Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jul 9.
Published in final edited form as: Am J Physiol Heart Circ Physiol. 2025 Jun 4;329(1):H241–H257. doi: 10.1152/ajpheart.00010.2025

Cellular and molecular mechanisms underlying hemodialysis arteriovenous fistula dysfunction and approaches to promote maturation: A vascular perspective

Yan-Ting Shiu 1,2,3,*, Hannah Northrup 2, Yufeng Huang 2, Monique Cho 1,2, Kanokwan Bunsawat 4,5,6,*
PMCID: PMC12237592  NIHMSID: NIHMS2088174  PMID: 40465509

Abstract

Hemodialysis requires functional vascular access, which serves as the conduit for blood flow between the patient and the hemodialysis machine. The arteriovenous fistula (AVF), created by surgically connecting an artery to a nearby vein in the upper extremity, is the preferred form of vascular access for maintenance hemodialysis. Newly created AVFs must undergo a maturation process, during which the fistula vein enlarges and develops sufficient lumen size and blood flow to be used effectively for dialysis. However, since the invention of AVFs 60 years ago, rates of AVF maturation failure have remained high. Currently, no proven therapies exist to promote maturation or prevent maturation failure. This review examines the current understanding of the cellular and molecular mechanisms underlying AVF maturation failure, with particular focus on the impact of the chronic kidney disease (CKD) environment on the vascular system. In CKD, patients often have elevated levels of uremic toxins, increased oxidative stress, and chronic inflammation, all of which adversely affect the function of vascular wall cells (such as endothelial cells, vascular smooth muscle cells, and fibroblasts) and circulating cells (such as platelets and immune/inflammatory cells). The goal of this review is to explore the mechanisms influencing both native and AVF vasculature in the context of CKD. Additionally, we will discuss current therapies aimed at improving native vasculature in CKD patients, as well as investigational approaches to promote AVF maturation.

Keywords: Chronic kidney disease, End-stage kidney disease, Vascular cells, Vascular functions

1. Introduction

The global prevalence of end-stage kidney disease (ESKD), also known as kidney failure and Stage 5 chronic kidney disease (CKD) requiring kidney replacement therapy, is estimated to be between 4.9 and 7.1 million people needing kidney replacement therapy.(1) Hemodialysis is the most common form of kidney replacement therapy in the United States (U.S.) and many developed countries.(2) Currently, more than 808,000 people in the U.S. are living with ESKD, with approximately 68% of them undergoing hemodialysis. The remaining patients either use peritoneal dialysis or have received kidney transplants.(3) Hemodialysis requires functional vascular access, which serves as the conduit for transporting blood from the patient to the dialysis machine and back. Autologous arteriovenous fistulas (AVFs), compared to other types of vascular access (i.e., synthetic arteriovenous grafts and central venous catheters), are more durable in long-term use and require less interventions, if they successfully mature (i.e., develop a sufficiently large lumen and high blood flow rate needed for dialysis).(4) For these reasons, AVFs have been the recommended form of vascular access for most patients since the Fistula First Initiative in 2003.(5) The Dialysis Outcomes and Practice Patterns Study (DOPPS) found that, in the U.S., AVF use increased from 20.7% to 26.5% between 1997 and 2003 (DOPPS I and II)(6) and to 63% and 68% in 2010 and 2013, respectively (DOPPS IV and V).(7)

In 1965, the surgeon Dr. Appel pioneered the first autologous AVF, a procedure later named the Brescia-Cimino fistula to honor the nephrologists who contributed to the concept and development of the AVF.(8) AVFs are surgically created by connecting an artery and a nearby vein directly, usually in the upper extremity. They are commonly constructed at the radiocephalic, brachiocephalic, and brachiobasilic locations (Figure 1). Approximately 49% of AVFs were located in the upper arm and 26% were in the forearm between 2009 and 2015 (DOPPS IV and V).(9) AVFs must successfully mature before they can be used for dialysis. A typical mature and functional AVF vein sees its lumen diameter growing from a preoperative value of ~2 mm to ~6 mm and the blood flow rate increasing from a preoperative value of 25 mL/min to >500 mL/ min.(1012) AVF maturation typically requires an average time of 6-12 weeks or longer.(11) Unfortunately, a significant number of newly created AVFs do not mature. Analysis of recent data from the United States Renal Data System revealed an AVF maturation failure rate of 51%.(13)

Figure 1: Major vasculature of the arm.

Figure 1:

The first panel shows the native vasculature commonly used for arteriovenous fistula (AVF) creation. The remaining panels show the radiocephalic, brachiocephalic, and brachiobasilic AVFs, respectively. Created in BioRender. Northrup, H. (2025) https://BioRender.com/l42a457

Historically, AVF maturation failure has been largely attributed to venous stenosis, a process involving “neointimal hyperplasia” formation (often referred to as inward remodeling) (Figure 2), which narrows the lumen, leading to reduced blood flow and increased risk of thrombosis within the fistula.(1416) Consequently, most previous studies have focused on treatments aimed at preventing thrombosis, inflammation, and cell proliferation. In recent years, insufficient “lumen expansion” (often referred to as outward remodeling) (Figure 2) has garnered increased attention.(17, 18) Successful AVF maturation is now considered a balance between hyperplasia (inward remodeling) and lumen expansion (outward remodeling) that allows for the flow and cannulation required for dialysis. Recent research has explored treatments that modulate vasodilation and the extracellular matrix (ECM). Given the complexity of AVF maturation failure, further investigation is essential to gain a more comprehensive understanding of the underlying pathophysiology and develop effective therapies to ensure successful maturation for hemodialysis.

Figure 2: Successful and failed arteriovenous fistula (AVF) remodeling.

Figure 2:

A) Idealized native vein. B) Idealized AVF vein showing outward remodeling. C) Idealized AVF vein showing outward remodeling with limited neointimal hyperplasia. D) Idealized AVF vein showing lack of outward remodeling. E) Idealized AVF vein showing outward remodeling with large neointimal hyperplasia. Used with permission: Northrup, H. M. (2022). Effects of Treatments on the Geometry and Vascular Biomechanics of Arteriovenous Fistulas in Rodents and Patients (Doctoral dissertation, The University of Utah).

Indeed, current therapies for AVF maturation failure focus on intervention after the dysfunction has occurred (e.g., plain balloon angioplasty, stent grafts, and drug-coated balloons). There is a lack of effective treatments to prevent maturation failure or to promote successful maturation. Thus far, landmark multicenter randomized controlled trials have reported systemic therapies to be minimally effective (Table 1). The impact of this deficit in effective clinical therapies is reflected in the Medicare costs in the U.S. Of the $2.8 billion spent on establishing vascular access for hemodialysis, most of it stemmed from AVF dysfunction or primary patency loss.(19) The financial burden of ESKD care is considered the most expensive under Medicare, with the majority of the cost attributed to dialysis treatments, which average around $100,000 per year per patient.(20) To put this in context, the average annual cost for a cardiovascular disease patient on Medicare is around $20,000.(21) Accordingly, further research focused on therapies aimed at improving AVF maturation and longevity is necessary to reduce costs and adequately address this unmet clinical need.

Table 1:

Recent and landmark clinical or pivotal studies, reported at clinicaltrials.gov, for therapies to improve arteriovenous fistula maturation.

Sponsor/National Clinical Trial Number (NCT) Year start Projected End Targeted cell type Therapy Delivery Mechanism of action Phase
Focus on platelet effects
National Institute of Diabetes and Digestive and Kidney Diseases
NCT03289520
2003 2007 Platelets Clopidogrel Oral Antiplatelet (binds platelet P2RY12 purinergic receptor) 3
Australasian Kidney Trials Network
*CTRN12607000569404
2008 2015 Many cell types Fish oil Oral Pleiotropic effects (decreases platelet aggregation, blood viscosity, and inflammation; promotes vasodilation) 3
HMG-CoA reductase inhibition
National Cheng-Kung University Hospital
NCT01863914
2012 2019 Many cell types Rosuvastatin Oral HMG-CoA reductase inhibitor Phase 2
Albany Medical College
NCT03188978
2018 2019 Many cell types Atorvastatin Oral HMG-CoA reductase inhibitor Phase 1, Phase 2
Pancreatic elastase
Proteon Therapeutics
NCT02110901
2014 2018 Smooth muscle cells PRT-201 Perivascular Recombinant human type 1 pancreatic elastase Phase 3
PDE5A inhibition
University of Alabama at Birmingham
NCT02414204
2017 2018 Smooth muscle cells Sildenafil Oral PDE5A inhibitor N/A
VEGF-A inhibition
Mayo Clinic
NCT02695641
2019 2021 Endothelial cells Bevacizumab Intravenous VEGF-A inhibition Early Phase 1
mTOR inhibition
Vascular Therapies, Inc
NCT05425056
2022 2025 Immune cells Sirolimus eluting collagen implant (SeCl) Perivascular mTOR inhibition Phase 3
Hand or arm exercise/movement
University Health Network, Toronto
NCT02205944
2014 2018 Many cell types Two types of presurgical exercise N/A Handgrip exercise N/A
Changi General Hospital
NCT03137680
2017 2019 Many cell types Pre-operative forearm exercises N/A Softball hand exercise N/A
Hospital General Universitario Gregorio Maranon
NCT03213756
2017 2019 Many cell types Preoperative isometric preoperative exercise N/A Isometric hand and arm exercises N/A
National Taiwan University Hospital
NCT03077815
2017 2018 Many cell types Arm exercise N/A Gripping arm movements N/A
Singapore General Hospital
NCT03886116
2018 2021 Many cell types Pre-operative forearm exercises N/A Forearm exercises N/A
Guangdong Provincial People’s Hospital
NCT03741985
2018 2019 Many cell types Dumbbell exercise N/A Dumbbell exercise N/A
Hospices Civils de Lyon
NCT04034433
2019 2026 Many cell types Perioperative handgrip training N/A Handgrip exercise N/A
Taipei Medical University
NCT05493046
2022 2026 Many cell types Interactive hand exercise game N/A Increase handgrip strength N/A
Physical therapy
Herlev and Gentofte Hospital
NCT04011072
2019 2023 Endothelial cells, fibroblasts Far infrared therapy N/A increased molecular vibration by photon absorption N/A
Suzhou Municipal Hospital
NCT06249373
2024 2026 Many cell types Low-intensity pulse ultrasound N/A mechanical vibrations in tissues and cells N/A
Stem cell therapy
Houssam Farres
NCT04392206
2020 2025 Many cell types Allogeneic adipose derived mesenchymal stem cells Perivascular Stem cell therapy Phase 1
Mechanical support
VenoStent
NCT05418816
2021 2024 Many cell types Bioabsorbable perivascular wrap, SelfWrap Perivascular biomimetic mechanical support N/A
VenoStent
NCT06001827
2023 2027 Many cell types Bioabsorbable perivascular wrap, SelfWrap Perivascular biomimetic mechanical support N/A
*

anzctr.org au identifier

The lack of therapies for AVF maturation highlights a gap in our understanding of the underlying mechanisms. The AVF milieu is unique. Cells in AVFs are exposed to shear stress and flow conditions that are much higher and more disturbed than under normal circumstances, due to the low resistance in the arteriovenous shunt circuit. Additionally, AVFs are almost exclusively created in patients with advanced CKD (i.e., Stage 4 CKD) and ESKD (i.e., Stage 5 CKD). These patients have uremic toxins in their bloodstream, increased oxidative stress, and chronic inflammation, all of which affect cells in the vascular wall (such as endothelial cells, vascular smooth muscle cells, and fibroblasts), as well as cells in circulation (such as platelets and immune/inflammatory cells) (Figure 3).

Figure 3: Overview of the arteriovenous fistula (AVF).

Figure 3:

Key cell types involved in AVF remodeling include vascular wall cells (such as endothelial cells, vascular smooth muscle cells, and fibroblasts) and circulating cells (such as platelets and immune/inflammatory cells). Blood flow is represented by black arrows. Neointimal hyperplasia is shown as limiting blood flow in the AVF vein. Created in BioRender. Northrup, H. (2025) https://BioRender.com/v70n709

Direct anastomosis of the low-pressure vein to the high-pressure artery allows for the redirection of arterial blood through the vein. Not only does this redirection cause an increase in venous flow rate, which is needed for the dialysis machine, but it also elevates pressure and induces flow disturbances—particularly at the arteriovenous anastomosis. AVF flow has been quantified using various parameters, including wall shear stress (WSS) and oscillatory shear index (OSI). WSS is the frictional force exerted by blood flow parallel to the vessel wall and plays a critical role in regulating endothelial cell signaling cascades. OSI is commonly used to quantify disturbed flow, with high OSI indicating oscillatory, non-unidirectional flow, and low OSI reflecting more uniform, unidirectional flow. In a prospective, multi-center clinical study (n=120), He et al. demonstrated that early WSS (1 day after AVF creation) was positively associated with greater subsequent lumen expansion, whereas OSI was negatively associated with lumen expansion.(22) This positive association between early WSS and later lumen diameter was corroborated in a larger, multi-center study (n=602).(23) Importantly, AVF location (Figure 1), postoperative venous diameter, and the presence of stenosis have also been found to influence AVF maturation.(12) These three parameters (AVF location, diameter, stenosis) altered the anastomotic geometry, thereby affecting local hemodynamics, such as WSS and OSI. While the study of hemodynamic parameters and their influence on AVF remodeling is making progress, significant gaps remain in our understanding of the underlying mechanisms. Notably, He et al.(22) reported that WSS in AVFs can reach magnitudes on the order of 1,000 dyn/cm2—far exceeding those observed in native veins (<10 dyn/cm2) and arteries (<100 dyn/cm2). These findings highlight the extreme mechanical environment of the AVF and underscore the importance of vascular mechanobiology in guiding successful AVF maturation and function.

This review aims to explore the current understanding of cell and molecular mechanisms in both native and AVF vasculature within the context of CKD. We will also discuss existing therapies aimed at improving the native vasculature in CKD patients, as well as investigational treatments designed to promote AVF maturation. The effects of abnormal blood flow on AVF maturation failure and the underlying mechanisms have been comprehensively reviewed elsewhere.(2426)

2. Key cellular players involved in AVF remodeling

Vascular disease is a major complication of CKD and can significantly hinder the maturation of AVFs used for hemodialysis. Characterized by endothelial dysfunction, arterial stiffness, vascular calcification, chronic inflammation, and increased oxidative stress, these changes create a hostile environment for the AVF maturation process. This process involves complex interactions among various cell types, including endothelial cells (ECs), vascular smooth muscle cells (SMCs), fibroblasts, platelets, and immune/inflammatory cells (Figure 3). Crosstalk between these key cellular players promotes a pro-thrombotic, pro-inflammatory, and pro-fibrotic environment, further exacerbating vascular dysfunction and AVF maturation failure.

2.1. Endothelial cells

ECs line the inner most layer of both arteries and veins, but their functions and the hemodynamic environments they experience differ.(27) Normally, arterial ECs are adapted to high-pressure, pulsatile blood flow and play a role in regulating vascular tone and preventing thrombosis by releasing vasodilators such as nitric oxide (NO). In contrast, venous ECs are exposed to lower pressure, slower and more laminar blood flow, and lower oxygen levels.(28, 29)

In CKD, vascular endothelial damage occurs early and worsens as the disease progresses.(30) This damage is driven by systemic inflammation, oxidative stress, the accumulation of uremic toxins, and pre-existing risk factors such as hypertension, diabetes mellitus, and hyperlipidemia.(31) The main types of structural damage to the endothelium in CKD include loss of endothelial integrity, increased permeability, endothelial activation by inflammation, osteogenic changes in ECs, endothelial-to-mesenchymal transition, and EC detachment.(32) Damaged ECs in CKD exhibit impaired abilities to regulate vascular tone, inflammation, coagulation, calcification, and angiogenesis. Moreover, the number and function of progenitor endothelial cells, responsible for repairing damaged endothelium and promoting new blood vessel formation, are significantly reduced in CKD.(33)

A key mechanism underlying endothelial dysfunction in CKD is reduced NO production. L- arginine, the precursor for NO synthesis, is converted to NO by endothelial nitric oxide synthase (eNOS) in vascular ECs. Once produced, NO diffuses across cell membranes and activates guanylate cyclase in various cell types, including vascular SMCs and circulating platelets and leukocytes.(34) In vascular SMCs, guanylate cyclase activation leads to myosin light chain dephosphorylation and vasorelaxation. In platelets, NO inhibits activation, adhesion, and aggregation, while it reduces leukocyte adhesiveness. In the CKD milieu, uremic toxins such as asymmetric dimethylarginine inhibit eNOS, while excessive reactive oxygen species (ROS) react readily with NO to form peroxynitrite, a harmful compound that further reduces NO availability.

Chronic oxidative stress in CKD also leads to increased expression of adhesion molecules such as vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) on ECs, promoting the adhesion and infiltration of immune cells such monocytes and lymphocytes. This creates a vicious cycle of endothelial damage, with ECs becoming more activated and releasing more pro-thrombotic and pro-inflammatory factors.(35) Furthermore, ECs in CKD can undergo a phenotypic switch toward a more osteogenic (bone-like) state. This shift leads to the secretion of endothelial microparticles containing adhesion molecules, microRNAs, and osteogenic factors (such as osteocalcin and bone morphogenetic proteins), which can stimulate both ECs and vascular SMCs, contributing to vascular calcification.(36) Damaged ECs also express more pro-thrombotic factors, including tissue factor and von Willebrand factor, while producing less tissue plasminogen activator, promoting a pro-thrombotic state and fibrosis.(37)

In the AVF context, animal studies show that eNOS overexpression in mice improve AVF remodeling (i.e., limited neointimal hyperplasia and improved blood flow in AVFs), whereas eNOS knockout worsens AVF remodeling.(3841) Jumonji domain-containing protein-3 (JMJD3), a histone H3 lysine 27 demethylase, promotes endothelial regeneration. It has been found that endothelial JMJD3 expression was negatively associated with neointimal hyperplasia of AVFs in patients, and knockout of endothelial JMJD3 decreased eNOS expression/NO production and increased neointimal hyperplasia formation in a mouse AVF model.(42) Bulk RNA sequencing of human AVF samples, obtained from patients undergoing 2-stage AVF creation surgery, revealed 17 endothelium- predominant genes that were downregulated in AVFs that failed compared to AVFs that matured.(43) Notable downregulated genes were eNOS, ICAM2, SELL, APOLD1, ARG2, SELP, PALMD, and SRGN.(43) The authors speculated that this downregulation was related to a lower level of endothelial specific transcription factors or due to vastatin, a collagen VIII–derived matrikine, from the upregulation in collagen VIII which was also prominent in human AVFs that failed to mature.(43)

2.2. Vascular smooth muscle cells

Vascular SMCs are the main constitutive stromal cells found in the medial layer of blood vessels where they regulate vascular tone and maintain structural integrity.(44, 45) In arteries, SMCs are more contractile, responding to vasoactive signals to regulate blood pressure. Conversely, veins contain fewer SMCs and are more compliant, designed to accommodate larger blood volumes at lower pressures.

In CKD, vascular SMCs undergo several changes, including phenotypic modulation, increased proliferation, and migration, which collectively lead to pathological vascular remodeling and medial calcification.(36) Medial calcification, along with intimal calcification (also known as atherosclerotic calcification and thought to originate from atherosclerosis), is prominent and is generally considered an irreversible process in CKD.(36) These pathological changes are driven by factors such as hypercalcemia, hyperphosphatemia, hyperthyroidism, oxidative stress, inflammation, alpha-Klotho deficiency, and uremia.(36)

The pathogenesis of vascular SMC changes in CKD have been extensively reviewed, and the phenotypic switching of vascular SMCs is central to theses pathological outcomes.(36, 4648) In their healthy state, vascular SMCs maintain a contractile phenotype, characterized by the expression of smooth muscle alpha-actin and myosin heavy chain.(49) However, in patients with CKD, vascular SMCs shift to a synthetic phenotype, marked by increased expression of ECM proteins such as collagen, elastin, vimentin, and matrix metalloproteinases, as well as pro-inflammatory cytokines and osteogenic genes.(46, 4952) This transition leads to the development of various vascular SMC subtypes, including myofibroblast, macrophage-like, osteogenic, and senescent vascular SMCs, all of which contribute to arterial fibrosis, stiffness, and calcification.

In the AVF context, a recent animal study identified a long non-coding RNA, LncDACH1, as a regulator of neointimal hyperplasia in the AVF.(53) Using smooth muscle-specific knockout and overexpression mouse models, the researchers found that KLF9 downregulates LncDACH1; in turn, LncDACH1 promotes the development of neointimal hyperplasia via the AKT/SRPK1/HSP90 signaling pathway.(53) Another group explored the role of the mineralocorticoid receptor (MR) in AVF remodeling in human and mouse AVFs.(54) Dysfunctional fistulas, collected from patients undergoing surgical revision, had increased MR expression and decreased smooth muscle myosin heavy chain, along with elevated osteopontin and cellular proliferation.(54) In rat AVF models, pharmacologic targeting of MR with finerenone significantly improved AVF outcomes, characterized by increased lumen diameters and flow volume, reduced medial fibrosis, and inhibited SMC phenotypic shifting from contractile to synthetic. These beneficial effects of finerenone were attributed to reduced activity of the EGFR-ERK1/2 signaling pathway.(54)

Using a vascular SMC lineage tracing reporter mouse model, researchers have tracked vascular SMC fate in a rat AVF model.(55) They induced the medial layer SMCs with tamoxifen to express green fluorescent protein (GFP) prior to AVF surgery. Four weeks after AVF creation, they observed medial layer thickening in the vein, composed primarily of differentiated GFP+/MYH11+/Ki67+ SMCs. In contrast, the neointimal hyperplasia at the anastomosis was made up of dedifferentiated GFP+/MYH11− VSMCs. These findings suggest that vascular SMC phenotype switching differs between medial wall thickening and neointimal hyperplasia.(55) The anatomical sources of neointimal cells in AVFs have been comprehensively reviewed elsewhere.(56)

2.3. Fibroblasts

Fibroblasts are specialized cells essential for the formation of connective tissue, which provides structural support to various organs and tissues throughout the body. These cells secrete collagen, a key protein that helps maintain the strength and integrity of tissues.(57) In blood vessels, fibroblasts are predominantly located in the adventitia, the outermost layer of the blood vessel, where they are typically in a quiescent state and lack contractile stress fibers (microfilaments)(58). Although few studies have explored their embryonic origins, these fibroblasts are thought to arise from local mesenchymal cell populations.(58) Vascular fibroblasts are challenging to define in vivo due to the variability in gene expression, even in their resting state. Like interstitial fibroblasts, activated adventitial fibroblasts proliferate, deposit ECM proteins, and secrete pro-inflammatory cytokines and chemokines.(58) These activated fibroblasts—often referred to as myofibroblasts—express contractile proteins like smooth muscle alpha-actin, which is also found in vascular SMCs as mentioned above.(57)

Dysfunctional vascular fibroblasts play a critical role in the progression of arterial diseases, including atherosclerosis, hypertension, and aortic dissection.(59) In the vein, these fibroblasts can be activated in response to changes in wall tension and acquire a contractile apparatus, including microfilaments, fibronexus junctions, and other features commonly found in vascular SMCs.(56) CKD significantly affects vascular fibroblasts, primarily by causing an increase in the level of fibroblast growth factor 23 (FGF-23) which is associated with vascular calcification in CKD patients.(60)

In the context of AVF, the ECM modifications induced by myofibroblasts play a crucial role in maintaining hemostasis during the final stages of AVF remodeling when the lumen size and wall thickness have grown adequately, allowing adaptation to arterial flow.(61) Evidence from in vitro cultures and animal models of restenosis suggests that adventitial myofibroblasts have a remarkable ability to migrate toward the intima in response to injury, contributing to the formation of neointimal hyperplasia.(56) However, vascular SMCs can also dedifferentiate into myofibroblasts. It remains unclear whether the different origins of these myofibroblasts (from SMCs or fibroblasts or else) results in identical or distinct phenotypes and contributions to the AVF maturation process. Additional AVF studies have looked into FGF21, which is a secreted protein associated with various disease states including oxidative stress, inflammation, and atherosclerosis.(62) The serum levels of FGF21 were associated with AVF functional patency loss in patients.(63) In animal studies, parathyroid hormone was found to increase vascular wall thickness and myofibroblasts in a mouse AVF model, and parathyroid hormone increased integrin beta-6. The authors speculated that it may indicate a transition of fibroblasts to myofibroblasts.(64) Given the established role of parathyroid hormone in regulating FGF-23,(65) it is possible that parathyroid hormone led to increased FGF-23 levels, which—as previously discussed in the context of CKD—is associated with vascular calcification. However, the authors did not investigate FGF-23 or vascular calcification in relation to parathyroid hormone treatment.(64)

2.4. Platelets

Platelets are small, anucleate cells that circulate in the blood in large quantities and play a vital role in hemostasis.(66) In addition to their role in clot formation, platelets also contribute to immune and inflammatory responses. These platelets remain inactive under a healthy state but become activated upon injury (i.e., vascular damage). Upon injury, platelets adhere to the subendothelial surface through the interactions between the platelet receptor GPIb-IX and von Willebrand factor, forming a plug, which is further stabilized by fibrin strands through the coagulation cascade. Activated platelets release various chemokines and cytokines, including CXCL4, CXCL5, CXCL8, CCL5, IL-1β, TGFβ and tumor necrosis factor-alpha, as well as signaling molecules like P-selectin, damage-associated molecular patterns, and growth factors (vascular endothelial growth factor (VEGF) and platelet-derived growth factor). These molecules promote leukocyte recruitment, differentiation, and phagocytosis, aiding tissue repair and protecting from pathogens.(66)

In patients with CKD, changes in platelet number and function have been inconsistently reported. Most patients with CKD who are not receiving dialysis do not exhibit a significant reduction in platelet number, while those on maintenance hemodialysis often experience a 20% decrease.(67) The cause of this reduction is unclear but may be due to increased consumption related to the extracorporeal circuit or dialysis membrane-induced platelet aggregation, or potentially insufficient platelet production by megakaryocytes. In addition to reduced platelet numbers, patients with CKD exhibit platelet dysfunction with impaired adhesion, aggregation, granule secretion, thromboxane A2 generation, and platelet-mediated clot retraction.(6870) These abnormalities can result in either thrombotic or hemorrhagic complications.(6870) CKD is also associated with endothelial injury as described above. These changes reduce antiplatelet mediators, promoting platelet activation and aggregation. Consequently, patients with CKD, particularly before starting dialysis, may have a higher thrombotic tendency, with platelet hyperactivity exacerbating inflammatory and immune responses.(68, 71)

During AVF creation, the blood vessel wall is injured, leading to platelet recruitment. Platelet factor 4, a protein expressed in platelets, plays a crucial role in promoting coagulation. Platelet factor 4 is also implicated in fibrosis and has been shown to be upregulated in AVFs, particularly those that fail to mature.(72) While stenosis and thrombosis are the primary causes of AVF maturation failure in clinical practice, there is limited mechanistic research on platelets. Clinical trials have investigated the use of antithrombotic drugs, such as dipyridamole and clopidogrel, to prevent thrombosis and improve AVF outcomes. Although both drugs were effective in reducing thrombosis, they did not significantly enhance AVF maturation.(73, 74) Similarly, neither aspirin or fish oil treatment, which has platelet aggregation inhibitory properties, had significant effects on AVF usability.(75) This limited impact from clinical trials may explain the relative paucity of studies on platelet mechanisms in AVF dysfunction.

2.5. Immune/inflammatory cells

Inflammatory cells circulating in the bloodstream primarily include neutrophils, monocytes, lymphocytes (T cells, B cells, and natural killer cells), eosinophils, and basophils. In patients with CKD, there are often increases in circulating neutrophils, monocytes, and pro-inflammatory lymphocytes, while regulatory T cells, particularly central memory CD4+ and CD8+ T cells, are significantly depleted in patients with ESKD.(7679). Additionally, the proportion of B cells decreases.(77)

As mentioned above, ECs in CKD express elevated levels of adhesion molecules such as VCAM-1, ICAM-1, and E-selectin, which promote the recruitment and adhesion of inflammatory cells, including neutrophils, monocytes, and lymphocytes.(35, 80) These adhered cells secrete pro- inflammatory cytokines and ROS, exacerbating endothelial dysfunction and amplifying vascular inflammation. In CKD, monocytes and macrophages are highly activated, producing elevated pro- inflammatory cytokines such as IL-1β, TNF-α, and monocyte chemoattractant protein-1. They also express angiotensin converting enzyme and secrete TGFβ, which stimulate vascular cells to produce excessive amounts of ECM, leading to vascular fibrosis and stenosis.(79, 81, 82) Additionally, activated monocytes and macrophages release cytokines and growth factors that induce vascular SMC de-differentiation or activation, driving abnormal vessel remodeling such as neointimal hyperplasia.(79) Platelet activation often parallels this process, promoting thrombotic occlusion and further exacerbating vascular damage in CKD.(83) Many studies on AVF remodeling have reported increased inflammatory markers in both human AVF samples and animal models of AVFs; consequently, several excellent reviews focusing on immune and inflammatory cells are available.(84, 85)

3. Vascular function and AVF remodeling

3.1. Association between preoperative vascular function and postoperative venous remodeling in AVFs in patients

Vasodilation and outward remodeling of both the artery and vein are essential for the successful maturation of a newly created AVF (Figure 2).(86) Endothelial dysfunction (characterized by a shift toward proinflammatory, prothrombotic, and reduced vasodilatory states)(87) and increased vascular stiffness (defined as a reduction in the flexibility of the vascular wall to expansion when blood volume increases) are commonly observed in patients with ESKD.(30, 71) The Hemodialysis Fistula Maturation (HFM) Study was a prospective, multicenter cohort study on AVF maturation, funded by the National Institute of Diabetes and Digestive and Kidney Diseases. It was designed to identify clinical and biological factors associated with fistula maturation outcomes.(88) The HFM Study proposed that pre-existing impairments in endothelial function, vascular SMC function, arterial compliance, and venous distensibility disrupt normal arterial and venous dilation processes and impede the vascular remodeling required for successful fistula maturation.(89) While a couple of earlier studies explored the association between arterial elasticity and fistula outcomes,(90, 91) and one study examined the link between venous capacitance and fistula maturation,(92) these studies were limited by small sample sizes (<100 patients), restricting their ability to account for vascular variability, adjust for clinical confounders, and generalize findings (23,24). In contrast, the HFM Study incorporated a wide range of measurements that assessed various vessel types and sizes, as well as both endothelium-dependent and endothelium-independent vasodilation.(89)

The HFM Study participants (n = 602, 7 centers) underwent standardized preoperative ultrasounds of the upper-extremity arteries and veins, along with five vascular function tests within 45 days before AVF surgical creation: (i) endothelium-dependent vasodilation via brachial artery flow- mediated dilation (FMD), (ii) endothelium-independent vasodilation via brachial artery nitroglycerin- mediated dilation (NMD), which assesses the capacity of arterial SMCs to respond to the NO donor nitroglycerin, (iii) stiffness of the elastic aorta via carotid-femoral pulse wave velocity, (iv) stiffness of the muscular arteries in the upper extremity via carotid-radial pulse wave velocity, and (v) venous capacitance in the upper extremity via venous occlusion plethysmography. Participants then underwent standardized postoperative AVF ultrasounds at 1 day, 2 weeks, and 6 weeks to measure AVF vein flow rate and lumen diameter. The relationship of each of the preoperative vascular functional parameters with subsequent AVF characteristics was examined by using mixed-effects multiple regression analyses.(89) After controlling for AVF location, preoperative ultrasound measurements (preoperative inflow artery diameter, minimum vein diameter, and brachial artery flow), and demographic factors (age, sex, race, and dialysis status), greater brachial artery FMD was correlated with enhanced 6-week AVF blood flow and diameter (for every 10% difference in FMD: change in blood flow rate = 11.6%, 95% CI, 0.6% to 23.9%, P=0.04; change in diameter = 0.31 mm, 95% CI, 0.05 to 0.57 mm, P=0.02). Similarly, higher brachial artery NMD was also associated with increased 6-week AVF blood flow and diameter (for every 10% difference in NMD: change in blood flow rate = 14.0%, 95% CI, 3.7% to 25.3%, P<0.01; change in diameter = 0.45 mm, 95% CI, 0.25 to 0.65 mm, P<0.001). In contrast, no significant or consistent relationships were found between other preoperative vascular functional parameters and postoperative AVF characteristics. Specifically, although carotid-femoral pulse wave velocity exhibited a trend toward an inverse relationship with 6- week AVF diameter, neither carotid-femoral nor carotid-radial pulse wave velocity showed statistically significant relationships with AVF blood flow. Although there was a weak positive relationship between venous capacitance and AVF diameter at 2 weeks, there was no consistent relationship of venous capacitance with AVF blood flow or diameter across the three consecutive postoperative assessments.

It is important to recognize that the inability to detect associations between AVF outcomes and pre-operative vascular functional parameters assessed by pulse wave velocity and venous occlusion plethysmography may reflect the inherent limitations and relative insensitivity of these tests. Pulse wave velocity techniques are indirect measures of arterial stiffness, influenced by the combined stiffness of multiple arteries, which may vary in their individual stiffness profiles. Similarly, venous occlusion plethysmography has limited reliability in assessing the compliance of large veins. Consequently, more advanced and precise technologies are needed to accurately measure the stiffness of the arteries and veins used for AVF creation. Additionally, it is important to note that a small preoperative arterial diameter (e.g., 3 mm) can result in high AVF flow volumes. This is often observed in high-flow AVFs that require banding to reduce excessive flow rate. A flow rate greater than 1500 mL/min can lead to complications such as cardiac overload, post-dialysis bleeding, and elevated venous pressure. While techniques vary, banding is typically performed near the arterial juxta-anastomosis to reduce the arterial diameter to between 2.5 and 6 mm, thereby lowering flow rate to a normal (600–1500 mL/min) or low (<600 mL/min) range, while preserving AVF patency and usability for hemodialysis.(93) Therefore, when evaluating vessels for AVF creation, the smallest preoperative inflow arterial diameter should be taken into consideration.

Nonetheless, the observed association of preoperative brachial artery FMD and NMD with favorable changes in the AVF vein after creation highlights the critical role of vasodilatory response integrity in promoting AVF maturation, at least by the 6-week postoperative mark.(94) Therefore, a better understanding of impaired conduit artery dilation in ESKD patients may be key to uncovering the underlying causes of AVF maturation failure and developing effective treatments to promote AVF maturation.

3.2. Clinical evidence for impaired conduit artery endothelium-dependent dilation in CKD and ESKD

Brachial artery FMD and NMD are commonly used, non-invasive assessments of macrovascular endothelial and smooth muscle cell functions, respectively.(95) These tests are typically expressed as the percentage changes in brachial artery diameter after ischemia (FMD) and after nitroglycerin administration (NMD). Both have been extensively used to investigate vascular function in patients with CKD and ESKD, establishing clear evidence of endothelial dysfunction in CKD and ESKD patients, while vascular smooth muscle dilation in response to NO appears preserved.

Yilmaz et al. (96) documented a progressive decline in endothelial function as CKD progresses from Stage 1 to Stage 5, despite preserved endothelium-independent vasodilatory response as shown by NMD, which suggests that arterial smooth muscle dilation in response to NO remains intact. Katulka et al. (97) also found a moderate association between endothelial function and estimated glomerular filtration rate in patients with Stage 3-5 CKD, though endothelium-independent vasodilation was not assessed in their study.

Conduit artery endothelial dysfunction, as assessed by brachial artery FMD, is also present in patients with ESKD who are on maintenance hemodialysis compared to healthy controls.(98101) Joannides et al. (102) reported similar findings using radial artery FMD testing in ESKD patients compared to healthy controls. Importantly, endothelium-independent vasodilation remains intact in patients with dialysis-dependent ESKD.(98, 102, 103) The HFM Study did not include healthy, non- CKD controls; however, it reported that FMD was higher among patients undergoing maintenance dialysis than in those not yet on dialysis.(104) This finding, where FMD was greater in dialysis patients compared to pre-dialysis patients, is consistent with the dialytic removal of mediators of endothelial dysfunction. Importantly, it suggests that endothelial function may be modifiable in the ESKD population.

3.3. Approach to improve conduit artery dilation in CKD and ESKD

Several pharmacological agents commonly prescribed to manage CKD-related comorbidities have demonstrated direct and/or indirect protective effects on endothelial function.(105) However, their safety in ESKD patients require further investigation in future studies. Several of these studies have explored whether these pharmacological agents can improve conduit artery endothelial function in patients with CKD Stages 1-5.(106109) Approaches for improving AVFs are discussed in Section 4.

3.3.1. Statins.

Statins, commonly prescribed for managing proteinuria-related hyperlipidemia in CKD patients, are known for their lipid-lowering and anti-inflammatory effects.(105, 110) For instance, atorvastatin has been shown to improve lipid profiles and enhance conduit artery endothelial function in CKD patients, partly by increasing eNOS activity and NO production.(111, 112)

3.3.2. Anti-inflammatory and antioxidant therapies.

Several drugs targeting inflammation and ROS have been studied in CKD patients. For example, treatment with rilonacept, an IL-1 blocker, improves conduit artery endothelial function and reduces high-sensitivity C-reactive protein concentrations and endothelial cell expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in patients with Stage 3-4 CKD, suggesting both anti-inflammatory and antioxidant benefits.(113) Allopurinol, a xanthine oxidase inhibitor used to manage hyperuricemia, has yielded mixed results regarding its effects on endothelial function. Some studies report no significant changes,(114, 115) while others have shown improvements in conduit artery endothelial function.(116, 117) These discrepancies for allopurinol may be due to variations in treatment duration, disease severity, and patient populations, and have been observed for vitamin C. For example, Ghiadoni et al. (109) documented that oral vitamin C administration (2 g) to patients with non-dialysis- dependent Stage 3-5 CKD and hemodialysis-dependent ESKD reduced oxidative stress biomarkers and enhanced antioxidant biomarkers in their plasma. However, these favorable changes were accompanied by an improvement in conduit artery endothelial function only in patients with hemodialysis-dependent ESKD, without affecting endothelium-independent vasodilation. In contrast, Cross et al. (118) found no improvement in conduit artery endothelial function, assessed in both the brachial and radial arteries, after parenteral administration of vitamin C in patients with non-dialysis- dependent Stage 4-5 CKD or hemodialysis-dependent ESKD, despite increases in serum antioxidant activity. The lack of improvement may, in part, be explained by differences in the method and dosage of vitamin C administration. In their study, vitamin C was administered intra-arterially (25 mg/min) for patients with non-dialysis-dependent Stage 4-5 CKD and intravenously (3 g) for patients with hemodialysis-dependent ESKD.(118)

3.3.3. Anti-hypertensive medications.

Anti-hypertensive agents are also commonly prescribed to manage hypertension, a prevalent comorbidity in CKD.(105) In CKD patients with type 2 diabetes mellitus (T2DM), angiotensin converting enzyme inhibition with ramipril improved conduit artery endothelial function,(119) suggesting that renin-angiotensin system activation plays a crucial role in endothelial function in CKD patients with T2DM. Similar improvements in endothelial function have also been observed in non-dialysis-dependent CKD patients treated with either ramipril or valsartan (an angiotensin II receptor blocker).(120) Spironolactone, which inhibits aldosterone by binding to mineralocorticoid receptors, has been shown to improve forearm microvascular endothelial function in ESKD patients on hemodialysis,(121) though a role of microvascular endothelial function in AVF remodeling has not yet been investigated.

3.3.4. Sodium-glucose cotransporter-2 (SGLT2) inhibitors.

Given the high prevalence of T2DM in CKD patients,(105) treatment with pharmacologic agents such as SGLT2 inhibitors may offer an attractive therapeutic option. Recent studies have shown that SGLT2 inhibitors like empagliflozin (122) and dapagliflozin (123) improve conduit artery endothelial function in T2DM patients, suggesting that these drugs may have vasoprotective effects that extend to CKD and/or ESKD patients.

4. Investigational treatments to improve AVF remodeling

While many potential treatments for AVF maturation have been explored, none have yet achieved definitive success in clinical trials (Table 1). However, ongoing research continues to offer promising avenues for future breakthroughs. AVF maturation involves complex vascular remodeling, and several investigational treatments are targeting the underlying mechanisms that hinder fistula development, with the goal of enhancing this process.

4.1. Pharmacological agents

4.1.1. Promote vasodilation.

Nitric oxide is essential for vasodilation, and therapies aimed at increasing its bioavailability could help promote AVF maturation. NO-releasing gels applied to the AVF anastomosis (Figure 4) have been shown to promote outward remodeling and inhibit neointimal hyperplasia in a rat AVF model.(38) A trial sponsored by Duke University aimed to enhance NO bioavailability in patients using forearm exercises or nitroglycerin ointment; however, the study was terminated due to recruitment challenges (ClinicalTrials.gov ID: NCT02164318). Sildenafil, a phosphodiesterase type 5A (PDE5A) inhibitor, facilitates smooth muscle relaxation and vessel dilation through the cyclic guanosine monophosphate pathway. In a rat AVF model, sildenafil was added to drinking water, improving hemodynamics and promoting AVF vein remodeling.(124) A small clinical trial in 2019 suggested that sildenafil treatment improved FMD in AVF patients, although the trial was limited by a small sample size and did not progress to phase trial status (ClinicalTrials.gov ID: NCT02414204).

Figure 4: Local drug delivery mechanisms.

Figure 4:

Localized wrap (left panel) and gel (right panel) are placed around or at the perivascular anastomosis of the arteriovenous fistula during creation surgery. Created in BioRender. Northrup, H. (2025) https://BioRender.com/p91d282

4.1.2. Anti-thrombosis.

Clopidogrel, a thienopyridine derivative, binds specifically and irreversibly to the platelet P2RY12 purinergic receptor, inhibiting adenosine diphosphate-mediated platelet activation and aggregation. Funded by the National Institute of Diabetes and Digestive and Kidney Diseases, the Dialysis Access Consortium clopidogrel study was a multicenter, randomized, double-blind, placebo-controlled trial conducted from 2003 to 2007 to examine the effects of clopidogrel on AVF thrombosis and suitability for dialysis.(125) Clopidogrel reduces the frequency of early thrombosis in newly created AVFs but does not increase the proportion of fistulas that mature and become suitable for dialysis.(73) Vorapaxar, a platelet thrombin receptor inhibitor, was studied for its potential therapeutic effects in a clinical trial conducted from 2015 to 2017. While only a few AVFs matured successfully, the results suggested that vorapaxar’s efficacy in promoting AVF maturation may be limited (ClinicalTrials.gov ID: NCT02475837).(126) Omega-3 fatty acids such as in fish oils are known to reduce thrombosis through the inhibition of platelet aggregation. Additionally, it has been shown to decrease blood viscosity, promoted vasodilation, inhibit smooth muscle cell proliferation, improve red blood cell flexibility, and has anti-inflammatory effects. The Omega-3 Fatty Acids and Aspirin in Vascular Access Outcomes in Renal Disease, or FAVOURED trial, investigated not only fish oil but aspirin as well on their effects on AVF failure. Like many of the previously mentioned studies, they found that neither fish oil nor aspirin reduced AVF maturation failure within 12 months after AVF creation (127)(anzctr.org au identifier: CTRN12607000569404).

4.1.3. Anti-proliferation, anti-inflammation, and anti-oxidative stress.

Neointimal hyperplasia in AVF causes stenosis and results from excessive cell proliferation, which may be driven by inflammation and/or oxidative stress. Matsubara et al. demonstrated the role of inflammation in AVF by inhibiting T-cells by Cyclosporine in mice, finding that T-cells regulate the accumulation of macrophages in the AVF venous wall.(128) Paclitaxel, an anti-proliferative drug used in drug-coated balloons, has been successful in preventing restenosis but has not yet proven effective in improving AVF maturation.(129132) Similarly, known for use in drug-coated balloon angioplasty, sirolimus has not shown significant success for AVF maturation. However, a sirolimus-eluting collagen implant is currently undergoing clinical testing (ClinicalTrials.gov ID: NCT05425056).(133) This implant is placed around the anastomosis of the AVF during its creation surgery (Figure 4). Prednisolone, a corticosteroid that reduces inflammation, has been tested but has not demonstrated lasting success.(134) Despite the lack of successful pharmacological treatments targeting proliferation and inflammation, several novel therapies continue to show potential for improving AVF maturation. For example, administration of tempol, a superoxide anion scavenger, decreased neointima formation in the juxta-anastomotic venous segment and improved AVF blood flow in rat AVFs.(135) Further, the silencing of molecules such as VEGF-A, ADAM metallopeptidase with thrombospondin type 1 motif 1, and microRNA-21—which promote cell proliferation—using lentiviral vectors, has been shown to reduce venous stenosis in mouse AVFs.(136138) A Phase 1 clinical trial using the VEGF-A monoclonal antibody bevacizumab was initiated in 2019 but was withdrawn due to enrollment issues (ClinicalTrials.gov ID: NCT02695641).

4.1.4. ECM modification.

AVF stenosis can result from the loss of compliance in fibrotic areas of the fistula, which converts neointimal hyperplasia into an occlusive feature.(139) Vonapanitase, which degrades elastin, is applied to the external surface of the blood vessel during AVF surgery to break down elastin fibers within the vessel wall. This degradation process is thought to promote outward remodeling, but testing of this therapeutic approach in Phase II and III clinical trials has not shown any benefit.(140) Delivery of beta-aminopropionitrile, a fibrosis inhibitor that blocks lysyl oxidase, by vascular wraps (Figure 4) increased AVF blood flow in a rat AVF model.(141) However, local application of a natural vascular scaffolding compound (4-amino-1,8-naphthalamide), which interlinks collagen and elastin via photoactivation to preserve vascular integrity, at the anastomosis during creation surgery increased AVF lumen diameter in a rat AVF model.(142) These findings from animal models suggest that ECM dynamics and/or turnover play an important role in the AVF maturation process.

4.1.5. Statin.

A previous retrospective study suggested that atorvastatin improved AVF primary patency compared to other statins or no statin use.(143) Additionally, use of atorvastatin was associated with favorable outward remodeling, preserved blood flow, and longer duration of primary AVF patency in a mouse model.(144) In a clinical trial aimed at assessing its effect on AVF maturation, patients were to receive atorvastatin daily for 2 weeks before AVF creation and for 6 weeks following creation, with the primary endpoints being AVF diameter and blood flow 6 weeks after creation. However, the trial was withdrawn due to recruitment issues (ClinicalTrials.gov ID: NCT03188978). In a clinical trial in Taiwan, 60 patients with T2DM undergoing AVF creation surgery were given either rosuvastatin or a placebo daily, starting 1 week before surgery and continuing for 3 weeks after. However, rosuvastatin therapy did not show any significant benefits compared to the placebo group (ClinicalTrials.gov ID: NCT01863914).(145) A retrospective, longitudinal cohort study found that statin did not have a beneficial effect on AVF patency.(74)

4.1.6. Senolytics.

Senescence has been reported in mouse and rat AVFs.(146, 147) Senescence markers (p16, p21, SA-β-gal) and senescence-associated secretory phenotype features occurred in both arterial and venous limbs shortly after anastomosis; over time, senescence diminished as the fistula remodeled. The senolytic fisetin increased AVF blood flow and lumen area over 3 weeks, suggesting therapeutic potential.(146)

4.2. Non-pharmacological therapies

4.2.1. Low-intensity pulsed ultrasound (LIPUS).

In this trial conducted in China, patients wear a portable LIPUS device at the anastomotic site of the AVF for 20 minutes, three times a week, for 12 weeks. The study started in February 2024 and is currently recruiting patients (ClinicalTrials.gov ID: NCT06249373). To the best of our knowledge, there are currently no published papers on LIPUS therapy for AVF. However, there have been studies showing the benefits of LIPUS on myocardial ischemia in a porcine model (148) and peripheral arterial disease in patients with Buerger disease.(149) The rationale for using LIPUS in AVF may stem from studies showing that ultrasound can induce NO release via increasing shear stress.(149) However, LIPUS has also been shown to increase endothelial cell extracellular matrix section and angiogenesis,(149) which are sometimes viewed as a hinderance to AVF maturation.(150)

4.2.2. Far infrared therapy (FIT).

In a recent clinical trial, patients received 40 minutes of infrared radiation at the site of their AVF during dialysis for one year. This trial has just been completed, and the results have not yet been released. Lindhard et al. recently reported that after one treatment with FIT during dialysis, there was a significant reduction in soluble VCAM-1 and soluble ICAM-1 levels. However, the authors concluded that their findings did not support the hypothesis that FIT during dialysis provides a vasoprotective effect after a single treatment (ClinicalTrials.gov ID: NCT04011072).(151)

4.2.3. Exercise interventions.

There has been a significant amount of research on exercise as a therapeutic approach. The most common exercise studied is handgrip exercise. A quick search on ClinicalTrials.gov reveals at least seven different handgrip trials for AVF maturation therapy, starting as early as 2008 (ClinicalTrials.gov ID: NCT05493046, NCT04034433, NCT03886116, NCT03741985, NCT03137680, NCT02205944, NCT01061008). These trials have taken place, or are ongoing, in the United Kingdom, Canada, Singapore, China, France, and Taiwan. Most of these studies involved using a ball that patients would grip for a specified duration, generally 20-60 minutes per day, over a set number of weeks, either preoperatively, postoperatively, or both. Some studies, however, used different types of exercises and compared them to ball exercises as a control. In 2022, Nantakool et al. published a review article on upper limb exercise as a therapy for AVF maturation, which included data from nine studies. They found that there was insufficient data to analyze the effects of preoperative exercise on AVF maturation. Regarding postoperative exercise, they concluded that study parameters made the effect of exercise on AVF maturation unclear.(152) The 2019 Kidney Disease Outcomes Quality Initiative (KDOQI) guidelines state: “There is inadequate evidence for KDOQI to make a recommendation on the use of upper extremity exercise to facilitate postoperative AVF maturation”.(153)

4.2.4. Mechanical support devices.

Alternative therapies under investigation include those offering mechanical support for the AVF. A pivotal trial sponsored by Venostent in the U.S. is currently ongoing to assess the safety and efficacy of a bioabsorbable perivascular wrap called SelfWrap. The wrap is applied around the perivascular area of the AVF (Figure 4) during surgery to provide mechanical support. This study is currently enrolling patients (ClinicalTrials.gov ID: NCT06001827). VasQ is a device that also provides mechanical support. It is a nitinol-based device implanted around the artery and vein during the surgical creation of an AVF. Numerous studies on VasQ have led to its FDA clearance for use in AVF. The data show that fistulas with VasQ led to a high percentage of being used for dialysis.(154157)

4.2.5. Biological therapies.

A Phase 1 clinical study is currently recruiting patients to evaluate the safety of allogeneic adipose-derived mesenchymal stem cell therapy. Mesenchymal stem cells, derived from the patient’s adipose tissue, are isolated and cultured. During AVF creation surgery, the stem cells are administered in Lactated Ringer’s solution to the perivasculature of the distal artery near the anastomosis (ClinicalTrials.gov ID: NCT04392206).(158) The rationale for this study is that MSCs can limit the inflammatory process and consequently reduce neointimal hyperplasia formation. A preclinical study in mice showed that administration of human adipose-derived MSCs improved AVF patency by reducing neointimal hyperplasia development as hypothesized.(158)

5. Conclusions

The AVF environment is unique and complicated. The vascular system and its key cell players in both arteries and veins (such as ECs, vascular SMCs, fibroblasts, platelets, and immune/inflammatory cells) are significantly compromised in CKD, creating substantial challenges for successful AVF maturation. Vascular dysfunction, in particular endothelial dysfunction, has been shown to play a central role in the maturation process. While no therapies have yet been proven to definitively and effectively prevent AVF maturation failure or promote successful maturation, several promising approaches are currently under investigation. Recent clinical trials have shifted focus from earlier strategies that targeted inhibiting thrombosis and neointimal hyperplasia, to now prioritizing the improvement of vascular function and ECM. Ongoing research continues to provide valuable insights, offering hope that these evolving strategies may ultimately lead to successful therapies for AVF maturation, a significant unmet clinical need.

7.Funding

This work was funded, in part, by the National Institutes of Health (R01DK123727 to Y.H.; R01DK129299 to Y.T.S.) and the U.S. Department of Veterans Affairs (IK2RX003670 to K.B.; I01BX006269 and I01BX006078 to Y.T.S.).

Footnotes

6. Disclosure

No conflicts of interest, financial or otherwise, are declared by the authors.

11. References

  • 1.Lv JC, and Zhang LX. Prevalence and Disease Burden of Chronic Kidney Disease. Adv Exp Med Biol 1165: 3–15, 2019. [DOI] [PubMed] [Google Scholar]
  • 2.Bello AK, Okpechi IG, Osman MA, Cho Y, Htay H, Jha V, Wainstein M, and Johnson DW. Epidemiology of haemodialysis outcomes. Nat Rev Nephrol 18: 378–395, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Pecoits-Filho R, Okpechi IG, Donner JA, Harris DCH, Aljubori HM, Bello AK, Bellorin-Font E, Caskey FJ, Collins A, Cueto-Manzano AM, Feehally J, Goh BL, Jager KJ, Nangaku M, Rahman M, Sahay M, Saleh A, Sola L, Turan Kazancioglu R, Walker RC, Walker R, Yao Q, Yu X, Zhao MH, and Johnson DW. Capturing and monitoring global differences in untreated and treated end-stage kidney disease, kidney replacement therapy modality, and outcomes. Kidney Int Suppl (2011) 10: e3–e9, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Allon M, and Robbin ML. Increasing arteriovenous fistulas in hemodialysis patients: problems and solutions. Kidney Int 62: 1109–1124, 2002. [DOI] [PubMed] [Google Scholar]
  • 5.Lee T Fistula First Initiative: Historical Impact on Vascular Access Practice Patterns and Influence on Future Vascular Access Care. Cardiovasc Eng Technol 8: 244–254, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Rayner HC, Besarab A, Brown WW, Disney A, Saito A, and Pisoni RL. Vascular access results from the Dialysis Outcomes and Practice Patterns Study (DOPPS): performance against Kidney Disease Outcomes Quality Initiative (K/DOQI) Clinical Practice Guidelines. Am J Kidney Dis 44: 22–26, 2004. [DOI] [PubMed] [Google Scholar]
  • 7.Pisoni RL, Zepel L, Port FK, and Robinson BM. Trends in US Vascular Access Use, Patient Preferences, and Related Practices: An Update From the US DOPPS Practice Monitor With International Comparisons. Am J Kidney Dis 65: 905–915, 2015. [DOI] [PubMed] [Google Scholar]
  • 8.Brescia MJ, Cimino JE, Appel K, and Hurwich BJ. Chronic hemodialysis using venipuncture and a surgically created arteriovenous fistula. N Engl J Med 275: 1089–1092, 1966. [DOI] [PubMed] [Google Scholar]
  • 9.Pisoni RL, Zepel L, Fluck R, Lok CE, Kawanishi H, Süleymanlar G, Wasse H, Tentori F, Zee J, Li Y, Schaubel D, Burke S, and Robinson B. International Differences in the Location and Use of Arteriovenous Accesses Created for Hemodialysis: Results From the Dialysis Outcomes and Practice Patterns Study (DOPPS). Am J Kidney Dis 71: 469–478, 2018. [DOI] [PubMed] [Google Scholar]
  • 10.MacRae JM, Oliver M, Clark E, Dipchand C, Hiremath S, Kappel J, Kiaii M, Lok C, Luscombe R, Miller LM, and Moist L. Arteriovenous Vascular Access Selection and Evaluation. Can J Kidney Health Dis 3: 2054358116669125, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Robbin ML, Greene T, Cheung AK, Allon M, Berceli SA, Kaufman JS, Allen M, Imrey PB, Radeva MK, Shiu YT, Umphrey HR, and Young CJ. Arteriovenous Fistula Development in the First 6 Weeks after Creation. Radiology 279: 620–629, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Robbin ML, Greene T, Allon M, Dember LM, Imrey PB, Cheung AK, Himmelfarb J, Huber TS, Kaufman JS, Radeva MK, Roy-Chaudhury P, Shiu YT, Vazquez MA, Umphrey HR, Alexander L, Abts C, Beck GJ, Kusek JW, and Feldman HI. Prediction of Arteriovenous Fistula Clinical Maturation from Postoperative Ultrasound Measurements: Findings from the Hemodialysis Fistula Maturation Study. J Am Soc Nephrol 29: 2735–2744, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lee T, Qian J, Thamer M, and Allon M. Tradeoffs in Vascular Access Selection in Elderly Patients Initiating Hemodialysis With a Catheter. Am J Kidney Dis 72: 509–518, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Beathard GA, Settle SM, and Shields MW. Salvage of the nonfunctioning arteriovenous fistula. Am J Kidney Dis 33: 910–916, 1999. [DOI] [PubMed] [Google Scholar]
  • 15.Asif A, Lenz O, Merrill D, Cherla G, Cipleu CD, Ellis R, Francois B, Epstein DL, and Pennell P. Percutaneous management of perianastomotic stenosis in arteriovenous fistulae: results of a prospective study. Kidney Int 69: 1904–1909, 2006. [DOI] [PubMed] [Google Scholar]
  • 16.Vazquez-Padron RI, Duque JC, Tabbara M, Salman LH, and Martinez L. Intimal Hyperplasia and Arteriovenous Fistula Failure: Looking Beyond Size Differences. Kidney360 2: 1360–1372, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Rothuizen TC, Wong C, Quax PH, van Zonneveld AJ, Rabelink TJ, and Rotmans JI. Arteriovenous access failure: more than just intimal hyperplasia? Nephrol Dial Transplant 28: 1085–1092, 2013. [DOI] [PubMed] [Google Scholar]
  • 18.Shiu YT, Rotmans JI, Geelhoed WJ, Pike DB, and Lee T. Arteriovenous conduits for hemodialysis: how to better modulate the pathophysiological vascular response to optimize vascular access durability. Am J Physiol Renal Physiol 316: F794–f806, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Thamer M, Lee TC, Wasse H, Glickman MH, Qian J, Gottlieb D, Toner S, and Pflederer TA. Medicare Costs Associated With Arteriovenous Fistulas Among US Hemodialysis Patients. Am J Kidney Dis 72: 10–18, 2018. [DOI] [PubMed] [Google Scholar]
  • 20.Jha V, Al-Ghamdi SMG, Li G, Wu MS, Stafylas P, Retat L, Card-Gowers J, Barone S, Cabrera C, and Garcia Sanchez JJ. Global Economic Burden Associated with Chronic Kidney Disease: A Pragmatic Review of Medical Costs for the Inside CKD Research Programme. Adv Ther 40: 4405–4420, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nichols GA, Bell TJ, Pedula KL, and O’Keeffe-Rosetti M. Medical care costs among patients with established cardiovascular disease. Am J Manag Care 16: e86–e93, 2010. [PubMed] [Google Scholar]
  • 22.He Y, Shiu YT, Imrey PB, Radeva MK, Beck GJ, Gassman JJ, Northrup HM, Roy-Chaudhury P, Berceli SA, and Cheung AK. Association of Shear Stress with Subsequent Lumen Remodeling in Hemodialysis Arteriovenous Fistulas. Clin J Am Soc Nephrol 18: 72–83, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.He Y, Wei G, Greene T, Imrey PB, Northrup H, Radeva MK, Beck GJ, Gassman JJ, Kraiss LW, Robbin M, Roy-Chaudhury P, Cheung AK, Berceli SA, and Shiu YT. Hemodynamics are associated with subsequent lumen remodeling and clinical maturation of hemodialysis arteriovenous fistula. Sci Rep 15: 6131, 2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Fitts MK, Pike DB, Anderson K, and Shiu YT. Hemodynamic Shear Stress and Endothelial Dysfunction in Hemodialysis Access. Open Urol Nephrol J 7: 33–44, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Remuzzi A, Bozzetto M, and Brambilla P. Is shear stress the key factor for AVF maturation? J Vasc Access 18: 10–14, 2017. [DOI] [PubMed] [Google Scholar]
  • 26.White NA, Xiao Z, EP DEW, Li M, MR DEV, KE VDB, and Rotmans JI. Hemodynamic considerations in arteriovenous vascular access modalities for hemodialysis. J Cardiovasc Surg (Torino) 2024. [DOI] [PubMed] [Google Scholar]
  • 27.Sturtzel C Endothelial Cells. Adv Exp Med Biol 1003: 71–91, 2017. [DOI] [PubMed] [Google Scholar]
  • 28.Sandoo A, van Zanten JJ, Metsios GS, Carroll D, and Kitas GD. The endothelium and its role in regulating vascular tone. Open Cardiovasc Med J 4: 302–312, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sriram K, Laughlin JG, Rangamani P, and Tartakovsky DM. Shear-Induced Nitric Oxide Production by Endothelial Cells. Biophys J 111: 208–221, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Roumeliotis S, Mallamaci F, and Zoccali C. Endothelial Dysfunction in Chronic Kidney Disease, from Biology to Clinical Outcomes: A 2020 Update. J Clin Med 9: 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Vila Cuenca M, Hordijk PL, and Vervloet MG. Most exposed: the endothelium in chronic kidney disease. Nephrol Dial Transplant 35: 1478–1487, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Harlacher E, Wollenhaupt J, Baaten C, and Noels H. Impact of Uremic Toxins on Endothelial Dysfunction in Chronic Kidney Disease: A Systematic Review. Int J Mol Sci 23: 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ozkok A, and Yildiz A. Endothelial Progenitor Cells and Kidney Diseases. Kidney Blood Press Res 43: 701–718, 2018. [DOI] [PubMed] [Google Scholar]
  • 34.Tran N, Garcia T, Aniqa M, Ali S, Ally A, and Nauli SM. Endothelial Nitric Oxide Synthase (eNOS) and the Cardiovascular System: in Physiology and in Disease States. Am J Biomed Sci Res 15: 153–177, 2022. [PMC free article] [PubMed] [Google Scholar]
  • 35.Cunha RSD, Santos AF, Barreto FC, and Stinghen AEM. How do Uremic Toxins Affect the Endothelium? Toxins (Basel) 12: 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Dube P, DeRiso A, Patel M, Battepati D, Khatib-Shahidi B, Sharma H, Gupta R, Malhotra D, Dworkin L, Haller S, and Kennedy D. Vascular Calcification in Chronic Kidney Disease: Diversity in the Vessel Wall. Biomedicines 9: 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Oe Y, and Takahashi N. Tissue Factor, Thrombosis, and Chronic Kidney Disease. Biomedicines 10: 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Somarathna M, Hwang PT, Millican RC, Alexander GC, Isayeva-Waldrop T, Sherwood JA, Brott BC, Falzon I, Northrup H, Shiu YT, Stubben CJ, Totenhagen J, Jun HW, and Lee T. Nitric oxide releasing nanomatrix gel treatment inhibits venous intimal hyperplasia and improves vascular remodeling in a rodent arteriovenous fistula. Biomaterials 280: 121254, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pike D, Shiu YT, Cho YF, Le H, Somarathna M, Isayeva T, Guo L, Symons JD, Kevil CG, Totenhagen J, and Lee T. The effect of endothelial nitric oxide synthase on the hemodynamics and wall mechanics in murine arteriovenous fistulas. Sci Rep 9: 4299, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Falzon I, Northrup H, Guo L, Totenhagen J, Lee T, and Shiu YT. The geometry of arteriovenous fistulas using endothelial nitric oxide synthase mouse models. Kidney360 1: 925–935, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Baltazar S, Northrup H, Chang J, Somarathna M, Isayeva Waldrop T, Lee T, and Shiu YT. Effects of endothelial nitric oxide synthase on mouse arteriovenous fistula hemodynamics. Sci Rep 13: 22786, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Feng S, Peden EK, Guo Q, Lee TH, Li Q, Yuan Y, Chen C, Huang F, and Cheng J. Downregulation of the endothelial histone demethylase JMJD3 is associated with neointimal hyperplasia of arteriovenous fistulas in kidney failure. J Biol Chem 298: 101816, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Martinez L, Rojas MG, Tabbara M, Pereira-Simon S, Santos Falcon N, Rauf MA, Challa A, Zigmond ZM, Griswold AJ, Duque JC, Lassance-Soares RM, Velazquez OC, Salman LH, and Vazquez- Padron RI. The Transcriptomics of the Human Vein Transformation After Arteriovenous Fistula Anastomosis Uncovers Layer-Specific Remodeling and Hallmarks of Maturation Failure. Kidney Int Rep 8: 837–850, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wang G, Jacquet L, Karamariti E, and Xu Q. Origin and differentiation of vascular smooth muscle cells. J Physiol 593: 3013–3030, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yap C, Mieremet A, de Vries CJM, Micha D, and de Waard V. Six Shades of Vascular Smooth Muscle Cells Illuminated by KLF4 (Krüppel-Like Factor 4). Arterioscler Thromb Vasc Biol 41: 2693–2707, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Cao G, Xuan X, Hu J, Zhang R, Jin H, and Dong H. How vascular smooth muscle cell phenotype switching contributes to vascular disease. Cell Communication and Signaling 20: 180, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Yang S, Zeng Z, Yuan Q, Chen Q, Wang Z, Xie H, and Liu J. Vascular calcification: from the perspective of crosstalk. Mol Biomed 4: 35, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zhang YX, Tang RN, Wang LT, and Liu BC. Role of crosstalk between endothelial cells and smooth muscle cells in vascular calcification in chronic kidney disease. Cell Prolif 54: e12980, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gomez D, and Owens GK. Smooth muscle cell phenotypic switching in atherosclerosis. Cardiovasc Res 95: 156–164, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Monroy MA, Fang J, Li S, Ferrer L, Birkenbach MP, Lee IJ, Wang H, Yang XF, and Choi ET. Chronic kidney disease alters vascular smooth muscle cell phenotype. Front Biosci (Landmark Ed) 20: 784–795, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Hénaut L, Mary A, Chillon JM, Kamel S, and Massy ZA. The Impact of Uremic Toxins on Vascular Smooth Muscle Cell Function. Toxins (Basel) 10: 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Cazaña-Pérez V, Cidad P, Donate-Correa J, Martín-Núñez E, López-López JR, Pérez-García MT, Giraldez T, Navarro-González JF, and Alvarez de la Rosa D. Phenotypic Modulation of Cultured Primary Human Aortic Vascular Smooth Muscle Cells by Uremic Serum. Front Physiol 9: 89, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Li Z, Zhao Y, Pan Z, Cai B, Zhang C, and Jiao J. LncRNA-LncDACH1 mediated phenotypic switching of smooth muscle cells during neointimal hyperplasia in male arteriovenous fistulas. Nat Commun 15: 3743, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Liu Y, Chen B, Chen K, Wang Y, Zhou C, Liang X, Wang K, and Wang P. Mineralocorticoid receptor antagonism attenuates arteriovenous fistula stenosis by modulating the phenotype of vascular smooth muscle cells. Nephrol Dial Transplant 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zhao J, Jourd’heuil FL, Xue M, Conti D, Lopez-Soler RI, Ginnan R, Asif A, Singer HA, Jourd’heuil D, and Long X. Dual Function for Mature Vascular Smooth Muscle Cells During Arteriovenous Fistula Remodeling. J Am Heart Assoc 6: 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Vazquez-Padron RI, Martinez L, Duque JC, Salman LH, and Tabbara M. The anatomical sources of neointimal cells in the arteriovenous fistula. J Vasc Access 24: 99–106, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Plikus MV, Wang X, Sinha S, Forte E, Thompson SM, Herzog EL, Driskell RR, Rosenthal N, Biernaskie J, and Horsley V. Fibroblasts: Origins, definitions, and functions in health and disease. Cell 184: 3852–3872, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Tinajero MG, and Gotlieb AI. Recent Developments in Vascular Adventitial Pathobiology: The Dynamic Adventitia as a Complex Regulator of Vascular Disease. Am J Pathol 190: 520–534, 2020. [DOI] [PubMed] [Google Scholar]
  • 59.Kuwabara JT, and Tallquist MD. Tracking Adventitial Fibroblast Contribution to Disease: A Review of Current Methods to Identify Resident Fibroblasts. Arterioscler Thromb Vasc Biol 37: 1598–1607, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jimbo R, Kawakami-Mori F, Mu S, Hirohama D, Majtan B, Shimizu Y, Yatomi Y, Fukumoto S, Fujita T, and Shimosawa T. Fibroblast growth factor 23 accelerates phosphate-induced vascular calcification in the absence of Klotho deficiency. Kidney Int 85: 1103–1111, 2014. [DOI] [PubMed] [Google Scholar]
  • 61.Laboyrie SL, de Vries MR, Bijkerk R, and Rotmans JI. Building a Scaffold for Arteriovenous Fistula Maturation: Unravelling the Role of the Extracellular Matrix. Int J Mol Sci 24: 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Tucker W, Tucker B, Rye KA, and Ong KL. Fibroblast growth factor 21 in heart failure. Heart Fail Rev 28: 261–272, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hu X, Ding H, Wei Q, Chen R, Zhao W, Jiang L, Wang J, Liu H, Cao J, Liu H, and Wang B. Fibroblast growth factor 21 predicts arteriovenous fistula functional patency loss and mortality in patients undergoing maintenance hemodialysis. Ren Fail 46: 2302407, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Liu CT, Hsu SC, Hsieh HL, Chen CH, Chen CY, Sue YM, Lin FY, Shih CM, Shiu YT, and Huang PH. Parathyroid Hormone Induces Transition of Myofibroblasts in Arteriovenous Fistula and Increases Maturation Failure. Endocrinology 162: 2021. [DOI] [PubMed] [Google Scholar]
  • 65.López I, Rodríguez-Ortiz ME, Almadén Y, Guerrero F, de Oca AM, Pineda C, Shalhoub V, Rodríguez M, and Aguilera-Tejero E. Direct and indirect effects of parathyroid hormone on circulating levels of fibroblast growth factor 23 in vivo. Kidney Int 80: 475–482, 2011. [DOI] [PubMed] [Google Scholar]
  • 66.Gremmel T, Frelinger AL 3rd, and Michelson AD. Platelet Physiology. Semin Thromb Hemost 50: 1173–1186, 2024. [DOI] [PubMed] [Google Scholar]
  • 67.Baaten C, Schröer JR, Floege J, Marx N, Jankowski J, Berger M, and Noels H. Platelet Abnormalities in CKD and Their Implications for Antiplatelet Therapy. Clin J Am Soc Nephrol 17: 155–170, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Baaten C, Sternkopf M, Henning T, Marx N, Jankowski J, and Noels H. Platelet Function in CKD: A Systematic Review and Meta-Analysis. J Am Soc Nephrol 32: 1583–1598, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Jain N, Corken AL, Kumar A, Davis CL, Ware J, and Arthur JM. Role of Platelets in Chronic Kidney Disease. J Am Soc Nephrol 32: 1551–1558, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gomchok D, Ge RL, and Wuren T. Platelets in Renal Disease. Int J Mol Sci 24: 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Baaten C, Vondenhoff S, and Noels H. Endothelial Cell Dysfunction and Increased Cardiovascular Risk in Patients With Chronic Kidney Disease. Circ Res 132: 970–992, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Xiao Y, Vazquez-Padron RI, Martinez L, Singer HA, Woltmann D, and Salman LH. Role of platelet factor 4 in arteriovenous fistula maturation failure: What do we know so far? J Vasc Access 25: 390–406, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Dember LM, Beck GJ, Allon M, Delmez JA, Dixon BS, Greenberg A, Himmelfarb J, Vazquez MA, Gassman JJ, Greene T, Radeva MK, Braden GL, Ikizler TA, Rocco MV, Davidson IJ, Kaufman JS, Meyers CM, Kusek JW, and Feldman HI. Effect of clopidogrel on early failure of arteriovenous fistulas for hemodialysis: a randomized controlled trial. Jama 299: 2164–2171, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chang TI, Chen CH, Hsieh HL, Chen CY, Hsu SC, Cheng HS, Huang WC, Sue YM, Hsu YH, Lin FY, Shih CM, Lin SJ, Huang PH, and Liu CT. Effects of cardiovascular medications on primary patency of hemodialysis arteriovenous fistula. Sci Rep 10: 12135, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Viecelli AK, Polkinghorne KR, Pascoe EM, Paul-Brent PA, Hawley CM, Badve SV, Cass A, Hooi LS, Kerr PG, Mori TA, Ong LM, Voss D, Johnson DW, and Irish AB. Fish oil and aspirin effects on arteriovenous fistula function: Secondary outcomes of the randomised omega-3 fatty acids (Fish oils) and Aspirin in Vascular access OUtcomes in REnal Disease (FAVOURED) trial. PLoS One 14: e0213274, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Okyay GU, Inal S, Oneç K, Er RE, Paşaoğlu O, Paşaoğlu H, Derici U, and Erten Y. Neutrophil to lymphocyte ratio in evaluation of inflammation in patients with chronic kidney disease. Ren Fail 35: 29– 36, 2013. [DOI] [PubMed] [Google Scholar]
  • 77.Xiang FF, Zhu JM, Cao XS, Shen B, Zou JZ, Liu ZH, Zhang H, Teng J, Liu H, and Ding XQ. Lymphocyte depletion and subset alteration correlate to renal function in chronic kidney disease patients. Ren Fail 38: 7–14, 2016. [DOI] [PubMed] [Google Scholar]
  • 78.Zhao WM, Tao SM, and Liu GL. Neutrophil-to-lymphocyte ratio in relation to the risk of all-cause mortality and cardiovascular events in patients with chronic kidney disease: a systematic review and meta-analysis. Ren Fail 42: 1059–1066, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Girndt M, Trojanowicz B, and Ulrich C. Monocytes in Uremia. Toxins (Basel) 12: 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Jansen F, Yang X, Baumann K, Przybilla D, Schmitz T, Flender A, Paul K, Alhusseiny A, Nickenig G, and Werner N. Endothelial microparticles reduce ICAM-1 expression in a microRNA-222-dependent mechanism. J Cell Mol Med 19: 2202–2214, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Trojanowicz B, Ulrich C, Seibert E, Fiedler R, and Girndt M. Uremic conditions drive human monocytes to pro-atherogenic differentiation via an angiotensin-dependent mechanism. PLoS One 9: e102137, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Nikolic-Paterson DJ, Wang S, and Lan HY. Macrophages promote renal fibrosis through direct and indirect mechanisms. Kidney Int Suppl (2011) 4: 34–38, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Liang S, Wang YL, Ji Y, Na SP, Jia XB, Zhao SL, Lyu HY, Yuan XY, and Bao YS. Circulating monocyte-platelet aggregates with different monocyte subsets and their association with disease severity in chronic kidney disease. Am J Med Sci 365: 443–449, 2023. [DOI] [PubMed] [Google Scholar]
  • 84.Zhang Y, Kong X, Liang L, and Xu D. Regulation of vascular remodeling by immune microenvironment after the establishment of autologous arteriovenous fistula in ESRD patients. Front Immunol 15: 1365422, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Samra G, Rai V, and Agrawal DK. Innate and adaptive immune cells associate with arteriovenous fistula maturation and failure. Can J Physiol Pharmacol 100: 716–727, 2022. [DOI] [PubMed] [Google Scholar]
  • 86.Dixon BS. Why don’t fistulas mature? Kidney Int 70: 1413–1422, 2006. [DOI] [PubMed] [Google Scholar]
  • 87.Endemann DH, and Schiffrin EL. Endothelial dysfunction. J Am Soc Nephrol 15: 1983–1992, 2004. [DOI] [PubMed] [Google Scholar]
  • 88.Dember LM, Imrey PB, Beck GJ, Cheung AK, Himmelfarb J, Huber TS, Kusek JW, Roy- Chaudhury P, Vazquez MA, Alpers CE, Robbin ML, Vita JA, Greene T, Gassman JJ, and Feldman HI. Objectives and design of the hemodialysis fistula maturation study. Am J Kidney Dis 63: 104–112, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Allon M, Greene T, Dember LM, Vita JA, Cheung AK, Hamburg NM, Imrey PB, Kaufman JS, Robbin ML, Shiu YT, Terry CM, Umphrey HR, and Feldman HI. Association between Preoperative Vascular Function and Postoperative Arteriovenous Fistula Development. J Am Soc Nephrol 27: 3788– 3795, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kheda MF, Brenner LE, Patel MJ, Wynn JJ, White JJ, Prisant LM, Jones SA, and Paulson WD. Influence of arterial elasticity and vessel dilatation on arteriovenous fistula maturation: a prospective cohort study. Nephrol Dial Transplant 25: 525–531, 2010. [DOI] [PubMed] [Google Scholar]
  • 91.Masengu A, Hanko JB, and Maxwell AP. Arterial stiffness and arteriovenous fistula failure of maturation. J Vasc Access 17: 477–482, 2016. [DOI] [PubMed] [Google Scholar]
  • 92.van der Linden J, Lameris TW, van den Meiracker AH, de Smet AA, Blankestijn PJ, and van den Dorpel MA. Forearm venous distensibility predicts successful arteriovenous fistula. Am J Kidney Dis 47: 1013–1019, 2006. [DOI] [PubMed] [Google Scholar]
  • 93.Miller GA, Goel N, Friedman A, Khariton A, Jotwani MC, Savransky Y, Khariton K, Arnold WP, and Preddie DC. The MILLER banding procedure is an effective method for treating dialysis-associated steal syndrome. Kidney Int 77: 359–366, 2010. [DOI] [PubMed] [Google Scholar]
  • 94.Nath KA, and Katusic ZS. Predicting the Functionality and Form of a Dialysis Fistula. J Am Soc Nephrol 27: 3508–3510, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Maruhashi T, Kajikawa M, Kishimoto S, Hashimoto H, Takaeko Y, Yamaji T, Harada T, Han Y, Aibara Y, Mohamad Yusoff F, Hidaka T, Kihara Y, Chayama K, Nakashima A, Goto C, Tomiyama H, Takase B, Kohro T, Suzuki T, Ishizu T, Ueda S, Yamazaki T, Furumoto T, Kario K, Inoue T, Koba S, Watanabe K, Takemoto Y, Hano T, Sata M, Ishibashi Y, Node K, Maemura K, Ohya Y, Furukawa T, Ito H, Ikeda H, Yamashina A, and Higashi Y. Diagnostic Criteria of Flow-Mediated Vasodilation for Normal Endothelial Function and Nitroglycerin-Induced Vasodilation for Normal Vascular Smooth Muscle Function of the Brachial Artery. J Am Heart Assoc 9: e013915, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Yilmaz MI, Stenvinkel P, Sonmez A, Saglam M, Yaman H, Kilic S, Eyileten T, Caglar K, Oguz Y, Vural A, Cakar M, Altun B, Yenicesu M, and Carrero JJ. Vascular health, systemic inflammation and progressive reduction in kidney function; clinical determinants and impact on cardiovascular outcomes. Nephrol Dial Transplant 26: 3537–3543, 2011. [DOI] [PubMed] [Google Scholar]
  • 97.Katulka EK, Hirt AE, Kirkman DL, Edwards DG, and Witman MAH. Altered vascular function in chronic kidney disease: evidence from passive leg movement. Physiol Rep 7: e14075, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.van Guldener C, Lambert J, Janssen MJ, Donker AJ, and Stehouwer CD. Endothelium-dependent vasodilatation and distensibility of large arteries in chronic haemodialysis patients. Nephrol Dial Transplant 12 Suppl 2: 14–18, 1997. [PubMed] [Google Scholar]
  • 99.Recio-Mayoral A, Banerjee D, Streather C, and Kaski JC. Endothelial dysfunction, inflammation and atherosclerosis in chronic kidney disease--a cross-sectional study of predialysis, dialysis and kidney-transplantation patients. Atherosclerosis 216: 446–451, 2011. [DOI] [PubMed] [Google Scholar]
  • 100.Oflaz H, Pusuroglu H, Genchallac H, Demirel S, Bugra Z, Sever MS, and Yildiz A. Endothelial function is more impaired in hemodialysis patients than renal transplant recipients. Clin Transplant 17: 528–533, 2003. [DOI] [PubMed] [Google Scholar]
  • 101.Kocak H, Ceken K, Dinckan A, Mahsereci E, Yavuz A, Yucetin L, Akbas SH, Gurkan A, Erdogan O, Ersoy F, Yakupoglu G, Demirbas A, and Tuncer M. Assessment and comparison of endothelial function between dialysis and kidney transplant patients. Transplant Proc 38: 416–418, 2006. [DOI] [PubMed] [Google Scholar]
  • 102.Joannides R, Bakkali EH, Le Roy F, Rivault O, Godin M, Moore N, Fillastre JP, and Thuillez C. Altered flow-dependent vasodilatation of conduit arteries in maintenance haemodialysis. Nephrol Dial Transplant 12: 2623–2628, 1997. [DOI] [PubMed] [Google Scholar]
  • 103.van Guldener C, Janssen MJ, Lambert J, Steyn M, Donker AJ, and Stehouwer CD. Endothelium- dependent vasodilatation is impaired in peritoneal dialysis patients. Nephrol Dial Transplant 13: 1782– 1786, 1998. [DOI] [PubMed] [Google Scholar]
  • 104.Dember LM, Imrey PB, Duess MA, Hamburg NM, Larive B, Radeva M, Himmelfarb J, Kraiss LW, Kusek JW, Roy-Chaudhury P, Terry CM, Vazquez MA, Vongpatanasin W, Beck GJ, and Vita JA. Vascular Function at Baseline in the Hemodialysis Fistula Maturation Study. J Am Heart Assoc 5: 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Levin A, Ahmed SB, Carrero JJ, Foster B, Francis A, Hall RK, Herrington WG, Hill G, Inker LA, Kazancioglu R, Lamb E, Lin P, Madero M, McIntyre N, Morrow K, Roberts G, Sabanayagam D, Schaeffner E, Shlipak M, Shroff R, Tangri N, Thanachayanont T, Ulasi I, Wong G, Yang CW, Zhang L, Robinson KA, Wilson L, Wilson RF, Kasiske BL, Cheung M, Earley A, and Stevens PE. Executive summary of the KDIGO 2024 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease: known knowns and known unknowns. Kidney Int 105: 684–701, 2024. [DOI] [PubMed] [Google Scholar]
  • 106.Massy ZA, Borderie D, Nguyen-Khoa T, Drueke TB, Ekindjian OG, and Lacour B. Increased plasma S-nitrosothiol levels in chronic haemodialysis patients. Nephrol Dial Transplant 18: 153–157, 2003. [DOI] [PubMed] [Google Scholar]
  • 107.Taylor JE, Scott N, Bridges A, Henderson IS, Stewart WK, and Belch JJ. Lipid peroxidation and antioxidants in continuous ambulatory dialysis patients. Perit Dial Int 12: 252–256, 1992. [PubMed] [Google Scholar]
  • 108.Witko-Sarsat V, Friedlander M, Capeillere-Blandin C, Nguyen-Khoa T, Nguyen AT, Zingraff J, Jungers P, and Descamps-Latscha B. Advanced oxidation protein products as a novel marker of oxidative stress in uremia. Kidney Int 49: 1304–1313, 1996. [DOI] [PubMed] [Google Scholar]
  • 109.Ghiadoni L, Cupisti A, Huang Y, Mattei P, Cardinal H, Favilla S, Rindi P, Barsotti G, Taddei S, and Salvetti A. Endothelial dysfunction and oxidative stress in chronic renal failure. J Nephrol 17: 512– 519, 2004. [PubMed] [Google Scholar]
  • 110.Jain MK, and Ridker PM. Anti-inflammatory effects of statins: clinical evidence and basic mechanisms. Nat Rev Drug Discov 4: 977–987, 2005. [DOI] [PubMed] [Google Scholar]
  • 111.Takenaka T, Takane H, Kikuta T, Watanabe Y, and Suzuki H. Statin improves flow-mediated vasodilation in chronic kidney diseases. Int J Hypertens 2013: 876865, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chen WH, Chen CH, Hsu MC, Chang RW, Wang CH, and Lee TS. Advances in the molecular mechanisms of statins in regulating endothelial nitric oxide bioavailability: Interlocking biology between eNOS activity and L-arginine metabolism. Biomed Pharmacother 171: 116192, 2024. [DOI] [PubMed] [Google Scholar]
  • 113.Nowak KL, Chonchol M, Ikizler TA, Farmer-Bailey H, Salas N, Chaudhry R, Wang W, Smits G, Tengesdal I, Dinarello CA, and Hung AM. IL-1 Inhibition and Vascular Function in CKD. J Am Soc Nephrol 28: 971–980, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Jalal DI, Decker E, Perrenoud L, Nowak KL, Bispham N, Mehta T, Smits G, You Z, Seals D, Chonchol M, and Johnson RJ. Vascular Function and Uric Acid-Lowering in Stage 3 CKD. J Am Soc Nephrol 28: 943–952, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Rutherford E, Ireland S, Mangion K, Stewart GA, MacGregor MS, Roditi G, Woodward R, Gandy SJ, Houston JG, Jardine AG, Rauchhaus P, Witham MD, Mark PB, and Struthers AD. A Randomized, Controlled Trial of the Effect of Allopurinol on Left Ventricular Mass Index in Hemodialysis Patients. Kidney Int Rep 6: 146–155, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Kao MP, Ang DS, Gandy SJ, Nadir MA, Houston JG, Lang CC, and Struthers AD. Allopurinol benefits left ventricular mass and endothelial dysfunction in chronic kidney disease. J Am Soc Nephrol 22: 1382–1389, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Yelken B, Caliskan Y, Gorgulu N, Altun I, Yilmaz A, Yazici H, Oflaz H, and Yildiz A. Reduction of uric acid levels with allopurinol treatment improves endothelial function in patients with chronic kidney disease. Clin Nephrol 77: 275–282, 2012. [DOI] [PubMed] [Google Scholar]
  • 118.Cross JM, Donald AE, Nuttall SL, Deanfield JE, Woolfson RG, and Macallister RJ. Vitamin C improves resistance but not conduit artery endothelial function in patients with chronic renal failure. Kidney Int 63: 1433–1442, 2003. [DOI] [PubMed] [Google Scholar]
  • 119.Yilmaz MI, Axelsson J, Sonmez A, Carrero JJ, Saglam M, Eyileten T, Caglar K, Kirkpantur A, Celik T, Oguz Y, Vural A, Yenicesu M, Lindholm B, and Stenvinkel P. Effect of renin angiotensin system blockade on pentraxin 3 levels in type-2 diabetic patients with proteinuria. Clin J Am Soc Nephrol 4: 535– 541, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Yilmaz MI, Saglam M, Sonmez A, Caglar K, Cakir E, Kurt Y, Eyileten T, Tasar M, Acikel C, Oguz Y, Vural A, and Yenicesu M. Improving proteinuria, endothelial functions and asymmetric dimethylarginine levels in chronic kidney disease: ramipril versus valsartan. Blood Purif 25: 327–335, 2007. [DOI] [PubMed] [Google Scholar]
  • 121.Flevari P, Kalogeropoulou S, Drakou A, Leftheriotis D, Panou F, Lekakis J, Kremastinos D, and Vlahakos DV. Spironolactone improves endothelial and cardiac autonomic function in non heart failure hemodialysis patients. J Hypertens 31: 1239–1244, 2013. [DOI] [PubMed] [Google Scholar]
  • 122.Irace C, Cutruzzola A, Parise M, Fiorentino R, Frazzetto M, Gnasso C, Casciaro F, and Gnasso A. Effect of empagliflozin on brachial artery shear stress and endothelial function in subjects with type 2 diabetes: Results from an exploratory study. Diab Vasc Dis Res 17: 1479164119883540, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Balleza Alejandri LR, Grover Paez F, Gonzalez Campos E, Ramos Becerra CG, Cardona Munoz EG, Pascoe Gonzalez S, Ramos Zavala MG, Reynoso Roa AS, Suarez Rico DO, Beltran Ramirez A, Garcia Galindo JJ, Cardona Muller D, and Galan Ruiz CY. Empagliflozin and Dapagliflozin Improve Endothelial Function in Mexican Patients with Type 2 Diabetes Mellitus: A Double-Blind Clinical Trial. J Cardiovasc Dev Dis 11: 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Northrup H, Somarathna M, Corless S, Falzon I, Totenhagen J, Lee T, and Shiu YT. Analysis of Geometric and Hemodynamic Profiles in Rat Arteriovenous Fistula Following PDE5A Inhibition. Front Bioeng Biotechnol 9: 779043, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Lok CE, and Moist L. More than reducing early fistula thrombosis is required: lessons from the Dialysis Access Consortium clopidogrel fistula study. Am J Kidney Dis 52: 834–838, 2008. [DOI] [PubMed] [Google Scholar]
  • 126.Olivier CB, Sundaram V, Chertow GM, Shashidhar S, McDonnell LK, Ding VY, Desai M, Mahaffey KW, and Mell M. A double-blind, randomized, placebo-controlled pilot trial to evaluate safety and efficacy of vorapaxar on arteriovenous fistula maturation. J Vasc Access 21: 467–474, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Irish AB, Viecelli AK, Hawley CM, Hooi LS, Pascoe EM, Paul-Brent PA, Badve SV, Mori TA, Cass A, Kerr PG, Voss D, Ong LM, and Polkinghorne KR. Effect of Fish Oil Supplementation and Aspirin Use on Arteriovenous Fistula Failure in Patients Requiring Hemodialysis: A Randomized Clinical Trial. JAMA Intern Med 177: 184–193, 2017. [DOI] [PubMed] [Google Scholar]
  • 128.Matsubara Y, Kiwan G, Liu J, Gonzalez L, Langford J, Gao M, Gao X, Taniguchi R, Yatsula B, Furuyama T, Matsumoto T, Komori K, and 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 41: e160–e174, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Fransson T, Gottsäter A, Abdulrasak M, Malina M, and Resch T. Drug-eluting balloon (DEB) versus plain old balloon angioplasty (POBA) in the treatment of failing dialysis access: A prospective randomized trial. J Int Med Res 50: 3000605221081662, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Böhme T, Noory E, Beschorner U, Jacques B, Bürgelin K, Hirstein S, and Zeller T. Combined treatment of dysfunctional dialysis access with cutting balloon and paclitaxel-coated balloon in real world. Vasa 52: 284–289, 2023. [DOI] [PubMed] [Google Scholar]
  • 131.Katsanos K, Ho P, Tang TY, Vlachou E, Yap CJQ, Kitrou PM, and Karnabatidis D. Polymer-coated paclitaxel-eluting stents for the treatment of stenosed native arteriovenous fistulas: Long-term results from the ELUDIA study. J Vasc Access 25: 1593–1600, 2024. [DOI] [PubMed] [Google Scholar]
  • 132.AlQubaisi A, Arabi M, AlZahrani Y, and Bashir O. Drug-coated balloons versus conventional percutaneous transluminal angioplasty for treatment-naïve dysfunctional arteriovenous fistulas.: A 10- year single center retrospective study. Saudi Med J 45: 741–744, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Fereydooni A, Guo X, Gorecka J, Isaji T, Ono S, Taniguchi R, Nassiri N, and Dardik A. Inhibition of the Akt1-mTORC1 Axis Alters Venous Remodeling to Improve Arteriovenous Fistula Patency. Journal of Vascular Surgery 70: e43–e44, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Voorzaat BM, van der Bogt KEA, Bezhaeva T, van Schaik J, Eefting D, van der Putten K, van Nieuwenhuizen RC, Groeneveld JO, Hoogeveen EK, van der Meer IM, Statius van Eps RG, Vogt L, Huisman L, Gabreëls B, Boom H, Verburgh CA, Boon D, Metselaar JM, Weijmer MC, and Rotmans JI. A Randomized Trial of Liposomal Prednisolone (LIPMAT) to Enhance Radiocephalic Fistula Maturation: A Pilot Study. Kidney Int Rep 5: 1327–1332, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Tsapenko MV, d’Uscio LV, Grande JP, Croatt AJ, Hernandez MC, Ackerman AW, Katusic ZS, and Nath KA. Increased production of superoxide anion contributes to dysfunction of the arteriovenous fistula. Am J Physiol Renal Physiol 303: F1601–1607, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Janardhanan R, Yang B, Kilari S, Leof EB, Mukhopadhyay D, and Misra S. The Role of Repeat Administration of Adventitial Delivery of Lentivirus-shRNA-Vegf-A in Arteriovenous Fistula to Prevent Venous Stenosis Formation. J Vasc Interv Radiol 27: 576–583, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Nieves Torres EC, Yang B, Roy B, Janardhanan R, Brahmbhatt A, Leof E, Mukhopadhyay D, and Misra S. Adventitial delivery of lentivirus-shRNA-ADAMTS-1 reduces venous stenosis formation in arteriovenous fistula. PLoS One 9: e94510, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kilari S, Cai C, Zhao C, Sharma A, Chernogubova E, Simeon M, Wu CC, Song HL, Maegdefessel L, and Misra S. The Role of MicroRNA-21 in Venous Neointimal Hyperplasia: Implications for Targeting miR-21 for VNH Treatment. Mol Ther 27: 1681–1693, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Martinez L, Duque JC, Tabbara M, Paez A, Selman G, Hernandez DR, Sundberg CA, Tey JCS, Shiu YT, Cheung AK, Allon M, Velazquez OC, Salman LH, and Vazquez-Padron RI. Fibrotic Venous Remodeling and Nonmaturation of Arteriovenous Fistulas. J Am Soc Nephrol 29: 1030–1040, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Bleyer AJ, Scavo VA, Wilson SE, Browne BJ, Ferris BL, Ozaki CK, Lee T, Peden EK, Dixon BS, Mishler R, O’Connor TP, Kidd K, and Burke SK. A randomized trial of vonapanitase (PATENCY-1) to promote radiocephalic fistula patency and use for hemodialysis. J Vasc Surg 69: 507–515, 2019. [DOI] [PubMed] [Google Scholar]
  • 141.Hernandez DR, Applewhite B, Martinez L, Laurito T, Tabbara M, Rojas MG, Wei Y, Selman G, Knysheva M, Velazquez OC, Salman LH, Andreopoulos FM, Shiu YT, and Vazquez-Padron RI. Inhibition of Lysyl Oxidase with β-aminopropionitrile Improves Venous Adaptation after Arteriovenous Fistula Creation. Kidney360 2: 270–278, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Shiu YT, He Y, Tey JCS, Knysheva M, Anderson B, and Kauser K. Natural Vascular Scaffolding Treatment Promotes Outward Remodeling During Arteriovenous Fistula Development in Rats. Front Bioeng Biotechnol 9: 622617, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Martinez L, Duque JC, Escobar LA, Tabbara M, Asif A, Fayad F, Vazquez-Padron RI, and Salman LH. Distinct impact of three different statins on arteriovenous fistula outcomes: a retrospective analysis. J Vasc Access 17: 471–476, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Cui J, Kessinger CW, Jhajj HS, Grau MS, Misra S, Libby P, McCarthy JR, and Jaffer FA. Atorvastatin Reduces In Vivo Fibrin Deposition and Macrophage Accumulation, and Improves Primary Patency Duration and Maturation of Murine Arteriovenous Fistula. J Am Soc Nephrol 31: 931–945, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Roan JN, Lin WH, Tsai MT, Kuo TH, Lin TW, Chen DY, Fang SY, Hsieh YP, Li CY, and Lam CF. Rosuvastatin Failed to Improve Arteriovenous Fistula Patency for Hemodialysis in Diabetic Patients - A Randomized Clinical Trial. Acta Cardiol Sin 37: 18–29, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Nath KA, Juncos LA, Singh RD, Grande JP, Croatt AJ, Ackerman AW, Kanamori KS, Adams CM, Tchkonia T, Kirkland JL, and Katusic ZS. The Occurrence of Senescence in the Arteriovenous Fistula in the Rat. Kidney360 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Nath KA, O’Brien DR, Croatt AJ, Grande JP, Ackerman AW, Nath MC, Yamada S, Terzic A, Tchkonia T, Kirkland JL, and Katusic ZS. The murine dialysis fistula model exhibits a senescence phenotype: pathobiological mechanisms and therapeutic potential. Am J Physiol Renal Physiol 315: F1493–f1499, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Hanawa K, Ito K, Aizawa K, Shindo T, Nishimiya K, Hasebe Y, Tuburaya R, Hasegawa H, Yasuda S, Kanai H, and Shimokawa H. Low-intensity pulsed ultrasound induces angiogenesis and ameliorates left ventricular dysfunction in a porcine model of chronic myocardial ischemia. PLoS One 9: e104863, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Mohamad Yusoff F, Kajikawa M, Yamaji T, Kishimoto S, Maruhashi T, Nakashima A, Tsuji T, and Higashi Y. Low-intensity pulsed ultrasound improves symptoms in patients with Buerger disease: a double-blinded, randomized, and placebo-controlled study. Scientific Reports 14: 13704, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Xia F, Rai V, and Agrawal DK. Vascular and Perivascular Role in the Regulation of Angiogenesis: Impact on Arteriovenous Fistula Maturation. Arch Intern Med Res 7: 284–296, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Lindhard K, Jensen BL, Pedersen BL, Meyer-Olesen C, Rix M, Hansen HP, Schalkwijk C, Waarenburg M, Heaf J, and Hansen D. Far infrared treatment on the arteriovenous fistula induces changes in sVCAM and sICAM in patients on hemodialysis. Nephrol Dial Transplant 38: 1752–1760, 2023. [DOI] [PubMed] [Google Scholar]
  • 152.Nantakool S, Reanpang T, Prasannarong M, Pongtam S, and Rerkasem K. Upper limb exercise for arteriovenous fistula maturation in people requiring permanent haemodialysis access. Cochrane Database Syst Rev 10: Cd013327, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Lok CE, Huber TS, Lee T, Shenoy S, Yevzlin AS, Abreo K, Allon M, Asif A, Astor BC, Glickman MH, Graham J, Moist LM, Rajan DK, Roberts C, Vachharajani TJ, and Valentini RP. KDOQI Clinical Practice Guideline for Vascular Access: 2019 Update. Am J Kidney Dis 75: S1–s164, 2020. [DOI] [PubMed] [Google Scholar]
  • 154.Shahverdyan R, Meyer T, and Matoussevitch V. Patency and functionality of radiocephalic arteriovenous fistulas with an external support device (VasQ™): Real-world single-center experience. J Vasc Access 22: 166–172, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Shahverdyan R, Tabbi P, and Mestres G. Multicenter European real-world utilization of VasQ anastomotic external support device for arteriovenous fistulae. J Vasc Surg 75: 248–254, 2022. [DOI] [PubMed] [Google Scholar]
  • 156.Shahverdyan R, and Hentschel DM. Achieving high maturation and cannulation rates of radial- cephalic arteriovenous fistulas with VasQ™ device. Semin Dial 36: 147–154, 2023. [DOI] [PubMed] [Google Scholar]
  • 157.Bozzetto M, Soliveri L, Poloni S, Brambilla P, Curtò D, Condemi GC, Cefalì P, Spina I, Villa A, Caroli A, and Remuzzi A. Arteriovenous fistula creation with VasQ(TM) device: A feasibility study to reveal hemodynamic implications. J Vasc Access 25: 60–70, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Piryani AK, Kilari S, Takahashi E, DeMartino RR, Mandrekar J, Dietz AB, and 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 2: 1945–1952, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES