The molecular mechanisms of hemodialysis vascular access failure

Akshaar Brahmbhatt, Andrea Remuzzi, Marco Franzoni, Sanjay Misra, Akshaar Brahmbhatt, Andrea Remuzzi, Marco Franzoni, Sanjay Misra

Abstract

The arteriovenous fistula has been used for more than 50 years to provide vascular access for patients undergoing hemodialysis. More than 1.5 million patients worldwide have end stage renal disease and this population will continue to grow. The arteriovenous fistula is the preferred vascular access for patients, but its patency rate at 1 year is only 60%. The majority of arteriovenous fistulas fail because of intimal hyperplasia. In recent years, there have been many studies investigating the molecular mechanisms responsible for intimal hyperplasia and subsequent thrombosis. These studies have identified common pathways including inflammation, uremia, hypoxia, sheer stress, and increased thrombogenicity. These cellular mechanisms lead to increased proliferation, migration, and eventually stenosis. These pathways work synergistically through shared molecular messengers. In this review, we will examine the literature concerning the molecular basis of hemodialysis vascular access malfunction.

Keywords: arteriovenous fistula; murine model; restenosis; vascular biology; venous neointimal hyperplasia.

Copyright © 2016 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
(a, b) Fistulograms showing a stenosis at the polytetrafluoroethylene graft anastomosis to the basilic vein. (c) A gross specimen from a different patient showing thickening of the vein to graft anastomosis. Hematoxylin and eosin stain (d) at 10× magnification demonstrating a thickened neointima and (e) at 40× magnification (red box in d) showing increased cellular proliferation. ePTFE, expanded polytetrafluoro-ethylene.
Figure 2. Schematic of vascular injuries contributing…
Figure 2. Schematic of vascular injuries contributing to stenosis formation in hemodialysis vascular access
IH, intimal hyperplasia.
Figure 3. TNF-α, MCP-1, and IL-1β expression…
Figure 3. TNF-α, MCP-1, and IL-1β expression by qRT-PCR
Tissue necrosis factor-alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), and interlukin-1 beta (IL-1β) expression by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) in graft veins and control veins at 3 and 7 days after arteriovenous fistula placement in mice with established chronic kidney disease. There is a significant increase in the mean TNF-α, MCP-1, and IL-1β expression at 3 days in graft veins when compared with control veins (P < 0.05). Each bar shows the mean ± SEM of 3 samples per group. Two-way analysis of variance with Student t test with post hoc Bonferroni correction was performed. *P < 0.05.
Figure 4. iNOS and Arg-1 expression using…
Figure 4. iNOS and Arg-1 expression using qRT-PCR
Inducible nitric oxide synthase (iNOS) and arginase-1 (Arg-1) expression using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) in graft veins and control veins at 3 and 7 days after arteriovenous fistula placement in mice with established chronic kidney disease. There is a significant increase in the mean Arg-1 expression with a decrease in iNOS between day 3 and 7. Each bar shows the mean ± SEM of 3 samples per group. Two-way analysis of variance with Student t test with post hoc Bonferroni correction was performed. P < 0.05.
Figure 5. Flow cytometry for bone marrow…
Figure 5. Flow cytometry for bone marrow Ly-6C Hi and Ly-6C Lo cells and splenic F4/80(+) cells from animals treated with CLO and PBS at day 14
(a) Representative flow diagram from a clodronate (CLO)-treated animal. (b) Representative flow diagram from a control–phosphate-buffered saline (PBS)-treated–animal. (c) Pooled data from 2 PBS-treated animals and 3 CLO-treated animals at day 14. Each bar shows the mean ± SEM of 2 to 3 samples per group. Two-way analysis of variance with Student t test with post hoc Bonferroni correction was performed. *P < 0.05.
Figure 6. H&E staining of outflow veins…
Figure 6. H&E staining of outflow veins removed from animals treated with PBS and CLO at day 14
Hematoxylin and eosin (H&E) staining of outflow veins removed from PBS-treated and CLO-treated animals at day 14 after arteriovenous fistula placement. There is a reduction in the neointima (n) of the CLO-treated animals when compared with PBS control animals. Asterisk (*) shows the lumen of the vessel. Scale bars = 50 μM. CLO, clodronate; m + a, media/adventitia; PBS, phosphate-buffered saline.
Figure 7. Inflammatory cytokines in A-treated versus…
Figure 7. Inflammatory cytokines in A-treated versus control animals
Inflammatory cytokines are elevated in Avastin (A)-treated vessels when compared with control animals (IgG) at days 7 and 14 after arteriovenous fistula placement. Vascular endothelial growth factor-A (VEGF-A), MCP-1, TNF-α, and IL-1β expression by qRT-PCR in graft veins and control veins at 7 and 14 days after arteriovenous fistula placement in mice with established chronic kidney disease. Each bar shows the mean ± SEM of 4 to 6 samples per group. Two-way analysis of variance with Student t test with post hoc Bonferroni correction was performed. *P < 0.05. IL-1β, interlukin-1 beta; MCP-1, monocyte chemoattractant protein-1; qRT-PCR, reverse transcriptase polymerase chain reaction; TNF-α, tissue necrosis factor-alpha.
Figure 8. Schematic representation of molecular mechanism…
Figure 8. Schematic representation of molecular mechanism of IH development in native AVF
Section of a side-to-end arteriovenous fistula (AVF). Laminar blood flow coming from the proximal artery stimulates the endothelial cells (ECs) with unidirectional wall shear stress (WSS) until the anastomosis level where the blood flow splits in 2 directions. At the vein curvature, the blood flow becomes unstable with disturbed and oscillating WSS and reverse flows at the inner curvature of the anastomosis. After the curvature, blood flow oscillations decrease and WSS returns to almost unidirectional. The different WSS patterns generated on the endothelium lead wall remodeling. At (a), the unidirectional WSS maintains vessel patency while at (b) oscillating and reversing WSS impair ECs quiescence leading to intimal hyperplasia (IH). ALK-5, activin receptor-like kinases 1/5; Ang-II, angiotensin II; ECM, extracellular matrix; ET-1, endothelin 1; GCX, glycocalyx; IL-8, interleukin 8; KLF-2, Krüppel-like factor 2; MCP-1 monocytes chemoattractant protein 1; MFs, myofibroblasts; MMP, metalloproteinase; NO, nitric oxide; SMCs, smooth muscle cells; TGF-β, transforming growth factor β; VCAM, vascular cell adhesion protein; VE, vascular endothelial; VSMC, vascular smooth muscle cells.

References

    1. Sands JJ. Increasing AV fistulas: revisiting a time-tested solution. Semin Dial. 2000;13:351–353.
    1. Roy-Chaudhury P, Lee TC. Vascular stenosis: biology and interventions. Curr Opin Nephrol Hypertens. 2007;16:516–522.
    1. Schinstock CA, Albright RC, Williams AW, et al. Outcomes of arteriovenous fistula creation after the Fistula First Initiative. Clin J Am Soc Nephrol. 2011;6:1996–2002.
    1. Rayner HC, Besarab A, Brown WW, et al. 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. 2004;44(suppl 2):22–26.
    1. Vassalotti JA, Jennings WC, Beathard GA, et al. for the Fistula First Breakthrough Initiative Community Education Committee Fistula first breakthrough initiative: targeting catheter last in fistula first. Semin Dial. 2012;25:303–310.
    1. Manook M, Calder F. Practical aspects of arteriovenous fistula formation in the pediatric population. Pediatr Nephrol. 2013;28:885–893.
    1. Zachara BA. Selenium and selenium-dependent antioxidants in chronic kidney disease. Adv Clin Chem. 2015;68:131–151.
    1. Rayner HC, Pisoni RL. The increasing use of hemodialysis catheters: evidence from the DOPPS on its significance and ways to reverse it. Semin Dial. 2010;23:6–10.
    1. Lopez-Vargas PA, Craig JC, Gallagher MP, et al. Barriers to timely arteriovenous fistula creation: a study of providers and patients. Am J Kidney Dis. 2011;57:873–882.
    1. Rooijens PP, Tordoir JH, Stijnen T, et al. Radiocephalic wrist arteriovenous fistula for hemodialysis: meta-analysis indicates a high primary failure rate. Eur J Vasc Endovasc Surg. 2004;28:583–589.
    1. Huijbregts HJT, Bots ML, Wittens CHA, et al. for the CIMINO Study Group Hemodialysis arteriovenous fistula patency revisited: results of a prospective, multicenter initiative. Clin J Am Soc Nephrol. 2008;3:714–719.
    1. Al-Jaishi AA, Oliver MJ, Thomas SM, et al. Patency rates of the arteriovenous fistula for hemodialysis: a systematic review and meta-analysis. Am J Kidney Dis. 2014;63:464–478.
    1. Wong V, Ward R, Taylor J, et al. Factors associated with early failure of arteriovenous fistulae for haemodialysis access. Eur J Vasc Endovascr Surg. 1996;12:207–213.
    1. Feldman HI, Joffe M, Rosas SE, et al. Predictors of successful arteriovenous fistula maturation. AmJ Kidney Dis. 2003;42:1000–1012.
    1. Aitken E, Jackson A, Kong C, et al. Renal function, uraemia and early arteriovenous fistula failure. BMC Nephrol. 2014;15:179.
    1. Gagliardi GM, Rossi S, Condino F, et al. Malnutrition, infection and arteriovenous fistula failure: is there a link? J Vasc Access. 2011;12:57–62.
    1. Smith GE, Gohil R, Chetter IC. Factors affecting the patency of arteriovenous fistulas for dialysis access. J Vasc Surg. 2012;55:849–855.
    1. Wasse H, Huang R, Naqvi N, et al. Inflammation, oxidation and venous neointimal hyperplasia precede vascular injury from AVF creation in CKD patients. J Vasc Access. 2012;13:168–174.
    1. Roy-Chaudhury P, Wang Y, Krishnamoorthy M, et al. Cellular phenotypes in human stenotic lesions from haemodialysis vascular access. Nephrol Dial Transplant. 2009;24:2786–2791.
    1. Roy-Chaudhury P, Arend L, Zhang J, et al. Neointimal hyperplasia in early arteriovenous fistula failure. Am J Kidney Dis. 2007;50:782–790.
    1. Stracke S, Konner K, Kostlin I, et al. Increased expression of TGF-beta1 and IGF-I in inflammatory stenotic lesions of hemodialysis fistulas. Kidney Int. 2002;61:1011–1019.
    1. Simone S, Loverre A, Cariello M, et al. Arteriovenous fistula stenosis in hemodialysis patients is characterized by an increased adventitial fibrosis. J Nephrol. 2014;27:555–562.
    1. Campos B, Lee T, Roy-Chaudhury P. Arteriovenous fistula failure: is there a role for epigenetic regulation? Semin Nephrol. 2013;33:400–406.
    1. Rotmans JI, Pattynama PM, Verhagen HJ, et al. Sirolimus-eluting stents to abolish intimal hyperplasia and improve flow in porcine arteriovenous grafts: a 4-week follow-up study. Circulation. 2005;111:1537–1542.
    1. Rotmans JI, Verhagen HJ, Velema E, et al. Local overexpression of C-type natriuretic peptide ameliorates vascular adaptation of porcine hemodialysis grafts. Kidney Int. 2004;65:1897–1905.
    1. Misra S, Fu AA, Anderson JL, et al. The rat femoral arteriovenous fistula model: increased expression of matrix metalloproteinase-2 and -9 at the venous stenosis. J Vasc Interv Radiol. 2008;19:587–594.
    1. Yang B, Shergill U, Fu AA, et al. The mouse arteriovenous fistula model. J Vasc Interv Radiol. 2009;20:946–950.
    1. Lee T, Chauhan V, Krishnamoorthy M, et al. Severe venous neointimal hyperplasia prior to dialysis access surgery. Nephrol Dial Transplant. 2011;26:2264–2270.
    1. Misra S, Gordon JD, Fu AA, et al. The porcine remnant kidney model of chronic renal insufficiency. J Surg Res. 2006;135:370–379.
    1. Hughes D, Fu AA, Puggioni A, et al. Adventitial transplantation of blood outgrowth endothelial cells in porcine haemodialysis grafts alleviates hypoxia and decreases neointimal proliferation through a matrix metalloproteinase-9-mediated pathway–a pilot study. Nephrol Dials Transplant. 2008;24:85–96.
    1. Misra S, Fu AA, Anderson JL, et al. Fetuin-A expression in early venous stenosis formation in a porcine model of hemodialysis graft failure. J Vasc Interv Radiol. 2008;19:1477–1482.
    1. Chamberlain RM, Shirley DG. Time course of the renal functional response to partial nephrectomy: measurements in conscious rats. Exp Physiol. 2007;92:251–262.
    1. Misra S, Shergill U, Yang B, et al. Increased expression of HIF-1alpha, VEGF-A and its receptors, MMP-2, TIMP-1, and ADAMTS-1 at the venous stenosis of arteriovenous fistula in a mouse model with renal insufficiency. J Vasc Interv Radiol. 2010;21:1255–1261.
    1. Ene-Iordache B, Cattaneo L, Dubini G, Remuzzi A. Effect of anastomosis angle on the localization of disturbed flow in ’side-to-end’ fistulae for haemodialysis access. Nephrol Dial Transplant. 2013;28:997–1005.
    1. Ene-Iordache B, Remuzzi A. Disturbed flow in radial-cephalic arteriovenous fistulae for haemodialysis: low and oscillating shear stress locates the sites of stenosis. Nephrol Dial Transplant. 2012;27:358–368.
    1. Brahmbhatt A, NievesTorres E, Yang B, et al. The Role of Iex-1 in the pathogenesis of venous neointimal hyperplasia associated with hemodialysis arteriovenous fistula. PLoS One. 2014;9:e102542.
    1. Riella MC, Roy-Chaudhury P. Vascular access in haemodialysis: strengthening the Achilles’ heel. Nat Rev Nephrol. 2013;9:348–357.
    1. Liang A, Wang Y, Han G, et al. Chronic kidney disease accelerates endothelial barrier dysfunction in a mouse model of an arteriovenous fistula. Am J Physiol Renal Physiol. 2013;304:F1413–F1420.
    1. Misra S, Fu AA, Rajan DK, et al. Expression of hypoxia inducible factor-1 alpha, macrophage migration inhibition factor, matrix metalloproteinase-2 and -9, and their inhibitors in hemodialysis grafts and arteriovenous fistulas. J Vasc Interv Radiol. 2008;19:252–259.
    1. Asare Y, Schmitt M, Bernhagen J. The vascular biology of macrophage migration inhibitory factor (MIF): expression and effects in inflammation, atherogenesis and angiogenesis. Thromb Haemost. 2013;109:391–398.
    1. Stangenberg S, Nguyen LT, Chen H, et al. Oxidative stress, mitochondrial perturbations and fetal programming of renal disease induced by maternal smoking. Int J Biochem Cell Biol. 2015;64:81–90.
    1. van der Weerd NC, Grooteman MP, Nube MJ, et al. Hepcidin in chronic kidney disease: not an anaemia management tool, but promising as a cardiovascular biomarker. Neth J Med. 2015;73:108–118.
    1. Virzì GM, Clementi A, de Cal M, et al. Oxidative stress: dual pathway induction in cardiorenal syndrome type 1 pathogenesis. Oxid Med Cell Longev. 2015;2015:391790.
    1. Veillat V, Carli C, Metz CN, et al. Macrophage migration inhibitory factor elicits an angiogenic phenotype in human ectopic endometrial cells and triggers the production of major angiogenic factors via CD44, CD74, and MAPK signaling pathways. J Clin Endocrinol Metab. 2010;95:E403–412.
    1. Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res. 2009;29:313–326.
    1. Carbo C, Arderiu G, Escolar G, et al. Differential expression of proteins from cultured endothelial cells exposed to uremic versus normal serum. Am J Kidney Dis. 2008;51:603–612.
    1. Martin-Rodriguez S, Caballo C, Gutierrez G, et al. TLR4 and NALP3 inflammasome in the development of endothelial dysfunction in uraemia. Eur J Clin Invest. 2015;45:160–169.
    1. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821–832.
    1. Heine GH, Ulrich C, Sester U, et al. Transforming growth factor beta1 genotype polymorphisms determine AV fistula patency in hemodialysis patients. Kidney Int. 2003;64:1101–1107.
    1. Ohtsubo K, Marth JD. Glycosylation in cellular mechanisms of health and disease. Cell. 2006;126:855–867.
    1. Zhao YY, Takahashi M, Gu JG, et al. Functional roles of N-glycans in cell signaling and cell adhesion in cancer. Cancer Sci. 2008;99:1304–1310.
    1. Shen N, Lin H, Wu T, et al. Inhibition of TGF-β1-receptor posttranslational core fucosylation attenuates rat renal interstitial fibrosis. Kidney Int. 2013;84:64–77.
    1. Takeda R, Suzuki E, Satonaka H, et al. Blockade of endogenous cytokines mitigates neointimal formation in obese Zucker rats. Circulation. 2005;111:1398–1406.
    1. Dixon BS. Why don't fistulas mature? Kidney Int. 2006;70:1413–1422.
    1. Sener EF, Taheri S, Korkmaz K, et al. Association of TNF-α -308 G > A and ACE I/D gene polymorphisms in hemodialysis patients with arteriovenous fistula thrombosis. Int Urol Nephrol. 2014;46:1419–1425.
    1. Sidawy AN, Gray R, Besarab A, et al. Recommended standards for reports dealing with arteriovenous hemodialysis accesses. J Vasc Surg. 2002;35:603–610.
    1. Kokubo T, Ishikawa N, Uchida H, et al. CKD accelerates development of neointimal hyperplasia in arteriovenous fistulas. J Am Soc Nephrol. 2009;20:1236–1245.
    1. Yang B, Vohra PK, Janardhanan R, et al. Expression of profibrotic genes in a murine remnant kidney model. J Vasc Interv Radiol. 2011;22:1765–1772. e1761.
    1. Weiss MF, Scivittaro V, Anderson JM. Oxidative stress and increased expression of growth factors in lesions of failed hemodialysis access. Am J Kidney Dis. 2001;37:970–980.
    1. Jankovic A, Damjanovic T, Djuric Z, et al. Impact of vascular calcifications on arteriovenous fistula survival in hemodialysis patients: a five-year follow-up. Nephron. 2015;129:247–252.
    1. DeFronzo RA, Alvestrand A, Smith D, et al. Insulin resistance in uremia. J Clin Invest. 1981;67:563–568.
    1. Rostand SG, Drueke TB. Parathyroid hormone, vitamin D, and cardiovascular disease in chronic renal failure. Kidney Int. 1999;56:383–392.
    1. Buzello M, Tornig J, Faulhaber J, et al. The apolipoprotein e knockout mouse: a model documenting accelerated atherogenesis in uremia. J Am Soc Nephrol. 2003;14:311–316.
    1. Georgiadis GS, Georgakarakos EI, Antoniou GA, et al. Correlation of preexisting radial artery macrocalcifications with late patency of primary radiocephalic fistulas in diabetic hemodialysis patients. J Vasc Surg. 2014;60:462–470.
    1. Balci M, Kirkpantur A, Turkvatan A, et al. Sclerostin as a new key player in arteriovenous fistula calcification. Herz. 2015;40:289–297.
    1. Mitsuhashi T, Morris RC, Ives HE. 1,25-dihydroxyvitamin D3 modulates growth of vascular smooth muscle cells. J Clin Invest. 1991;87:1889–1895.
    1. Danziger J. Vitamin K-dependent proteins, warfarin, and vascular calcification. Clin J Am Soc Nephrol. 2008;3:1504–1510.
    1. Hruska KA, Mathew S, Memon I, et al. The pathogenesis of vascular calcification in the chronic kidney disease mineral bone disorder: the links between bone and the vasculature. Semin Nephrol. 2009;29:156–165.
    1. Yamanouchi D, Takei Y, Komori K. Balanced mineralization in the arterial system: possible role of osteoclastogenesis/osteoblastogenesis in abdominal aortic aneurysm and stenotic disease. Circ J. 2012;76:2732–2737.
    1. Lu L, Erhard P, Salomon RG, Weiss MF. Serum vitamin E and oxidative protein modification in hemodialysis: a randomized clinical trial. Am J Kidney Dis. 2007;50:305–313.
    1. Wu M, Rementer C, Giachelli CM. Vascular calcification: an update on mechanisms and challenges in treatment. Calcif Tissue Int. 2013;93:365–373.
    1. Moe SM, Reslerova M, Ketteler M, et al. Role of calcification inhibitors in the pathogenesis of vascular calcification in chronic kidney disease (CKD). Kidney Int. 2005;67:2295–2304.
    1. Misra S, Fu AA, Puggioni A, et al. Increased expression of hypoxiainducible factor-1 alpha in venous stenosis of arteriovenous polytetrafluoroethylene grafts in a chronic renal insufficiency porcine model. J Vasc Interv Radiol. 2008;19:260–265.
    1. Zhu Y, Lawton MT, Du R, et al. Expression of hypoxia-inducible factor-1 and vascular endothelial growth factor in response to venous hypertension. Neurosurgery. 2006;59:687–696. [discussion: 687-696]
    1. Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3:721–732.
    1. Wan J, Lata C, Santilli A, et al. Supplemental oxygen reverses hypoxia induced smooth muscle cell proliferation by modulating HIF-alpha and VEGF levels in a rabbit arteriovenous fistula model. Ann Vasc Surg. 2014;28:725–736.
    1. Yang B, Janardhanan R, Vohra P, et al. Adventitial transduction of lentivirus-shRNA-VEGF-A in arteriovenous fistula reduces venous stenosis formation. Kidney Int. 2014;85:289–306.
    1. Candan F, Yildiz G, Kayatas M. Role of the VEGF 936 gene polymorphism and VEGF-A levels in the late-term arteriovenous fistula thrombosis in patients undergoing hemodialysis. Int Urol Nephrol. 2014;46:1815–1823.
    1. Ohtani K, Egashira K, Hiasa K, et al. Blockade of vascular endothelial growth factor suppresses experimental restenosis after intraluminal injury by inhibiting recruitment of monocyte lineage cells. Circulation. 2004;110:2444–2452.
    1. Shibuya M. Differential roles of vascular endothelial growth factor receptor-1 and receptor-2 in angiogenesis. J Biochem Mol Biol. 2006;39:469–478.
    1. Huusko J, Merentie M, Dijkstra MH, et al. The effects of VEGF-R1 and VEGF-R2 ligands on angiogenic responses and left ventricular function in mice. Cardiovasc Res. 2010;86:122–130.
    1. Janda K, Krzanowski M, Gajda M, et al. Cardiovascular risk in chronic kidney disease patients: intima-media thickness predicts the incidence and severity of histologically assessed medial calcification in radial arteries. BMC Nephrol. 2015;16:78.
    1. Fragoso A, Silva AP, Gundlach K, et al. Magnesium and FGF-23 are independent predictors of pulse pressure in pre-dialysis diabetic chronic kidney disease patients. Clinical Kidney J. 2014;7:161–166.
    1. Vempati P, Popel AS, Mac Gabhann F. Extracellular regulation of VEGF: isoforms, proteolysis, and vascular patterning. Cytokine Growth Factor Rev. 2014;25:1–19.
    1. Kikuchi R, Nakamura K, MacLauchlan S, et al. An antiangiogenic isoform of VEGF-A contributes to impaired vascularization in peripheral artery disease. Nat Med. 2014;20:1464–1471.
    1. Nowak DG, Woolard J, Amin EM, et al. Expression of pro- and anti-angiogenic isoforms of VEGF is differentially regulated by splicing and growth factors. J Cell Sci. 2008;121:3487–3495.
    1. Geng L, Chaudhuri A, Talmon G, et al. TGF-Beta suppresses VEGFA-mediated angiogenesis in colon cancer metastasis. PLoS One. 2013;8:e59918.
    1. Nakagawa T, Li JH, Garcia G, et al. TGF-beta induces proangiogenic and antiangiogenic factors via parallel but distinct Smad pathways. Kidney Int. 2004;66:605–613.
    1. Shi X, Guo LW, Seedial SM, et al. TGF-β/Smad3 inhibit vascular smooth muscle cell apoptosis through an autocrine signaling mechanism involving VEGF-A. Cell Death Dis. 2014;5:e1317.
    1. Lata C, Green D, Wan J, et al. The role of short-term oxygen administration in the prevention of intimal hyperplasia. J Vasc Surg. 2013;58:452–459.
    1. Modaresi A, Nafar M, Sahraei Z. Oxidative stress in chronic kidney disease. Iranian J Kidney Dis. 2015;9:165–179.
    1. Himmelfarb J, Stenvinkel P, Ikizler TA, Hakim RM. The elephant in uremia: oxidant stress as a unifying concept of cardiovascular disease in uremia. Kidney Int. 2002;62:1524–1538.
    1. Puchades MJ, Saez G, Munoz MC, et al. Study of oxidative stress in patients with advanced renal disease and undergoing either hemodialysis or peritoneal dialysis. Clin Nephrol. 2013;80:177–186.
    1. Lehoux S. Redox signalling in vascular responses to shear and stretch. Cardiovasc Res. 2006;71:269–279.
    1. Sakoh T, Nakayama M, Tanaka S, et al. Association of serum total bilirubin with renal outcome in Japanese patients with stages 3-5 chronic kidney disease. Metabolism. 2015;69:1096–1102.
    1. Dursun B, Dursun E, Suleymanlar G, et al. Carotid artery intima-media thickness correlates with oxidative stress in chronic haemodialysis patients with accelerated atherosclerosis. Nephrol Dial Transplant. 2008;23:1697–1703.
    1. Byon CH, Chen Y. Molecular mechanisms of vascular calcification in chronic kidney disease: the link between bone and the vasculature. Curr Osteoporos Rep. 2015;13:206–215.
    1. Ene-Iordache B, Mosconi L, Antiga L, et al. Radial artery remodeling in response to shear stress increase within arteriovenous fistula for hemodialysis access. Endothelium. 2003;10:95–102.
    1. Tucker PS, Scanlan AT, Dalbo VJ. Chronic kidney disease influences multiple systems: describing the relationship between oxidative stress, inflammation, kidney damage, and concomitant disease. Oxid Med Cell Longev. 2015;2015:806358.
    1. Manini S, Passera K, Huberts W, et al. Computational model for simulation of vascular adaptation following vascular access surgery in haemodialysis patients. Comput Methods Biomech Biomed Engin. 2014;17:1358–1367.
    1. Ene-Iordache B, Semperboni C, Dubini G, Remuzzi A. Disturbed flow in a patient-specific arteriovenous fistula for hemodialysis: multidirectional and reciprocating near-wall flow patterns. J Biomech. 2015;48:2195–2200.
    1. Nishikawa M, Ishimori N, Takada S, et al. AST-120 ameliorates lowered exercise capacity and mitochondrial biogenesis in the skeletal muscle from mice with chronic kidney disease via reducing oxidative stress. Nephrol Dial Transplant. 2015;30:934–942.
    1. Guijarro C, Egido J. Transcription factor-kappa B (NF-kappa B) and renal disease. Kidney Int. 2001;59:415–424.
    1. Assis RP, Castro JF, Gutierres VO, et al. Effects of uremic solutes on reactive oxygen species in vitro model systems as a possibility of support the renal function management. BMC Nephrol. 2015;16:50.
    1. Chevalier RL. Congenital urinary tract obstruction: the long view. Adv Chronic Kidney Dis. 2015;22:312–319.
    1. Ruiz S, Pergola PE, Zager RA, Vaziri ND. Targeting the transcription factor Nrf2 to ameliorate oxidative stress and inflammation in chronic kidney disease. Kidney Int. 2013;83:1029–1041.
    1. Zaniew M, Zachwieja J, Warzywoda A, et al. [Influence of vitamin E and N-acetylcysteine on intracellular oxidative stress in T lymphocytes in children treated with dialysis]. Wiad Lek. 2005;58(suppl 1):58–65. in Polish.
    1. Zhang L, Coombes J, Pascoe EM, et al. for the HERO Study Collaborative Group The effect of pentoxifylline on oxidative stress in chronic kidney disease patientswith erythropoiesis-stimulating agent hyporesponsiveness: sub-study of the HERO trial [e-pub ahead of print]. Redox Rep. doi: 10.1179/ 1351000215Y.0000000022, accessed December 22, 2015.
    1. Locatelli F, Canaud B, Eckardt KU, et al. Oxidative stress in end-stage renal disease: an emerging threat to patient outcome. Nephrol Dial Transplant. 2003;18:1272–1280.
    1. Cachofeiro V, Goicochea M, de Vinuesa SG, et al. Oxidative stress and inflammation, a link between chronic kidney disease and cardiovascular disease. Kidney Int Suppl. 2008;111:S4–S9.
    1. Fassett RG, Robertson IK, Ball MJ, et al. Effects of atorvastatin on oxidative stress in chronic kidney disease. Nephrology (Carlton) 2015;20:697–705.
    1. Vaziri ND, Liu S, Farzaneh SH, et al. Dose-dependent deleterious and salutary actions of the Nrf2 inducer dh404 in chronic kidney disease. Free Radic Biol Med. 2015;86:374–381.
    1. Vaziri ND, Zhao YY, Pahl MV. Altered intestinal microbial flora and impaired epithelial barrier structure and function in CKD: the nature, mechanisms, consequences and potential treatment [e-pub ahead of print]. Nephrol Dial Transplant. doi: 10.1093/ndt/gfv095, accessed December 22, 2015.
    1. Huang TH, Chen YT, Sung PH, et al. Peripheral blood-derived endothelial progenitor cell therapy prevented deterioration of chronic kidney disease in rats. Am J Transl Res. 2015;7:804–824.
    1. Schelling JR. Tubular atrophy in the pathogenesis of chronic kidney disease progression. Pediatr Nephrol. 2015:1–4.
    1. Zafrani L, Ince C. Microcirculation in acute and chronic kidney diseases. Am J Kidney Dis. 2015;66:1083–1094.
    1. Granata S, Zaza G, Simone S, et al. Mitochondrial dysregulation and oxidative stress in patients with chronic kidney disease. BMC Genomics. 2009;10:388.
    1. Chiu JJ, Chien S. Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol Rev. 2011;91:327–387.
    1. Sellin L, Kielstein JT, de Groot K. [Hyperuricemia—more than gout: impact on cardiovascular risk and renal insufficiency]. Z Rheumatol. 2015;74:322–328. in German.
    1. Gross P, Massy ZA, Henaut L, et al. Para-cresyl sulfate acutely impairs vascular reactivity and induces vascular remodeling. J Cell Physiol. 2015;230:2927–2935.
    1. Tschopp J. Mitochondria: sovereign of inflammation? Eur J Immunol. 2011;41:1196–1202.
    1. Oberg BP, McMenamin E, Lucas FL, et al. Increased prevalence of oxidant stress and inflammation in patients with moderate to severe chronic kidney disease. Kidney Int. 2004;65:1009–1016.
    1. Ma Y, Zhou L, Dong J, et al. Arterial stiffness and increased cardiovascular risk in chronic kidney disease. Int Urol Nephrol. 2015;47:1157–1164.
    1. Ali BH, Adham SA, Al Za'abi M, et al. Ameliorative effect of chrysin on adenine-induced chronic kidney disease in rats. PLoS One. 2015;10:e0125285.
    1. Roy-Chaudhury P, Spergel LM, Besarab A, et al. Biology of arteriovenous fistula failure. J Nephrol. 2007;20:150–163.
    1. Juncos JP, Grande JP, Kang L, et al. MCP-1 contributes to arteriovenous fistula failure. J Am Soc Nephrol. 2011;22:43–48.
    1. Juncos JP, Tracz MJ, Croatt AJ, et al. Genetic deficiency of heme oxygenase-1 impairs functionality and form of an arteriovenous fistula in the mouse. Kidney Int. 2008;74:47–51.
    1. Kang L, Grande JP, Farrugia G, et al. Functioning of an arteriovenous fistula requires heme oxygenase-2. Am J Physiol Renal Physiol. 2013;305:F545–F552.
    1. Lin CC, Yang WC, Lin SJ, et al. Length polymorphism in heme oxygenase-1 is associated with arteriovenous fistula patency in hemodialysis patients. Kidney Int. 2006;69:165–172.
    1. Zhou Q, Liao JK. Pleiotropic effects of statins: basic research and clinical perspectives. Circ J. 2010;74:818–826.
    1. Freidja ML, Toutain B, Caillon A, et al. Heme oxygenase 1 is differentially involved in blood flow-dependent arterial remodeling: role of inflammation, oxidative stress, and nitric oxide. Hypertension. 2011;58:225–231.
    1. Machowska A, Carrero JJ, Lindholm B, Stenvinkel P. Therapeutics targeting persistent inflammation in chronic kidney disease. Transl Res. 2016;167:204–213.
    1. Zhang MH, Pan MM, Ni HF, et al. [Effect of Cordyceps sinensis powder on renal oxidative stress and mitochondria functions in 5/6 nephrectomized rats]. Zhongguo Zhong Xi Yi Jie He Za Zhi. 2015;35:443–449. in Chinese.
    1. Chokhandre MK, Mahmoud MI, Hakami T, et al. Vitamin D & its analogues in type 2 diabetic nephropathy: a systematic review. J Diabetes Metab Disord. 2015;14:58.
    1. Liu MC, Lee YW, Lee PT, et al. Cyclophilin A is associated with peripheral artery disease and chronic kidney disease in geriatrics: the Tianliao Old People (TOP) study. Sci Rep. 2015;5:9937.
    1. Yazdi PG, Moradi H, Yang JY, et al. Skeletal muscle mitochondrial depletion and dysfunction in chronic kidney disease. Int J Clin Exp Med. 2013;6:532539.
    1. Uribarri J, He JC. The low AGE diet: a neglected aspect of clinical nephrology practice? Nephron. 2015;130:48–53.
    1. Egorova AD, Van der Heiden K, Van de Pas S, et al. Tgfβ/Alk5 signaling is required for shear stress induced klf2 expression in embryonic endothelial cells. Dev Dyn. 2011;240:1670–1680.
    1. Moonen JA, Lee ES, Schmidt M, et al. Endothelial-to-mesenchymal transition contributes to fibro-proliferative vascular disease and is modulated by fluid shear stress. Cardiovasc Res. 2015;108:377–386.
    1. Barros AF, Borges NA, Ferreira DC, et al. Is there interaction between gut microbial profile and cardiovascular risk in chronic kidney disease patients? Future Microbiol. 2015;10:517–526.
    1. Textor SC, Lerman LO. Paradigm shifts in atherosclerotic renovascular disease: where are we now? J Am Soc Nephrol. 2015;26:2074–2080.
    1. Chen NX, O'Neill KD, Allen MR, et al. Low bone turnover in chronic kidney disease is associated with decreased VEGF-A expression and osteoblast differentiation. Am J Nephrol. 2015;41:464–473.
    1. Wu X, Guan Y, Yan J, et al. ShenKang injection suppresses kidney fibrosis and oxidative stress via transforming growth factor-β/Smad3 signalling pathway in vivo and in vitro. J Pharm Pharmacol. 2015;67:1054–1065.
    1. van Hinsbergh VW. Endothelium—role in regulation of coagulation and inflammation. Semin Immunopathol. 2012;34:93–106.
    1. Rios DR, Carvalho M, Lwaleed BA, et al. Hemostatic changes in patients with end stage renal disease undergoing hemodialysis. Clin Chim Acta. 2010;411:135–139.
    1. Turitto VT, Hall CL. Mechanical factors affecting hemostasis and thrombosis. Thromb Res. 1998;92(suppl 2):S25–S31.
    1. Corti R, Hutter R, Badimon JJ, Fuster V. Evolving concepts in the triad of atherosclerosis, inflammation and thrombosis. J Thromb Thrombolysis. 2004;17:35–44.
    1. Serrano A, Garcia F, Serrano M, et al. IgA antibodies against β2 glycoprotein I in hemodialysis patients are an independent risk factor for mortality. Kidney Int. 2012;81:1239–1244.
    1. Fischer MJ, Rauch J, Levine JS. The antiphospholipid syndrome. Semin Nephrol. 2007;27:35–46.
    1. Costa E, Rocha S, Rocha-Pereira P, et al. Cross-talk between inflammation, coagulation/fibrinolysis and vascular access in hemodialysis patients. J Vasc Access. 2008;9:248–253.
    1. Erdem Y, Haznedaroglu IC, Celik I, et al. Coagulation, fibrinolysis and fibrinolysis inhibitors in haemodialysis patients: contribution of arteriovenous fistula. Nephrol Dial Transplant. 1996;11:1299–1305.
    1. Milburn JA, Ford I, Cassar K, et al. Platelet activation, coagulation activation and C-reactive protein in simultaneous samples from the vascular access and peripheral veins of haemodialysis patients. Int J Lab Hematol. 2012;34:52–58.
    1. Milburn JA, Ford I, Mutch NJ, et al. Thrombin-Anti-thrombin levels and patency of arterio-venous fistula in patients undergoing haemodialysis compared to healthy volunteers: a prospective analysis. PLoS One. 2013;8:e67799.
    1. Smits JH, van der Linden J, Blankestijn PJ, Rabelink TJ. Coagulation and haemodialysis access thrombosis. Nephrol Dial Transplant. 2000;15:1755–1760.
    1. Chien CT, Fan SC, Lin SC, et al. Glucagon-like peptide-1 receptor agonist activation ameliorates venous thrombosis-induced arteriovenous fistula failure in chronic kidney disease. Thromb Haemost. 2014;112:1051–1064.
    1. Martinovic Z, Basic-Jukic N, Pavlovic DB, Kes P. Importance of platelet aggregation in patients with end-stage renal disease. Acta Clin Croat. 2013;52:472–477.
    1. Klamroth R, Orlovic M, Fritsche I, et al. The influence of thrombophilic risk factors on vascular access survival in chronic dialysis patients in a retrospective evaluation. Vasa. 2013;42:32–39.
    1. Rios DR, Fernandes AP, Carvalho MG, et al. Hemodialysis vascular access thrombosis: the role of factor V Leiden, prothrombin gene mutation and ABO blood groups. Clin Chim Acta. 2011;412:425–429.
    1. Jamshid R, Reza SA, Abbas G, Raha A. Incidence of arteriovenous thrombosis and the role of anticardiolipin antibodies in hemodialysis patients. Int Urol Nephrol. 2003;35:275–282.
    1. Salmela B, Hartman J, Peltonen S, et al. Thrombophilia and arteriovenous fistula survival in ESRD. Clin J Am Soc Nephrol. 2013;8:962–968.
    1. Knoll GA, Wells PS, Young D, et al. Thrombophilia and the risk for hemodialysis vascular access thrombosis. J Am Soc Nephrol. 2005;16:1108–1114.
    1. Verschuren JJW, Ocak G, Dekker FW, et al. Candidate gene analysis of arteriovenous fistula failure in hemodialysis patients. Clin J Am Soc Nephrol. 2013;8:1358–1366.
    1. Allon M, Zhang L, Maya ID, et al. Association of factor V Leiden gene polymorphism with arteriovenous graft failure. Am J Kidney Dis. 2012;59:682–688.
    1. Serati AR, Roozbeh J, Sagheb MM. Serum LDL levels are a major prognostic factor for arteriovenous fistula thrombosis (AVFT) in hemodialysis patients. J Vasc Access. 2007;8:109–114.
    1. Milburn JA, Cassar K, Ford I, et al. Prothrombotic changes in platelet, endothelial and coagulation function following hemodialysis. Int J Artif Organs. 2011;34:280–287.
    1. Bartels PC, Schoorl M, Schoorl M, et al. Activation of coagulation during treatment with haemodialysis. Scand J Clin Lab Invest. 2000;60:283–290.
    1. Hadhri S, Rejeb MB, Belarbia A, et al. Hemodialysis duration, human platelet antigen HPA-3 and IgA isotype of anti-β2glycoprotein I antibodies are associated with native arteriovenous fistula failure in Tunisian hemodialysis patients. Thromb Res. 2013;131:e202–e209.
    1. Wu JH, Zhang DW, Cheng XL, et al. Platelet glycoprotein IIb HPA-3 a/b polymorphism is associated with native arteriovenous fistula thrombosis in chronic hemodialysis patients. Ren Fail. 2012;34:960–963.
    1. Pisoni R, Barker-Finkel J, Allo M. Statin therapy is not associated with improved vascular access outcomes. Clin J Am Soc Nephrol. 2010;5:1447–1450.
    1. Janardhanan R, Yang B, Vohra P, et al. Simvastatin reduces venous stenosis formation in a murine hemodialysis vascular access model. Kidney Int. 2013;84:338–352.
    1. Birch N, Fillaus J, Florescu MC. The effect of statin therapy on the formation of arteriovenous fistula stenoses and the rate of reoccurrence of previously treated stenoses. Hemodial Int. 2013;17:586–593.
    1. Florescu MC, Birch N. Statin therapy and hemodialysis vascular access—were we bringing a knife to a gunfight and were hoping to win? Semin Dial. 2012;25:700–702.
    1. Sigovan M, Rayz V, Gasper W, et al. Vascular remodeling in autogenous arterio-venous fistulas by MRI and CFD. Ann Biomed Eng. 2013;41:657–668.
    1. Kwon H, Choi JY, Ko HK, et al. Comparison of surgical and endovascular salvage procedures for juxta-anastomotic stenosis in autogenous wrist radiocephalic arteriovenous fistula. Ann Vasc Surg. 2014;28:1840–1846.

Source: PubMed

3
Abonneren