Nothing Special   »   [go: up one dir, main page]

Next Article in Journal
Bioprinting of Cells, Organoids and Organs-on-a-Chip Together with Hydrogels Improves Structural and Mechanical Cues
Previous Article in Journal
Endothelial Myosin IIA Is Required for the Maintenance of Blood–Brain Barrier Integrity
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Cardio-Renal Inflammation in Deciding the Fate of the Arteriovenous Fistula in Haemodialysis Therapy

Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
*
Author to whom correspondence should be addressed.
Cells 2024, 13(19), 1637; https://doi.org/10.3390/cells13191637
Submission received: 7 August 2024 / Revised: 12 September 2024 / Accepted: 27 September 2024 / Published: 1 October 2024
(This article belongs to the Section Cells of the Cardiovascular System)

Abstract

:
Vascular access is an indispensable component of haemodialysis therapy for end-stage kidney disease patients. The arteriovenous fistula (AVF) is most common, but importantly, two-year failure rates are greater than fifty percent. AVF failure can occur due to a lack of suitable vascular remodelling, and inappropriate inflammation preventing maturation, or alternatively neointimal hyperplasia and vascular stenosis preventing long-term use. A comprehensive mechanistic understanding of these processes is still lacking, but recent studies highlight an essential role for inflammation from uraemia and the AVF itself. Inflammation affects each cell in the cascade of AVF failure, the endothelium, the infiltrating immune cells, and the vascular smooth muscle cells. This review examines the role of inflammation in each cell step by step and the influence on AVF failure. Inflammation resulting in AVF failure occurs initially via changes in endothelial cell activation, permeability, and vasoprotective chemokine secretion. Resultingly, immune cells can extravasate into the subendothelial space to release inflammatory cytokines and cause other deleterious changes to the microenvironment. Finally, all these changes modify vascular smooth muscle cell function, resulting in excessive and unchecked hyperplasia and proliferation, eventually leading to stenosis and the failure of the AVF. Finally, the emerging therapeutic options based off these findings are discussed, including mesenchymal stem cells, small-molecule inhibitors, and far-infrared therapies. Recent years have clearly demonstrated a vital role for inflammation in deciding the fate of the AVF, and future works must be centred on this to develop therapies for a hitherto unacceptably underserved patient population.

1. Introduction

Chronic kidney disease (CKD) is a serious public health issue in the United States, causing progressive loss of kidney function [1]. CKD is defined in terms of the estimated glomerular filtration rate (eGFR) and/or albuminuria where eGFR has fallen below the threshold value of 60 mL/min/1.73 m2 and albumin levels are greater than 30 mg/g [2]. In 2023, 14% of US adults had a CKD diagnosis. Most people with CKD do not progress towards end-stage kidney disease (ESKD), where eGFR falls below 15 mL/min/1.73 m2, but some 2219 cases of ESKD per million Americans were reported in 2021 [1]. Kidney replacement therapies are considered for people with continuously progressive ESKD, and in the absence of readily available transplant options, the main modality chosen to extend their life is haemodialysis therapy (HD) [3]. This therapy involves the haemodialysis machine filtering the blood. The system has two circuits, which never directly meet, a blood circuit and a dialysate circuit. The connection between the two sides is via the dialyser, which comprises a dialysing membrane which filters out uraemic toxins. Most often, this technique is performed in-centre and requires 3–4 sessions per week [2,4].
Vascular access must be created to enable haemodialysis therapy, and this can be in the form of an arteriovenous fistula (AVF), arteriovenous graft (AVG), or central venous catheter (CVC) [5]. AVFs are the most common choice where 60% of prevalent American adults receiving HD do so via an AVF access [1]. Utilising an AVF is favourable because of longer-term patency and fewer vascular events like thrombosis, stenosis, and infections as compared to AVGs or CVCs [6]. Before the AVF can be used, it must be mature, meaning that it is easily cannulated while providing sufficient blood flow and having low flow resistance—establishing these characteristics is a complex and multi-faceted process. However, all three vascular access methods are highly prone to complications impinging on the efficacy of HD [7].

AVF Complications and Failure Are Common in Haemodialysis

Despite AVF creation being the preferred choice in HD, AVF failure is common, and the cumulative two-year loss of primary unassisted patency is at 47.3% [1]. Non-maturation, thrombosis, stenosis, or eventual failure can occur in the absence of appropriate vascular remodelling [8]. Percutaneous transluminal angioplasty (PTA) is considered the gold standard for treating AVF stenoses and is indicated once the reduction in lumen size begins to affect dialysis efficacy [5]. However, even with these treatments, restenosis is common and the AVF often still fails [9,10]. Subsequently, further access points (possibly of other access types) must be created, or the person may be transferred to other dialysis modalities like peritoneal dialysis [11]. The maturation or the failure of an AVF can be thought of as a balancing act between desired vascular responses and several failure risk factors and can be delineated to two phases—deleterious vascular remodelling in the maturation phase and unwanted vascular responses to ongoing dialysis [12,13,14]. Following surgical creation, AVF maturation depends on an appropriate response to the intervention with a progressive increase in arterial and venous diameters and subsequent blood flow. These changes are, in successful AVFs, a suitable response following haemodynamic changes secondary to the anastomosis such as increased blood flow, blood pressure, and shear stress [15,16,17]. Suitable vascular responses to these injuries are the release of endothelium-derived vasodilators such as nitric oxide (NO) and prostacyclin. These molecules promote increases in luminal area, block thrombus formation, and promote vascular smooth muscle cell (VSMC) differentiation and transmigration. Subsequently, NO interacts with the matrix metalloproteinases 2 and 9 (MMP-2, MMP-9) and promotes further dilation [18]. Consequently, desirable outward structural remodelling occurs with elastin fibre degradation, VSMC hypertrophy, and realignment of endothelial cells (ECs) to the new flow direction [16,19,20]. These changes must take place without compromising the luminal area, as shown in Figure 1. In the context of non-maturing AVFs and later AVF failure, one or several of these processes fail, resulting in venous neointimal hyperplasia (VNH), unfavourable remodelling, and subsequent decreases in luminal area beyond a point of suitability for HD therapy [21]. Vascular remodelling is required for AVF maturation, but the processes carry with them risk for AVF failure as do subsequent inflammatory events induced by HD itself like repeated needling [22]. Inflammatory burden associates with AVF failure. The precise molecular mechanisms underpinning AVF failure are complex and poorly understood. A substantial part of AVF failure is the postulated association with the inflammatory state [23,24,25], which will form the focus of this review. In people undergoing AVF creation, interleukin-6 (IL-6) serum levels [26], C-reactive protein (CRP) levels [27], IL-1β, and C-X-C chemokine receptor type 4 [28] (CXCR-4) were significantly increased in those with dysfunctional versus functional fistulas. Additionally, increases in the neutrophil-to-lymphocyte and platelet-to-lymphocyte ratio are also strongly predictive of AVF maturation [29,30,31]. Relatedly, throughout several animal studies in our lab [23,24,32,33] and from other groups [34,35,36], the essential role of inflammation in controlling the development and maturation of the AVF has become clear, as has the pivotal role anti-inflammatory therapeutics may play. Therefore, this review will describe and discuss the impact of inflammation on each of the cell types involved in AVF failure, and then go on to discuss how therapeutic intervention in these therapies may be beneficial and what they can reveal about the mechanistic underpinnings.

2. The Role of Inflammation in Each Cell Type in the Cascade of AVF Failure

2.1. Endothelial Cells

The endothelial layer is incumbent in two important roles—managing the infiltration of immune cells into the vessel walls and offering VSMC-enhancing delivery of nitric oxide (NO). Firstly, the degree and scale of endothelial dysfunction in the early stages of AVF development predict future VNH and VS. Specifically to the juxta-anastomotic site, there is an early increase in the endothelial permeability and expression of vascular cell adhesion molecule-1 (VCAM-1) [37], which is then strongly correlated to the greatest degree of stenosis [37]. More broadly, impairment of endothelial function associates with less remodelling, a reduced venous diameter, and a reduced rate of successful AVF maturation [38].
This includes microvascular dysfunction in addition to the established macrovascular dysfunction of the AVF itself. Korsheed et al. already described some 10 years ago how the creation of an AVF associates with both local and remote changes in the microcirculation, as assessed by laser doppler and endothelial-dependent vasodilation assays [39]. The exact changes remain unclear; however, a recent study found no change in microvascular endothelial function from baseline up to 6 weeks after AVF creation, but a better baseline microvascular function did associate with a better probability of successful AVF maturation [40]. The major changes induced by the haemodynamics of the AVF, and the trauma involved in its creation, may therefore be inducing inflammation in distant microvasculature sites, leading to further potentiated inflammatory responses.
The other role for ECs is the delivery of NO. Mechanistically, endothelial injury, such as that induced by TNF-β secondary to repeated needling, results in mitochondrial reactive oxygen species (ROS) generation [41,42,43]. As a result, there is reduced endothelial NO production and less vasodilation. A recent study from the Lee group described the protective effects of eNOS overexpression on the natural development of the AVF. The overexpression of eNOS was able to modify the flow dynamics in the AVF, and the presence of more eNOS reduced wall shear stress and vorticity as compared to wild-type mice and eNOS knockout mice [44]. The modulation of EC function and NO delivery may therefore be an approach to improve response to physical stressors of haemodialysis and in the inflammatory cascade secondary to AVF creation and ongoing uraemia.
These effects may be modifiable directly via the modulation of the vitamin D receptor (VDR) [33,45]. VDR expression was strongly decreased in venous tissue samples of patients with AVF stenosis, whilst the expression of P66Shc, P-P66Shc, fibronectin, collagen-1, and 8-OHdG were increased significantly. These findings were mirrored with concomitant increases in mitochondrial ROS species, following the stimulation of HUVECs with TGF-β. Relatedly, in this in vitro model, the overexpression of VDR and the modulation of Pin1, a regulator of P66, rescued TGF-β-induced injury. Therefore, the modulation of VDR signalling may be a new therapeutic avenue. Improvements to endothelial function may be directly achievable by the administration of vitamin D. Specifically to CKD, human endothelial cells cultured in a CKD-like environment showed reduced inflammatory cytokine levels and increased eNOS expression following vitamin D3 administration [46]. In vivo, uraemic rat paricalcitol administration was able to attenuate endothelial dysfunction by restoring cell–cell contacts and reducing permeability, a finding also mirrored in human endothelial cell lines [47].
Similarly, in a recent study from our own group specifically on the AVF context [33], we administered 1α, 25 (OH)2D3 encapsulated in nanoparticles to the adventitia of a surgically created AVF in uraemic mice and pigs [33,48] and then after a PTA was also performed on the porcine AVF [49]. This treatment reduced the presence of the key immune markers MCP-1 and CD68 in the outflow vein. Blood flow and lumen area were significantly increased whilst maximal velocity, VNH, and shear stress were reduced. In line with the previously discussed studies, RNA sequencing revealed an upregulation for TGFβ1 pathways. Taken together, these data support an essential role for the vascular endothelium, possibly modulated by VDR expression.
Patients receiving haemodialysis very often have several comorbidities, which may predispose them to endothelial injury and a predication for therefore problematic vascular rebuilding following AVF creation. A major comorbidity is atherosclerosis [50], which is triggered in its initial stages by endothelial dysfunction [51,52] and accelerated in dialysis [50,53]. The causes and pathobiology are beyond the scope of this review, but atherosclerotic endothelia are activated and express high levels of adhesion molecules to facilitate an easy transmigration of immune cells [54]. These processes have clear parallels to the outlined process of AVF failure and may be interrelated, particularly given how inflammation in chronic disease may be one and the same [55]. Vascular calcification is the other major comorbidity in people receiving haemodialysis therapy [56]. During pre-operative screening, the surgeon aims to select against calcified arteries, but given the close-to-universal prevalence of some cardiovascular calcification in HD patients [57], there are still up to 23% of AVFs that have calcifications [58]. A preliminary event in calcification forming is the over-formation of calciprotein particles (CPPs), which have recently been shown to impair endothelial function. CPPs reduce NO bioavailability via reducing the expression of eNOS, leading to increased exposure to reactive oxygen species [59,60]. Taken together, the baseline state of the vessels of a patient in which an AVF is created may itself influence resulting AVF failure due to endothelial dysfunction and inflammation.
Recently, there was an association described between the atherosclerotic risk score and AVF failure, identifying this as a potential new risk score, but also supporting the role for a tendency toward endothelial dysfunction in driving unwanted vascular outcomes. Despite the best intentions of the surgeon, pre-existing atherosclerotic changes in the target vessel, underpinned by endothelial dysfunction, may influence AVF failure [61]. Separately to the main AVF itself, the microvasculature may play a role; a recent study found microvascular endothelial function to be predictive of AVF success and subsequent function, possibly a supporting determinant of the ability to dilate in response to new shear stress following surgery [62].
Endothelial damage may also be due to the process of performing dialysis itself. In addition to the turbulent stress the endothelium experiences due to surgery, repeated needling is associated with endothelial damage. This needling can activate the endothelium, potentiate dysfunction, and lead to downstream failure and damage to the AVF from such direct physical hits [63]. Lastly, the uraemic milieu experienced by those receiving haemodialysis can be responsible for further endothelial dysfunction. Cell viability, proliferation, migration, and healing capacity are negatively impacted by uraemic serum [64,65,66,67], which in turn impinge upon suitable endothelial function. A downstream result may be a disposition to allow immune cell infiltration from the circulation to the subendothelial space. Infiltrating immune cells and their behaviour are the second lynchpin in deciding the fate of an AVF.

2.2. Immune Cell Infiltration

The exaggerated process of neointimal hyperplasia inside a vessel is a complex process initiated by endothelial damage, exposing the vascular smooth muscle cells to the blood components. This complex process is further affected by the proliferative and inflammatory responses [68]. In this section, we will elaborate upon different immune cells and how their infiltration affects the progression of the VNH. Immune cell infiltration in AVF progression can be divided into three distinct categories: firstly, the initial inflammation necessary for AVF maturation; secondly, the unchecked runaway inflammation causing the failure of AVF; and finally, the therapeutic strategy targeting the inflammation, which results in VHS/VS.

2.3. AVF Maturation

The histological findings from the initial creation of AVF are clearly indicative of inflammatory cell infiltration as early as 4 h, which attenuates vein graft endothelial cells and elevates subendothelial oedema [69,70]. Kuwahara et al. reported that M2 macrophage infiltration predominantly supports the extracellular matrix (ECM) deposition (hyaluronic acid) required for AVF maturation mainly via interaction with a widely expressed cellular adhesion molecule, CD44. This ECM:CD44 interaction supports the adhesion of leukocytes to endothelial cells, inducing the macrophage secretion and regulating VSMC proliferation and migration [71]. Other studies have also suggested that the role of M2 macrophages is important in vascular wall thickening and supporting the AVF maturation. The important immune cells that help in facilitating an adequate blood flow and guiding the outward remodelling are the CD4+ T cells [72,73,74]. Despite the fact that immune infiltration helps in vascular modelling, there needs to be a balance between pro-inflammatory and anti-inflammatory cells/response as both excessive and insufficient responses lead towards AVF failure [25].

2.4. AVF Failure

AVF failure is attributed to one of two stages, either early-stage failure linked with immaturity of AVF or later-stage/chronic failure mostly due to uncontrolled immune responses, leading to negative remodelling and reduced patency of AVF [13]. The pathological steps around adventitial remodelling have been reported to start with the infiltration of myofibroblasts into the adventitia [75,76], while further reports suggest endothelial-to-mesenchymal transitions (EndoMETs) as the main phenomenon involved, leading towards the fibrotic vessel. These mechanisms mainly are driven by the activations of signalling cascades associated with PI3K/Akt/mTOR pathways. These signalling pathways are activated by several cytokine/growth factors such as TGFβ, which is secreted by T cells and macrophages. The role of macrophages and T cells is not yet defined clearly as they play a role in both maturation (positive remodelling) and failure (negative remodelling) [30,72,74,77]. Samra et al. investigated the major immune cells by IHC and qRT-PCR, in the miniswine model, to further add to the strong association of immune response in AVF creation and maturation [78]. Our group also composed a review earlier about various vascular molecular messengers associated with haemodialysis vascular access malfunctions at early local inflammation and later a systemic rise in inflammation, which helps us to lead and design different therapeutic approaches to increase the patency of AVF by targeting these inflammatory cells/cytokines [79,80,81,82].

2.5. Therapeutics

The research work from our lab and other groups has established the strong role of macrophages in the progression of the VNH/VS. Blocking CX3CR1, an active player involved with macrophage survival and migration, significantly reduced neointima with increased lumen vessel area and patency in a mice model of AVF [23]. In another study, genetic deficiency of MCP-1 enhanced fistula patency by 6 weeks after its formation with reduced venous cell wall thickness and increased luminal area in MCP1−/− compared with MCP-1 +/+ mice [83]. A transcriptomic study identified a role for differentially expressed genes like tripartite motif-containing protein 55 (TRIM55), myoblast determination protein 1 (MYOD1), and several other genes. These play a crucial role in inflammation and the regulation of inflammatory pathways, and immune regulation has strong association with vascular cuffing and chronic inflammation-mediated early thrombosis in AVF [84]. A bioinformatic study identified significant elevation in PTGS2 in AVF failure. In a murine AVF model, a selective inhibitor of PTGS2 significantly altered different immune cells and the most significant ones had a positive correlation coefficient with mast cell activation, and the highest negative correlation coefficient was observed for resting mast cells [85]. The role of matrix metalloproteinase has been well established in the maturation and failure of AVFs. In an MMP-9 knockout mouse model, AVF fistula stenosis was attenuated mainly by reducing perioperative vascular inflammation [86]. TNFα is another key regulator of inflammation, and drugs targeting TNFα have been well studied in several diseases; however, they have not yet been approved to treat vascular stenosis [87]. Future studies are needed in appropriate study models to define the definitive subtype of macrophage (M1/M2) and T cell (Tregs/Th1/Th2/TH17) roles in both AVF maturation and AVF failure to effectively target the specific immune regulators and thus enhance the patency of the AVF.

3. VSMCs

Vascular smooth muscle cells (VSMCs) are an essential component of the vascular system. VSMCs are located intervascularly in the medial layer and are essential for contraction, vessel stability, and consistent blood pressure to allow transport of blood and other cell types. Mature VSMCs can dedifferentiate unlike most cells in a vascular setting [88,89,90,91,92,93,94]. This dedifferentiation is identifiable by protein markers of α-smooth muscle actin and calponin, which are needed to continue a contractile state [95]. The dedifferentiation will not occur unless triggered by injury or illness [93,95,96,97].
In the absence of injury, the cells stay in a contractile state [95]. This contractile state is where proliferation is lowest, and the cells continue to express the protein markers α-smooth muscle actin and calponin [95]. Whilst these proteins continue to be expressed, low proliferation will continue until dedifferentiation begins.
The protein levels of SM22 and alpha smooth muscle actin (αSMA) remain low as dedifferentiation has not occurred. Once dedifferentiation occurs, these proteins are secreted [95,98]. Endothelial cells (ECs) and VSMCs are mutually supportive and assist in the continual support throughout the vascular system, offering enhancing functions for the other. ECs create a contractile state for the VSMCs and secrete glycosaminoglycans; this includes heparin and other heparin-like substances [95,98]. The ECs will continue to excrete the substance until an injury occurs. When an injury occurs, the ECs stop secreting the substance and allow the VSMCs to inundate the wound site. The state of the VSMCs evolves when glycosaminoglycan ceases; this new state is called a synthetic one [89,91,92,93,95,98]. The synthetic state is where the most proliferation and cell differentiation take place for VSMCs that have dedifferentiated from the contractile state [89,91,92,93,95,98]. After the VSMCs have started to dedifferentiate into a synthetic state, an opening has occurred that allows macrophages to infiltrate into the cell wall and affect how the smooth muscle cells interact with each other [89,91,92,93,95,98].

3.1. VSMC Dysfunction in AVF Inflammation and Failure

VSMCs are known to have crosstalk between ECs, immune cells, and macrophages, which allows for research to be performed on what cell or cytokine causes specific changes in phenotypes of the smooth muscle cells [89,94,95]. Rai et al. studied the ability of VSMCs to crosstalk and how that could affect atherosclerosis maturation and plaque growth. They also investigated what cells are involved before and after VSMC phenotype switching. Rai et al. found that changing the cytokines downstream of where phenotypic switching occurs can change how the VSMCs are modulated. The authors state that future studies on immune cell infiltration will show a decrease in inflammation and mitigate the damage being done to AVFs as they mature.
In one study, Bezhaeva et al. looked specifically at TLR4 in response to inflammation in a murine AVF model [99]. As TLR4 is a common precipitator of inflammation in microbial infection, atherosclerosis, and vascular remodelling, they targeted a known regulator on inflammatory immune cells and VSMCs called RP105 (also known as CD180). The study allowed them to see how RP105 knockdown affected the αSMA levels immunobiologically in an AVF. They saw a marked decrease in αSMA between a wild-type mouse and an RP105−/− mouse [99]. They also saw a marked change in the lumen area and number of VSMCs located in the intima. As the VSMCs have a similar regulatory factor, TLR4, as an immune cell, removing RP105 affected them both in similar ways [99].
After AVF placement, studies have shown that VSMCs polarise to a synthetic phenotype, and extensive smooth muscle cell proliferation can occur [89,91,92,93,95,98]. The VSMCs can migrate to the location of injury and create a pro-inflammatory setting. Such a setting can cause a number of complications to an AVF’s long-term usage, including stenosis, oedema, thrombosis, and eventual AVF failure [94]. This migration is activated when the TGF-β pathway changes due to stress and the overall injury [92]. The changing in the VSMCs after their movement from the medial layer into all layers of a cell causes an oscillating effect. This oscillating effect has the VSMCs reacting to the changes in metabolism and inflammation, and the proliferation of more synthetic cells occurs due to the continual change in TGF-β [92].
This change creates a pro-inflammatory type of synthetic VSMC that interacts with ECs and allows for negative effects on the AVF site [92,94,98,100]. This synthetic state straddles a thin line between helpful and detrimental to any injury to the vascular system. More research into the versatile status of VSMCs is needed for long-term success of many types of vascular intervention. Research is currently being performed on metabolism [92,95], hormones [95,100], uraemia levels [96], and suppression [88,91,94]. Finally, there may be a crucial sex-based difference determining the fate of the AVF. Female AVFs have a decreased expression of BMP7 and IL17R and increased TGFβ-1 and its receptor [101]. Similarly, in the uraemic mouse with an AVF model, female mice had more negative vascular remodelling, reduced peak blood flow velocity, loss of proliferation, and increased apoptosis [101]. These sex differences should be investigated further to expand on their clinical impact and relevance.

3.2. Non-Inflammation-Based Factors Controlling AVF Failure

Whilst the focus of this review has been on cellular inflammation, there are other factors controlling the success of the AVF, which will be briefly discussed. The age of the patient is an important consideration, as typically older people have even more comorbidities than the general ESKD population [1], and represent the fastest growing haemodialysis group. Broadly, however, increasing age in and of itself does not lend itself to worsened AVF outcomes; rather, it is the relationship between age and other relevant comorbidities like the aforementioned atherosclerosis and vascular calcification that may drive increased failure rates [102,103,104]. The US Renal Database shows that once these hurdles are managed, the actual AVF failure rate is comparable between age groups [1]; what remains clear, however, is that older patients must have a unique specific treatment plan and approach to optimise their outcomes. Sex of the patient may play a role, as females are at a greater (2.42x) risk of AVF failure compared to males [105]. US patient statistics show a somewhat mixed state, as females have higher overall rates of fistula maturation and yet the duration of patency is shorter as compared to males [1]. The precise etiological reasons for this discrepancy are beyond the scope of this review but must be considered.
The Dialysis Outcomes and Practice Patterns Study reported data from 12 countries and determined that surgical prowess and experience was a major factor determining eventual AVF success. The more AVFs a surgeon had already placed was directly correlated to the maturation rate of the placed AVF. In nations or regions where the training for doctors focused more on vascular access, the eventual outcomes for AVFs were strongly improved [106]. Other socioeconomic factors may have a mixed effect on the outcomes of AVFs, and patients from a lower socioeconomic background may have restricted access to healthcare and are less likely to receive an AVF and when they do, the outcome is worse [107], but these findings are not consistent across nations; the National Health Service in the UK does not report these changes [108], and similar findings in the US vary between some effect and no effect of the socioeconomic group [109,110]. Taken together, there is a wide complicated collection of factors controlling and modulating AVF success or failure, which vary nation to nation and centre to centre. Therefore, in seeking to ameliorate the burden placed by AVF failure, the focus must be placed onto universally treatable factors, which ideally involve therapies aimed at easing inflammatory burden locally in the AVF and systemically.

3.3. Therapeutic Insights and Progress

The advancements in knowledge and insight we have so far outlined have generated several new lines of inquiry in developing effective methods to treat and prevent stenosis and ease the burden on people with ESKD. As recently as 2006, the vascular access guidelines from the national kidney guidelines did not address pharmaceutical intervention to resolve access failure, instead relying on PTA or surgical relocation [111]; beneficially, there are now some emerging therapies of choice [5]. Far-infrared therapy (FIR) has in recent years found favour in abrogating inflammatory responses in the AVF. The mechanism remains unclear, but the non-thermal effects are likely via modifying the oxidative state and subsequent inflammatory milieu. FIR increases the expression of hemoxygenase-1, resultingly increasing antioxidant levels, reducing the TNF-α-induced expression of endothelial adhesion molecules, and restoring endothelial nitric oxide synthase and subsequent nitric oxide levels and endothelial function. FIR is seemingly efficacious in patient groups at each stage of the HD process, in those patients with a high HD vintage and several interventions on pre-existing AVFs [112], in patients with a high HD vintage but no interventions [113,114], and in those new to HD with a new AVF [115].
Figure 2. Uncontrolled runaway inflammation underpins and drives the failure of the arteriovenous fistula. Inflammation in the AVF involving the endothelial layer, subsequent immune cell infiltration, and cytokine changes therein, and smooth muscle changes are shown. These are essential processes in determining the fate of the arteriovenous fistula. Reactive oxygen species (ROS). Toll-like receptors (TLRs). Tumour necrosis factor-alpha (TNF-α). Monocyte chemoattract protein-1 (MCP-1). Interleukin-6 (IL-6). Vascular cell adhesion molecule-1 (VCAM-1). Intercellular adhesion molecule-1 (ICAM-1). Made with BioRender.
Figure 2. Uncontrolled runaway inflammation underpins and drives the failure of the arteriovenous fistula. Inflammation in the AVF involving the endothelial layer, subsequent immune cell infiltration, and cytokine changes therein, and smooth muscle changes are shown. These are essential processes in determining the fate of the arteriovenous fistula. Reactive oxygen species (ROS). Toll-like receptors (TLRs). Tumour necrosis factor-alpha (TNF-α). Monocyte chemoattract protein-1 (MCP-1). Interleukin-6 (IL-6). Vascular cell adhesion molecule-1 (VCAM-1). Intercellular adhesion molecule-1 (ICAM-1). Made with BioRender.
Cells 13 01637 g002
A more recent study by Lindhard [116] and colleagues draws this into question at least with a single treatment course where the vasoprotective effects could not be seen, but changes in soluble VCAM and ICAM were present. Whilst an emerging and promising choice [117], FIR is clearly in need of much more work before it can be developed into a potent clinical tool.
Small-molecule inhibitors have found favour—in a study from our own group, we demonstrated that a nanobody blockade of CX3C motif chemokine receptor 1 (CX3CR1) in a murine AVF model reduced neointima formation and increased lumen vessel area in the outflow vein. These changes were associated with reductions in CD68 and CX3CR1 expression in both mice and human samples [23]. In experimental murine AVF, a week of 1.14 mg/kg/day of atorvastatin potently decreased venous outflow inflammation [82], likely by reduced macrophage infiltration and fibrin deposition [118], and statin use in humans is associated with an 18% reduction in the risk for AVF failure [119]. Lastly, a retrospective cohort study predicted utility for statins in AVF treatment following creation [120]. The reliability in these findings may be doubtful; however, a more recent randomised clinical trial found no efficacy in maintaining AVFs following treatment with rosuvastatin [121], but importantly the trial was underpowered, finished prematurely, and only utilised a low dose—so a follow-up study is essential to confirm these findings.
Lastly, mesenchymal stem cells have emerged as promising candidates for modulating inflammatory responses in the AVF. Firstly, we treated mouse AVFs at the time of surgical creation with human adipose tissue-derived mesenchymal stem cells (MSCs) and saw reduced intimal hyperplasia and MCP-1 expression [122]. These findings were similar when MSCs were administered at the time of PTA instead; there was less IL-1β and TNF-α, fibrosis, proliferation, and smooth muscle actin deposition [123]. These changes were further associated with improved haemodynamics, with increased blood flow and lumen vessel area and reduced neointima [123]. Currently, the MEST AVF trial is ongoing to determine if this approach can extend into patient groups [124].

4. Conclusions

There is now a large pool of experimental evidence supporting a key and indispensable role for inflammation in deciding the fate of an AVF. These changes are a complex interplay between the local environment, individual cell types, the interactions between them, the patient, and the wider clinical setting. Despite a mounting collection of evidence describing AVF failure secondary to inflammation which are summarised in Figure 2, there is still an unacceptably sparse number of therapeutic options available. Investigating and developing the more promising solutions must become a key research focus in the coming years to ease the burden of AVF failure in haemodialysis therapy.

Author Contributions

J.K., G.B. and S.M. designed and conceived the manuscript. J.K., A.L. and G.B. wrote the manuscript. S.M. reviewed and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by NIH R01 HL098967 and DK135407 and a Minnesota Regenerative Medicine grant to SM.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analysed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors have no conflicts of interest to declare.

References

  1. United States Renal Data System. 2023 USRDS Annual Data Report: Epidemiology of Kidney Disease in the United States; United States Renal Data System: Bethesda, MD, USA, 2023. [Google Scholar]
  2. Levin, A.; Stevens, P.E.; Ahmed, S.B.; Carrero, J.J.; Foster, B.; Francis, A.; Hall, R.K.; Herrington, W.G.; Hill, G.; Inker, L.A.; et al. KDIGO 2024 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney Int. 2024, 105, S117–S314. [Google Scholar] [CrossRef] [PubMed]
  3. Thurlow, J.S.; Joshi, M.; Yan, G.; Norris, K.C.; Agodoa, L.Y.; Yuan, C.M.; Nee, R. Global Epidemiology of End-Stage Kidney Disease and Disparities in Kidney Replacement Therapy. Am. J. Nephrol. 2021, 52, 98–107. [Google Scholar] [CrossRef] [PubMed]
  4. Nunes, S.; Culfaz-Emecen, P.; Ramon, G.; Visser, T.; Koops, G.; Jin, W.; Ulbricht, M. Thinking the future of membranes: Perspectives for advanced and new membrane materials and manufacturing processes. J. Membr. Sci. 2020, 598, 117761. [Google Scholar] [CrossRef]
  5. Lok, C.E.; Huber, T.S.; Lee, T.; Shenoy, S.; Yevzlin, A.S.; Abreo, K.; Allon, M.; Asif, A.; Astor, B.C.; Glickman, M.H.; et al. KDOQI Clinical Practice Guideline for Vascular Access: 2019 Update. Am. J. Kidney Dis. 2020, 75, S1–S164. [Google Scholar] [CrossRef]
  6. Lok, C.E.; Huber, T.S.; Orchanian-Cheff, A.; Rajan, D.K. Arteriovenous Access for Hemodialysis. JAMA 2024, 331, 1307. [Google Scholar] [CrossRef] [PubMed]
  7. Siddiqui, M.A.; Ashraff, S.; Santos, D.; Carline, T.; Siddiqui, M.A.; Ashraff, S.; Santos, D.; Carline, T. An overview of AVF maturation and endothelial dysfunction in an advanced renal failure. Ren. Replace. Ther. 2017, 3, 42. [Google Scholar] [CrossRef]
  8. Sabiu, G.; Gallieni, M. Pathophysiology of Arteriovenous Fistula Maturation and Nonmaturation. Clin. J. Am. Soc. Nephrol. CJASN 2023, 18, 8–10. [Google Scholar] [CrossRef]
  9. Tepe, G.; Laird, J.; Schneider, P.; Brodmann, M.; Krishnan, P.; Micari, A.; Metzger, C.; Scheinert, D.; Zeller, T.; Cohen, D.J.; et al. Drug-Coated Balloon Versus Standard Percutaneous Transluminal Angioplasty for the Treatment of Superficial Femoral and Popliteal Peripheral Artery Disease. Circulation 2015, 131, 495–502. [Google Scholar] [CrossRef]
  10. Ratnam, L.; Karunanithy, N.; Mailli, L.; Diamantopoulos, A.; Morgan, R.A. Dialysis Access Maintenance: Plain Balloon Angioplasty. Cardiovasc. Interv. Radiol. 2023, 46, 1136–1143. [Google Scholar] [CrossRef]
  11. Al-Balas, A.; Lee, T.; Young, C.J.; Allon, M. Choice of a second vascular access in hemodialysis patients whose initial arteriovenous fistula failed to mature. J. Vasc. Surg. 2018, 68, 1858–1864.e1. [Google Scholar] [CrossRef]
  12. Viecelli, A.; Mori, T.; Roy-Chaudhury, P.; Polkinghorne, K.R.; Hawley, C.M.; Johnson, D.W.; Pascoe, E.M.; Irish, A. The pathogenesis of hemodialysis vascular access failure and systemic therapies for its prevention: Optimism unfulfilled. Semin. Dial. 2018, 31, 244–257. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, Y.; Kong, X.; Liang, L.; Xu, D. Regulation of vascular remodeling by immune microenvironment after the establishment of autologous arteriovenous fistula in ESRD patients. Front. Immunol. 2024, 15, 1365422. [Google Scholar] [CrossRef] [PubMed]
  14. Soliveri, L.; Bozzetto, M.; Brambilla, P.; Caroli, A.; Remuzzi, A. Hemodynamics in AVF over time: A protective role of vascular remodeling toward flow stabilization. Int. J. Artif. Organs 2023, 46, 547–554. [Google Scholar] [CrossRef] [PubMed]
  15. He, Y.; Shiu, Y.-T.; Pike, D.B.; Roy-Chaudhury, P.; Cheung, A.K.; Berceli, S.A. Comparison of hemodialysis arteriovenous fistula blood flow rates measured by Doppler ultrasound and phase-contrast magnetic resonance imaging. J. Vasc. Surg. 2018, 68, 1848–1857. [Google Scholar] [CrossRef] [PubMed]
  16. Zonnebeld, N.; Huberts, W.; van Loon, M.M.; Delhaas, T.; Tordoir, J.H. Natural Vascular Remodelling After Arteriovenous Fistula Creation in Dialysis Patients With and Without Previous Ipsilateral Vascular Access. Eur. J. Vasc. Endovasc. Surg. 2020, 59, 277–287. [Google Scholar] [CrossRef]
  17. Browne, L.D.; Bashar, K.; Griffin, P.; Kavanagh, E.G.; Walsh, S.R.; Walsh, M.T. The Role of Shear Stress in Arteriovenous Fistula Maturation and Failure: A Systematic Review. PLoS ONE 2015, 10, e0145795. [Google Scholar] [CrossRef]
  18. Tronc, F.O.; Mallat, Z.; Lehoux, S.P.; Wassef, M.; Esposito, B.; Tedgui, A. Role of Matrix Metalloproteinases in Blood Flow–Induced Arterial Enlargement. Arterioscler. Thromb. Vasc. Biol. 2000, 20, e120–e126. [Google Scholar] [CrossRef]
  19. Shemesh, D.; Goldin, I.; Berelowitz, D.; Zaghal, I.; Zigelman, C.; Olsha, O. Blood Flow Volume Changes in the Maturing Arteriovenous Access for Hemodialysis. Ultrasound Med. Biol. 2007, 33, 727–733. [Google Scholar] [CrossRef]
  20. Malovrh, M. Postoperative Assessment of Vascular Access. J. Vasc. Access 2014, 15, S10–S14. [Google Scholar] [CrossRef]
  21. Rothuizen, T.C.; Wong, C.; Quax, P.H.A.; van Zonneveld, A.J.; Rabelink, T.J.; Rotmans, J.I. Arteriovenous access failure: More than just intimal hyperplasia? Nephrol. Dial. Transplant. 2013, 28, 1085–1092. [Google Scholar] [CrossRef]
  22. Roy-Chaudhury, P.; Kruska, L. Future directions for vascular access for hemodialysis. Semin. Dial. 2015, 28, 107–113. [Google Scholar] [CrossRef] [PubMed]
  23. Misra, S.; Kilari, S.; Yang, B.; Sharma, A.; Wu, C.-C.; Vazquez-Padron, R.I.; Broadwater, J. Anti Human CX3CR1 VHH Molecule Attenuates Venous Neointimal Hyperplasia of Arteriovenous Fistula in Mouse Model. J. Am. Soc. Nephrol. JASN 2021, 32, 1630–1648. [Google Scholar] [CrossRef] [PubMed]
  24. Singh, A.K.; Kilari, S.; Cai, C.; Misra, S. Bindarit encapsulated nanoparticles prevent venous neointimal hyperplasia and restenosis in a murine angioplasty model. Transl. Res. 2022, 248, 68–86. [Google Scholar] [CrossRef] [PubMed]
  25. Samra, G.; Rai, V.; Agrawal, D.K. Innate and adaptive immune cells associate with arteriovenous fistula maturation and failure. Can. J. Physiol. Pharmacol. 2022, 100, 215. [Google Scholar] [CrossRef] [PubMed]
  26. Baek, J.; Lee, H.; Yang, T.; Lee, S.-Y.; Kim, Y.G.; Kim, J.S.; Ahn, S.; Kim, K.; Kang, S.H.; Lee, M.-J.; et al. Plasma Interleukin-6 Level Predicts the Risk of Arteriovenous Fistula Dysfunction in Patients Undergoing Maintenance Hemodialysis. J. Pers. Med. 2023, 13, 151. [Google Scholar] [CrossRef]
  27. Stirbu, O.; Gadalean, F.; Pitea, I.V.; Ciobanu, G.; Schiller, A.; Grosu, I.; Nes, A.; Bratescu, R.; Olariu, N.; Timar, B.; et al. C-reactive protein as a prognostic risk factor for loss of arteriovenous fistula patency in hemodialyzed patients. J. Vasc. Surg. 2019, 70, 208–215. [Google Scholar] [CrossRef]
  28. Lu, P.; Wang, T.; Wan, Z.; Wang, M.; Zhou, Y.; He, Z.; Liao, S.; Liu, H.; Shu, C. Immune-Related Genes and Immune Cell Infiltration Characterize the Maturation Status of Arteriovenous Fistulas: An Integrative Bioinformatics Study and Experimental Validation Based on Transcriptome Sequencing. J. Inflamm. Res. 2024, 17, 137–152. [Google Scholar] [CrossRef]
  29. Wongmahisorn, Y. Role of neutrophil-to-lymphocyte ratio as a prognostic indicator for hemodialysis arteriovenous fistula failure. J. Vasc. Access 2019, 20, 608–614. [Google Scholar] [CrossRef]
  30. Kaygin, M.A.; Halici, U.; Aydin, A.; Dag, O.; Binici, D.N.; Limandal, H.K.; Arslan, Ü.; Kiymaz, A.; Kahraman, N.; Calik, E.S.; et al. The relationship between arteriovenous fistula success and inflammation. Ren. Fail. 2013, 35, 1085–1088. [Google Scholar] [CrossRef]
  31. Kaller, R.; Arbănași, E.M.; Mureșan, A.V.; Voidăzan, S.; Arbănași, E.M.; Horváth, E.; Suciu, B.A.; Hosu, I.; Halmaciu, I.; Brinzaniuc, K.; et al. The Predictive Value of Systemic Inflammatory Markers, the Prognostic Nutritional Index, and Measured Vessels’ Diameters in Arteriovenous Fistula Maturation Failure. Life 2022, 12, 1447. [Google Scholar] [CrossRef]
  32. Yang, B.; Kilari, S.; Brahmbhatt, A.; McCall, D.L.; Torres, E.N.; Leof, E.B.; Mukhopadhyay, D.; Misra, S. CorMatrix Wrapped Around the Adventitia of the Arteriovenous Fistula Outflow Vein Attenuates Venous Neointimal Hyperplasia. Sci. Rep. 2017, 7, 14298. [Google Scholar] [CrossRef] [PubMed]
  33. Singh, A.K.; Cai, C.; Kilari, S.; Zhao, C.; Simeon, M.L.; Takahashi, E.; Edelman, E.R.; Kong, H.J.; Macedo, T.; Singh, R.J.; et al. 1α,25-Dihydroxyvitamin D3 Encapsulated in Nanoparticles Prevents Venous Neointimal Hyperplasia and Stenosis in Porcine Arteriovenous Fistulas. J. Am. Soc. Nephrol. JASN 2021, 32, 866–885. [Google Scholar] [CrossRef]
  34. Wong, C.-Y.; Bezhaeva, T.; Rothuizen, T.C.; Metselaar, J.M.; Vries, M.R.d.; Verbeek, F.P.R.; Vahrmeijer, A.L.; Wezel, A.; Zonneveld, A.-J.v.; Rabelink, T.J.; et al. Liposomal prednisolone inhibits vascular inflammation and enhances venous outward remodeling in a murine arteriovenous fistula model. Sci. Rep. 2016, 6, 30439. [Google Scholar] [CrossRef]
  35. Kiwan, G.; Gonzalez, L.; Langford, J.; Yatsula, B.; Dardik, A. Inhibition of T-Cells by Cyclosporine A Reduces Macrophage Accumulation to Regulate Venous Adaptive Remodeling and Increase Arteriovenous Fistula Maturation. Arterioscler. Thromb. Vasc. Biol. 2021, 41, E160–E174. [Google Scholar] [CrossRef]
  36. Wang, Y.; Krishnamoorthy, M.; Banerjee, R.; Zhang, J.; Rudich, S.; Holland, C.; Arend, L.; Roy-Chaudhury, P. Venous stenosis in a pig arteriovenous fistula model—Anatomy, mechanisms and cellular phenotypes. Nephrol. Dial. Transplant. 2008, 23, 525–533. [Google Scholar] [CrossRef] [PubMed]
  37. Cui, J.; Kessinger, C.W.; McCarthy, J.R.; Sosnovik, D.E.; Libby, P.; Thadhani, R.I.; Jaffer, F.A. In Vivo Nanoparticle Assessment of Pathological Endothelium Predicts the Development of Inflow Stenosis in Murine Arteriovenous Fistula. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 189–196. [Google Scholar] [CrossRef]
  38. Owens, C.; Wake, N.; Kim, J.; Hentschel, D.; Conte, M.; Schanzer, A. Endothelial function predicts positive arterial-venous fistula remodeling in subjects with stage IV and V chronic kidney disease. J. Vasc. Access 2010, 11, 329–334. [Google Scholar] [CrossRef] [PubMed]
  39. Korsheed, S.; Crowley, L.; Fluck, R.; McIntyre, C. Creation of an arteriovenous fistula is associated with significant acute local and systemic changes in microvascular function. Nephron Clin. Pract. 2013, 123, 173–179. [Google Scholar] [CrossRef]
  40. Chai, S.; Sanip, Z.; Rasool, A.; Shokri, A.; Halim, A.; Saad, A.; Sulaiman, W. Systemic microvascular endothelial function with arteriovenous fistula creation in chronic kidney disease. J. Res. Med. Sci. 2022, 27, 46. [Google Scholar] [CrossRef]
  41. Werner, N.; Junk, S.; Laufs, U.; Link, A.; Walenta, K.; Bohm, M.; Nickenig, G. Intravenous transfusion of endothelial progenitor cells reduces neointima formation after vascular injury. Circ. Res. 2003, 93, 17–24. [Google Scholar] [CrossRef]
  42. Lee, T.; Haq, N.U. New Developments in Our Understanding of Neointimal Hyperplasia. Adv. Chronic Kidney Dis. 2015, 22, 431–437. [Google Scholar] [CrossRef] [PubMed]
  43. Huynh, T.N.; Chacko, B.K.; Teng, X.; Brott, B.C.; Allon, M.; Kelpke, S.S.; Thompson, J.A.; Patel, R.P.; Anayiotos, A.S. Effects of venous needle turbulence during ex vivo hemodialysis on endothelial morphology and nitric oxide formation. J. Biomech. 2007, 40, 2158–2166. [Google Scholar] [CrossRef] [PubMed]
  44. Baltazar, S.; Northrup, H.; Chang, J.; Somarathna, M.; Isayeva Waldrop, T.; Lee, T.; Shiu, Y.T. Effects of endothelial nitric oxide synthase on mouse arteriovenous fistula hemodynamics. Sci. Rep. 2023, 13, 22786. [Google Scholar] [CrossRef] [PubMed]
  45. Han, Y.-C.; Liu, Y.-T.; Zhang, H.; Xu, Y.; Liu, J.; Chen, H.; Song, N.; Qin, D.-L.; Yang, S. VDR alleviates endothelial cell injury in arteriovenous fistula through inhibition of P66Shc-mediated mitochondrial ROS. Sci. Rep. 2023, 13, 22786. [Google Scholar] [CrossRef]
  46. Talmor-Barkan, Y.; Bernheim, J.; Green, J.; Benchertrit, S.; Rasdhid, G. Calcitriol counteracts endothelial cell pro-inflammatory processes in a chronic kidney disease-like environment. J. Steroid Biochem. Mol. Biol. 2011, 124, 19–24. [Google Scholar] [CrossRef]
  47. Cuenca, M.V.; Ferrantelli, E.; Meinster, E.; Pouw, S.M.; Kovacevic, I.; Menezes, R.X.d.; Niessen, H.W.; Beelen, R.H.J.; Hordijk, P.L.; Vervloet, M.G. Vitamin D Attenuates Endothelial Dysfunction in Uremic Rats and Maintains Human Endothelial Stability. J. Am. Heart Assoc. 2018, 7, e008776. [Google Scholar] [CrossRef] [PubMed]
  48. Brahmbhatt, A.; Nieves-Torres, E.; Yang, B.; Edwards, W.D.; Chaudhury, P.R.; Lee, M.K.; Kong, H.; Mukhopadhyay, D.; Kumar, R.; Misra, S. The role of Iex-1 in the pathogenesis of venous neointimal hyperplasia associated with hemodialysis arteriovenous fistula. PLoS ONE 2014, 9, e102542. [Google Scholar] [CrossRef]
  49. Cai, C.; Kilari, S.; Zhao, C.; Singh, A.K.; Simeon, M.L.; Misra, A.; Li, Y.; Takahashi, E.; Kumar, R.; Misra, S. Adventitial delivery of nanoparticles encapsulated with 1α, 25-dihydroxyvitamin D3 attenuates restenosis in a murine angioplasty model. Sci. Rep. 2021, 11, 4772. [Google Scholar] [CrossRef] [PubMed]
  50. Valdivielso, J.M.; Rodríguez-Puyol, D.; Pascual, J.; Barrios, C.; Bermúdez-López, M.; Sánchez-Niño, M.D.; Pérez-Fernández, M.; Ortiz, A. Atherosclerosis in Chronic Kidney Disease: More, Less, or Just Different? Arterioscler. Thromb. Vasc. Biol. 2019, 39, 1938–1966. [Google Scholar] [CrossRef]
  51. Chen, P.; Qin, L.; Li, G.; Wang, Z.; Dahlman, J.; Malagon-Lopez, J.; Gujja, S.; Clifone, N.; Kauffman, K.; Sun, L.; et al. Endothelial TGF-β signalling drives vascular inflammation and atherosclerosis. Nat. Metab. 2019, 1, 912–926. [Google Scholar] [CrossRef]
  52. Kluza, E.; Beldman, T.; Shami, A.; Scholl, E.; Malinova, T.; Grootemaat, A.; van der Wel, N.; Gonçalves, I.; Huveneers, S.; Mulder, W.J.M.; et al. Diverse ultrastructural landscape of atherosclerotic endothelium. Atherosclerosis 2021, 339, 35–45. [Google Scholar] [CrossRef] [PubMed]
  53. Kane, J.; Vos, W.G.; Bosmans, L.A.; van Os, B.W.; den Toom, M.; Hoeksema-Hackmann, S.; Moen-de Wit, D.; Gijbels, M.; Beckers, L.; Grefhorst, A.; et al. Peritoneal Dialysis Aggravates and Accelerates Atherosclerosis in Uremic ApoE-/- Mice. J. Am. Heart Assoc. 2024, 13, e034066. [Google Scholar] [CrossRef] [PubMed]
  54. Dzobo, K.E.; Cupido, A.; Mol, B.; Stiekema, L.; Versloot, M.; Winkelmeijer, M.; Peter, J.; Pennekamp, A.; Havik, S.; Vaz, F.; et al. Diacylglycerols and Lysophosphatidic Acid, Enriched on Lipoprotein(a), Contribute to Monocyte Inflammation. Arterioscler. Thromb. Vasc. Biol. 2024, 44, 720–740. [Google Scholar] [CrossRef] [PubMed]
  55. Libby, P.; Smith, R.; Rubin, E.J.; Glassberg, M.K.; Farkouh, M.E.; Rosenson, R.S. Inflammation unites diverse acute and chronic diseases. Eur. J. Clin. Investig. 2024, 24, e14280. [Google Scholar] [CrossRef]
  56. Bikbov, B.; Purcell, C.A.; Levey, A.S.; Smith, M.; Abdoli, A.; Abebe, M.; Adebayo, O.M.; Afarideh, M.; Agarwal, S.K.; Agudelo-Botero, M.; et al. Global, regional, and national burden of chronic kidney disease, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2020, 395, 709–733. [Google Scholar] [CrossRef]
  57. Kraus, M.A.; Kalra, P.A.; Hunter, J.; Menoyo, J.; Stankus, N. The prevalence of vascular calcification in patients with end-stage renal disease on hemodialysis: A cross-sectional observational study. Ther. Adv. Chronic Dis. 2015, 6, 84–96. [Google Scholar] [CrossRef]
  58. Toussaint, N.D.; Lau, K.K.; Polkinghorne, K.R.; Kerr, P.G. Measurement of vascular calcification using CT fistulograms. Nephrol. Dial. Transplant. 2007, 22, 484–490. [Google Scholar] [CrossRef]
  59. Feenstra, L.; Kutikhin, A.G.; Shishkova, D.K.; Buikema, H.; Zeper, L.W.; Bourgonje, A.R.; Krenning, G.; Hillebrands, J.-L. Calciprotein Particles Induce Endothelial Dysfunction by Impairing Endothelial Nitric Oxide Metabolism. Arterioscler. Thromb. Vasc. Biol. 2023, 43, 443–455. [Google Scholar] [CrossRef]
  60. Shishkova, D.; Lobov, A.; Repkin, E.; Markova, V.; Markova, Y.; Sinitskaya, A.; Sinitsky, M.; Kondratiev, E.; Torgunakova, E.; Kutikhin, A.; et al. Calciprotein Particles Induce Cellular Compartment-Specific Proteome Alterations in Human Arterial Endothelial Cells. J. Cardiovasc. Dev. Dis. 2023, 11, 5. [Google Scholar] [CrossRef]
  61. Lee, J.Y.; Ok, K.Y. Pre-existing arterial pathologic changes affecting arteriovenous fistula patency and cardiovascular mortality in hemodialysis patients. Korean J. Intern. Med. 2017, 32, 790–797. [Google Scholar] [CrossRef]
  62. MacRae, J.; Ahmed, S.; Hemmelgam, B.; Sun, Y.; Martin, B.; Roifman, I.; Anderson, T. Role of vascular function in predicting arteriovenous fistula outcomes: An observational pilot study. Can. J. Kidney Health Dis. 2015, 2, 55. [Google Scholar] [CrossRef] [PubMed]
  63. Erdem, Y.; Haznedaroglu, I.; Celik, I.; Yalcin, A.; Yasavul, U.; Turgan, C.; Caglar, S. Coagulation, fibrinolysis and fibrinolysis inhibitors in haemodialysis patients: Contribution of arteriovenous fistula. Nephrol. Dial. Transpl. 1996, 11, 1299–1305. [Google Scholar] [CrossRef]
  64. Harlacher, E.; Wollenhaupt, J.; Baaten, C.; Noels, H. Impact of Uremic Toxins on Endothelial Dysfunction in Chronic Kidney Disease: A Systematic Review. Int. J. Mol. Sci. 2022, 23, 531. [Google Scholar] [CrossRef]
  65. Cardinal, H.; Raymond, M.; Hebert, M.; Madore, F. Uraemic plasma decreases the expression of ABCA1, ABCG1 and cell-cycle genes in human coronary arterial endothelial cells. Nephrol. Dial. Transpl. 2007, 22, 409–416. [Google Scholar] [CrossRef]
  66. Juni, R.P.; Al-Shama, R.; Kuster, D.W.D.; van der Velden, J.; Hamer, H.M.; Vervloet, M.G.; Eringa, E.C.; Koolwijk, P.; van Hinsbergh, V.W.M. Empagliflozin restores chronic kidney disease-induced impairment of endothelial regulation of cardiomyocyte relaxation and contraction. Kidney Int. 2021, 99, 1088–1101. [Google Scholar] [CrossRef] [PubMed]
  67. Juni, R.P.; Kuster, D.W.D.; Goebel, M.; Helmes, M.; Musters, R.J.P.; van der Velden, J.; Koolwijk, P.; Paulus, W.J.; van Hinsbergh, V.W.M. Cardiac Microvascular Endothelial Enhancement of Cardiomyocyte Function Is Impaired by Inflammation and Restored by Empagliflozin. JACC Basic Transl. Sci. 2019, 4, 575–591. [Google Scholar] [CrossRef]
  68. Tian, D.; Jin, X.; Zeng, X.; Wang, Y. Notch Signaling in Endothelial Cells: Is It the Therapeutic Target for Vascular Neointimal Hyperplasia? Int. J. Mol. Sci. 2017, 18, 1615. [Google Scholar] [CrossRef]
  69. Owens, C.; Gasper, W.J.; Rahman, A.S.; Conte, M.S. Vein graft failure. J. Vasc. Surg. 2015, 61, 203–216. [Google Scholar] [CrossRef]
  70. Sharony, R.; Pintucci, G.; Saunders, P.; Grossi, E.; Baumann, F.; Galloway, A.; Mignatti, P. Matrix metalloproteinase expression in vein grafts: Role of inflammatory mediators and extracellular signal-regulated kinases-1 and -2. Am. J. Physiol.-Heart Circ. Physiol. 2006, 290, 1651–1659. [Google Scholar] [CrossRef]
  71. Kuwahara, G.; Hashimoto, T.; Tsuneki, M.; Yamamoto, K.; Assi, R.; Foster, T.R.; Hanisch, J.J.; Bai, H.; Hu, H.; Protack, C.D.; et al. CD44 Promotes Inflammation and Extracellular Matrix Production During Arteriovenous Fistula Maturation. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1147–1156. [Google Scholar] [CrossRef]
  72. Matsubara, Y.; Kiwan, G.; Fereydooni, A.; Langford, J.; Dardik, A. Distinct subsets of T cells and macrophages impact venous remodeling during arteriovenous fistula maturation. JVS Vasc. Sci. 2020, 1, 207–218. [Google Scholar] [CrossRef] [PubMed]
  73. Yao, J.; Zhao, X.; Tan, F.; Cao, X.; Guo, S.; Li, X.; Huang, Z.; Diabakte, K.; Wang, L.; Liu, M.; et al. Early modulation of macrophage ROS-PPARγ-NF-κB signalling by sonodynamic therapy attenuates neointimal hyperplasia in rabbits. Sci. Rep. 2020, 10, 11638. [Google Scholar] [CrossRef] [PubMed]
  74. Ratti, S.; Mauro, R.; Rocchi, C.; Mongiorgi, S.; Ramazzotti, G.; Gargiulo, M.; Manzoli, L.; Cocco, L.; Fiume, R. Roles of PI3K/AKT/mTOR Axis in Arteriovenous Fistula. Biomolecules 2022, 12, 350. [Google Scholar] [CrossRef]
  75. Misra, S.; Doherty, M.G.; Woodrum, D.; Homburger, J.; Mandrekar, J.N.; Elkouri, S.; Sabater, E.A.; Bjarnason, H.; Fu, A.A.; Glockner, J.F.; et al. Adventitial remodeling with increased matrix metalloproteinase-2 activity in a porcine arteriovenous polytetrafluoroethylene grafts. Kidney Int. 2005, 68, 2890–2900. [Google Scholar] [CrossRef] [PubMed]
  76. Yang, B.; Janardhanan, R.; Vohra, P.; Greene, E.L.; Bhattacharya, S.; Withers, S.; Roy, B.; Torres, E.C.N.; Mandrekar, J.; Leof, E.B.; et al. Adventitial transduction of lentivirus-shRNA-VEGF-A in arteriovenous fistula reduces venous stenosis formation. Kidney Int. 2014, 85, 289–306. [Google Scholar] [CrossRef]
  77. Piera-Velazquez, S.; Li, Z.; Jimenez, S. Role of endothelial-mesenchymal transition (EndoMT) in the pathogenesis of fibrotic disorders. Am. J. Pathol. 2011, 179, 1074–1080. [Google Scholar] [CrossRef]
  78. Samra, G.; Rai, V.; Agrawal, D.K. Heterogeneous Population of Immune cells Associated with Early Thrombosis in Arteriovenous Fistula. J. Surg. Res. 2022, 5, 423–434. [Google Scholar] [CrossRef]
  79. Brahmbhatt, A.; Remuzzi, A.; Franzoni, M.; Misra, S. The molecular mechanisms of hemodialysis vascular access failure. Kidney Int. 2016, 89, 303–316. [Google Scholar] [CrossRef]
  80. Janardhanan, R.; Yang, B.; Vohra, P.; Roy, B.; Withers, S.; Bhattacharya, S.; Mandrekar, J.; Kong, H.; Leof, E.B.; Mukhopadhyay, D.; et al. Simvastatin reduces venous stenosis formation in a murine hemodialysis vascular access model. Kidney Int. 2013, 84, 338–352. [Google Scholar] [CrossRef]
  81. Zhao, C.; Zuckerman, S.T.; Cai, C.; Kilari, S.; Singh, A.; Simeon, M.; Recum, H.A.v.; Korley, J.N.; Misra, S. Periadventitial Delivery of Simvastatin-Loaded Microparticles Attenuate Venous Neointimal Hyperplasia Associated with Arteriovenous Fistula. J. Am. Heart Assoc. 2020, 9, e018418. [Google Scholar] [CrossRef]
  82. Cui, J.; Jhajj, H.; Kessinger, C.; Jaffer, F. Atorvastatin decreases venous outflow inflammation and prolonged AVF patency in a murine arteriovenous fistula model. J. Vasc. Interv. Radiol. 2017, 28, 147. [Google Scholar] [CrossRef]
  83. Juncos, J.P.; Grande, J.P.; Kang, L.; Ackerman, A.W.; Croatt, A.J.; Katusic, Z.S.; Nath, K.A. MCP-1 Contributes to Arteriovenous Fistula Failure. J. Am. Soc. Nephrol. 2011, 22, 43–48. [Google Scholar] [CrossRef] [PubMed]
  84. Rai, V.; Agrawal, D.K. Transcriptomic Analysis Identifies Differentially Expressed Genes Associated with Vascular Cuffing and Chronic Inflammation Mediating Early Thrombosis in Arteriovenous Fistula. Biomedicines 2022, 10, 433. [Google Scholar] [CrossRef] [PubMed]
  85. Hu, K.; Guo, Y.; Li, Y.; Zhou, S.; Lu, C.; Cai, C.; Yang, H.; Li, Y.; Wang, W. Identification and Validation of PTGS2 Gene as an Oxidative Stress-Related Biomarker for Arteriovenous Fistula Failure. Antioxidants 2024, 13, 5. [Google Scholar] [CrossRef]
  86. Shih, Y.; Chen, P.; Ko, T.; Huang, P.H.; Ma, H.; Tarng, D.C. MMP-9 Deletion Attenuates Arteriovenous Fistula Neointima through Reduced Perioperative Vascular Inflammation. Int. J. Mol. Sci. 2021, 22, 5448. [Google Scholar] [CrossRef]
  87. Boosani, C.S.; Burela, L. The Exacerbating Effects of the Tumor Necrosis Factor in Cardiovascular Stenosis: Intimal Hyperplasia. Cancers 2024, 16, 1435. [Google Scholar] [CrossRef]
  88. Liang, M.; Wang, Y.; Liang, A.; Mitch, W.E.; Roy-Chaudhury, P.; Han, G.; Cheng, J. Migration of smooth muscle cells from the arterial anastomosis of arteriovenous fistulas requires Notch activation to form neointima. Kidney Int. 2015, 88, 490–502. [Google Scholar] [CrossRef] [PubMed]
  89. Worssam, M.D.; Jørgensen, H.F. Mechanisms of vascular smooth muscle cell investment and phenotypic diversification in vascular diseases. Biochem. Soc. Trans. 2021, 49, 2101–2111. [Google Scholar] [CrossRef]
  90. Zhao, J.; Jourd’heuil, F.L.; Xue, M.; Conti, D.; Lopez-Soler, R.I.; Ginnan, R.; Asif, A.; Singer, H.A.; Jourd’heuil, D.; Long, X. Dual Function for Mature Vascular Smooth Muscle Cells During Arteriovenous Fistula Remodeling. J. Am. Heart Assoc. 2017, 6, e004891. [Google Scholar] [CrossRef]
  91. Tang, H.-Y.; Chen, A.-Q.; Zhang, H.; Gao, X.-F.; Kong, X.-Q.; Zhang, J.-J. Vascular Smooth Muscle Cells Phenotypic Switching in Cardiovascular Diseases. Cells 2022, 11, 4060. [Google Scholar] [CrossRef]
  92. Xin, Y.; Zhang, Z.; Lv, S.; Xu, S.; Liu, A.; Li, H.; Li, P.; Han, H.; Liu, Y. Elucidating VSMC phenotypic transition mechanisms to bridge insights into cardiovascular disease implications. Front. Cardiovasc. Med. 2024, 11, 1400780. [Google Scholar] [CrossRef] [PubMed]
  93. Chen, R.; McVey, D.G.; Shen, D.; Huang, X.; Ye, S. Phenotypic Switching of Vascular Smooth Muscle Cells in Atherosclerosis. J. Am. Heart Assoc. 2023, 12, e031121. [Google Scholar] [CrossRef]
  94. Sorokin, V.; Vickneson, K.; Kofidis, T.; Woo, C.C.; Lin, X.Y.; Foo, R.; Shanahan, C.M. Role of Vascular Smooth Muscle Cell Plasticity and Interactions in Vessel Wall Inflammation. Front. Immunol. 2020, 11, 599415. [Google Scholar] [CrossRef]
  95. Shi, J.; Yang, Y.; Cheng, A.; Xu, G.; He, F. Metabolism of vascular smooth muscle cells in vascular diseases. Am. J. Physiol.-Heart Circ. Physiol. 2020, 319, 613–631. [Google Scholar] [CrossRef] [PubMed]
  96. Monroy, M.A.; Fang, J.; Li, S.; Ferrer, L.; Birkenbach, M.P.; Lee, I.J.; Wang, H.; Yang, X.-F.; Choi, E.T. Chronic kidney disease alters vascular smooth muscle cell phenotype. Front. Biosci. 2015, 20, 784–795. [Google Scholar] [CrossRef]
  97. Hsieh, T.-B.; Jin, J.-P. Evolution and function of calponin and transgelin. Front. Cell Dev. Biol. 2023, 11, 1206147. [Google Scholar] [CrossRef]
  98. Elmarasi, M.; Elmakaty, I.; Elsayed, B.; Elsayed, A.; Zein, J.A.; Boudaka, A.; Eid, A.H. Phenotypic switching of vascular smooth muscle cells in atherosclerosis, hypertension, and aortic dissection. J. Cell. Physiol. 2024, 239, e31200. [Google Scholar] [CrossRef] [PubMed]
  99. Bezhaeva, T.; Wong, C.-Y.; Vries, M.R.d.; Veer, E.P.v.d.; Alem, C.M.A.v.; Que, I.; Lalai, R.A.; Zonneveld, A.-J.v.; Rotmans, J.I.; Quax, P.H.A. Deficiency of TLR4 homologue RP105 aggravates outward remodeling in a murine model of arteriovenous fistula failure. Sci. Rep. 2017, 7, 10269. [Google Scholar] [CrossRef]
  100. Rai, V.; Singh, H.; Agrawal, D.K. Targeting the Crosstalk of Immune Response and Vascular Smooth Muscle Cells Phenotype Switch for Arteriovenous Fistula Maturation. Int. J. Mol. Sci. 2022, 23, 12012. [Google Scholar] [CrossRef]
  101. Cai, C.; Kilari, S.; Singh, A.K.; Zhao, C.; Simeon, M.L.; Misra, A.; Li, Y.; Misra, S. Differences in Transforming Growth Factor-β1/BMP7 Signaling and Venous Fibrosis Contribute to Female Sex Differences in Arteriovenous Fistulas. J. Am. Heart Assoc. 2020, 9, e017420. [Google Scholar] [CrossRef]
  102. Zhang, F.; Li, J.; Yu, J.; Jiang, Y.; Xiao, H.; Yang, Y.; Liang, Y.; Liu, K.; Luo, X.; Zhang, F.; et al. Risk factors for arteriovenous fistula dysfunction in hemodialysis patients: A retrospective study. Sci. Rep. 2023, 13, 21325. [Google Scholar] [CrossRef] [PubMed]
  103. Ahmed, F.A.; Catic, A.G. Decision-Making in Geriatric Patients with End-Stage Renal Disease: Thinking Beyond Nephrology. J. Clin. Med. 2019, 8, 5. [Google Scholar] [CrossRef] [PubMed]
  104. Chou, A.; Li, K.C.; Brown, M.A. Survival of Older Patients With Advanced CKD Managed Without Dialysis: A Narrative Review. Kidney Med. 2022, 4, 100447. [Google Scholar] [CrossRef]
  105. Peterson, W.J.; Barker, J.; Allon, M. Disparities in fistula maturation persist despite preoperative vascular mapping. Clin. J. Am. Soc. Nephrol. 2008, 3, 437–441. [Google Scholar] [CrossRef]
  106. Saran, R.; Elder, S.J.; Goodkin, D.A.; Akiba, T.; Ethier, J.; Rayner, H.C.; Saito, A.; Young, E.W.; Gillespie, B.W.; Merion, R.M.; et al. Enhanced training in vascular access creation predicts arteriovenous fistula placement and patency in hemodialysis patients: Results from the Dialysis Outcomes and Practice Patterns Study. Ann. Surg. 2008, 247, 885–891. [Google Scholar] [CrossRef]
  107. Ethier, J.; Mendelssohn, D.C.; Elder, S.J.; Hasegawa, T.; Akizawa, T.; Akiba, T.; Canaud, B.J.; Pisoni, R.L. Vascular access use and outcomes: An international perspective from the Dialysis Outcomes and Practice Patterns Study. Nephrol. Dial. Transplant. 2008, 23, 3219–3226. [Google Scholar] [CrossRef]
  108. Wilmink, T.; Wijewardane, A.; Lee, K.; Murley, A.; Hollingworth, L.; Powers, S.; Baharani, J. Effect of ethnicity and socioeconomic status on vascular access provision and performance in an urban NHS hospital. Clin. Kidney J. 2017, 10, 62–67. [Google Scholar] [CrossRef] [PubMed]
  109. Hall, Y.N. Racial and ethnic disparities in end stage renal disease: Access failure. Clin. J. Am. Soc. Nephrol. 2012, 7, 196–198. [Google Scholar] [CrossRef]
  110. Patibandla, B.K.; Narra, A.; Desilva, R.; Chawla, V.; Vin, Y.; Brown, R.S.; Goldfarb-Rumyantzev, A.S. Disparities in arteriovenous fistula placement in older hemodialysis patients. Hemodial. Int. 2014, 18, 118–126. [Google Scholar] [CrossRef]
  111. Navuluri, R.; Regalado, S. The KDOQI 2006 Vascular Access Update and Fistula First Program Synopsis. Semin. Interv. Radiol. 2009, 26, 122–124. [Google Scholar] [CrossRef]
  112. Lai, C.C.; Fang, H.C.; Mar, G.Y.; Liou, J.C.; Tseng, C.; Liu, C.P. Post-angioplasty Far Infrared Radiation Therapy Improves 1-Year Angioplasty-Free Hemodialysis Access Patency of Recurrent Obstructive Lesions. Eur. J. Vasc. Endovasc. Surg. 2013, 46, 726–732. [Google Scholar] [CrossRef]
  113. Lin, C.-C.; Chang, C.-F.; Lai, M.-Y.; Chen, T.-W.; Lee, P.-C.; Yang, W.-C. Far-infrared therapy: A novel treatment to improve access blood flow and unassisted patency of arteriovenous fistula in hemodialysis patients. J. Am. Soc. Nephrol. 2007, 18, 985–992. [Google Scholar] [CrossRef]
  114. Lin, C.-C.; Yang, W.-C.; Chen, M.-C.; Liu, W.-S.; Yang, C.-Y.; Lee, P.-C. Effect of Far Infrared Therapy on Arteriovenous Fistula Maturation: An Open-Label Randomized Controlled Trial. Am. J. Kidney Dis. 2013, 62, 304–311. [Google Scholar] [CrossRef]
  115. Lin, C.-C.; Chung, M.-Y.; Yang, W.-C.; Lin, S.-J.; Lee, P.-C. Length polymorphisms of heme oxygenase-1 determine the effect of far-infrared therapy on the function of arteriovenous fistula in hemodialysis patients: A novel physicogenomic study. Nephrol. Dial. Transplant. 2013, 28, 1284–1293. [Google Scholar] [CrossRef] [PubMed]
  116. Lindhard, K.; Jensen, B.L.; Pedersen, B.L.; Meyer-Olesen, C.; Rix, M.; Hansen, H.P.; Schalkwijk, C.; Waarenburg, M.; Heaf, J.; Hansen, D. Far infrared treatment on the arteriovenous fistula induces changes in sVCAM and sICAM in patients on hemodialysis. Nephrol. Dial. Transplant. 2023, 38, 1752–1760. [Google Scholar] [CrossRef]
  117. Bashar, K.; Healy, D.; Browne, L.D.; Kheirelseid, E.A.H.; Walsh, M.T.; Moloney, M.C.; Burke, P.E.; Kavanagh, E.G.; Walsh, S.R. Role of Far Infra-Red Therapy in Dialysis Arterio-Venous Fistula Maturation and Survival: Systematic Review and Meta-Analysis. PLoS ONE 2014, 9, e104931. [Google Scholar] [CrossRef] [PubMed]
  118. Cui, J.; Kessinger, C.W.; Jhajj, H.S.; Grau, M.S.; Misra, S.; Libby, P.; McCarthy, J.R.; Jaffer, F.A. Atorvastatin Reduces In Vivo Fibrin Deposition and Macrophage Accumulation, and Improves Primary Patency Duration and Maturation of Murine Arteriovenous Fistula. J. Am. Soc. Nephrol. JASN 2020, 31, 931–945. [Google Scholar] [CrossRef]
  119. Chang, H.-H.; Chang, Y.-K.; Lu, C.-W.; Huang, C.-T.; Chien, C.-T.; Hung, K.-Y.; Huang, K.-C.; Hsu, C.-C.; Chang, H.-H.; Chang, Y.-K.; et al. Statins Improve Long Term Patency of Arteriovenous Fistula for Hemodialysis. Sci. Rep. 2016, 6, 22197. [Google Scholar] [CrossRef] [PubMed]
  120. Sanada, S.; Miyasaka, Y.; Kanno, A.; Sato, K.; Sato, M.; Sugai, H.; Kitamura, H.; Sato, T.; Taguma, Y. Efficacy of statin on vascular access patency in diabetic hemodialysis patients. J. Vasc. Access 2017, 18, 295–300. [Google Scholar] [CrossRef]
  121. Roan, J.-N.; Lin, W.-H.; Tsai, M.-T.; Kuo, T.-H.; Lin, T.-W.; Chen, D.-Y.; Fang, S.-Y.; Hsieh, Y.-P.; Li, C.-Y.; Lam, C.-F. Rosuvastatin Failed to Improve Arteriovenous Fistula Patency for Hemodialysis in Diabetic Patients—A Randomized Clinical Trial. Acta Cardiol. Sin. 2021, 37, 18–29. [Google Scholar] [CrossRef]
  122. Yang, B.; Brahmbhatt, A.; Torres, E.N.; Thielen, B.; McCall, D.L.; Engel, S.; Bansal, A.; Pandey, M.K.; Dietz, A.B.; Leof, E.B.; et al. Tracking and Therapeutic Value of Human Adipose Tissue-derived Mesenchymal Stem Cell Transplantation in Reducing Venous Neointimal Hyperplasia Associated with Arteriovenous Fistula. Radiology 2016, 279, 513–522. [Google Scholar] [CrossRef] [PubMed]
  123. Cai, C.; Kilari, S.; Zhao, C.; Simeon, M.L.; Misra, A.; Li, Y.; van Wijnen, A.J.; Mukhopadhyay, D.; Misra, S. Therapeutic Effect of Adipose Derived Mesenchymal Stem Cell Transplantation in Reducing Restenosis in a Murine Angioplasty Model. J. Am. Soc. Nephrol. 2020, 31, 1781–1795. [Google Scholar] [CrossRef] [PubMed]
  124. Piryani, A.K.; Kilari, S.; Takahashi, E.; DeMartino, R.R.; Mandrekar, J.; Dietz, A.B.; Misra, S. Rationale and Trial Design of MesEnchymal Stem Cell Trial in Preventing Venous Stenosis of Hemodialysis Vascular Access Arteriovenous Fistula (MEST AVF Trial). Kidney360 2021, 2, 1945–1952. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Suitable vascular remodelling determines initial AVF maturation and long-term patency. Following AVF creation, the vessel must thicken and remodel outwardly, increasing the internal diameter to allow for appropriate haemodialysis. Failure immediately following surgery involves no vascular remodelling or increases in diameter. Alternatively, failure over a longer term typically involves venous stenosis secondary to neointimal hyperplasia. Made with BioRender.
Figure 1. Suitable vascular remodelling determines initial AVF maturation and long-term patency. Following AVF creation, the vessel must thicken and remodel outwardly, increasing the internal diameter to allow for appropriate haemodialysis. Failure immediately following surgery involves no vascular remodelling or increases in diameter. Alternatively, failure over a longer term typically involves venous stenosis secondary to neointimal hyperplasia. Made with BioRender.
Cells 13 01637 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kane, J.; Lemieux, A.; Baranwal, G.; Misra, S. The Role of Cardio-Renal Inflammation in Deciding the Fate of the Arteriovenous Fistula in Haemodialysis Therapy. Cells 2024, 13, 1637. https://doi.org/10.3390/cells13191637

AMA Style

Kane J, Lemieux A, Baranwal G, Misra S. The Role of Cardio-Renal Inflammation in Deciding the Fate of the Arteriovenous Fistula in Haemodialysis Therapy. Cells. 2024; 13(19):1637. https://doi.org/10.3390/cells13191637

Chicago/Turabian Style

Kane, Jamie, Alaura Lemieux, Gaurav Baranwal, and Sanjay Misra. 2024. "The Role of Cardio-Renal Inflammation in Deciding the Fate of the Arteriovenous Fistula in Haemodialysis Therapy" Cells 13, no. 19: 1637. https://doi.org/10.3390/cells13191637

APA Style

Kane, J., Lemieux, A., Baranwal, G., & Misra, S. (2024). The Role of Cardio-Renal Inflammation in Deciding the Fate of the Arteriovenous Fistula in Haemodialysis Therapy. Cells, 13(19), 1637. https://doi.org/10.3390/cells13191637

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop