolites. It circulates primarily in combination with albumin and is taken up by body cells via OAT1 and/or OAT3.[72] Wu et al.[73] use a prospective cohort study to investigate how serum IS and PCS are related with all-cause mortality and cardiovascular mortality in elderly hemodialysis patients. It is concluded that serum free PCS levels can help predict all-cause and cardiovascular mortality risk in elderly hemodialysis patients beyond traditional and uremia-related risk factors. It is also reported that PCS is associated with adverse clinical effects.[74] Lin et al.[75] demonstrated that serum PCS and IS levels are associated with PAD. In vitro cell experiments reveal that p-cresol's major in vivo metabolite, p-cresol sulfate, has a pro-inflammatory effect on unstimulated leukocytes.[76] Gross et al.[72] demonstrated that PCS induces ROS production in vascular smooth muscle cells (VSMCs), confirming its prooxidative effect on endothelial cells. They also point out that PCS can directly activate Rho kinase to induce constriction of mouse thoracic aorta. Pletinck et al.[77] assessed the effects of acute and sustained exposure to uremic levels of IS, PCS, and p-cresol glucuronide (PCG) on the recruitment of circulating leukocytes in rat peritoneal vascular bed in rats using intravital microscopy. The results show that IS, PCS, and PCG exert pro-inflammatory effects by stimulating crosstalk between leukocytes and blood vessels, leading to vascular injury.

Phosphate

Hyperphosphatemia is very common in patients with ESRD. For many years, hyperphosphatemia has been recognized as an important cause of high CVD morbidity and mortality in CKD patients.[78,79] However, its common pathogenicity of promoting vascular calcification does not explain recent epidemiological findings that higher serum phosphate levels within the normal range may also be a risk factor for CVD in individuals with normal renal function.[80,81] Six et al.[78] found that phosphate had a rapid, concentration-dependent vasoconstrictive effect on the mouse aorta and that this constriction is abrogated by the hydroxyl radical scavenger dimethylthiourea. Furthermore, phosphate increases H2O2 production in human vascular smooth muscle cells (HVSMCs). Thus, uremic toxins such as phosphate can enhance vasoconstriction by releasing oxygen free radicals. Shuto et al.[80] studied the acute effects of phosphorus loading on endothelial function in vitro and in vivo. It is found that exposure of bovine aortic endothelial cells to phosphorus loading increases production of ROS dependent on phosphorus influx through the sodium-dependent phosphate transporter, and meanwhile, reduces NO through inhibition of phosphorylation of eNOS generation. Further studies by Peng et al.[82] pointed out that both hyperphosphatemia and hypophosphatemia decrease eNOS activity by decreasing intracellular calcium and increasing PKCβ2. Hyperphosphatemia also decrease eNOS transcription via signaling through the PI3K/Akt/NF-κB and mitogen-activated protein kinases (MAPK)/NF-κB pathways. It also shows that hyperphosphatemia can increase oxidative stress through increased ROS generation leading to apoptosis of endothelial cells, a process that compromises endothelial cell integrity.[83] Di Marco et al.[84] demonstrated that high phosphate levels unrelated to vascular calcification play a role in the pathophysiology of cardiovascular burden by interfering with endothelial function, thereby impairing microvascular function, angiogenic capacity, and inducing endothelial stiffness.

Sevelamer is a calcium-free phosphate binder that lowers blood phosphate and has pleiotropic effects including correction of lipid abnormalities and reduction of oxidative stress and inflammation. Caglar et al.[85] conducted a small, randomized, prospective study showing that short-term treatment with sevelamer significantly increases fetuin A levels and improves flow-mediated dilation (FMD) in nondiabetic stage 4 CKD patients. Six et al.[78] use multiple approaches to test the therapeutic potential of Sevelamer to prevent phosphate- and CKD-induced endothelial dysfunction and reveal that it can reduce the expression of adhesion molecules (ICAM-1 and VCAM-1).

Fibroblast growth factor 23 (FGF-23)

FGF-23, a bone-derived phosphoturic hormone, reduces renal synthesis of 1,25-dihydroxy vitamin D3 (1,25[OH]2D3). FGF-23 acts primarily by binding to a receptor complex consisting of fibroblast growth factor receptors (FGFRs) and a co-receptor α-Klotho (known as Klotho).[86] In CKD, FGF-23 serum levels gradually increase with a decline in GFR and renal phosphate excretion capacity.[87] Several epidemiological studies note an association between serum FGF-23 levels and systemic atherosclerosis.[8890] Animal experiments show that FGF-23 increases superoxide, inhibits the bioavailability of NO, and affects endothelial vasodilation, leading to aortic endothelial dysfunction in mice.[91] Klotho deficiency in mice can also lead to decreased level of NO synthesis in vascular endothelial cells, affecting endothelial function and causing arteriosclerosis.[86,92]

Asymmetric dimethylarginine (ADMA)

ADMA is an endogenous inhibitor of eNOS. It is that elevated ADMA levels during CKD may be associated with decreased renal excretion and increased production due to dysfunction of the endothelial L-arginine/NO pathway.[93] Meinitzer et al.[94] noted that ADMA concentrations can predict all-cause and cardiovascular mortality in patients with CAD. Mangiacapra et al. demonstrated that serum ADMA levels are an independent predictor of the extent of coronary atherosclerosis. Lee et al.[95] reported that pathological concentrations of UA increase intracellular levels of ADMA, disrupt the balance of the ADMA/dimethylarginine dimethylaminohydrolase 2 (DDAH-2) axis, and result in endothelial cell dysfunction, thereby accelerating atherosclerosis. Several in vitro cell experiments prove that ADMA can enhance the adhesion of monocytes to endothelial cells in vitro and mediate the proinflammatory state of endothelial cells.[9699] There are also animal cell studies showing that ADMA contributes to eNOS suppression through multiple mechanisms, increases ROS burden, and can also reduce ROS disposal, thereby establishing a pathogenic redox state in the vasculature.[100104] Furthermore, studies show that ADMA can significantly upregulate the expression of LOX-1 in monocytes, upregulate cholesterol acyltransferase (ACAT), elevate the uptake of oxidized low density lipoprotein (oxLDL), and drive the formation of foam cells in atherosclerotic plaques.[99,103]

Gut microbiome-related metabolites

Trimethylamine N-oxide (TMAO) is a gut microbiota-derived metabolite formed from the conversion of trimethylamine (TMA) by hepatic flavin monooxygenase 3 (FMO3). Mammals cannot metabolize TMAO, and about 95% of it are excreted by the kidneys. Multiple epidemiological investigations in recent years show that the serum TMAO concentration of CKD patients is elevated, which is strongly negatively correlated with eGFR, indicating that the long-term survival rate is reduced.[105108]

Multiple studies show that TMAO can promote the progression of atherosclerosis.[109] A large clinical cohort study is the first to establish that TMAO can predict CVD risk,[110] and in subsequent animal experiments, a significant increase in atherosclerosis is observed in mice supplemented with TMAO.[110,111] Wang et al.[112] noted that diet-induced atherosclerosis can be affected by using specific inhibitors of TMAO's precursor metabolites, further supporting the pro-atherogenic properties of TMAO. Dietary supplementation of the TMAO precursor choline results in increased atherosclerotic lesion burden, increases aortic expression of scavenger receptors (CD36 and scavenger receptor A), and increases foam cell formation by cholesterol-rich macrophages.[113] Studies show that TMAO can promote vascular endothelial cell pyroptosis through ROS induced by upregulation of succinate dehydrogenase complex subunit B (SDHB),[114] and can also induce vascular injury by affecting the distribution of bile acids.[115] Interestingly, although TMAO has increasing evidence of its association with CVD events, its role in atherosclerosis remains controversial. In two acknowledged mouse models of atherosclerosis, apolipoprotein E (ApoE)-/- and low density lipoprotein receptor (Ldlr)-/- mice, dietary choline supplementation does not alter atherosclerotic lesion size.[116,117] In addition, in some recent population-based clinical studies, the positive association between cardiovascular risk and TMAO levels is not confirmed.[118,119] There are many reasons for the controversy, such as the difference in animal cells used by different research groups, the duration of the experiment, etc., which are also affected by many factors in human research. Overall, TMAO is only one of many factors that affect atherosclerosis and its complications.

Phenylacetylglutamine (PAGln) is also a metabolite produced by intestinal microorganisms and mainly excreted by the kidneys. When the kidney function is impaired, it can accumulate in the body. Like TMAO, it belongs to the gut-derived uremic toxin (GDUT).[120,121] Multiple epidemiological studies point out an independent association between PAGln and the burden of coronary atherosclerosis, as well as its correlation with major adverse cardiovascular events.[122124]

Advanced glycation end products (AGEs)

AGEs are a heterogeneous group of compounds. They are representative metabolites of T2DM and are derived from non-enzymatic glycosylation of proteins, lipids, and nucleic acids through a complex reaction called the Maillard reaction, followed by further sugar oxidation reactions under oxidative stress conditions.[125] AGEs are metabolized and cleared by the kidneys, filtered through the glomeruli, and reabsorbed by the renal proximal tubules, so the gradual retention of AGEs that occurs with the decline of renal function will cause a vicious cycle of renal damage and accelerated decline of renal function.[10] Epidemiological investigations show that AGE and circulating receptor for AGEs (RAGE) levels are independently associated with decreased GFR.[126]

Several studies manifest that AGEs are highly correlated with adverse cardiovascular events caused by atherosclerosis.[127,128] Modifications of extracellular proteins such as collagen, elastin, and laminin by AGEs can alter the structure, function, and properties of normal tissues and trigger inflammatory responses. Haitoglou et al.[129] demonstrated that glycosylation of laminin and collagen IV impairs endothelial cell adhesion and migration by disrupting cell attachment sites. The formation of AGEs on vascular wall and myocardial collagen leads to mutual cross-linking of collagen molecules, which leads to the loss of collagen elasticity, which in turn leads to a decrease in vascular wall flexibility and vascular stiffness.[130] In cells, AGEs can accumulate in the ER and bind to mitochondrial proteins, increasing superoxide and ROS production.[131,132]

Metabolic disturbance caused by renal insufficiency can induce vascular smooth muscle cell type transformation and abnormal function

VSMCs reside in the media of arterial vessels, maintaining arterial structural integrity and vascular tone. A range of phenotypes are identified in VSMCs, exhibiting a contractile phenotype under physiological conditions. When challenged with noxious stimuli, VSMCs lose their contractile properties and switch to a synthetic phenotype, which is critical for atherosclerotic plaque formation and progression.[133]

Ok et al.[134] demonstrated that cLDL induces dose-dependent vascular cell injury associated with atherosclerosis, including vascular smooth muscle cell proliferation and endothelial cell death. Yuan et al.[135] use in vitro cell experiments to prove that AGEs can activate the PI3K/Akt pathway through RAGE, thereby promoting the proliferation and migration of human aortic vascular smooth muscle cells (HASMC). Studies by Xing et al.[133] showed that AGEs can induce M1-type polarization of plaque macrophages through activation of the RAGE/toll-like receptor 4 (TLR4) pathway. In this process, activated RAGE/TLR4 signaling promotes Dll4 expression in this plaque M1 macrophage subset through the extracellular regulated protein kinases/forkhead box protein c2 (ERK/FOXC2) pathway. Through direct cell-to-cell contact, Dll4 is expressed on macrophage-mediated VSMCs to achieve contractile phenotype switching through the Notch pathway, thereby promoting atherosclerosis.

Metabolic disorders caused by renal insufficiency can lead to vascular calcification and thrombosis

Vascular calcification is a mineral metabolism disorder that is very common in patients with CKD,[136] occurring in the early stages of CKD and becoming increasingly severe as the disease progresses. It can lead to the development of atherosclerosis and plaque rupture,[137,138] and is one of the main risk factors for cardiovascular death.[139] Vascular calcification is the abnormal deposition of phosphate (Pi) salts in vascular tissue (including blood vessels, valves, and the heart), and its development involves complex pathological mechanisms, including osteogenic differentiation and apoptosis of vascular smooth muscle cells, instability and release of calcium and phosphate from extracellular vesicles, and elastic fiber degradation.[136]

Multiple studies have shown that there is a pathogenic link between hyperphosphatemia and the associated development of vascular calcification.[140,141] Phosphate-induced endothelial dysfunction and vascular calcification are considered key inducing factors for atherosclerosis in patients with CKD.[142] With the decline in renal function and glomerular unit mass, parathyroid hormone (PTH) and FGF-23 are synthesized and secreted as early as CKD stages 2 and 3 in response to relative phosphate overload. Although it is widely believed that FGF-23 levels are elevated in CKD and are associated with renal function impairment and mineral metabolism disorders, the role of FGF-23 in vascular smooth muscle cell (SMC) matrix calcification remains controversial.[136,143] Zhao et al.[139] demonstrated that tumor necrosis factor (TNF) activates NF-κB promotes inorganic phosphate induced calcification of human aortic smooth muscle cells. Thi Nguyen's team demonstrated that high Pi increases mitochondrial phosphate carrier (PiC) abundance by activating protein translation dependent on ERK1/2-mechanistic target of rapamycin (mTOR) signaling.[144] High Pi can upregulate the abundance of plasma cell Pi transporter PiT-1/2 through ERK1/2, mTOR, and p70 ribosomal protein S6 kinase (p70S6K), and can also stimulate its plasma membrane transport.[145] Upregulation of Pi transporter protein can increase cytoplasmic and mitochondrial ROS, which helps NF-κB inhibitor alpha (inhibitor kappa B alpha, IKB-α) release NF-κB. Enable its transport to the nucleus of VSMCs. NF-κB, as a transcription activator of many osteogenic genes, including Runt-related transcription factor 2 (Runx2) and Osteopontin, plays a core role in vascular calcification.[144]

Multiple clinical studies demonstrate a significant association between circulating TMAO levels and the risk of thrombotic events.[146,147] The mechanism of TMAO in thrombus formation has been studied a lot in recent years. Skye et al.[148] found that transplantation of a subject's human fecal microbiota produces differential TMA and TMAO in the host, accompanying increased platelet aggregation assay reactivity and in vivo thrombotic potential. Zhu et al.[149] pointed out that direct exposure of platelets to TMAO can enhance multiple agonist stimulus-dependent platelet activation by increasing the release of intracellular calcium ions. It is also demonstrated that the key enzyme FMO3, which regulates the last step of the TMAO pathway, can affect platelet reactivity and thrombosis rate.[150] However, since the nature, if any, of the "TMAO receptor" remains unknown, more detailed molecular mechanisms of the exact pathways by which TMAO exerts its pro-inflammatory and pro-thrombotic effects remain elusive.[151] There is also a positive correlation between PAGln levels and thrombotic events in humans. On the basis of previous studies, Nemet et al.[124] verified that PAGln can enhance platelet activation-related phenotypes and thrombosis potential in animal models of arterial injury, and that PAGln can mediate cellular events through G protein-coupled receptors including α2A, α2B, and β2-adrenergic receptors.

HEART FAILURE AND RENAL METABOLISM

Heart failure is a chronic and progressive disease resulting from structural or functional cardiac abnormalities that lead to reduced pumping capacity. It can be categorized as either reduced or preserved left ventricular ejection fraction (LVEF).[152,153] Heart failure, as the primary syndrome, can accompany secondary CKD, and vice versa. Both can coexist based on shared risk factors or systemic disease. In a recent international academic conference on the relationship between heart failure and CKD, it was pointed out that as the severity of CKD increases, the prevalence of heart failure also increases, and that the decrease in eGFR is associated with all-cause mortality, cardiovascular mortality, and an increased risk of hospitalization in patients with heart failure with preserved ejection fraction (HFpEF) or heart failure with reduced ejection fraction (HFrEF).[154]

Heart failure is a multi-dimensional lesion. Changes in systemic circulation volume and blood pressure, organic changes in the heart itself, and abnormalities in circulating endocrine metabolites are all part of it. CKD, which occurs bidirectionally with heart failure, effects all three levels above. In CKD and ESRD, as CKD progresses, blood pressure control is generally disrupted. Simultaneously, salt and water retention leading to excessive preload, and left ventricular hypertrophy (LVH) and fibrosis are expected to develop sequentially. In addition, there are CKD- and end-stage kidney disease (ESKD)-specific factors that affect cardiac systolic and diastolic capacity.[154156] Next, we will mainly explore the mechanism of heart failure due to altered renal metabolism.

The influence of abnormal renal metabolism on circulating volume

Hypertension is one of the main causes of heart failure. Studies show that long-term hypertension (refractory hypertension/renal hypertension) is an important factor leading to heart failure,[157] and one of the most common complications of CKD is renal hypertension.[158] The retention of water and sodium caused by renal insufficiency can cause increased blood volume and increased blood pressure. In addition, the metabolites retained by the decreased renal clearance rate are also confirmed to cause hypertension. Previously, we have discussed that various kidney-related metabolites can cause endothelial dysfunction through impaired NO production. Studies show that endothelial dysfunction, oxidative stress, and elevated endothelin levels are all associated with the pathogenesis of hypertension in CKD patients.[158] Angiotensin II (Ang II) can directly constrict blood vessels, increase systemic vascular resistance and blood pressure. Jiang et al.[159] demonstrated that TMAO promotes Ang II-induced vasoconstriction and the consequent hypertension, which involves the protein kinase R-like endoplasmic reticulum kinase/ROS/calcium–calmodulin dependent protein kinase II/phosphatidylinositol specific phosphoesterase Cβ3 (PERK/ROS/CaMKII/PLCβ3) axis.

Anemia is a common feature of CKD, and the kidney is the main source of erythropoietin. For many years, the hypothesis that erythropoietin deficiency or resistance is the main cause of anemia in CKD patients has been confirmed.[160] Several studies show that anemia affects the prognosis of patients with heart failure.[161163] Thus, under such pathophysiological background, several small clinical studies show that the administration of erythropoietic-stimulating agents (ESAs) to patients with anemic heart failure improves outcome events.[162] However, ESAs do not improve systolic heart failure in a large randomized, double-blind trial.[164] This indicates that anemia may not be a mechanism causing heart failure, but just a mark of heart failure.

Abnormal metabolism of the kidneys can lead to organic changes in the heart

LVH is not a disease in essence. It can be a natural response of the myocardium to aerobic exercise and strength training, but it is often a precursor to heart disease and a pathological response to CVD and hypertension. LVH is an important manifestation in the progression of heart failure, and multiple studies have shown a strong correlation between CKD and LVH.[165] Anemia, as well as sodium and water retention secondary to decreased renal function contribute to volume overload. Metabolic abnormalities associated with CKD are also shown to be risk factors for LVH. In the early stage of CKD, FGF-23 prevents the increase in serum phosphate levels, thereby attenuating phosphate-induced vascular calcification. Whereas in ESRD, FGF-23 can no longer maintain serum phosphate homeostasis. Hyperphosphatemia and elevated FGF-23 levels can promote the development of hypertension, vascular calcification, and LVH through different mechanisms.[166,167] Faul et al.[168] reported that chronically elevated FGF-23 level directly contributes to high LVH morbidity and mortality in CKD patients in a large, ethnically diverse CKD cohort. As renal function declines, serum phosphate levels rise, which in turn induces the secretion of the phosphate hormone FGF-23. This further stimulates FGFR, which can lead to cardiac hypertrophy, alter cardiomyocyte contractility, and increase the risk of cardiac arrhythmic events in cardiomyocytes.[169] Bao et al.[170] found that CKD-related factors such as FGF-23, uremic toxin, and Ang II inhibit the expression of cardiac microRNA-30, an important regulator affecting LVH. This could serve as a new therapeutic target for LVH in CKD, which is beneficial to block the induction of CKD on ventricular hypertrophy.

Another aspect of the organic change of the heart is interstitial fibrosis. Interstitial fibrosis reduces the compliance of the ventricle Since the myocardial contractility cannot exert its proper ejection effect, a vicious circle is formed. Eventually, it leads to irreversible end-stage heart failure. In clinical and animal studies, the severity of renal failure is closely and directly correlated with the degree of cardiac fibrosis.[171,172] Extensive studies point out that transforming growth factor-β1 (TGF-β1) promotes renal fibrosis through the TGF-β1/Smad signaling pathway in CKD.[173] Plus, its elevated level also induces cardiac fibrosis. Targeted therapy for this mechanism can attenuate CKD-induced cardiac fibrosis.[172]

The influence of abnormal renal metabolism on endocrine regulation

In the process of heart failure, the activation of the renin-angiotensin-aldosterone system (RAAS) is one of the neurohumoral regulation mechanisms in the compensatory period of heart failure. Although the RAAS has a compensatory effect, it will also activate and promote the remodeling of the heart and blood vessels. This can aggravate myocardial injury and worsen cardiac function.[174] RAAS activation in CKD also increases absolute mortality and the occurrence of high-risk adverse events.[175] RAAS activation leads to increased aldosterone secretion, and aldosterone-induced downregulation of transient receptor potential melastatin 7 (TRPM7) and associated hypomagnesemia can cause CVD and kidney injury.[176] According to previous studies, TRPM7 mediates anti-inflammatory and anti-fibrotic effects through magnesium ions.[177] In recent years, there have been many studies on targeted therapy for RAAS. Chung et al.[178] pointed out that aldosterone antagonists can reduce proteinuria and systolic blood pressure in adults with mild to moderate CKD, but may increase the risk of hyperkalemia, acute kidney injury, and gynecological coma if combined with angiotensin converting enzyme inhibitors (ACEI) and/or angiotensin II receptor blockers (ARB). It is proved that aldosterone is an enhancer of vascular calcification progression. Hammer et al.[179] noticed a protective effect of aldosterone on vascular calcification in CKD in a prospective randomized controlled clinical trial. Since aldosterone acts as a mineralocorticoid, the mineralocorticoid receptor antagonists, spironolactone and eplerenone, are proved to improve both heart failure and CKD.[180] Another factor in the RAAS that can induce heart failure progression is Ang II. Studies indicate that Ang II can regulate ROS production, mitochondrial dysfunction, expression of pro-inflammatory cytokines, autophagy, apoptosis, and cardiovascular system pathogenesis including hypertension and heart failure.[181] Several clinical studies evaluate the therapeutic effect of angiotensin-neprilysin inhibition.[182,183]

There are two forms of vitamin D in the human body, vitamin D3 (cholecalciferol) and vitamin D2 (ergocalciferol). Vitamin D3 is converted to 25-hydroxyvitamin D3 (25-OH-D3) by hydroxylation in the liver, and then converted to active 1,25-(OH)2D3 in the kidney. Therefore, when kidney function is impaired, vitamin D activation will be severely affected. The role of 1,25-(OH)2D in the regulation of calcium and phosphate homeostasis is central. Heart failure is associated with decreased circulating 1,25-(OH)2D and elevated FGF-23 level.[184] Raising serum vitamin D level have beneficial effects on proteinuria, blood pressure, inflammation, and cardiovascular outcomes.[185187] It is also pointed out that one of the mechanisms of action of vitamin D is that it serves as a negative regulator of the RAAS.[188] Another metabolic disorder brought about by CKD is secondary hyperparathyroidism.[189] Serum parathyroid hormone level correlates significantly with the severity of heart failure.[190,191] Clinically, drug treatments for this symptom, such as the benefits and harms of calcimimetic agents, are also being evaluated continuously.[192]

CONCLUSION

The synergistic effect between cardiovascular and kidney is prominent. Under pathological conditions, there is a significant correlation between a variety of CVDs and renal diseases. This pathophysiological connection between the heart and kidneys led to the first proposal of the concept of "Cardiorenal Syndrome (CRS)" by Bongartz et al.[193] A few years later, European scholars from many countries further refined the definition of CRS, which was divided into five subtypes. In this review, we discussed the multi-mechanism effects of the accumulation of metabolites caused by kidney disease on atherosclerosis from the whole process of atherosclerosis formation and development. These metabolites can be mainly divided into uremic toxins including UA, urea, IS, p-cresol sulfate, phosphate, and ADMA, gut-derived metabolites including TMAO and PAGln, as well as FGF-23 and the end product of advanced glycation. Then we summarized the impact of renal metabolism-related metabolites based on three different levels of biological changes manifested in patients with heart failure.

In fact, besides CAD and heart failure, metabolic abnormalities caused by kidney disease are also risk factors for other CVDs. CKD confers a risk of uremic cardiomyopathy, which is characterized by cardiac hypertrophy, fibrosis, and functional impairment.[194] Accumulating evidence suggests that increased production of FGF-23 and Klotho deficiency are potential major drivers of cardiac remodeling in patients with uremic cardiomyopathy.[195,196] Due to the adverse cardiomyopathy and vasculopathy environment in CKD, the occurrence of arrhythmias, conduction abnormalities, and sudden cardiac death may be exacerbated by electrolyte shifts, divalent ion abnormalities, diabetes, sympathetic overactivity, and inflammation and iron deposition.[197]

Cardiovascular problems and kidney diseases significantly contribute to the global medical burden. Although the etiology and pathogenic mechanism are constantly improving, there is still a long way to go for diagnosis, treatment, and prevention. Because of the inextricable connection between the heart and the kidneys, reasonable and effective treatment should be taken into consideration. A recent meta-analysis indicates a significant benefit of sodium-glucose cotransporter-2 inhibitors (SGLT2i) in reducing both heart failure hospitalizations and renal disease progression.[198] In the future, the development and application of drugs should reduce the types and quantities while achieving more comprehensive, more precise, and longer-lasting clinical effects.

DECLARATION

Author contributions

Deng SY: Conceptualization, Investigation, Resources, Writing—Original draft. Zheng LM: Writing—Review and Editing, Visualization, Supervision. Huang W: Writing—Review and Editing, Visualization, Supervision.

Source of funding

This research received no external funding.

Informed consent

Not applicable.

Ethical approval

Not applicable.

Conflict of interest

The authors declare no conflict of interest.

Data sharing

Not applicable.

REFERENCES

  1. GBD 2019 Diseases and injuries collaborators. Global burden of 369 diseases and injuries in 204 countries and territories, 1990-2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet. 2020;396(10258):1204–1222.    DOI: 10.1016/S0140-6736(20)30925-9    PMID: 33069326
  2. Roth GA, Mensah GA, Johnson CO, et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990-2019: Update From the GBD 2019 Study. J Am Coll Cardiol. 2020;76(25):2982–3021.    DOI: 10.1016/j.jacc.2020.11.010
  3. Ussher JR, Elmariah S, Gerszten RE, Dyck JRB. The emerging role of metabolomics in the diagnosis and prognosis of cardiovascular disease. J Am Coll Cardiol. 2016;68(25):2850–2870.    DOI: 10.1016/j.jacc.2016.09.972    PMID: 28007146
  4. McGarrah RW, Crown SB, Zhang GF, Shah SH, Newgard CB. Cardiovascular metabolomics. Circ Res. 2018;122(9):1238–1258.    DOI: 10.1161/CIRCRESAHA.117.311002    PMID: 29700070
  5. Cases and observations, illustrative of renal disease, accompanied with the secretion of albuminous urine. Br Foreign Med Rev. 1840;10(20):301–329.    PMID: 30161702
  6. Kottgen A, Russell SD, Loehr LR, et al. Reduced kidney function as a risk factor for incident heart failure: the atherosclerosis risk in communities (ARIC) study. J Am Soc Nephrol. 2007;18(4):1307–1315.    DOI: 10.1681/ASN.2006101159    PMID: 17344421
  7. Abramson JL, Jurkovitz CT, Vaccarino V, Weintraub WS, McClellan W. Chronic kidney disease, anemia, and incident stroke in a middle-aged, community-based population: the ARIC Study. Kidney Int. 2003;64(2):610–615.    DOI: 10.1046/j.1523-1755.2003.00109.x    PMID: 12846757
  8. Wattanakit K, Folsom AR, Selvin E, Coresh J, Hirsch AT, Weatherley BD. Kidney function and risk of peripheral arterial disease: results from the Atherosclerosis Risk in Communities (ARIC) Study. J Am Soc Nephrol. 2007;18(2):629–636.    DOI: 10.1681/ASN.2005111204    PMID: 17215445
  9. Gansevoort RT, Correa-Rotter R, Hemmelgarn BR, et al. Chronic kidney disease and cardiovascular risk: epidemiology, mechanisms, and prevention. Lancet. 2013;382(9889):339–352.    DOI: 10.1016/S0140-6736(13)60595-4    PMID: 23727170
  10. Wojtaszek E, Oldakowska-Jedynak U, Kwiatkowska M, Glogowski T, Malyszko J. Uremic toxins, oxidative stress, atherosclerosis in chronic kidney disease, and kidney transplantation. Oxid Med Cell Longev. 2021;2021:6651367.    DOI: 10.1155/2021/6651367    PMID: 33628373
  11. European Guidelines on cardiovascular disease prevention in clinical practice (version 2012): the Fifth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of nine societies and by invited experts). Eur J Prev Cardiol. 2012;19(4):585–667.    DOI: 10.1177/2047487312450228    PMID: 22763626
  12. Wanner C, Lachin JM, Inzucchi SE, et al. Empagliflozin and clinical outcomes in patients with type 2 diabetes mellitus, established cardiovascular disease, and chronic kidney disease. Circulation. 2018;137(2):119–129.    DOI: 10.1161/CIRCULATIONAHA.117.028268    PMID: 28904068
  13. Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med. 2004;351(13):1296–1305.    DOI: 10.1056/NEJMoa041031    PMID: 15385656
  14. Reboldi G, Verdecchia P, Fiorucci G, et al. Glomerular hyperfiltration is a predictor of adverse cardiovascular outcomes. Kidney Int. 2018;93(1):195–203.    DOI: 10.1016/j.kint.2017.07.013    PMID: 28935213
  15. Altay S, Onat A, Özpamuk-Karadeniz F, Karadeniz Y, Kemaloğlu-Öz T, Can G. Renal hyperfiltrators are at elevated risk of death and chronic diseases. BMC Nephrol. 2014;15:160.    DOI: 10.1186/1471-2369-15-160    PMID: 25278185
  16. Adeva-Andany MM, Fernández-Fernández C, Carneiro-Freire N, Castro-Quintela E, Pedre-Piñeiro A, Seco-Filgueira M. Insulin resistance underlies the elevated cardiovascular risk associated with kidney disease and glomerular hyperfiltration. Rev Cardiovasc Med. 2020;21(1):41–56.    DOI: 10.31083/j.rcm.2020.01.5102    PMID: 32259903
  17. Cox HJ, Bhandari S, Rigby AS, Kilpatrick ES. Mortality at low and high estimated glomerular filtration rate values: a ‘U’ shaped curve. Nephron Clin Pract. 2008;110(2):c67–c72.    DOI: 10.1159/000151720    PMID: 18758185
  18. Pesavento TE. Kidney transplantation in the context of renal replacement therapy. Clin J Am Soc Nephrol. 2009;4(12):2035–2039.    DOI: 10.2215/CJN.05500809    PMID: 19850770
  19. Shaya GE, Leucker TM, Jones SR, Martin SS, Toth PP. Coronary heart disease risk: low-density lipoprotein and beyond. Trends Cardiovasc Med. 2022;32(4):181–194.    DOI: 10.1016/j.tcm.2021.04.002    PMID: 33872757
  20. Bauersachs R, Zeymer U, Brière JB, Marre C, Bowrin K, Huelsebeck M. Burden of coronary artery disease and peripheral artery disease: A literature review. Cardiovasc Ther. 2019;2019:8295054.    DOI: 10.1155/2019/8295054    PMID: 32099582
  21. Libby P, Buring JE, Badimon L, et al. Atherosclerosis. Nat Rev Dis Primers. 2019;5:56.    DOI: 10.1038/s41572-019-0106-z
  22. Libby P, Ridker PM, Hansson GK. Progress and challenges in translating the biology of atherosclerosis. Nature. 2011;473(7347):317–325.    DOI: 10.1038/nature10146    PMID: 21593864
  23. Nakano T, Ninomiya T, Sumiyoshi S, et al. Association of kidney function with coronary atherosclerosis and calcification in autopsy samples from Japanese Elders: the Hisayama study. Am J Kidney Dis. 2010;55(1):21–30.    DOI: 10.1053/j.ajkd.2009.06.034    PMID: 19765871
  24. Bae EH, Lim SY, Cho KH, et al. GFR and cardiovascular outcomes after acute myocardial infarction: results from the Korea Acute Myocardial Infarction Registry. Am J Kidney Dis. 2012;59(6):795–802.    DOI: 10.1053/j.ajkd.2012.01.016    PMID: 22445708
  25. Tonelli M, Karumanchi SA, Thadhani R. Epidemiology and mechanisms of uremia-related cardiovascular disease. Circulation. 2016;133(5):518–536.    DOI: 10.1161/CIRCULATIONAHA.115.018713    PMID: 26831434
  26. Tonelli M, Muntner P, Lloyd A, et al. Risk of coronary events in people with chronic kidney disease compared with those with diabetes: a population-level cohort study. Lancet. 2012;380(9844):807–814.    DOI: 10.1016/S0140-6736(12)60572-8    PMID: 22717317
  27. Attman PO, Samuelsson O, Alaupovic P. Lipoprotein metabolism and renal failure. Am J Kidney Dis. 1993;21(6):573–592.    DOI: 10.1016/s0272-6386(12)80030-8    PMID: 8503411
  28. Vaziri ND. Dyslipidemia of chronic renal failure: the nature, mechanisms, and potential consequences. Am J Physiol Renal Physiol. 2006;290(2):F262–F272.    DOI: 10.1152/ajprenal.00099.2005    PMID: 16403839
  29. Heuck CC, Liersch M, Ritz E, Stegmeier K, Wirth A, Mehls O. Hyperlipoproteinemia in experimental chronic renal insufficiency in the rat. Kidney Int. 1978;14(2):142–150.    DOI: 10.1038/ki.1978.101    PMID: 691973
  30. Afshinnia F, Pennathur S. Lipids and cardiovascular risk with CKD. Clin J Am Soc Nephrol. 2020;15(1):5–7.    DOI: 10.2215/CJN.13531119    PMID: 31846933
  31. Cholesterol Treatment Trialists' (CTT) Collaboration, Baigent C, Blackwell L, et al. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet. 2010;376(9753):1670–1681.    DOI: 10.1016/S0140-6736(10)61350-5    PMID: 21067804
  32. Baigent C, Landray MJ, Reith C, et al. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): A randomised placebo-controlled trial. Lancet. 2011;377(9784):2181–2192.    DOI: 10.1016/S0140-6736(11)60739-3    PMID: 21663949
  33. Shepherd J, Kastelein JJ, Bittner V, et al. Intensive lipid lowering with atorvastatin in patients with coronary heart disease and chronic kidney disease: the TNT (Treating to New Targets) study. J Am Coll Cardiol. 2008;51(15):1448–1454.    DOI: 10.1016/j.jacc.2007.11.072    PMID: 18402899
  34. Ting RD, Keech AC, Drury PL, et al. Benefits and safety of long-term fenofibrate therapy in people with type 2 diabetes and renal impairment: the FIELD Study. Diabetes Care. 2012;35(2):218–225.    DOI: 10.2337/dc11-1109    PMID: 22210576
  35. Kalil RS, Wang JH, de Boer IH, et al. Effect of extended-release niacin on cardiovascular events and kidney function in chronic kidney disease: a post hoc analysis of the AIM-HIGH trial. Kidney Int. 2015;87(6):1250–1257.    DOI: 10.1038/ki.2014.383    PMID: 25651367
  36. Barter PJ, Caulfield M, Eriksson M, et al. Effects of torcetrapib in patients at high risk for coronary events. N Engl J Med. 2007;357(21):2109–2122.    DOI: 10.1056/NEJMoa0706628    PMID: 17984165
  37. Schwartz GG, Olsson AG, Barter PJ. Dalcetrapib in patients with an acute coronary syndrome. N Engl J Med. 2013;368(9):869–870.    DOI: 10.1056/NEJMc1300057    PMID: 23445098
  38. HPS2-THRIVE Collaborative Group, Landray MJ, Haynes R, et al. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med. 2014;371(3):203–212.    DOI: 10.1056/NEJMoa1300955    PMID: 25014686
  39. Aitman TJ. CD36, insulin resistance, and coronary heart disease. Lancet. 2001;357(9257):651–652.    DOI: 10.1016/S0140-6736(00)04149-0    PMID: 11247544
  40. Dugani SB, Moorthy MV, Li C, et al. Association of lipid, inflammatory, and metabolic biomarkers with age at onset for incident coronary heart disease in women. JAMA Cardiol. 2021;6(4):437–447.    DOI: 10.1001/jamacardio.2020.7073    PMID: 33471027
  41. de Boer IH, Katz R, Chonchol MB, et al. Insulin resistance, cystatin C, and mortality among older adults. Diabetes Care. 2012;35(6):1355–1360.    DOI: 10.2337/dc11-1657    PMID: 22432118
  42. Klarin D, Zhu QM, Emdin CA, et al. Genetic analysis in UK Biobank links insulin resistance and transendothelial migration pathways to coronary artery disease. Nat Genet. 2017;49(9):1392–1397.    DOI: 10.1038/ng.3914    PMID: 28714974
  43. Ormazabal V, Nair S, Elfeky O, Aguayo C, Salomon C, Zuñiga FA. Association between insulin resistance and the development of cardiovascular disease. Cardiovasc Diabetol. 2018;17(1):122.    DOI: 10.1186/s12933-018-0762-4    PMID: 30170598
  44. Hill MA, Yang Y, Zhang L, et al. Insulin resistance, cardiovascular stiffening and cardiovascular disease. Metabolism. 2021;119:154766.    DOI: 10.1016/j.metabol.2021.154766    PMID: 33766485
  45. de Boer IH, Mehrotra R. Insulin resistance in chronic kidney disease: a step closer to effective evaluation and treatment. Kidney Int. 2014;86(2):243–245.    DOI: 10.1038/ki.2014.123    PMID: 25079023
  46. Gungor O, Ulu S, Hasbal NB, Anker SD, Kalantar-Zadeh K. Effects of hormonal changes on sarcopenia in chronic kidney disease: where are we now and what can we do? J Cachexia Sarcopenia Muscle. 2021;12(6):1380–1392.    DOI: 10.1002/jcsm.12839    PMID: 34676694
  47. Mima A, Ohshiro Y, Kitada M, et al. Glomerular-specific protein kinase C-β-induced insulin receptor substrate-1 dysfunction and insulin resistance in rat models of diabetes and obesity. Kidney Int. 2011;79(8):883–896.    DOI: 10.1038/ki.2010.526    PMID: 21228767
  48. Huh JH, Yadav D, Kim JS, et al. An association of metabolic syndrome and chronic kidney disease from a 10-year prospective cohort study. Metabolism. 2017;67:54–61.    DOI: 10.1016/j.metabol.2016.11.003    PMID: 28081778
  49. Xu S, Ilyas I, Little PJ, et al. Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: From mechanism to pharmacotherapies. Pharmacol Rev. 2021;73(3):924–967.    DOI: 10.1124/pharmrev.120.000096    PMID: 34088867
  50. Chong KC, Owens CD, Park M, et al. Relationship between kidney disease and endothelial function in peripheral artery disease. J Vasc Surg. 2014;60(6):1605–1611.    DOI: 10.1016/j.jvs.2014.08.105    PMID: 25441679
  51. Maesaka JK, Fishbane S. Regulation of renal urate excretion: a critical review. Am J Kidney Dis. 1998;32(6):917–933.    DOI: 10.1016/s0272-6386(98)70067-8    PMID: 9856507
  52. Yanai H, Adachi H, Hakoshima M, Katsuyama H. Molecular biological and clinical understanding of the pathophysiology and treatments of hyperuricemia and its association with metabolic syndrome, cardiovascular diseases and chronic kidney disease. Int J Mol Sci. 2021;22(17):9221.    DOI: 10.3390/ijms22179221    PMID: 34502127
  53. Ma M, Wang L, Huang W, et al. Meta-analysis of the correlation between serum uric acid level and carotid intima-media thickness. PLoS One. 2021;16(2):e0246416.    DOI: 10.1371/journal.pone.0246416    PMID: 33571245
  54. Li M, Hu X, Fan Y, et al. Hyperuricemia and the risk for coronary heart disease morbidity and mortality a systematic review and dose-response meta-analysis. Sci Rep. 2016;6:19520.    DOI: 10.1038/srep19520    PMID: 26814153
  55. Li P, Zhang L, Zhang M, Zhou C, Lin N. Uric acid enhances PKC-dependent eNOS phosphorylation and mediates cellular ER stress: a mechanism for uric acid-induced endothelial dysfunction. Int J Mol Med. 2016;37(4):989–997.    DOI: 10.3892/ijmm.2016.2491    PMID: 26935704
  56. Cai W, Duan XM, Liu Y, et al. Uric acid induces endothelial dysfunction by activating the HMGB1/RAGE signaling pathway. Biomed Res Int. 2017;2017:4391920.    DOI: 10.1155/2017/4391920    PMID: 28116308
  57. Sincer I, Kurtoglu E, Calıskan M, et al. Significant correlation between uric acid levels and flow-mediated dilatation in patients with masked hypertension. Clin Exp Hypertens. 2014;36(5):315–320.    DOI: 10.3109/10641963.2013.827694    PMID: 24555420
  58. Tomiyama H, Higashi Y, Takase B, et al. Relationships among hyperuricemia, metabolic syndrome, and endothelial function. Am J Hypertens. 2011;24(7):770–774.    DOI: 10.1038/ajh.2011.55    PMID: 21490690
  59. Berg AH, Drechsler C, Wenger J, et al. Carbamylation of serum albumin as a risk factor for mortality in patients with kidney failure. Sci Transl Med. 2013;5(175):175ra29.    DOI: 10.1126/scitranslmed.3005218    PMID: 23467560
  60. Wang Z, Nicholls SJ, Rodriguez ER, et al. Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nat Med. 2007;13(10):1176–1184.
  61. Gross ML, Piecha G, Bierhaus A, et al. Glycated and carbamylated albumin are more nephrotoxic than unmodified albumin in the amphibian kidney. Am J Physiol Renal Physiol. 2011;301(3):F476–F485.    DOI: 10.1152/ajprenal.00342.2010    PMID: 21367923
  62. Apostolov EO, Shah SV, Ray D, Basnakian AG. Scavenger receptors of endothelial cells mediate the uptake and cellular proatherogenic effects of carbamylated LDL. Arterioscler Thromb Vasc Biol. 2009;29(10):1622–1630.    DOI: 10.1161/ATVBAHA.109.189795    PMID: 19696406
  63. Apostolov EO, Shah SV, Ok E, Basnakian AG. Carbamylated low-density lipoprotein induces monocyte adhesion to endothelial cells through intercellular adhesion molecule-1 and vascular cell adhesion molecule-1. Arterioscler Thromb Vasc Biol. 2007;27(4):826–832.    DOI: 10.1161/01.ATV.0000258795.75121.8a    PMID: 17255534
  64. Schreier SM, Hollaus M, Hermann M, et al. Carbamoylated free amino acids in uremia: HOCl generates volatile protein modifying and cytotoxic oxidant species from N-carbamoyl-threonine but not threonine. Biochimie. 2012;94(11):2441–2447.    DOI: 10.1016/j.biochi.2012.06.032    PMID: 22776434
  65. Kalim S, Ortiz G, Trottier CA, et al. The effects of parenteral amino acid therapy on protein carbamylation in maintenance hemodialysis patients. J Ren Nutr. 2015;25(4):388–392.    DOI: 10.1053/j.jrn.2015.01.019    PMID: 25753604
  66. Watanabe H, Miyamoto Y, Otagiri M, Maruyama T. Update on the pharmacokinetics and redox properties of protein-bound uremic toxins. J Pharm Sci. 2011;100(9):3682–3695.    DOI: 10.1002/jps.22592    PMID: 21538353
  67. Barreto FC, Barreto DV, Liabeuf S, et al. Serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin J Am Soc Nephrol. 2009;4(10):1551–1558.    DOI: 10.2215/CJN.03980609    PMID: 19696217
  68. Itoh Y, Ezawa A, Kikuchi K, Tsuruta Y, Niwa T. Protein-bound uremic toxins in hemodialysis patients measured by liquid chromatography/tandem mass spectrometry and their effects on endothelial ROS production. Anal Bioanal Chem. 2012;403(7):1841–1850.    DOI: 10.1007/s00216-012-5929-3    PMID: 22447217
  69. Ito S, Osaka M, Higuchi Y, Nishijima F, Ishii H, Yoshida M. Indoxyl sulfate induces leukocyte-endothelial interactions through up-regulation of E-selectin. J Biol Chem. 2010;285(50):38869–38875.    DOI: 10.1074/jbc.m110.166686    PMID: 20937831
  70. Matsuo K, Yamamoto S, Wakamatsu T, et al. Increased proinflammatory cytokine production and decreased cholesterol efflux due to downregulation of ABCG1 in macrophages exposed to indoxyl sulfate. Toxins. 2015;7(8):3155–3166.    DOI: 10.3390/toxins7083155    PMID: 26287243
  71. Nakano T, Katsuki S, Chen M, et al. Uremic toxin indoxyl sulfate promotes proinflammatory macrophage activation via the interplay of OATP2B1 and Dll4-Notch signaling. Circulation. 2019;139(1):78–96.    DOI: 10.1161/CIRCULATIONAHA.118.034588    PMID: 30586693
  72. Gross P, Massy ZA, Henaut L, et al. Para-cresyl sulfate acutely impairs vascular reactivity and induces vascular remodeling. J Cell Physiol. 2015;230(12):2927–2935.    DOI: 10.1002/jcp.25018    PMID: 25899466
  73. Wu IW, Hsu KH, Hsu HJ, et al. Serum free p-cresyl sulfate levels predict cardiovascular and all-cause mortality in elderly hemodialysis patients—a prospective cohort study. Nephrol Dial Transplant. 2012;27(3):1169–1175.    DOI: 10.1093/ndt/gfr453    PMID: 21891772
  74. Lin CJ, Pan CF, Chuang CK, et al. P-cresyl sulfate is a valuable predictor of clinical outcomes in pre-ESRD patients. Biomed Res Int. 2014;2014:526932.    DOI: 10.1155/2014/526932    PMID: 24592393
  75. Lin CJ, Pan CF, Liu HL, et al. The role of protein-bound uremic toxins on peripheral artery disease and vascular access failure in patients on hemodialysis. Atherosclerosis. 2012;225(1):173–179.    DOI: 10.1016/j.atherosclerosis.2012.07.012    PMID: 22981405
  76. Schepers E, Meert N, Glorieux G, Goeman J, Van der Eycken J, Vanholder R. P-cresylsulphate, the main in vivo metabolite of p-cresol, activates leucocyte free radical production. Nephrol Dial Transplant. 2007;22(2):592–596.    DOI: 10.1093/ndt/gfl584    PMID: 17040995
  77. Pletinck A, Glorieux G, Schepers E, et al. Protein-bound uremic toxins stimulate crosstalk between leukocytes and vessel wall. J Am Soc Nephrol. 2013;24(12):1981–1994.    DOI: 10.1681/ASN.2012030281    PMID: 24009240
  78. Six I, Maizel J, Barreto FC, et al. Effects of phosphate on vascular function under normal conditions and influence of the uraemic state. Cardiovasc Res. 2012;96(1):130–139.    DOI: 10.1093/cvr/cvs240    PMID: 22822101
  79. Ganesh SK, Stack AG, Levin NW, Hulbert-Shearon T, Port FK. Association of elevated serum PO4, Ca × PO4 product, and parathyroid hormone with cardiac mortality risk in chronic hemodialysis patients. J Am Soc Nephrol. 2001;12(10):2131–2138.    DOI: 10.1681/ASN.V12102131    PMID: 11562412
  80. Shuto E, Taketani Y, Tanaka R, et al. Dietary phosphorus acutely impairs endothelial function. J Am Soc Nephrol. 2009;20(7):1504–1512.    DOI: 10.1681/ASN.2008101106    PMID: 19406976
  81. Onufrak SJ, Bellasi A, Shaw LJ, et al. Phosphorus levels are associated with subclinical atherosclerosis in the general population. Atherosclerosis. 2008;199(2):424–431.    DOI: 10.1016/j.atherosclerosis.2007.11.004    PMID: 18093595
  82. Peng A, Wu T, Zeng C, et al. Adverse effects of simulated hyper- and hypo-phosphatemia on endothelial cell function and viability. PLoS One. 2011;6(8):e23268.    DOI: 10.1371/journal.pone.0023268    PMID: 21858050
  83. Di Marco GS, Hausberg M, Hillebrand U, et al. Increased inorganic phosphate induces human endothelial cell apoptosis in vitro. Am J Physiol Ren Physiol. 2008;294(6):F1381–F1387.    DOI: 10.1152/ajprenal.00003.2008    PMID: 18385273
  84. Di Marco GS, König M, Stock C, et al. High phosphate directly affects endothelial function by downregulating annexin II. Kidney Int. 2013;83(2):213–222.    DOI: 10.1038/ki.2012.300    PMID: 22913982
  85. Caglar K, Yilmaz MI, Saglam M, et al. Short-term treatment with sevelamer increases serum fetuin-a concentration and improves endothelial dysfunction in chronic kidney disease stage 4 patients. Clin J Am Soc Nephrol. 2008;3(1):61–68.    DOI: 10.2215/CJN.02810707    PMID: 18057307
  86. Richter B, Haller J, Haffner D, Leifheit-Nestler M. Klotho modulates FGF23-mediated NO synthesis and oxidative stress in human coronary artery endothelial cells. Pflügers Arch Eur J Physiol. 2016;468(9):1621–1635.    DOI: 10.1007/s00424-016-1858-x    PMID: 27448998
  87. Shimada T, Kakitani M, Yamazaki Y, et al. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest. 2004;113(4):561–568.    DOI: 10.1172/JCI19081    PMID: 14966565
  88. Mirza MAI, Hansen T, Johansson L, et al. Relationship between circulating FGF23 and total body atherosclerosis in the community. Nephrol Dial Transplant. 2009;24(10):3125–3131.    DOI: 10.1093/ndt/gfp205    PMID: 19429932
  89. Rodríguez-Ortiz ME, Alcalá-Díaz JF, Canalejo A, et al. Fibroblast growth factor 23 predicts carotid atherosclerosis in individuals without kidney disease. The CORDIOPREV study. Eur J Intern Med. 2020;74:79–85.    DOI: 10.1016/j.ejim.2019.12.008    PMID: 31899053
  90. Coban M, Inci A, Yılmaz U, Asilturk E. The association of fibroblast growth factor 23 with arterial stiffness and atherosclerosis in patients with autosomal dominant polycystic kidney disease. Kidney Blood Press Res. 2018;43(4):1160–1173.    DOI: 10.1159/000492244    PMID: 30064143
  91. Silswal N, Touchberry CD, Daniel DR, et al. FGF23 directly impairs endothelium-dependent vasorelaxation by increasing superoxide levels and reducing nitric oxide bioavailability. Am J Physiol Endocrinol Metab. 2014;307(5):E426–E436.    DOI: 10.1152/ajpendo.00264.2014    PMID: 25053401
  92. Saito Y, Nakamura T, Ohyama Y, et al. In vivo klotho gene delivery protects against endothelial dysfunction in multiple risk factor syndrome. Biochem Biophys Res Commun. 2000;276(2):767–772.    DOI: 10.1006/bbrc.2000.3470    PMID: 11027545
  93. Zanoli L, Lentini P, Briet M, et al. Arterial stiffness in the heart disease of CKD. J Am Soc Nephrol. 2019;30(6):918–928.    DOI: 10.1681/ASN.2019020117
  94. Meinitzer A, Seelhorst U, Wellnitz B, et al. Asymmetrical dimethylarginine independently predicts total and cardiovascular mortality in individuals with angiographic coronary artery disease (the Ludwigshafen Risk and Cardiovascular Health study). Clin Chem. 2007;53(2):273–283.    DOI: 10.1373/clinchem.2006.076711    PMID: 17185364
  95. Lee TS, Lu TM, Chen CH, Guo BC, Hsu CP. Hyperuricemia induces endothelial dysfunction and accelerates atherosclerosis by disturbing the asymmetric dimethylarginine/dimethylarginine dimethylaminotransferase 2 pathway. Redox Biol. 2021;46:102108.    DOI: 10.1016/j.redox.2021.102108    PMID: 34438260    PMCID: PMC8390558
  96. Konukoglu D, Firtina S, Serin O. The relationship between plasma asymmetrical dimethyl-L-arginine and inflammation and adhesion molecule levels in subjects with normal, impaired, and diabetic glucose tolerance. Metabolism. 2008;57(1):110–115.    DOI: 10.1016/j.metabol.2007.08.013    PMID: 18078867
  97. Guo WK, Zhang DL, Wang XX, et al. Actin cytoskeleton modulates ADMA-induced NF-kappaB nuclear translocation and ICAM-1 expression in endothelial cells. Med Sci Monit. 2011;17(9):BR242–BR247.    DOI: 10.12659/msm.881927    PMID: 21873936
  98. Guo W, Zhang D, Wang L, Zhang Y, Liu W. Disruption of asymmetric dimethylarginine-induced RelA/P65 association with actin in endothelial cells. Acta Biochim Biophys Sin. 2013;45(3):229–235.    DOI: 10.1093/abbs/gms120    PMID: 23296075
  99. Dowsett L, Higgins E, Alanazi S, Alshuwayer NA, Leiper FC, Leiper J. ADMA: A key player in the relationship between vascular dysfunction and inflammation in atherosclerosis. J Clin Med. 2020;9(9):3026.    DOI: 10.3390/jcm9093026    PMID: 32962225
  100. Böger RH, Bode-Böger SM, Tsao PS, Lin PS, Chan JR, Cooke JP. An endogenous inhibitor of nitric oxide synthase regulates endothelial adhesiveness for monocytes. J Am Coll Cardiol. 2000;36(7):2287–2295.    DOI: 10.1016/s0735-1097(00)01013-5    PMID: 11127475
  101. Toth J, Racz A, Kaminski PM, Wolin MS, Bagi Z, Koller A. Asymmetrical dimethylarginine inhibits shear stress–induced nitric oxide release and dilation and elicits superoxide-mediated increase in arteriolar tone. Hypertension. 2007;49(3):563–568.    DOI: 10.1161/01.HYP.0000256764.86208.3d    PMID: 17242303
  102. Antoniades C, Shirodaria C, Leeson P, et al. Association of plasma asymmetrical dimethylarginine (ADMA) with elevated vascular superoxide production and endothelial nitric oxide synthase uncoupling: implications for endothelial function in human atherosclerosis. Eur Heart J. 2009;30(9):1142–1150.    DOI: 10.1093/eurheartj/ehp061    PMID: 19297385
  103. Böger RH, Sydow K, Borlak J, et al. LDL cholesterol upregulates synthesis of asymmetrical dimethylarginine in human endothelial cells: Involvement of S-adenosylmethionine-dependent methyltransferases. Circ Res. 2000;87(2):99–105.    DOI: 10.1161/01.res.87.2.99    PMID: 10903992
  104. Pekarova M, Kubala L, Martiskova H, et al. Asymmetric dimethylarginine regulates the lipopolysaccharide-induced nitric oxide production in macrophages by suppressing the activation of NF-kappaB and iNOS expression. Eur J Pharmacol. 2013;713(1/2/3):68–77.    DOI: 10.1016/j.ejphar.2013.05.001    PMID: 23665490
  105. Tang WH, Wang Z, Kennedy DJ, et al. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ Res. 2015;116(3):448–455.    DOI: 10.1161/CIRCRESAHA.116.305360    PMID: 25599331
  106. Stubbs JR, House JA, Ocque AJ, et al. Serum trimethylamine-N-oxide is elevated in CKD and correlates with coronary atherosclerosis burden. J Am Soc Nephrol. 2016;27(1):305–313.    DOI: 10.1681/ASN.2014111063    PMID: 26229137
  107. Missailidis C, Hällqvist J, Qureshi AR, et al. Serum trimethylamine-N-oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PLoS One. 2016;11(1):e0141738.    DOI: 10.1371/journal.pone.0141738    PMID: 26751065
  108. Zeng Y, Guo M, Fang X, et al. Gut microbiota-derived trimethylamine N-oxide and kidney function: a systematic review and meta-analysis. Adv Nutr. 2021;12(4):1286–1304.    DOI: 10.1093/advances/nmab010    PMID: 33751019
  109. Wang M, Wang Z, Lee Y, et al. Dietary meat, trimethylamine N-oxide-related metabolites, and incident cardiovascular disease among older adults: the cardiovascular health study. Arterioscler Thromb Vasc Biol. 2022;42(9):e273–e288.    DOI: 10.1161/ATVBAHA.121.316533    PMID: 35912635
  110. Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57–63.    DOI: 10.1038/nature09922    PMID: 21475195
  111. Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–585.    DOI: 10.1038/nm.3145    PMID: 23563705
  112. Wang Z, Roberts AB, Buffa JA, et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell. 2015;163(7):1585–1595.    DOI: 10.1016/j.cell.2015.11.055    PMID: 26687352
  113. Witkowski M, Weeks TL, Hazen SL. Gut microbiota and cardiovascular disease. Circ Res. 2020;127(4):553–570.    DOI: 10.1161/CIRCRESAHA.120.316242    PMID: 32762536
  114. Wu P, Chen J, Chen J, et al. Trimethylamine N-oxide promotes apoE–/– mice atherosclerosis by inducing vascular endothelial cell pyroptosis via the SDHB/ROS pathway. J Cell Physiol. 2020;235(10):6582–6591.    DOI: 10.1002/jcp.29518    PMID: 32012263
  115. Ding L, Chang M, Guo Y, et al. Trimethylamine-N-oxide (TMAO)-induced atherosclerosis is associated with bile acid metabolism. Lipids Health Dis. 2018;17(1):286.    DOI: 10.1186/s12944-018-0939-6    PMID: 30567573
  116. Lindskog Jonsson A, Caesar R, Akrami R, et al. Impact of gut microbiota and diet on the development of atherosclerosis in apoe-/- mice. Arterioscler Thromb Vasc Biol. 2018;38(10):2318–2326.    DOI: 10.1161/ATVBAHA.118.311233    PMID: 29903735
  117. Aldana-Hernández P, Leonard KA, Zhao YY, Curtis JM, Field CJ, Jacobs RL. Dietary choline or trimethylamine N-oxide supplementation does not influence atherosclerosis development in ldlr-/- and apoe-/- male mice. J Nutr. 2020;150(2):249–255.    DOI: 10.1093/jn/nxz214    PMID: 31529091
  118. Meyer KA, Benton TZ, Bennett BJ, et al. Microbiota-dependent metabolite trimethylamine N-oxide and coronary artery calcium in the coronary artery risk development in young adults study (CARDIA). J Am Heart Assoc. 2016;5(10):e003970.    DOI: 10.1161/JAHA.116.003970    PMID: 27792658
  119. Koay YC, Chen YC, Wali JA, et al. Plasma levels of trimethylamine-N-oxide can be increased with ‘healthy’ and ‘unhealthy’ diets and do not correlate with the extent of atherosclerosis but with plaque instability. Cardiovasc Res. 2021;117(2):435–449.    DOI: 10.1093/cvr/cvaa094    PMID: 32267921
  120. Aronov PA, Luo FJG, Plummer NS, et al. Colonic contribution to uremic solutes. J Am Soc Nephrol. 2011;22(9):1769–1776.    DOI: 10.1681/ASN.2010121220    PMID: 21784895
  121. Bogiatzi C, Gloor G, Allen-Vercoe E, et al. Metabolic products of the intestinal microbiome and extremes of atherosclerosis. Atherosclerosis. 2018;273:91–97.    DOI: 10.1016/j.atherosclerosis.2018.04.015    PMID: 29702430
  122. Liu Y, Liu S, Zhao Z, Song X, Qu H, Liu H. Phenylacetylglutamine is associated with the degree of coronary atherosclerotic severity assessed by coronary computed tomographic angiography in patients with suspected coronary artery disease. Atherosclerosis. 2021;333:75–82.    DOI: 10.1016/j.atherosclerosis.2021.08.029    PMID: 34438323
  123. Tang Y, Zou Y, Cui J, et al. Analysis of two intestinal bacterial metabolites (trimethylamine N-oxide and phenylacetylglutamine) in human serum samples of patients with T2DM and AMI using a liquid chromatography tandem mass spectrometry method. Clin Chim Acta. 2022;536:162–168.    DOI: 10.1016/j.cca.2022.09.018    PMID: 36191613
  124. Nemet I, Saha PP, Gupta N, et al. A cardiovascular disease-linked gut microbial metabolite acts via adrenergic receptors. Cell. 2020;180(5):862–877.e22.    DOI: 10.1016/j.cell.2020.02.016    PMID: 32142679
  125. Reddy VP, Aryal P, Darkwah EK. Advanced glycation end products in health and disease. Microorganisms. 2022;10(9):1848.    DOI: 10.3390/microorganisms10091848    PMID: 36144449
  126. Semba RD, Ferrucci L, Fink JC, et al. Advanced glycation end products and their circulating receptors and level of kidney function in older community-dwelling women. Am J Kidney Dis. 2009;53(1):51–58.    DOI: 10.1053/j.ajkd.2008.06.018    PMID: 18789567
  127. de la Cruz-Ares S, Cardelo MP, Gutiérrez-Mariscal FM, et al. Endothelial dysfunction and advanced glycation end products in patients with newly diagnosed versus established diabetes: from the CORDIOPREV study. Nutrients. 2020;12(1):238.    DOI: 10.3390/nu12010238    PMID: 31963378
  128. Ying L, Shen Y, Zhang Y, et al. Advanced glycation end products via skin autofluorescence as potential marker of carotid atherosclerosis in patients with type 2 diabetes. Nutr Metab Cardiovasc Dis. 2021;31(12):3449–3456.    DOI: 10.1016/j.numecd.2021.09.005    PMID: 34688535
  129. Haitoglou CS, Tsilibary EC, Brownlee M, Charonis AS. Altered cellular interactions between endothelial cells and nonenzymatically glucosylated laminin/type IV collagen. J Biol Chem. 1992;267(18):12404–12407.    PMID: 1618745
  130. Aronson D. Cross-linking of glycated collagen in the pathogenesis of arterial and myocardial stiffening of aging and diabetes. J Hypertens. 2003;21(1):3–12.    DOI: 10.1097/00004872-200301000-00002    PMID: 12544424
  131. Fishman SL, Sonmez H, Basman C, Singh V, Poretsky L. The role of advanced glycation end-products in the development of coronary artery disease in patients with and without diabetes mellitus: a review. Mol Med. 2018;24(1):59.    DOI: 10.1186/s10020-018-0060-3    PMID: 30470170
  132. Liu Z, Zhu H, Ma Y, et al. AGEs exacerbates coronary microvascular dysfunction in NoCAD by activating endoplasmic reticulum stress-mediated PERK signaling pathway. Metabolism. 2021;117:154710.    DOI: 10.1016/j.metabol.2021.154710    PMID: 33485865
  133. Xing Y, Pan S, Zhu L, et al. Advanced glycation end products induce atherosclerosis via RAGE/TLR4 signaling mediated-M1 macrophage polarization-dependent vascular smooth muscle cell phenotypic conversion. Oxid Med Cell Longev. 2022;2022:9763377.    DOI: 10.1155/2022/9763377    PMID: 35069982
  134. Ok E, Basnakian AG, Apostolov EO, Barri YM, Shah SV. Carbamylated low-density lipoprotein induces death ofendothelial cells: a link to atherosclerosis in patients with kidney disease. Kidney Int. 2005;68(1):173–178.    DOI: 10.1111/j.1523-1755.2005.00391.x    PMID: 15954906
  135. Yuan G, Si G, Hou Q, et al. Advanced glycation end products induce proliferation and migration of human aortic smooth muscle cells through PI3K/AKT pathway. Biomed Res Int. 2020;2020:8607418.    DOI: 10.1155/2020/8607418    PMID: 32733959
  136. Yamada S, Giachelli CM. Vascular calcification in CKD-MBD: Roles for phosphate, FGF23, and klotho. Bone. 2017;100:87–93.    DOI: 10.1016/j.bone.2016.11.012    PMID: 27847254
  137. Aikawa E, Aikawa M, Libby P, et al. Arterial and aortic valve calcification abolished by elastolytic cathepsin S deficiency in chronic renal disease. Circulation. 2009;119(13):1785–1794.    DOI: 10.1161/CIRCULATIONAHA.108.827972    PMID: 19307473
  138. Voelkl J, Tuffaha R, Luong TTD, et al. Zinc inhibits phosphate-induced vascular calcification through TNFAIP3-mediated suppression of NF-κB. J Am Soc Nephrol. 2018;29(6):1636–1648.    DOI: 10.1681/ASN.2017050492    PMID: 29654213
  139. Zhao G, Xu MJ, Zhao MM, et al. Activation of nuclear factor-kappa B accelerates vascular calcification by inhibiting ankylosis protein homolog expression. Kidney Int. 2012;82(1):34–44.    DOI: 10.1038/ki.2012.40    PMID: 22437419
  140. Paloian NJ, Giachelli CM. A current understanding of vascular calcification in CKD. Am J Physiol Renal Physiol. 2014;307(8):F891–F900.    DOI: 10.1152/ajprenal.00163.2014    PMID: 25143458
  141. Ogata H, Fukagawa M, Hirakata H, et al. Effect of treating hyperphosphatemia with lanthanum carbonate vs calcium carbonate on cardiovascular events in patients with chronic kidney disease undergoing hemodialysis: the LANDMARK randomized clinical trial. JAMA. 2021;325(19):1946–1954.    DOI: 10.1001/jama.2021.4807    PMID: 34003226
  142. Hénaut L, Massy ZA. Phosphate meeting cholesterol—consequences for cardiovascular disease in chronic kidney disease? Kidney Int. 2021;99(6):1264–1267.    DOI: 10.1016/j.kint.2021.02.022    PMID: 34023025
  143. Miyazaki-Anzai S, Keenan AL, Blaine J, Miyazaki M. Targeted disruption of a proximal tubule-specific TMEM174 gene in mice causes hyperphosphatemia and vascular calcification. J Am Soc Nephrol. 2022;33(8):1477–1486.    DOI: 10.1681/ASN.2021121578    PMID: 35459732
  144. Thi Nguyen N, Thi Nguyen T, Nguyen HT, et al. Inhibition of mitochondrial phosphate carrier prevents high phosphate-induced superoxide generation and vascular calcification. Exp Mol Med. 2023;55(3):532–540.    DOI: 10.1038/s12276-023-00950-0    PMID: 36854772
  145. Nguyen NT, Nguyen TT, Da Ly D, et al. Oxidative stress by Ca2+ overload is critical for phosphate-induced vascular calcification. Am J Physiol Heart Circ Physiol. 2020;319(6):H1302–H1312.    DOI: 10.1152/ajpheart.00305.2020    PMID: 33095057
  146. Schiattarella G, Sannino A, Toscano E, et al. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis. Eur Heart J. 2017;38:2948–2956.    DOI: 10.1093/eurheartj/ehx342    PMID: 29020409
  147. Haghikia A, Li XS, Liman TG, et al. Gut microbiota-dependent trimethylamine N-oxide predicts risk of cardiovascular events in patients with stroke and is related to proinflammatory monocytes. Arterioscler Thromb Vasc Biol. 2018;38(9):2225–2235.    DOI: 10.1161/ATVBAHA.118.311023    PMID: 29976769
  148. Skye SM, Zhu W, Romano KA, et al. Microbial transplantation with human gut commensals containing CutC is sufficient to transmit enhanced platelet reactivity and thrombosis potential. Circ Res. 2018;123(10):1164–1176.    DOI: 10.1161/CIRCRESAHA.118.313142    PMID: 30359185
  149. Zhu W, Gregory JC, Org E, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016;165(1):111–124.    DOI: 10.1016/j.cell.2016.02.011    PMID: 26972052
  150. Zhu W, Buffa JA, Wang Z, et al. Flavin monooxygenase 3, the host hepatic enzyme in the metaorganismal trimethylamine N-oxide-generating pathway, modulates platelet responsiveness and thrombosis risk. J Thromb Haemost. 2018;16(9):1857–1872.    DOI: 10.1111/jth.14234    PMID: 29981269
  151. Tang WHW, Li DY, Hazen SL. Dietary metabolism, the gut microbiome, and heart failure. Nat Rev Cardiol. 2019;16(3):137–154.    DOI: 10.1038/s41569-018-0108-7    PMID: 30410105
  152. Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur J Heart Fail. 2016;18(8):891–975.    DOI: 10.1002/ejhf.592    PMID: 27207191
  153. Borlaug BA. Evaluation and management of heart failure with preserved ejection fraction. Nat Rev Cardiol. 2020;17(9):559–573.    DOI: 10.1038/s41569-020-0363-2    PMID: 32231333
  154. House AA, Wanner C, Sarnak MJ, et al. Heart failure in chronic kidney disease: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int. 2019;95(6):1304–1317.    DOI: 10.1016/j.kint.2019.02.022    PMID: 31053387
  155. Reddy YNV, Obokata M, Dean PG, Melenovsky V, Nath KA, Borlaug BA. Long-term cardiovascular changes following creation of arteriovenous fistula in patients with end stage renal disease. Eur Heart J. 2017;38(24):1913–1923.    DOI: 10.1093/eurheartj/ehx045    PMID: 28329100
  156. Eckardt KU, Bansal N, Coresh J, et al. Improving the prognosis of patients with severely decreased glomerular filtration rate (CKD G4+): conclusions from a Kidney Disease: improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int. 2018;93(6):1281–1292.    DOI: 10.1016/j.kint.2018.02.006    PMID: 29656903
  157. Messerli FH, Rimoldi SF, Bangalore S. The transition from hypertension to heart failure: contemporary update. JACC Heart Fail. 2017;5(8):543–551.    DOI: 10.1016/j.jchf.2017.04.012    PMID: 28711447
  158. Ku E, Lee BJ, Wei J, Weir MR. Hypertension in CKD: Core Curriculum 2019. Am J Kidney Dis. 2019;74(1):120–131.    DOI: 10.1053/j.ajkd.2018.12.044    PMID: 30898362
  159. Jiang S, Shui Y, Cui Y, et al. Gut microbiota dependent trimethylamine N-oxide aggravates angiotensin II-induced hypertension. Redox Biol. 2021;46:102115.    DOI: 10.1016/j.redox.2021.102115    PMID: 34474396
  160. Babitt JL, Lin HY. Mechanisms of anemia in CKD. J Am Soc Nephrol. 2012;23(10):1631–1634.    DOI: 10.1681/ASN.2011111078    PMID: 22935483
  161. Rizzo C, Carbonara R, Ruggieri R, Passantino A, Scrutinio D. Iron deficiency: A new target for patients with heart failure. Front Cardiovasc Med. 2021;8:709872.    DOI: 10.3389/fcvm.2021.709872    PMID: 34447793
  162. Lipšic E, van der Meer P, van Veldhuisen DJ. Erythropoiesis-stimulating agents and heart failure. Cardiovasc Ther. 2011;29(4):e52–e59.    DOI: 10.1111/j.1755-5922.2010.00240.x    PMID: 20973926
  163. Anand IS, Gupta P. Anemia and iron deficiency in heart failure: current concepts and emerging therapies. Circulation. 2018;138(1):80–98.    DOI: 10.1161/CIRCULATIONAHA.118.030099    PMID: 29967232
  164. Swedberg K, Young JB, Anand IS, et al. Treatment of anemia with darbepoetin alfa in systolic heart failure. N Engl J Med. 2013;368(13):1210–1219.    DOI: 10.1056/NEJMoa1214865    PMID: 23473338
  165. Taddei S, Nami R, Bruno RM, Quatrini I, Nuti R. Hypertension, left ventricular hypertrophy and chronic kidney disease. Heart Fail Rev. 2011;16(6):615–620.    DOI: 10.1007/s10741-010-9197-z    PMID: 21116711
  166. Vogt I, Haffner D, Leifheit-Nestler M. FGF23 and phosphate-cardiovascular toxins in CKD. Toxins. 2019;11(11):647.    DOI: 10.3390/toxins11110647    PMID: 31698866
  167. Ritter CS, Slatopolsky E. Phosphate toxicity in CKD: the killer among us. Clin J Am Soc Nephrol. 2016;11(6):1088–1100.    DOI: 10.2215/CJN.11901115    PMID: 26912542
  168. Faul C, Amaral AP, Oskouei B, et al. FGF23 induces left ventricular hypertrophy. J Clin Invest. 2011;121(11):4393–4408.    DOI: 10.1172/JCI46122    PMID: 21985788
  169. Prié D. FGF23 and Cardiovascular Risk. Ann Endocrinol (Paris). 2021;82(3-4):141–143.    DOI: 10.1016/j.ando.2020.03.007    PMID: 32950228
  170. Bao J, Lu Y, She Q, et al. microRNA-30 regulates left ventricular hypertrophy in chronic kidney disease. JCI Insight. 2021;6(10):e138027.    DOI: 10.1172/jci.insight.138027    PMID: 33848263
  171. Charytan DM, Padera R, Helfand AM, et al. Increased concentration of circulating angiogenesis and nitric oxide inhibitors induces endothelial to mesenchymal transition and myocardial fibrosis in patients with chronic kidney disease. Int J Cardiol. 2014;176(1):99–109.    DOI: 10.1016/j.ijcard.2014.06.062    PMID: 25049013
  172. Zhou Y, Ng DYE, Richards AM, Wang P. microRNA-221 inhibits latent TGF-β1 activation through targeting thrombospondin-1 to attenuate kidney failure-induced cardiac fibrosis. Mol Ther Nucleic Acids. 2020;22:803–814.    DOI: 10.1016/j.omtn.2020.09.041    PMID: 33230477
  173. Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-β: the master regulator of fibrosis. Nat Rev Nephrol. 2016;12(6):325–338.    DOI: 10.1038/nrneph.2016.48    PMID: 27108839
  174. Pugliese NR, Masi S, Taddei S. The renin-angiotensin-aldosterone system: a crossroad from arterial hypertension to heart failure. Heart Fail Rev. 2020;25(1):31–42.    DOI: 10.1007/s10741-019-09855-5    PMID: 31512149
  175. Fu EL, Evans M, Clase CM, et al. Stopping renin-angiotensin system inhibitors in patients with advanced CKD and risk of adverse outcomes: a nationwide study. J Am Soc Nephrol. 2021;32(2):424–435.    DOI: 10.1681/ASN.2020050682    PMID: 33372009
  176. Rios FJ, Zou ZG, Harvey AP, et al. TRPM7 deficiency exacerbates cardiovascular and renal damage induced by aldosterone-salt. Commun Biol. 2022;5(1):746.    DOI: 10.1038/s42003-022-03715-z    PMID: 35882956
  177. Rios FJ, Zou ZG, Harvey AP, et al. Chanzyme TRPM7 protects against cardiovascular inflammation and fibrosis. Cardiovasc Res. 2020;116(3):721–735.    DOI: 10.1093/cvr/cvz164    PMID: 31250885
  178. Chung EY, Ruospo M, Natale P, et al. Aldosterone antagonists in addition to renin angiotensin system antagonists for preventing the progression of chronic kidney disease. Cochrane Database Syst Rev. 2020;10(10):CD007004.    DOI: 10.1002/14651858.CD007004.pub4    PMID: 33107592
  179. Hammer F, Buehling SS, Masyout J, et al. Protective effects of spironolactone on vascular calcification in chronic kidney disease. Biochem Biophys Res Commun. 2021;582:28–34.    DOI: 10.1016/j.bbrc.2021.10.023    PMID: 34678593
  180. Flatt DM, Brown MC, Mizeracki AM, King BJ, Weber KT. Mineralocorticoid receptor antagonists in the management of heart failure and resistant hypertension: a review. JAMA Cardiol. 2016;1(5):607–612.    DOI: 10.1001/jamacardio.2016.1878    PMID: 27434136
  181. Hsieh CC, Li CY, Hsu CH, et al. Mitochondrial protection by simvastatin against angiotensin II-mediated heart failure. Br J Pharmacol. 2019;176(19):3791–3804.    DOI: 10.1111/bph.14781    PMID: 31265743
  182. Mc Causland FR, Lefkowitz MP, Claggett B, et al. Angiotensin–neprilysin inhibition and renal outcomes across the spectrum of ejection fraction in heart failure. European J Heart Fail. 2022;24(9):1591–1598.    DOI: 10.1002/ejhf.2421    PMID: 34989105
  183. Judge P, Haynes R, Landray MJ, Baigent C. Neprilysin inhibition in chronic kidney disease. Nephrol Dial Transplant. 2015;30(5):738–743.    DOI: 10.1093/ndt/gfu269    PMID: 25140014
  184. Zittermann A, Ernst JB. Calciotropic and phosphaturic hormones in heart failure. Nutr Metab Cardiovasc Dis. 2016;26(11):971–979.    DOI: 10.1016/j.numecd.2016.06.007    PMID: 27493144
  185. Sterling KA, Eftekhari P, Girndt M, Kimmel PL, Raj DS. The immunoregulatory function of vitamin D: implications in chronic kidney disease. Nat Rev Nephrol. 2012;8(7):403–412.    DOI: 10.1038/nrneph.2012.93    PMID: 22614789
  186. Chau YY, Kumar J. Vitamin D in chronic kidney disease. Indian J Pediatr. 2012;79(8):1062–1068.    DOI: 10.1007/s12098-012-0765-1    PMID: 22544696
  187. Panizo S, Martínez-Arias L, Alonso-Montes C, et al. Fibrosis in Chronic Kidney Disease: Pathogenesis and Consequences. Int J Mol Sci. 2021;22(1):408.    DOI: 10.3390/ijms22010408    PMID: 33401711
  188. Latic N, Erben RG. Interaction of vitamin D with peptide hormones with emphasis on parathyroid hormone, FGF23, and the renin-angiotensin-aldosterone system. Nutrients. 2022;14(23):5186.    DOI: 10.3390/nu14235186    PMID: 36501215
  189. Hyder R, Sprague SM. Secondary hyperparathyroidism in a patient with CKD. Clin J Am Soc Nephrol. 2020;15(7):1041–1043.    DOI: 10.2215/CJN.13411119    PMID: 32461217
  190. Altay H, Zorlu A, Binici S, et al. Relation of serum parathyroid hormone level to severity of heart failure. Am J Cardiol. 2012;109(2):252–256.    DOI: 10.1016/j.amjcard.2011.08.039    PMID: 21996143
  191. Li Y, Chen C, Liu HL, Qian G. Vitamin D, parathyroid hormone, and heart failure in a Chinese elderly population. Endocr Pract. 2015;21(1):30–40.    DOI: 10.4158/EP14123.OR    PMID: 25100378
  192. Palmer SC, Mavridis D, Johnson DW, Tonelli M, Ruospo M, Strippoli GFM. Comparative effectiveness of calcimimetic agents for secondary hyperparathyroidism in adults: A systematic review and network meta-analysis. Am J Kidney Dis. 2020;76(3):321–330.    DOI: 10.1053/j.ajkd.2020.02.439    PMID: 32475604
  193. Bongartz LG, Cramer MJ, Doevendans PA, Joles JA, Braam B. The severe cardiorenal syndrome: ‘Guyton revisited’. Eur Heart J. 2005;26(1):11–17.    DOI: 10.1093/eurheartj/ehi020    PMID: 15615794
  194. Soppert J, Frisch J, Wirth J, et al. A systematic review and meta-analysis of murine models of uremic cardiomyopathy. Kidney Int. 2022;101(2):256–273.    DOI: 10.1016/j.kint.2021.10.025    PMID: 34774555
  195. Law JP, Price AM, Pickup L, et al. Clinical Potential of Targeting Fibroblast Growth Factor-23 and αKlotho in the Treatment of Uremic Cardiomyopathy. J Am Heart Assoc. 2020;9(7):e016041.    DOI: 10.1161/JAHA.120.016041    PMID: 32212912
  196. Hu MC, Shi M, Gillings N, et al. Recombinant α-Klotho may be prophylactic and therapeutic for acute to chronic kidney disease progression and uremic cardiomyopathy. Kidney Int. 2017;91(5):1104–1114.    DOI: 10.1016/j.kint.2016.10.034    PMID: 28131398
  197. Shamseddin MK, Parfrey PS. Sudden cardiac death in chronic kidney disease: Epidemiology and prevention. Nat Rev Nephrol. 2011;7(3):145–154.    DOI: 10.1038/nrneph.2010.191    PMID: 21283136
  198. Zelniker TA, Wiviott SD, Raz I, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet. 2019;393(10166):31–39.    DOI: 10.1016/S0140-6736(18)32590-X    PMID: 30424892