Next Article in Journal
Implications for Self-Management among African Caribbean Adults with Noncommunicable Diseases and Mental Health Disorders: A Systematic Review
Next Article in Special Issue
New Insights into Molecular Mechanisms of Chronic Kidney Disease
Previous Article in Journal
Quantitative Proteomics Indicate Radical Removal of Non-Small Cell Lung Cancer and Predict Outcome
Previous Article in Special Issue
Stage II of Chronic Kidney Disease—A Tipping Point in Disease Progression?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tissue Factor, Thrombosis, and Chronic Kidney Disease

1
Division of Nephrology, Rheumatology, and Endocrinology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
2
Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA 92161, USA
3
VA San Diego Healthcare System, San Diego, CA 92161, USA
4
Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences, Sendai 980-8578, Japan
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(11), 2737; https://doi.org/10.3390/biomedicines10112737
Submission received: 9 September 2022 / Revised: 20 October 2022 / Accepted: 24 October 2022 / Published: 28 October 2022
(This article belongs to the Special Issue Pathophysiology of Chronic Kidney Disease and Its Complications)

Abstract

:
Coagulation abnormalities are common in chronic kidney disease (CKD). Tissue factor (TF, factor III) is a master regulator of the extrinsic coagulation system, activating downstream coagulation proteases, such as factor Xa and thrombin, and promoting fibrin formation. TF and coagulation proteases also activate protease-activated receptors (PARs) and are implicated in various organ injuries. Recent studies have shown the mechanisms by which thrombotic tendency is increased under CKD-specific conditions. Uremic toxins, such as indoxyl sulfate and kynurenine, are accumulated in CKD and activate TF and coagulation; in addition, the TF–coagulation protease–PAR pathway enhances inflammation and fibrosis, thereby exacerbating renal injury. Herein, we review the recent research studies to understand the role of TF in increasing the thrombotic risk and CKD progression.

1. Introduction

The number of patients with chronic kidney disease (CKD) has been increasing globally. The worldwide prevalence of CKD increased by approximately 29.3% between 1997 and 2017 [1]. CKD often results in renal replacement therapy and exacerbates the risk of stroke and cardiovascular events; therefore, patients with CKD have a poor prognosis and reduced quality of life [2,3,4].
Coagulation abnormalities are, generally, observed in patients with CKD and their risk of thrombosis is high [5]. The expression of markers associated with coagulation activities, such as von Willebrand factor, fibrinogen, factor VII (FVII), factor VIII, and D-dimer, is elevated in patients with CKD or those undergoing hemodialysis [6,7,8,9]. Patients undergoing dialysis have a higher mortality rate for pulmonary embolism, atherothrombosis, myocardial infarction, and stroke [10,11]. Reports have suggested that the risk of venous thromboembolism (VTE) increases by approximately 1.5–1.7 fold in patients with CKD and reduced renal function, such as those with stages 3 and 4 [12,13,14]. Another study showed an independent association of low estimated glomerular filtration rate (eGFR; <45 mL/min/1.73 m2) with VTE risk [15].
Tissue factor (TF, factor III) regulates the extrinsic coagulation system. TF dysregulation is linked to abnormal hemostasis and increases the risk of thrombotic events [16]. Typically, inflammatory mediators, such as cytokines, oxidative stress, and oxidized low-density lipoprotein, elevate TF levels, but recent studies have revealed the relationship between CKD-specific pathogenesis, including uremic toxins, and regulation of TF activity [17,18,19,20]. Therefore, new mechanisms underlying thrombosis development in patients with CKD are attracting increasing attention.
In addition to the pro-coagulant ability of TF, coagulation proteases, such as activated factor VII (FVIIa), factor X (FXa), and thrombin, which are located downstream of TF, activate protease-activated receptors (PARs) [21]. Signaling through PARs increases inflammation and kidney injury [21,22,23,24]; therefore, TF is a risk factor for thrombosis as well as an aggravating factor for renal injury.
Herein, we review recent findings related to TF and coagulation in the pathogenesis of CKD to understand the role of TF in increasing thrombotic risk and CKD progression.

2. TF and the Extrinsic Coagulation System

TF is a transmembrane protein with a molecular weight of 47 kDa and an initiator of the extrinsic coagulation system. It binds to FVII to convert FVIIa. The TF/FVIIa complex catalyzes the activation of FX and factor XI (FXI). FXa and activated cofactor V (FVa) form a prothrombin complex, which generates thrombin. Finally, thrombin converts fibrinogen to fibrin to form thrombi [16].
To prevent abnormal activation of coagulation, TF is not normally expressed in cells exposed to blood, such as endothelial cells or monocytes, but is highly expressed in subendothelial cells, including vascular smooth muscle cells (VSMCs) [16,25]; however, under pathological conditions, such as inflammatory diseases and dyslipidemia, activator protein 1 and nuclear factor-κB (NF-κB) increase TF gene expression in monocytes and endothelial cells, and this activates the extrinsic pathway, resulting in a thrombotic tendency [16,26,27].

3. Expression of TF in the Kidneys

In addition to the cells that make up blood cells and blood vessels, the expression of TF in kidney-derived cells has been previously characterized. In a study using human biopsy, the presence of immunoreactive TF was detected in podocytes and parietal epithelial cells [28,29]. Another study revealed that the level of TF in podocytes was increased via NF-κB signaling under hypoxic conditions [30]. In other kidney-derived cells, TF expression could be induced under pathological conditions; the expression of mesangial TF was increased in models of mesangial proliferative glomerulonephritis [31,32]. In addition, in vitro studies have shown that advanced glycation end products and elevated glucose levels upregulate TF expression in human mesangial cells [33]. Renal tubular cells also express TF, and the expression has been reported to be upregulated in models of unilateral ureteral obstruction and cisplatin-induced nephrotoxicity [34,35]. In vitro studies, various stimuli, including lipopolysaccharide, high glucose, and agonists of PARs, induce TF expression in renal tubular cells [36,37,38].

4. Relationship between Uremic Toxins and TF

Uremic toxins are harmful metabolites that accumulate in the body of patients with CKD. More than 100 uremic toxins have been identified thus far [39], and the pathological roles of protein-bound uremic toxins, such as indoxyl sulfate, have been actively studied. Accumulated uremic toxins are involved in CKD progression and complications, including cardiac dysfunction, sarcopenia, and cognitive function [19,40,41]. Notably, the relationship between the hemostasis system and uremic toxins in CKD pathogenesis has recently received considerable attention [17,18,19,20] (Figure 1).

4.1. Indoxyl Sulfate (IS) and Indole-3-Acetic Acid (IAA)

Indolic uremic solutes, such as IS and IAA, are tryptophan-derived uremic toxins. Indole is derived from the metabolism of tryptophan by intestinal bacterial tryptophanase and is absorbed through the intestine and metabolized to IS by cytochrome P450 2E1 and sulfotransferase in the liver. Similarly, IAA is metabolized from tryptophan by the gut microbiota [42,43,44].
IS and IAA have been reported to increase TF expression in endothelial cells, monocytes, and VSMCs [45,46,47,48]; moreover, indolic uremic solutes have been reported to induce the release of endothelial microparticles with TF pro-coagulant activity [46,49]. Mechanistically, indolic uremic solutes are ligands of the transcription factor aryl hydrocarbon receptor (AHR) [50]. Activated AHR translocates into the nucleus and regulates the expression of various genes, including that of TF. It has been reported that small interfering RNA or AHR inhibitors reduce the IS- and IAA-induced upregulation of TF expression in human endothelial cells and peripheral blood mononuclear cells [46]. Another study showed that IAA upregulated TF expression in human umbilical endothelial cells via the AHR/p38 mitogen-activated protein kinase (MAPK)/NF-κB pathway [51]. In addition, we demonstrated that IS and IAA induced TF expression in monocytic THP-1 cells, and this was suppressed by a MAPK inhibitor [47]. In vitro studies using VSMCs have revealed different mechanisms underlying the regulation of TF expression. Uremic serum, IS, and IAA upregulate TF levels by decreasing TF ubiquitination and increasing TF stability in VSMCs [52]. The IS–AHR pathway reduces the interaction between TF and the ubiquitin ligase STIP1 homology and U-box-containing protein 1 (STUB1), resulting in the inhibition of TF ubiquitination in VSMCs [48,53]. The IS-AHR axis may also induce TF expression by downregulating the Mas receptor expression or upregulating the pro-renin receptor expression in VSMCs [54,55]. The procoagulant effect of IS has also been demonstrated in animal studies; IS exposure increased arterial thrombotic tendency in rats [56,57] and mice [48], and this was suppressed by AHR inhibition.

4.2. Kynurenine (Kyn)

The Kyn pathway, another route of tryptophan metabolism, is catalyzed by the enzymes tryptophan dioxygenase (TDO) in the liver and indoleamine 2,3-dioxygenases (IDOs), which are inducible enzymes under pathological conditions, such as inflammation [58]. Kyn and its degradation products accumulate as a result of impaired renal function and play crucial roles in thrombotic activity during CKD pathogenesis [17,18,59]. In a previous study, it was demonstrated that the levels of plasma TF and other pro-coagulant markers were correlated with metabolites related to the Kyn pathway in patients undergoing dialysis or conservative treatment [60,61]. A study revealed that Kyn increased pro-coagulant TF activity in human VSMCs and arterial thrombosis formation in a carotid artery ferric chloride (FeCl3) injury model [62]. In addition, Kyn-induced TF expression and thrombotic activity are suppressed by AHR inhibition [62]. It has also been demonstrated that inhibition of IDO1 expression reduced blood Kyn levels, accompanied by a reduction in TF levels and thrombotic activity in a CKD model [63]. Moreover, the prothrombotic effect of Kyn on endothelial cells and venous thrombosis models via AHR has also been demonstrated [64]. Therefore, the Kyn–AHR–TF pathway is involved in the thrombotic tendency observed in patients with CKD.

4.3. p-Cresyl Sulfate (PCS)

PCS originates from phenolic compounds, such as p-cresol, which are converted from tyrosine and phenylalanine via bacterial fermentation [65]. Animal and human studies have demonstrated PCS to be associated with vascular dysfunction and cardiovascular events in CKD [66,67]; however, the effect of PCS on the induction of TF expression is still unclear. In vitro studies have shown that PCS fails to upregulate TF expression in human endothelial cells and monocytes [45,47]. In contrast, PCS increases the release of endothelial microparticles associated with thrombogenicity [68].

4.4. Trimethylamine N-Oxide (TMAO)

Trimethylamine (TMA) is generated by the gut microbiota from foods containing nutrients, such as choline, carnitine, and phosphatidylcholine. TMAO is biosynthesized from absorbed TMA by hepatic flavin-containing monooxygenases-3 in the liver [43]. Human studies have demonstrated that TMAO accumulates in the blood as renal function declines and that higher blood levels of TMAO are associated with CKD progression, CVD events, and mortality [69,70,71,72]. Prothrombotic roles of TMAO have been addressed in the literature; TMAO induces TF expression in human microvascular endothelial cells in a dose-dependent manner. An increase in aortic TF expression by TMAO was observed in choline-supplemented mice. In addition, TMAO enhances arterial thrombosis, which is inhibited by a TF-inhibitory antibody or a mechanism-based microbial choline TMA-lyase inhibitor in mouse models of arterial injury [72]. Another study showed that TMAO increased TF expression and activity via NF-κB signaling in vascular endothelial cells. In addition, TMAO and pro-inflammatory molecules, such as tumor necrosis factor α or high mobility group box 1 protein, synergistically increase TF expression in endothelial cells, suggesting its prothrombotic roles under inflammatory conditions [73].

5. Elevated Uremic Toxin-TF Axis and Risk of Thrombosis in CKD

A human study showed that blood TF levels were elevated with lower eGFR in patients with CKD and healthy controls [74]. In addition, blood TF levels were closely correlated with the levels of uremic toxins, such as IS and IAA, in patients with CKD [46,51]. Therefore, the uremic solute-TF axis, which increases with renal impairment, plays a crucial role in the thrombotic events in CKD. A previous study showed that a high serum IS level is associated with dialysis graft thrombosis after endovascular interventions [75]. Moreover, the Dialysis Access Consortium Clopidogrel Prevention of Early AV Fistula Thrombosis trial (DAC-Fistula) examined thrombotic complications after hemodialysis arteriovenous fistula creation. Participants with subsequent arteriovenous thrombosis had significantly higher levels of indoxyl sulfate and Kyn, and increased AHR and TF activity compared to those without thrombosis [62]. Similarly, subgroup analysis of the Thrombolysis in Myocardial Infarction II trial (TIMI-II trial), in which patients underwent percutaneous transluminal coronary angioplasty for myocardial infarction with ST-segment elevation, was performed. The thrombotic events in patients with milder CKD (stages 2 and 3) were associated with increased blood Kyn levels and AHR and TF activities [62].

6. Targeting Uremic Toxins for the Prevention of TF Activity and Thrombotic Risk

6.1. Role of Gut Microbiota in Reducing Uremic Toxins

Dysbiosis and alternation of the gut microbiota are observed in patients with CKD. For example, there is a decrease in the number of Lactobacillus groups, which are considered good bacteria, and an increase in the bacterial flora that contain urease, uricase, and tryptophanase (for example, that accelerates the production of toxins, such as indole or p-cresol) [76,77]. Dysbiosis has been suggested to be associated with the progression of renal damage as well as with cardiovascular complications [71,78,79]. Based on the relationship between TF and uremic toxins, intervention in the gut microbiota may help reduce uremic toxins and the risk of thrombosis.
Probiotics and prebiotics are typical therapies targeting intestinal bacterial function and composition. The former replenishes live microorganisms that have health benefits by improving and restoring intestinal microflora. The latter provides non-digestible compounds in foods that stimulate the growth and activity of beneficial bacteria, such as Bifidobacteria and Lactobacilli [80]. The impact of these approaches of the gut microbiota intervention on uremic toxins in patients with CKD has been demonstrated. Although the number of cases analyzed was small, probiotics reduced serum uremic toxin levels, such as IAA-O-glucuronide, in patients undergoing dialysis [81]. Another randomized study revealed that probiotic treatment for 24 weeks resulted in a reduction in plasma IS levels in patients undergoing dialysis [82]. These could be new therapies targeting uremic toxins; however, their effects on TF and coagulation activity, as well as on the risk of thrombosis, are unknown and require further investigation.

6.2. AST-120: An Oral Adsorbent of Uremic Toxin

AST-120 (Kremezin®) is an orally available adsorbent composed of porous carbon microspheres. These particles adsorb uremic toxin precursors in the intestinal lumen, allowing them to be expelled with stool [83]; thus, studies have shown that AST-120 ameliorates the accumulation of blood uremic toxins in patients with CKD [84,85] and in rat CKD models [86]. In addition, this drug reduces the accumulation of IS and PCS in several organs, including the kidney, skeletal muscle, and brain [87]. Whether the suppression of uremic toxins by AST-120 can reduce renal failure has also been examined; however, randomized controlled trials failed to demonstrate a treatment effect on the progression of renal impairment [88,89,90]. With regard to CVD complications, a basic study showed that AST-120 inhibited the progression of atherosclerosis in apolipoprotein E-deficient mice that underwent subtotal nephrectomy, suggesting an inhibitory effect on CVD events [91,92]. Whether the reduction in uremic toxins by AST-120 has the potential to reduce TF expression and coagulation is interesting and deserves further investigation.

7. Hypoxia and TF in CKD

Hypoxia is involved in the pathogenesis of CKD. Cortical hypoxia has been detected using blood oxygenation level-dependent magnetic resonance imaging in patients with CKD [93] or a rodent model of diabetic kidney disease (DKD) [94]. Hypoxia is sensed by hypoxia-inducible factor (HIF), a master regulator of oxygen homeostasis, that regulates the expression of several genes to adapt to hypoxic conditions [95]. Under normal conditions, HIF-α, which is prolyl hydroxylated by the proline hydroxylase domain, is recognized by the von Hippel-Lindau E3 ubiquitin ligase complex and undergoes ubiquitination and proteasomal degradation; however, during hypoxic conditions, hydroxylation of HIF-α is inhibited, and it translocates to the nucleus, dimerizes with HIF-β, binds to hypoxia response elements, and induces transcription of hypoxia-related target genes [95]. The pathological roles of HIF in CKD progression have been actively studied, and its protective and harmful roles have been demonstrated [96].
Hypoxia enhances blood coagulation and thrombotic events [97]. HIF has been shown to regulate the expression of factors in the coagulation-fibrinolytic system as a transcription factor. Induction of TF expression by HIF1α has been reported in cancer-related thrombosis [98]. It has also been shown that HIF2α suppresses tissue factor pathway inhibitor (TFPI) and exacerbates thrombosis [99], and that HIF1α/HIF2α may upregulate plasminogen activator inhibitor-1 expression and suppress the fibrinolytic system [100]. These responses to hypoxia exacerbate thrombosis; in vivo level analyses have also shown that thrombosis is exacerbated in rodent models reared under hypoxic conditions [101,102].
In recent years, hypoxia-inducible factor prolyl hydroxylase (HIF-PH) inhibitors have become popular treatments for renal anemia. This drug improves renal anemia by stabilizing HIF and inducing downstream erythropoietin (EPO) expression. Its therapeutic effects are comparable to those of conventional EPO-stimulating agents [103,104]. One side effect of concern with HIF-PH inhibitors is the risk of thrombosis; a recent meta-analysis including 30 studies comprising 13,146 patients reported an increased risk of thrombosis by HIF-PH inhibitors compared to conventional therapy (risk ratio of 1.31, 95% CI 1.05 to 1.63) [105]. The causes of thrombosis are diverse and involve not only elevated coagulation factors but also platelet activation and excessive elevation in hemoglobin levels. As noted above, the activation of HIF induces TF expression, which may contribute to the risk of thrombosis associated with this treatment. There are still many unknowns regarding these factors, and further analysis is required.

8. Targeting the Coagulation System Reduces Kidney Injury in CKD

In addition to its prothrombotic effects, there is a great interest in whether TF is involved in kidney injury in CKD. Coagulation proteases downstream of TF, such as FXa and thrombin, activate PARs. PARs are members of the G protein-coupled receptor superfamily, and four PAR proteins (PAR1-4) have been identified. PAR is activated when its N-terminus is cleaved by proteases and the new N-terminus acts as a ligand to transmit signals. Each PAR is activated by a specific coagulation protease. The TF-FVIIa complex activates PAR2, ternary TF-FVIIa-FXa complex targets PAR1 and PAR2, and thrombin targets PAR1, PAR3, and PAR4 [21,106]. Signaling through PARs activates NF-κB or MAPK, which increases cytokine/chemokine or pro-fibrotic mediators, thereby exacerbating inflammatory and fibrotic diseases, including CKD [21,23]. Anti-inflammatory effects via suppression of PARs by factor Xa or thrombin inhibitors, such as rivaroxaban and dabigatran, have also been reported [21]. TF, coagulation proteases, and PARs could be involved in CKD progression, and their roles in CKD have been examined primarily in animal studies (Figure 2).

8.1. Relationship among TF, eNOS, and Inflammation in DKD

DKD is a major etiology of CKD and is associated with increased TF expression and hypercoagulability [23]. Animal studies have shown that renal TF expression is increased in type 1 diabetic models, such as the streptozotocin model [107,108]. In addition, our series of studies demonstrated a relationship between TF, endothelial nitric oxide synthase (eNOS), and DKD [23,109,110,111,112]. eNOS is an endothelium-derived NO with vasorelaxant and anti-inflammatory properties [113,114]. Genetic polymorphisms of eNOS are associated with the prognosis of DKD [115] and eNOS-deficient diabetic mice exhibit severe glomerulosclerosis and massive proteinuria as a model of human advanced diabetic nephropathy [116]. Using Akita diabetic mice with various eNOS expressions (eNOS+/+, eNOS+/−, and eNOS−/−), we demonstrated that diabetic kidney injury worsened in response to reduced eNOS levels. Renal TF expression and activity are correlated with decreased eNOS levels and severe kidney injury [109]. In another study, we demonstrated that renal TF expression was elevated when a high-fat diet and eNOS deficiency were combined [111]. These findings indicate a crucial role of eNOS and dyslipidemia in hypercoagulability under diabetic conditions. A causal link between TF and diabetic kidney injury has also been demonstrated; short-term administration of anti-TF neutralizing antibodies reduces inflammatory markers in the kidneys of diabetic mice lacking eNOS [111]. We also tested the roles of FXa and PAR2 downstream of TF in eNOS-deficient mice [110], and showed that the administration of a FXa inhibitor improved nephrosclerosis and inflammatory markers in diabetic Akita mice lacking eNOS. Similarly, PAR2 deficiency showed therapeutic effects in the same model. Another study showed that PAR1 and PAR2 inhibitors improved kidney injury in Akita mice with reduced eNOS expression and that the combination of these inhibitors additively reduced albuminuria and glomerulosclerosis with a reduction in inflammation and fibrosis markers [112].

8.2. Deletion of Myeloid TF Reduces Kidney Injury in Adenine-Induced CKD Model

CKD progression is characterized by the loss of renal cells and accumulation of the extracellular matrix. Adenine-induced nephropathy causes damage to the tubular epithelium, resulting in inflammation and interstitial fibrosis; therefore, it is widely used as a model of CKD [117]. Adenine-induced CKD is associated with an increase in TF expression in the kidney and a prothrombotic state [118]. We demonstrated the role of monocyte/macrophage TF in this model; conditional knockout of myeloid TF attenuated histological damage, including tubular atrophy, and the expression of pro-inflammatory cytokines in the kidneys of mice with adenine-induced renal fibrosis [47]. Further studies have also shown an inhibitory effect of factors downstream of TF on CKD, as FXa inhibitors have been shown to improve renal fibrosis in models of unilateral ureteral obstruction (UUO) and 5/6 subtotal nephrectomy [119,120]. Dabigatran, a direct thrombin inhibitor, ameliorated the epithelial–mesenchymal transition in the UUO model [121]. In addition, studies have shown that pharmacological inhibition or deletion of PAR1 and PAR2 protects against tubular injury in renal fibrosis models, including UUO, and adenine treatment [122,123,124,125,126]. Therefore, the TF-dependent coagulation and PARs pathways are involved in inflammation and pro-fibrotic response, which are hallmarks of CKD pathogenesis.

8.3. Role of TF in Podocyte Injury

Podocytes and glomerular epithelial cells play crucial roles in the glomerular filtration barrier [127]. Podocyte injury is involved in the pathogenesis of many forms of glomerulopathy, including focal segmental glomerulosclerosis, DKD, and IgA nephropathy, and podocyte loss is an important factor in CKD progression [128,129]. As described above, TF is expressed in human podocytes. In addition, fibrin deposition in renal biopsy specimens is widely observed in glomerular disorders, suggesting a pathological role for intraglomerular coagulation in CKD [130]. An in vitro study using human immortalized podocytes demonstrated the harmful effects of TF in hypoxic podocyte injury. TF knockdown prevented the decrease in CD2-associated protein levels and actin reorganization under hypoxic conditions, suggesting that TF promotes reorganization of the actin cytoskeleton in podocytes and their injury [30]. In contrast, a unique role of TF in podocyte injury was shown; mice lacking the cytoplasmic domain of TF exhibited increased proteinuria under normal conditions or when nephritis was induced, accompanied by a reduction in the number of glomerular podocytes. An electron microscopy study demonstrated abnormal podocyte morphology due to a deficiency in cytoplasmic TF [131]. These findings suggest that the cytoplasmic domain of TF may contribute to the structural integrity of podocytes.

9. Conclusions and Future Perspectives

This review outlines two possible effects of TF in patients with CKD, namely, the risk of thrombosis and the mechanism of renal damage, suggesting that TF is a promising target for the prognosis of CKD. Inhibition of abnormally activated TF remains a challenge, and reducing uremic toxin levels using probiotics or AST-120 can be used as a treatment option; however, this finding requires further validation in clinical studies. Recently, AHR inhibitors have been developed and are expected to be applied to cancer therapy [132]. They may help to prevent the risk of thrombosis caused by uremic toxins. Moreover, the cell-specific effects of TF in renal disease remain elusive. For example, conflicting effects of TF on podocyte damage have been reported. The mechanisms by which podocyte- and tubule-derived TF or the cytoplasmic domain of TF affect renal injury need to be determined to facilitate the use of TF as a therapeutic target for CKD.

Author Contributions

Y.O. writing—original draft preparation, N.T. writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Gonryo Medical Foundation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Collaboration, G.C.K.D. Global, regional, and national burden of chronic kidney disease, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2020, 395, 709–733. [Google Scholar] [CrossRef] [Green Version]
  2. Go, A.S.; Chertow, G.M.; Fan, D.; McCulloch, C.E.; Hsu, C.Y. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N. Engl. J. Med. 2004, 351, 1296–1305. [Google Scholar] [CrossRef]
  3. Chen, T.K.; Knicely, D.H.; Grams, M.E. Chronic Kidney Disease Diagnosis and Management: A Review. JAMA 2019, 322, 1294–1304. [Google Scholar] [CrossRef]
  4. Gansevoort, R.T.; Correa-Rotter, R.; Hemmelgarn, B.R.; Jafar, T.H.; Heerspink, H.J.; Mann, J.F.; Matsushita, K.; Wen, C.P. Chronic kidney disease and cardiovascular risk: Epidemiology, mechanisms, and prevention. Lancet 2013, 382, 339–352. [Google Scholar] [CrossRef]
  5. Lutz, J.; Menke, J.; Sollinger, D.; Schinzel, H.; Thürmel, K. Haemostasis in chronic kidney disease. Nephrol. Dial. Transpl. 2014, 29, 29–40. [Google Scholar] [CrossRef] [Green Version]
  6. Huang, M.J.; Wei, R.B.; Wang, Y.; Su, T.Y.; Di, P.; Li, Q.P.; Yang, X.; Li, P.; Chen, X.M. Blood coagulation system in patients with chronic kidney disease: A prospective observational study. BMJ Open 2017, 7, e014294. [Google Scholar] [CrossRef] [Green Version]
  7. Dubin, R.; Cushman, M.; Folsom, A.R.; Fried, L.F.; Palmas, W.; Peralta, C.A.; Wassel, C.; Shlipak, M.G. Kidney function and multiple hemostatic markers: Cross sectional associations in the multi-ethnic study of atherosclerosis. BMC Nephrol. 2011, 12, 3. [Google Scholar] [CrossRef] [Green Version]
  8. Tran, L.; Pannier, B.; Lacolley, P.; Serrato, T.; Benetos, A.; London, G.M.; Bézie, Y.; Regnault, V. A case-control study indicates that coagulation imbalance is associated with arteriosclerosis and markers of endothelial dysfunction in kidney failure. Kidney Int. 2021, 99, 1162–1172. [Google Scholar] [CrossRef]
  9. Kamiński, T.W.; Pawlak, K.; Karbowska, M.; Myśliwiec, M.; Pawlak, D. Indoxyl sulfate—The uremic toxin linking hemostatic system disturbances with the prevalence of cardiovascular disease in patients with chronic kidney disease. BMC Nephrol. 2017, 18, 35. [Google Scholar] [CrossRef] [Green Version]
  10. Ocak, G.; van Stralen, K.J.; Rosendaal, F.R.; Verduijn, M.; Ravani, P.; Palsson, R.; Leivestad, T.; Hoitsma, A.J.; Ferrer-Alamar, M.; Finne, P.; et al. Mortality due to pulmonary embolism, myocardial infarction, and stroke among incident dialysis patients. J. Thromb. Haemost. 2012, 10, 2484–2493. [Google Scholar] [CrossRef]
  11. Ocak, G.; Noordzij, M.; Rookmaaker, M.B.; Cases, A.; Couchoud, C.; Heaf, J.G.; Jarraya, F.; De Meester, J.; Groothoff, J.W.; Waldum-Grevbo, B.E.; et al. Mortality due to bleeding, myocardial infarction and stroke in dialysis patients. J. Thromb. Haemost. 2018, 16, 1953–1963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Wattanakit, K.; Cushman, M.; Stehman-Breen, C.; Heckbert, S.R.; Folsom, A.R. Chronic kidney disease increases risk for venous thromboembolism. J. Am. Soc. Nephrol. 2008, 19, 135–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Folsom, A.R.; Lutsey, P.L.; Astor, B.C.; Wattanakit, K.; Heckbert, S.R.; Cushman, M.; Study, A.R.i.C. Chronic kidney disease and venous thromboembolism: A prospective study. Nephrol. Dial. Transpl. 2010, 25, 3296–3301. [Google Scholar] [CrossRef] [PubMed]
  14. Mahmoodi, B.K.; Gansevoort, R.T.; Næss, I.A.; Lutsey, P.L.; Brækkan, S.K.; Veeger, N.J.; Brodin, E.E.; Meijer, K.; Sang, Y.; Matsushita, K.; et al. Association of mild to moderate chronic kidney disease with venous thromboembolism: Pooled analysis of five prospective general population cohorts. Circulation 2012, 126, 1964–1971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Cheung, K.L.; Zakai, N.A.; Folsom, A.R.; Kurella Tamura, M.; Peralta, C.A.; Judd, S.E.; Callas, P.W.; Cushman, M. Measures of Kidney Disease and the Risk of Venous Thromboembolism in the REGARDS (Reasons for Geographic and Racial Differences in Stroke) Study. Am. J. Kidney Dis. 2017, 70, 182–190. [Google Scholar] [CrossRef]
  16. Grover, S.P.; Mackman, N. Tissue Factor: An Essential Mediator of Hemostasis and Trigger of Thrombosis. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 709–725. [Google Scholar] [CrossRef] [Green Version]
  17. Addi, T.; Dou, L.; Burtey, S. Tryptophan-Derived Uremic Toxins and Thrombosis in Chronic Kidney Disease. Toxins 2018, 10, 412. [Google Scholar] [CrossRef] [Green Version]
  18. Fryc, J.; Naumnik, B. Thrombolome and Its Emerging Role in Chronic Kidney Diseases. Toxins 2021, 13, 223. [Google Scholar] [CrossRef]
  19. Ravid, J.D.; Kamel, M.H.; Chitalia, V.C. Uraemic solutes as therapeutic targets in CKD-associated cardiovascular disease. Nat. Rev. Nephrol. 2021, 17, 402–416. [Google Scholar] [CrossRef]
  20. Shashar, M.; Francis, J.; Chitalia, V. Thrombosis in the uremic milieu--emerging role of “thrombolome”. Semin. Dial. 2015, 28, 198–205. [Google Scholar] [CrossRef]
  21. Posma, J.J.; Grover, S.P.; Hisada, Y.; Owens, A.P.; Antoniak, S.; Spronk, H.M.; Mackman, N. Roles of Coagulation Proteases and PARs (Protease-Activated Receptors) in Mouse Models of Inflammatory Diseases. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 13–24. [Google Scholar] [CrossRef] [PubMed]
  22. Palygin, O.; Ilatovskaya, D.V.; Staruschenko, A. Protease-activated receptors in kidney disease progression. Am. J. Physiol. Renal. Physiol. 2016, 311, F1140–F1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Oe, Y.; Miyazaki, M.; Takahashi, N. Coagulation, Protease-Activated Receptors, and Diabetic Kidney Disease: Lessons from eNOS-Deficient Mice. Tohoku J. Exp. Med. 2021, 255, 1–8. [Google Scholar] [CrossRef] [PubMed]
  24. Madhusudhan, T.; Kerlin, B.A.; Isermann, B. The emerging role of coagulation proteases in kidney disease. Nat. Rev. Nephrol. 2016, 12, 94–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Østerud, B.; Bjørklid, E. Sources of tissue factor. Semin. Thromb. Hemost. 2006, 32, 11–23. [Google Scholar] [CrossRef]
  26. Samad, F.; Ruf, W. Inflammation, obesity, and thrombosis. Blood 2013, 122, 3415–3422. [Google Scholar] [CrossRef]
  27. Owens, A.P.; Passam, F.H.; Antoniak, S.; Marshall, S.M.; McDaniel, A.L.; Rudel, L.; Williams, J.C.; Hubbard, B.K.; Dutton, J.A.; Wang, J.; et al. Monocyte tissue factor-dependent activation of coagulation in hypercholesterolemic mice and monkeys is inhibited by simvastatin. J. Clin. Investig. 2012, 122, 558–568. [Google Scholar] [CrossRef] [Green Version]
  28. Flössel, C.; Luther, T.; Müller, M.; Albrecht, S.; Kasper, M. Immunohistochemical detection of tissue factor (TF) on paraffin sections of routinely fixed human tissue. Histochemistry 1994, 101, 449–453. [Google Scholar] [CrossRef]
  29. Osterholm, C.; Veress, B.; Simanaitis, M.; Hedner, U.; Ekberg, H. Differential expression of tissue factor (TF) in calcineurin inhibitor-induced nephrotoxicity and rejection--implications for development of a possible diagnostic marker. Transpl. Immunol. 2005, 15, 165–172. [Google Scholar] [CrossRef] [Green Version]
  30. Narita, I.; Shimada, M.; Yamabe, H.; Kinjo, T.; Tanno, T.; Nishizaki, K.; Kawai, M.; Nakamura, M.; Murakami, R.; Nakamura, N.; et al. NF-κB-dependent increase in tissue factor expression is responsible for hypoxic podocyte injury. Clin. Exp. Nephrol. 2016, 20, 679–688. [Google Scholar] [CrossRef]
  31. Shimosawa, M.; Sakamoto, K.; Tomari, Y.; Kamikado, K.; Otsuka, H.; Liu, N.; Kitamura, H.; Uemura, K.; Nogaki, F.; Mori, N.; et al. Lipopolysaccharide-triggered acute aggravation of mesangioproliferative glomerulonephritis through activation of coagulation in a high IgA strain of ddY mice. Nephron Exp. Nephrol. 2009, 112, e81–e91. [Google Scholar] [CrossRef] [PubMed]
  32. Nomura, K.; Liu, N.; Nagai, K.; Hasegawa, T.; Kobayashi, I.; Nogaki, F.; Tanaka, M.; Arai, H.; Fukatsu, A.; Kita, T.; et al. Roles of coagulation pathway and factor Xa in rat mesangioproliferative glomerulonephritis. Lab. Investig. 2007, 87, 150–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ettelaie, C.; Su, S.; Li, C.; Collier, M.E. Tissue factor-containing microparticles released from mesangial cells in response to high glucose and AGE induce tube formation in microvascular cells. Microvasc. Res. 2008, 76, 152–160. [Google Scholar] [CrossRef] [PubMed]
  34. Wendt, T.; Zhang, Y.M.; Bierhaus, A.; Kriegsmann, J.; Deng, Y.; Waldherr, R.; Teske, T.; Luther, T.; Fünfstück, R.; Nawroth, P.P. Tissue factor expression in an animal model of hydronephrosis. Nephrol. Dial. Transpl. 1995, 10, 1820–1828. [Google Scholar]
  35. Watanabe, M.; Oe, Y.; Sato, E.; Sekimoto, A.; Sato, H.; Ito, S.; Takahashi, N. Protease-activated receptor 2 exacerbates cisplatin-induced nephrotoxicity. Am. J. Physiol. Renal. Physiol. 2019, 316, F654–F659. [Google Scholar] [CrossRef] [PubMed]
  36. Lwaleed, B.A.; Vayro, S.; Racusen, L.C.; Cooper, A.J. Tissue factor expression by a human kidney proximal tubular cell line in vitro: A model relevant to urinary tissue factor secretion in disease? J. Clin. Pathol. 2007, 60, 762–767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Sommeijer, D.W.; Hansen, H.R.; van Oerle, R.; Hamulyak, K.; van Zanten, A.P.; Meesters, E.; Spronk, H.M.; ten Cate, H. Soluble tissue factor is a candidate marker for progression of microvascular disease in patients with Type 2 diabetes. J. Thromb. Haemost. 2006, 4, 574–580. [Google Scholar] [CrossRef]
  38. Iyer, A.; Humphries, T.L.R.; Owens, E.P.; Zhao, K.N.; Masci, P.P.; Johnson, D.W.; Nikolic-Paterson, D.; Gobe, G.C.; Fairlie, D.P.; Vesey, D.A. PAR2 Activation on Human Kidney Tubular Epithelial Cells Induces Tissue Factor Synthesis, That Enhances Blood Clotting. Front. Physiol. 2021, 12, 615428. [Google Scholar] [CrossRef]
  39. Duranton, F.; Cohen, G.; De Smet, R.; Rodriguez, M.; Jankowski, J.; Vanholder, R.; Argiles, A.; Group, E.U.T.W. Normal and pathologic concentrations of uremic toxins. J. Am. Soc. Nephrol. 2012, 23, 1258–1270. [Google Scholar] [CrossRef] [Green Version]
  40. Liabeuf, S.; Pepin, M.; Franssen, C.F.M.; Viggiano, D.; Carriazo, S.; Gansevoort, R.T.; Gesualdo, L.; Hafez, G.; Malyszko, J.; Mayer, C.; et al. Chronic kidney disease and neurological disorders: Are uraemic toxins the missing piece of the puzzle? Nephrol. Dial. Transpl. 2021, 37, ii33–ii44. [Google Scholar] [CrossRef]
  41. Chalupsky, M.; Goodson, D.A.; Gamboa, J.L.; Roshanravan, B. New insights into muscle function in chronic kidney disease and metabolic acidosis. Curr. Opin. Nephrol. Hypertens. 2021, 30, 369–376. [Google Scholar] [CrossRef] [PubMed]
  42. Hubbard, T.D.; Murray, I.A.; Perdew, G.H. Indole and Tryptophan Metabolism: Endogenous and Dietary Routes to Ah Receptor Activation. Drug Metab. Dispos. 2015, 43, 1522–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Fernandez-Prado, R.; Esteras, R.; Perez-Gomez, M.V.; Gracia-Iguacel, C.; Gonzalez-Parra, E.; Sanz, A.B.; Ortiz, A.; Sanchez-Niño, M.D. Nutrients Turned into Toxins: Microbiota Modulation of Nutrient Properties in Chronic Kidney Disease. Nutrients 2017, 9, 489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Vanholder, R.; Nigam, S.K.; Burtey, S.; Glorieux, G. What If Not All Metabolites from the Uremic Toxin Generating Pathways Are Toxic? A Hypothesis. Toxins 2022, 14, 221. [Google Scholar] [CrossRef] [PubMed]
  45. Oe, Y.; Sato, E.; Sato, H.; Miyazaki, M.; Ito, S.; Takahashi, N. Uremic toxins alter coagulation and fibrinolysis-related genes expression in human endothelial cells. Thromb. Res. 2020, 186, 75–77. [Google Scholar] [CrossRef]
  46. Gondouin, B.; Cerini, C.; Dou, L.; Sallée, M.; Duval-Sabatier, A.; Pletinck, A.; Calaf, R.; Lacroix, R.; Jourde-Chiche, N.; Poitevin, S.; et al. Indolic uremic solutes increase tissue factor production in endothelial cells by the aryl hydrocarbon receptor pathway. Kidney Int. 2013, 84, 733–744. [Google Scholar] [CrossRef] [Green Version]
  47. Yamakage, S.; Oe, Y.; Sato, E.; Okamoto, K.; Sekimoto, A.; Kumakura, S.; Sato, H.; Yoshida, M.; Nagasawa, T.; Miyazaki, M.; et al. Myeloid cell-derived coagulation tissue factor is associated with renal tubular damage in mice fed an adenine diet. Sci. Rep. 2021, 11, 12159. [Google Scholar] [CrossRef]
  48. Shashar, M.; Belghasem, M.E.; Matsuura, S.; Walker, J.; Richards, S.; Alousi, F.; Rijal, K.; Kolachalama, V.B.; Balcells, M.; Odagi, M.; et al. Targeting STUB1-tissue factor axis normalizes hyperthrombotic uremic phenotype without increasing bleeding risk. Sci. Transl. Med. 2017, 9. [Google Scholar] [CrossRef] [Green Version]
  49. Faure, V.; Dou, L.; Sabatier, F.; Cerini, C.; Sampol, J.; Berland, Y.; Brunet, P.; Dignat-George, F. Elevation of circulating endothelial microparticles in patients with chronic renal failure. J. Thromb. Haemost. 2006, 4, 566–573. [Google Scholar] [CrossRef]
  50. Schroeder, J.C.; Dinatale, B.C.; Murray, I.A.; Flaveny, C.A.; Liu, Q.; Laurenzana, E.M.; Lin, J.M.; Strom, S.C.; Omiecinski, C.J.; Amin, S.; et al. The uremic toxin 3-indoxyl sulfate is a potent endogenous agonist for the human aryl hydrocarbon receptor. Biochemistry 2010, 49, 393–400. [Google Scholar] [CrossRef] [Green Version]
  51. Addi, T.; Poitevin, S.; McKay, N.; El Mecherfi, K.E.; Kheroua, O.; Jourde-Chiche, N.; de Macedo, A.; Gondouin, B.; Cerini, C.; Brunet, P.; et al. Mechanisms of tissue factor induction by the uremic toxin indole-3 acetic acid through aryl hydrocarbon receptor/nuclear factor-kappa B signaling pathway in human endothelial cells. Arch. Toxicol. 2019, 93, 121–136. [Google Scholar] [CrossRef]
  52. Chitalia, V.C.; Shivanna, S.; Martorell, J.; Balcells, M.; Bosch, I.; Kolandaivelu, K.; Edelman, E.R. Uremic serum and solutes increase post-vascular interventional thrombotic risk through altered stability of smooth muscle cell tissue factor. Circulation 2013, 127, 365–376. [Google Scholar] [CrossRef]
  53. Shivanna, S.; Kolandaivelu, K.; Shashar, M.; Belghasim, M.; Al-Rabadi, L.; Balcells, M.; Zhang, A.; Weinberg, J.; Francis, J.; Pollastri, M.P.; et al. The Aryl Hydrocarbon Receptor is a Critical Regulator of Tissue Factor Stability and an Antithrombotic Target in Uremia. J. Am. Soc. Nephrol. 2016, 27, 189–201. [Google Scholar] [CrossRef] [Green Version]
  54. Ng, H.Y.; Bolati, W.; Lee, C.T.; Chien, Y.S.; Yisireyili, M.; Saito, S.; Pei, S.N.; Nishijima, F.; Niwa, T. Indoxyl Sulfate Downregulates Mas Receptor via Aryl Hydrocarbon Receptor/Nuclear Factor-kappa B, and Induces Cell Proliferation and Tissue Factor Expression in Vascular Smooth Muscle Cells. Nephron 2016, 133, 205–212. [Google Scholar] [CrossRef]
  55. Yisireyili, M.; Saito, S.; Abudureyimu, S.; Adelibieke, Y.; Ng, H.Y.; Nishijima, F.; Takeshita, K.; Murohara, T.; Niwa, T. Indoxyl sulfate-induced activation of (pro)renin receptor promotes cell proliferation and tissue factor expression in vascular smooth muscle cells. PLoS ONE 2014, 9, e109268. [Google Scholar] [CrossRef] [Green Version]
  56. Karbowska, M.; Kaminski, T.W.; Znorko, B.; Domaniewski, T.; Misztal, T.; Rusak, T.; Pryczynicz, A.; Guzinska-Ustymowicz, K.; Pawlak, K.; Pawlak, D. Indoxyl Sulfate Promotes Arterial Thrombosis in Rat Model via Increased Levels of Complex TF/VII, PAI-1, Platelet Activation as Well as Decreased Contents of SIRT1 and SIRT3. Front. Physiol. 2018, 9, 1623. [Google Scholar] [CrossRef] [Green Version]
  57. Karbowska, M.; Kaminski, T.W.; Marcinczyk, N.; Misztal, T.; Rusak, T.; Smyk, L.; Pawlak, D. The Uremic Toxin Indoxyl Sulfate Accelerates Thrombotic Response after Vascular Injury in Animal Models. Toxins 2017, 9, 229. [Google Scholar] [CrossRef] [Green Version]
  58. Wang, Q.; Liu, D.; Song, P.; Zou, M.H. Tryptophan-kynurenine pathway is dysregulated in inflammation, and immune activation. Front. Biosci. (Landmark Ed.) 2015, 20, 1116–1143. [Google Scholar] [CrossRef] [Green Version]
  59. Zakrocka, I.; Załuska, W. Kynurenine pathway in kidney diseases. Pharmacol. Rep. 2022, 74, 27–39. [Google Scholar] [CrossRef]
  60. Pawlak, K.; Tankiewicz, J.; Mysliwiec, M.; Pawlak, D. Tissue factor/its pathway inhibitor system and kynurenines in chronic kidney disease patients on conservative treatment. Blood Coagul. Fibrinolysis. 2009, 20, 590–594. [Google Scholar] [CrossRef]
  61. Pawlak, K.; Mysliwiec, M.; Pawlak, D. Hypercoagulability is independently associated with kynurenine pathway activation in dialysed uraemic patients. Thromb. Haemost. 2009, 102, 49–55. [Google Scholar] [CrossRef] [PubMed]
  62. Kolachalama, V.B.; Shashar, M.; Alousi, F.; Shivanna, S.; Rijal, K.; Belghasem, M.E.; Walker, J.; Matsuura, S.; Chang, G.H.; Gibson, C.M.; et al. Uremic Solute-Aryl Hydrocarbon Receptor-Tissue Factor Axis Associates with Thrombosis after Vascular Injury in Humans. J. Am. Soc. Nephrol. 2018, 29, 1063–1072. [Google Scholar] [CrossRef] [Green Version]
  63. Walker, J.A.; Richards, S.; Whelan, S.A.; Yoo, S.B.; Russell, T.L.; Arinze, N.; Lotfollahzadeh, S.; Napoleon, M.A.; Belghasem, M.; Lee, N.; et al. Indoleamine 2,3-dioxygenase-1, a Novel Therapeutic Target for Post-Vascular Injury Thrombosis in CKD. J. Am. Soc. Nephrol. 2021, 32, 2834–2850. [Google Scholar] [CrossRef]
  64. Belghasem, M.; Roth, D.; Richards, S.; Napolene, M.A.; Walker, J.; Yin, W.; Arinze, N.; Lyle, C.; Spencer, C.; Francis, J.M.; et al. Metabolites in a mouse cancer model enhance venous thrombogenicity through the aryl hydrocarbon receptor-tissue factor axis. Blood 2019, 134, 2399–2413. [Google Scholar] [CrossRef] [PubMed]
  65. Gryp, T.; Vanholder, R.; Vaneechoutte, M.; Glorieux, G. p-Cresyl Sulfate. Toxins 2017, 9, 52. [Google Scholar] [CrossRef] [Green Version]
  66. Meijers, B.K.; Claes, K.; Bammens, B.; de Loor, H.; Viaene, L.; Verbeke, K.; Kuypers, D.; Vanrenterghem, Y.; Evenepoel, P. p-Cresol and cardiovascular risk in mild-to-moderate kidney disease. Clin. J. Am. Soc. Nephrol. 2010, 5, 1182–1189. [Google Scholar] [CrossRef] [Green Version]
  67. Opdebeeck, B.; Maudsley, S.; Azmi, A.; De Maré, A.; De Leger, W.; Meijers, B.; Verhulst, A.; Evenepoel, P.; D’Haese, P.C.; Neven, E. Indoxyl Sulfate and p-Cresyl Sulfate Promote Vascular Calcification and Associate with Glucose Intolerance. J. Am. Soc. Nephrol. 2019, 30, 751–766. [Google Scholar] [CrossRef]
  68. Meijers, B.K.; Van Kerckhoven, S.; Verbeke, K.; Dehaen, W.; Vanrenterghem, Y.; Hoylaerts, M.F.; Evenepoel, P. The uremic retention solute p-cresyl sulfate and markers of endothelial damage. Am. J. Kidney Dis. 2009, 54, 891–901. [Google Scholar] [CrossRef]
  69. Missailidis, C.; Hällqvist, J.; Qureshi, A.R.; Barany, P.; Heimbürger, O.; Lindholm, B.; Stenvinkel, P.; Bergman, P. Serum Trimethylamine-N-Oxide Is Strongly Related to Renal Function and Predicts Outcome in Chronic Kidney Disease. PLoS ONE 2016, 11, e0141738. [Google Scholar] [CrossRef] [Green Version]
  70. Pelletier, C.C.; Croyal, M.; Ene, L.; Aguesse, A.; Billon-Crossouard, S.; Krempf, M.; Lemoine, S.; Guebre-Egziabher, F.; Juillard, L.; Soulage, C.O. Elevation of Trimethylamine-N-Oxide in Chronic Kidney Disease: Contribution of Decreased Glomerular Filtration Rate. Toxins 2019, 11, 635. [Google Scholar] [CrossRef] [Green Version]
  71. Tang, W.H.; Wang, Z.; Kennedy, D.J.; Wu, Y.; Buffa, J.A.; Agatisa-Boyle, B.; Li, X.S.; Levison, B.S.; Hazen, S.L. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ. Res. 2015, 116, 448–455. [Google Scholar] [CrossRef] [PubMed]
  72. Witkowski, M.; Friebel, J.; Buffa, J.A.; Li, X.S.; Wang, Z.; Sangwan, N.; Li, L.; DiDonato, J.A.; Tizian, C.; Haghikia, A.; et al. Vascular endothelial Tissue Factor contributes to trimethylamine N-oxide-enhanced arterial thrombosis. Cardiovasc. Res. 2021, 118, 2367–2384. [Google Scholar] [CrossRef] [PubMed]
  73. Cheng, X.; Qiu, X.; Liu, Y.; Yuan, C.; Yang, X. Trimethylamine N-oxide promotes tissue factor expression and activity in vascular endothelial cells: A new link between trimethylamine N-oxide and atherosclerotic thrombosis. Thromb. Res. 2019, 177, 110–116. [Google Scholar] [CrossRef] [PubMed]
  74. Mercier, E.; Branger, B.; Vecina, F.; Al-Sabadani, B.; Berlan, J.; Dauzat, M.; Fourcade, J.; Gris, J.C. Tissue factor coagulation pathway and blood cells activation state in renal insufficiency. Hematol. J. 2001, 2, 18–25. [Google Scholar] [CrossRef]
  75. Wu, C.C.; Hsieh, M.Y.; Hung, S.C.; Kuo, K.L.; Tsai, T.H.; Lai, C.L.; Chen, J.W.; Lin, S.J.; Huang, P.H.; Tarng, D.C. Serum Indoxyl Sulfate Associates with Postangioplasty Thrombosis of Dialysis Grafts. J. Am. Soc. Nephrol. 2016, 27, 1254–1264. [Google Scholar] [CrossRef] [Green Version]
  76. Vaziri, N.D.; Wong, J.; Pahl, M.; Piceno, Y.M.; Yuan, J.; DeSantis, T.Z.; Ni, Z.; Nguyen, T.H.; Andersen, G.L. Chronic kidney disease alters intestinal microbial flora. Kidney Int. 2013, 83, 308–315. [Google Scholar] [CrossRef] [Green Version]
  77. Wong, J.; Piceno, Y.M.; DeSantis, T.Z.; Pahl, M.; Andersen, G.L.; Vaziri, N.D. Expansion of urease- and uricase-containing, indole- and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD. Am. J. Nephrol. 2014, 39, 230–237. [Google Scholar] [CrossRef] [Green Version]
  78. Ramezani, A.; Raj, D.S. The gut microbiome, kidney disease, and targeted interventions. J. Am. Soc. Nephrol. 2014, 25, 657–670. [Google Scholar] [CrossRef] [Green Version]
  79. Meijers, B.; Evenepoel, P.; Anders, H.J. Intestinal microbiome and fitness in kidney disease. Nat. Rev. Nephrol. 2019, 15, 531–545. [Google Scholar] [CrossRef]
  80. Markowiak, P.; Śliżewska, K. Effects of Probiotics, Prebiotics, and Synbiotics on Human Health. Nutrients 2017, 9, 1021. [Google Scholar] [CrossRef]
  81. Liu, S.; Liu, H.; Chen, L.; Liang, S.S.; Shi, K.; Meng, W.; Xue, J.; He, Q.; Jiang, H. Effect of probiotics on the intestinal microbiota of hemodialysis patients: A randomized trial. Eur. J. Nutr. 2020, 59, 3755–3766. [Google Scholar] [CrossRef] [PubMed]
  82. Lim, P.S.; Wang, H.F.; Lee, M.C.; Chiu, L.S.; Wu, M.Y.; Chang, W.C.; Wu, T.K. The Efficacy of Lactobacillus-Containing Probiotic Supplementation in Hemodialysis Patients: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Ren. Nutr. 2021, 31, 189–198. [Google Scholar] [CrossRef] [PubMed]
  83. Asai, M.; Kumakura, S.; Kikuchi, M. Review of the efficacy of AST-120 (KREMEZIN). Ren. Fail. 2019, 41, 47–56. [Google Scholar] [CrossRef] [Green Version]
  84. Schulman, G.; Agarwal, R.; Acharya, M.; Berl, T.; Blumenthal, S.; Kopyt, N. A multicenter, randomized, double-blind, placebo-controlled, dose-ranging study of AST-120 (Kremezin) in patients with moderate to severe CKD. Am. J. Kidney Dis. 2006, 47, 565–577. [Google Scholar] [CrossRef] [PubMed]
  85. Niwa, T.; Nomura, T.; Sugiyama, S.; Miyazaki, T.; Tsukushi, S.; Tsutsui, S. The protein metabolite hypothesis, a model for the progression of renal failure: An oral adsorbent lowers indoxyl sulfate levels in undialyzed uremic patients. Kidney Int. Suppl. 1997, 62, S23–S28. [Google Scholar]
  86. Kikuchi, K.; Itoh, Y.; Tateoka, R.; Ezawa, A.; Murakami, K.; Niwa, T. Metabolomic search for uremic toxins as indicators of the effect of an oral sorbent AST-120 by liquid chromatography/tandem mass spectrometry. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2010, 878, 2997–3002. [Google Scholar] [CrossRef]
  87. Sato, E.; Saigusa, D.; Mishima, E.; Uchida, T.; Miura, D.; Morikawa-Ichinose, T.; Kisu, K.; Sekimoto, A.; Saito, R.; Oe, Y.; et al. Impact of the Oral Adsorbent AST-120 on Organ-Specific Accumulation of Uremic Toxins: LC-MS/MS and MS Imaging Techniques. Toxins 2017, 10, 19. [Google Scholar] [CrossRef] [Green Version]
  88. Chen, Y.C.; Wu, M.Y.; Hu, P.J.; Chen, T.T.; Shen, W.C.; Chang, W.C.; Wu, M.S. Effects and Safety of an Oral Adsorbent on Chronic Kidney Disease Progression: A Systematic Review and Meta-Analysis. J. Clin. Med. 2019, 8, 1718. [Google Scholar] [CrossRef] [Green Version]
  89. Schulman, G.; Berl, T.; Beck, G.J.; Remuzzi, G.; Ritz, E.; Arita, K.; Kato, A.; Shimizu, M. Randomized Placebo-Controlled EPPIC Trials of AST-120 in CKD. J. Am. Soc. Nephrol. 2015, 26, 1732–1746. [Google Scholar] [CrossRef] [Green Version]
  90. Cha, R.H.; Kang, S.W.; Park, C.W.; Cha, D.R.; Na, K.Y.; Kim, S.G.; Yoon, S.A.; Han, S.Y.; Chang, J.H.; Park, S.K.; et al. A Randomized, Controlled Trial of Oral Intestinal Sorbent AST-120 on Renal Function Deterioration in Patients with Advanced Renal Dysfunction. Clin. J. Am. Soc. Nephrol. 2016, 11, 559–567. [Google Scholar] [CrossRef] [Green Version]
  91. Nakada, Y.; Onoue, K.; Nakano, T.; Ishihara, S.; Kumazawa, T.; Nakagawa, H.; Ueda, T.; Nishida, T.; Soeda, T.; Okayama, S.; et al. AST-120, an Oral Carbon Absorbent, Protects against the Progression of Atherosclerosis in a Mouse Chronic Renal Failure Model by Preserving sFlt-1 Expression Levels. Sci. Rep. 2019, 9, 15571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Yamamoto, S.; Zuo, Y.; Ma, J.; Yancey, P.G.; Hunley, T.E.; Motojima, M.; Fogo, A.B.; Linton, M.F.; Fazio, S.; Ichikawa, I.; et al. Oral activated charcoal adsorbent (AST-120) ameliorates extent and instability of atherosclerosis accelerated by kidney disease in apolipoprotein E-deficient mice. Nephrol. Dial. Transpl. 2011, 26, 2491–2497. [Google Scholar] [CrossRef] [PubMed]
  93. Pruijm, M.; Milani, B.; Pivin, E.; Podhajska, A.; Vogt, B.; Stuber, M.; Burnier, M. Reduced cortical oxygenation predicts a progressive decline of renal function in patients with chronic kidney disease. Kidney Int. 2018, 93, 932–940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Franzén, S.; Pihl, L.; Khan, N.; Gustafsson, H.; Palm, F. Pronounced kidney hypoxia precedes albuminuria in type 1 diabetic mice. Am. J. Physiol. Renal. Physiol. 2016, 310, F807–F809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Majmundar, A.J.; Wong, W.J.; Simon, M.C. Hypoxia-inducible factors and the response to hypoxic stress. Mol. Cell 2010, 40, 294–309. [Google Scholar] [CrossRef] [Green Version]
  96. Sugahara, M.; Tanaka, T.; Nangaku, M. Hypoxia-Inducible Factor and Oxygen Biology in the Kidney. Kidney360 2020, 1, 1021–1031. [Google Scholar] [CrossRef]
  97. Gupta, N.; Zhao, Y.Y.; Evans, C.E. The stimulation of thrombosis by hypoxia. Thromb. Res. 2019, 181, 77–83. [Google Scholar] [CrossRef]
  98. Sun, L.; Liu, Y.; Lin, S.; Shang, J.; Liu, J.; Li, J.; Yuan, S.; Zhang, L. Early growth response gene-1 and hypoxia-inducible factor-1α affect tumor metastasis via regulation of tissue factor. Acta Oncol. 2013, 52, 842–851. [Google Scholar] [CrossRef]
  99. Stavik, B.; Espada, S.; Cui, X.Y.; Iversen, N.; Holm, S.; Mowinkel, M.C.; Halvorsen, B.; Skretting, G.; Sandset, P.M. EPAS1/HIF-2 alpha-mediated downregulation of tissue factor pathway inhibitor leads to a pro-thrombotic potential in endothelial cells. Biochim. Biophys. Acta 2016, 1862, 670–678. [Google Scholar] [CrossRef]
  100. Ahn, Y.T.; Chua, M.S.; Whitlock, J.P.; Shin, Y.C.; Song, W.H.; Kim, Y.; Eom, C.Y.; An, W.G. Rodent-specific hypoxia response elements enhance PAI-1 expression through HIF-1 or HIF-2 in mouse hepatoma cells. Int. J. Oncol. 2010, 37, 1627–1638. [Google Scholar] [CrossRef]
  101. Gupta, N.; Sahu, A.; Prabhakar, A.; Chatterjee, T.; Tyagi, T.; Kumari, B.; Khan, N.; Nair, V.; Bajaj, N.; Sharma, M.; et al. Activation of NLRP3 inflammasome complex potentiates venous thrombosis in response to hypoxia. Proc. Natl. Acad. Sci. USA 2017, 114, 4763–4768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Lawson, C.A.; Yan, S.D.; Yan, S.F.; Liao, H.; Zhou, Y.S.; Sobel, J.; Kisiel, W.; Stern, D.M.; Pinsky, D.J. Monocytes and tissue factor promote thrombosis in a murine model of oxygen deprivation. J. Clin. Investig. 1997, 99, 1729–1738. [Google Scholar] [CrossRef] [PubMed]
  103. Gupta, N.; Wish, J.B. Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitors: A Potential New Treatment for Anemia in Patients With CKD. Am. J. Kidney Dis. 2017, 69, 815–826. [Google Scholar] [CrossRef] [Green Version]
  104. Kurata, Y.; Tanaka, T.; Nangaku, M. Hypoxia-inducible factor prolyl hydroxylase inhibitor in the treatment of anemia in chronic kidney disease. Curr. Opin. Nephrol. Hypertens. 2020, 29, 414–422. [Google Scholar] [CrossRef] [PubMed]
  105. Chen, H.; Cheng, Q.; Wang, J.; Zhao, X.; Zhu, S. Long-term efficacy and safety of hypoxia-inducible factor prolyl hydroxylase inhibitors in anaemia of chronic kidney disease: A meta-analysis including 13,146 patients. J. Clin. Pharm. Ther. 2021, 46, 999–1009. [Google Scholar] [CrossRef] [PubMed]
  106. Witkowski, M.; Landmesser, U.; Rauch, U. Tissue factor as a link between inflammation and coagulation. Trends Cardiovasc. Med. 2016, 26, 297–303. [Google Scholar] [CrossRef] [PubMed]
  107. Emekli-Alturfan, E.; Kasikci, E.; Yarat, A. Tissue factor activities of streptozotocin induced diabetic rat tissues and the effect of peanut consumption. Diabetes Metab. Res. Rev. 2007, 23, 653–658. [Google Scholar] [CrossRef]
  108. Sommeijer, D.W.; Florquin, S.; Hoedemaker, I.; Timmerman, J.J.; Reitsma, P.H.; Ten Cate, H. Renal tissue factor expression is increased in streptozotocin-induced diabetic mice. Nephron Exp. Nephrol. 2005, 101, e86–e94. [Google Scholar] [CrossRef]
  109. Wang, C.H.; Li, F.; Hiller, S.; Kim, H.S.; Maeda, N.; Smithies, O.; Takahashi, N. A modest decrease in endothelial NOS in mice comparable to that associated with human NOS3 variants exacerbates diabetic nephropathy. Proc. Natl. Acad. Sci. USA 2011, 108, 2070–2075. [Google Scholar] [CrossRef] [Green Version]
  110. Oe, Y.; Hayashi, S.; Fushima, T.; Sato, E.; Kisu, K.; Sato, H.; Ito, S.; Takahashi, N. Coagulation Factor Xa and Protease-Activated Receptor 2 as Novel Therapeutic Targets for Diabetic Nephropathy. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 1525–1533. [Google Scholar] [CrossRef] [Green Version]
  111. Li, F.; Wang, C.H.; Wang, J.G.; Thai, T.; Boysen, G.; Xu, L.; Turner, A.L.; Wolberg, A.S.; Mackman, N.; Maeda, N.; et al. Elevated tissue factor expression contributes to exacerbated diabetic nephropathy in mice lacking eNOS fed a high fat diet. J. Thromb. Haemost. 2010, 8, 2122–2132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Mitsui, S.; Oe, Y.; Sekimoto, A.; Sato, E.; Hashizume, Y.; Yamakage, S.; Kumakura, S.; Sato, H.; Ito, S.; Takahashi, N. Dual blockade of protease-activated receptor 1 and 2 additively ameliorates diabetic kidney disease. Am. J. Physiol. Renal. Physiol. 2020, 318, F1067–F1073. [Google Scholar] [CrossRef] [PubMed]
  113. Passauer, J.; Pistrosch, F.; Büssemaker, E. Nitric oxide in chronic renal failure. Kidney Int. 2005, 67, 1665–1667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Oe, Y.; Mitsui, S.; Sato, E.; Shibata, N.; Kisu, K.; Sekimoto, A.; Miyazaki, M.; Sato, H.; Ito, S.; Takahashi, N. Lack of Endothelial Nitric Oxide Synthase Accelerates Ectopic Calcification in Uremic Mice Fed an Adenine and High Phosphorus Diet. Am. J. Pathol. 2021, 191, 283–293. [Google Scholar] [CrossRef]
  115. Dong, J.; Ping, Y.; Wang, Y.; Zhang, Y. The roles of endothelial nitric oxide synthase gene polymorphisms in diabetes mellitus and its associated vascular complications: A systematic review and meta-analysis. Endocrine 2018, 62, 412–422. [Google Scholar] [CrossRef] [PubMed]
  116. Azushima, K.; Gurley, S.B.; Coffman, T.M. Modelling diabetic nephropathy in mice. Nat. Rev. Nephrol. 2018, 14, 48–56. [Google Scholar] [CrossRef]
  117. Jia, T.; Olauson, H.; Lindberg, K.; Amin, R.; Edvardsson, K.; Lindholm, B.; Andersson, G.; Wernerson, A.; Sabbagh, Y.; Schiavi, S.; et al. A novel model of adenine-induced tubulointerstitial nephropathy in mice. BMC Nephrol. 2013, 14, 116. [Google Scholar] [CrossRef] [Green Version]
  118. Makhloufi, C.; Crescence, L.; Darbousset, R.; McKay, N.; Massy, Z.A.; Dubois, C.; Panicot-Dubois, L.; Burtey, S.; Poitevin, S. Assessment of Thrombotic and Bleeding Tendency in Two Mouse Models of Chronic Kidney Disease: Adenine-Diet and 5/6th Nephrectomy. TH Open 2020, 4, e66–e76. [Google Scholar] [CrossRef] [Green Version]
  119. Horinouchi, Y.; Ikeda, Y.; Fukushima, K.; Imanishi, M.; Hamano, H.; Izawa-Ishizawa, Y.; Zamami, Y.; Takechi, K.; Miyamoto, L.; Fujino, H.; et al. Renoprotective effects of a factor Xa inhibitor: Fusion of basic research and a database analysis. Sci. Rep. 2018, 8, 10858. [Google Scholar] [CrossRef] [Green Version]
  120. Fang, L.; Ohashi, K.; Ogawa, H.; Otaka, N.; Kawanishi, H.; Takikawa, T.; Ozaki, Y.; Takahara, K.; Tatsumi, M.; Takefuji, M.; et al. Factor Xa inhibitor, edoxaban ameliorates renal injury after subtotal nephrectomy by reducing epithelial-mesenchymal transition and inflammatory response. Physiol. Rep. 2022, 10, e15218. [Google Scholar] [CrossRef]
  121. Saifi, M.A.; Annaldas, S.; Godugu, C. A direct thrombin inhibitor, dabigatran etexilate protects from renal fibrosis by inhibiting protease activated receptor-1. Eur. J. Pharmacol. 2021, 893, 173838. [Google Scholar] [CrossRef] [PubMed]
  122. Hayashi, S.; Oe, Y.; Fushima, T.; Sato, E.; Sato, H.; Ito, S.; Takahashi, N. Protease-activated receptor 2 exacerbates adenine-induced renal tubulointerstitial injury in mice. Biochem. Biophys. Res. Commun. 2017, 483, 547–552. [Google Scholar] [CrossRef] [PubMed]
  123. Chung, H.; Ramachandran, R.; Hollenberg, M.D.; Muruve, D.A. Proteinase-activated receptor-2 transactivation of epidermal growth factor receptor and transforming growth factor-β receptor signaling pathways contributes to renal fibrosis. J. Biol. Chem. 2013, 288, 37319–37331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Du, C.; Zhang, T.; Xiao, X.; Shi, Y.; Duan, H.; Ren, Y. Protease-activated receptor-2 promotes kidney tubular epithelial inflammation by inhibiting autophagy via the PI3K/Akt/mTOR signalling pathway. Biochem. J. 2017, 474, 2733–2747. [Google Scholar] [CrossRef]
  125. Lok, S.W.Y.; Yiu, W.H.; Li, H.; Xue, R.; Zou, Y.; Li, B.; Chan, K.W.; Chan, L.Y.Y.; Leung, J.C.K.; Lai, K.N.; et al. The PAR-1 antagonist vorapaxar ameliorates kidney injury and tubulointerstitial fibrosis. Clin. Sci. 2020, 134, 2873–2891. [Google Scholar] [CrossRef]
  126. Ha, S.; Chung, K.W.; Lee, J.; Chung, H.Y.; Moon, H.R. Renal tubular PAR2 promotes interstitial fibrosis by increasing inflammatory responses and EMT process. Arch. Pharm. Res. 2022, 45, 159–173. [Google Scholar] [CrossRef]
  127. Brinkkoetter, P.T.; Ising, C.; Benzing, T. The role of the podocyte in albumin filtration. Nat. Rev. Nephrol. 2013, 9, 328–336. [Google Scholar] [CrossRef]
  128. Leeuwis, J.W.; Nguyen, T.Q.; Dendooven, A.; Kok, R.J.; Goldschmeding, R. Targeting podocyte-associated diseases. Adv. Drug Deliv. Rev. 2010, 62, 1325–1336. [Google Scholar] [CrossRef]
  129. Trimarchi, H.; Coppo, R. Podocytopathy in the mesangial proliferative immunoglobulin A nephropathy: New insights into the mechanisms of damage and progression. Nephrol. Dial. Transpl. 2019, 34, 1280–1285. [Google Scholar] [CrossRef]
  130. Farquhar, A.; MacDonald, M.K.; Ireland, J.T. The role of fibrin deposition in diabetic glomerulosclerosis: A light, electron and immunofluorescence microscopy study. J. Clin. Pathol. 1972, 25, 657–667. [Google Scholar] [CrossRef] [Green Version]
  131. Apostolopoulos, J.; Moussa, L.; Tipping, P.G. The cytoplasmic domain of tissue factor restricts physiological albuminuria and pathological proteinuria associated with glomerulonephritis in mice. Nephron Exp. Nephrol. 2010, 116, e72–e83. [Google Scholar] [CrossRef] [PubMed]
  132. Labadie, B.W.; Bao, R.; Luke, J.J. Reimagining IDO Pathway Inhibition in Cancer Immunotherapy via Downstream Focus on the Tryptophan-Kynurenine-Aryl Hydrocarbon Axis. Clin. Cancer Res. 2019, 25, 1462–1471. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Uremic toxins increase TF in CKD. Kynurenine (Kyn) and related products are synthesized from dietary tryptophan by tryptophan dioxygenase (TDO) and indoleamine 2,3-dioxygenases (IDOs). Tryptophan is also converted to indole by bacterial tryptophanases and metabolized to indoxyl sulfate (IS) and indole-3-acetic acid (IAA). Tryptophan-derived uremic toxins accumulate in the body as renal function declines; they stimulate the aryl hydrocarbon receptor (AHR) and increase TF transcription. AHR also decreases the interaction of TF with the ubiquitin ligase STIP1 homology and U-box-containing protein 1 (STUB1), which inhibits TF ubiquitination and promotes TF stability. Foods containing nutrients, such as choline and carnitine, can be metabolized to trimethylamine (TMA) by the gut microbiota. Trimethylamine N-oxide (TMAO) is biosynthesized from absorbed TMA by hepatic flavin-containing monooxygenase-3 (FMO3). TMAO increases TF expression and activity in endothelial cells (ECs). VSMCs: vascular smooth muscle cells; and CYP2E1: cytochrome P450 2E1.
Figure 1. Uremic toxins increase TF in CKD. Kynurenine (Kyn) and related products are synthesized from dietary tryptophan by tryptophan dioxygenase (TDO) and indoleamine 2,3-dioxygenases (IDOs). Tryptophan is also converted to indole by bacterial tryptophanases and metabolized to indoxyl sulfate (IS) and indole-3-acetic acid (IAA). Tryptophan-derived uremic toxins accumulate in the body as renal function declines; they stimulate the aryl hydrocarbon receptor (AHR) and increase TF transcription. AHR also decreases the interaction of TF with the ubiquitin ligase STIP1 homology and U-box-containing protein 1 (STUB1), which inhibits TF ubiquitination and promotes TF stability. Foods containing nutrients, such as choline and carnitine, can be metabolized to trimethylamine (TMA) by the gut microbiota. Trimethylamine N-oxide (TMAO) is biosynthesized from absorbed TMA by hepatic flavin-containing monooxygenase-3 (FMO3). TMAO increases TF expression and activity in endothelial cells (ECs). VSMCs: vascular smooth muscle cells; and CYP2E1: cytochrome P450 2E1.
Biomedicines 10 02737 g001
Figure 2. TF is involved in CKD progression. TF is activated by a variety of stimuli, including uremic toxins, hyperglycemia, and endothelial damage, such as eNOS dysfunction, under CKD pathogenesis. TF can activate downstream coagulation proteases, such as factor VIIa (FVIIa), factor Xa (FXa), and thrombin, which activate protease-activated receptors (PARs). PARs increase production of pro-inflammatory and pro-fibrotic mediators via NF-κB and MAPK signaling, resulting in kidney injury. TF reduces the expression of slit diaphragm proteins in podocytes and is, possibly, involved in podocyte injury under hypoxia; in contrast, the cytoplasmic domain of TF may contribute to the maintenance of normal podocyte structure and function. MCP1: Monocyte chemoattractant protein-1; PAI1: plasminogen activator inhibitor-1; and TGFβ; transforming growth factor β.
Figure 2. TF is involved in CKD progression. TF is activated by a variety of stimuli, including uremic toxins, hyperglycemia, and endothelial damage, such as eNOS dysfunction, under CKD pathogenesis. TF can activate downstream coagulation proteases, such as factor VIIa (FVIIa), factor Xa (FXa), and thrombin, which activate protease-activated receptors (PARs). PARs increase production of pro-inflammatory and pro-fibrotic mediators via NF-κB and MAPK signaling, resulting in kidney injury. TF reduces the expression of slit diaphragm proteins in podocytes and is, possibly, involved in podocyte injury under hypoxia; in contrast, the cytoplasmic domain of TF may contribute to the maintenance of normal podocyte structure and function. MCP1: Monocyte chemoattractant protein-1; PAI1: plasminogen activator inhibitor-1; and TGFβ; transforming growth factor β.
Biomedicines 10 02737 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Oe, Y.; Takahashi, N. Tissue Factor, Thrombosis, and Chronic Kidney Disease. Biomedicines 2022, 10, 2737. https://doi.org/10.3390/biomedicines10112737

AMA Style

Oe Y, Takahashi N. Tissue Factor, Thrombosis, and Chronic Kidney Disease. Biomedicines. 2022; 10(11):2737. https://doi.org/10.3390/biomedicines10112737

Chicago/Turabian Style

Oe, Yuji, and Nobuyuki Takahashi. 2022. "Tissue Factor, Thrombosis, and Chronic Kidney Disease" Biomedicines 10, no. 11: 2737. https://doi.org/10.3390/biomedicines10112737

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

Article Metrics

Back to TopTop