Next Article in Journal
Nivalenol Mycotoxin Concerns in Foods: An Overview on Occurrence, Impact on Human and Animal Health and Its Detection and Management Strategies
Next Article in Special Issue
Postbiotics and Kidney Disease
Previous Article in Journal
Isolation and Aflatoxin B1-Degradation Characteristics of a Microbacterium proteolyticum B204 Strain from Bovine Faeces
Previous Article in Special Issue
Lipid Profile Is Negatively Associated with Uremic Toxins in Patients with Kidney Failure—A Tri-National Cohort
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Serum Trimethylamine N-Oxide Level Is Associated with Peripheral Arterial Stiffness in Advanced Non-Dialysis Chronic Kidney Disease Patients

1
Division of Nephrology, Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97010, Taiwan
2
School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
3
Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Toxins 2022, 14(8), 526; https://doi.org/10.3390/toxins14080526
Submission received: 29 June 2022 / Revised: 29 July 2022 / Accepted: 29 July 2022 / Published: 31 July 2022
(This article belongs to the Special Issue Kidney Disease-Gut Dysbiosis: What Is the Role of Uremic Toxins?)

Abstract

:
Trimethylamine N-oxide (TMAO) is a gut-derived uremic toxin involved in cardiovascular diseases (CVD). Peripheral arterial stiffness (PAS), measured by the brachial-ankle pulse wave velocity (baPWV) is a valuable indicator of the existence of CVD alongside other diseases. The study recruited 157 patients with chronic kidney disease (CKD) stages 3 to 5, and aimed to determine the correlation between serum TMAO and PAS, defined as a baPWV of >18.0 m/s. Patients with CKD who were diagnosed with PAS (68 patients, 43.3%) were older, had a higher percentage of hypertension or diabetes mellitus, higher systolic blood pressure, and higher fasting glucose, C-reactive protein, and TMAO levels. Furthermore, besides old age and SBP, patients with CKD who had higher serum TMAO were more likely to have PAS, with an odds ratio of 1.016 (95% confidence interval = 1.002–1.029, p = 0.021) by multivariate logistic regression analysis. Correlation analysis demonstrated that serum TMAO was positively correlated with C-reactive protein level and either left or right baPWV. Thus, we supposed that serum TMAO levels were associated with PAS in patients with advanced non-dialysis CKD.
Key Contribution: Serum TMAO was positively associated with baPWV and was indicative of a biomarker for the PAS occurrence in patients with stage 3–5 CKD.

1. Introduction

Patients with chronic kidney disease (CKD) were found to accumulate all kinds of waste products, including noxious metabolic by-products absorbed via the intestines as the kidneys lost their excretion abilities [1]. Existing evidence demonstrated that gut-derived metabolic harmful products could induce adverse effects such as cardiovascular diseases (CVD) and even CKD progression. Indoxyl sulfate and p-cresol sulfate, two well-known protein-bound uremic toxins, were produced from the intestinal bacterial flora, and could be associated with CKD progression and future CVD development in non-dialytic or dialytic CKD [2,3,4]. Similarly, trimethylamine N-oxide (TMAO), also known as another kind of gut microbiota-derived metabolite, originated from trimethylamine [1]. Over the past decade, TMAO had been implicated to have an important role in the occurrence of various diseases, including diabetes mellitus, hypertension, heart failure, inflammation, and most importantly, CKD development [5,6,7,8]. Previous studies reported that TMAO is physiologically filtrated and secreted unchanged by the kidneys and would accumulate as renal function worsened. The high TMAO concentration could, in turn, accelerate kidney dysfunction progression by modulating the progress of tubule-interstitial fibrosis and collagen deposition [9,10,11,12]. An animal study demonstrated that the TMAO of rats that underwent 5/6 nephrectomy could induce vascular oxidative stress and inflammation, resulting in endothelial dysfunction and further CVD [13]. Furthermore, evidence from meta-analysis and systemic review had shown that TMAO correlated with a higher risk of hypertension [14] and all-cause and adverse CV-associated events in the general population [15], geriatric population [16], and patients with CKD [17].
Previous studies have shown that vascular stiffening, as measured by pulse wave velocity (PWV), was correlated with CVD, which was long recognized as the main cause of long-term adverse outcomes in CKD through the possible mechanisms of oxidative stress, low-grade inflammation, or disordered mineral metabolism [18,19,20]. In this study, we measured peripheral arterial stiffness (PAS) by applying a noninvasive modality as brachial-ankle PWV (baPWV), which could be an indicator of CKD progression or mortality, to assess the association with the serum TMAO in patients with non-dialysis advanced CKD [21]. As we had reported, the gut-derived uremic toxins, indoxyl sulfate, and p-cresol sulfate, were correlated with endothelial dysfunction and arterial stiffness [22,23,24], we hypothesized that TMAO could have a potential role in the occurrence of peripheral arterial stiffness.

2. Results

A total of 68 (43.3%) and 89 (56.7%) patients were categorized in the PAS and control groups (Table 1). Compared to the control group, patients in the PAS group were older, had higher fasting glucose, systolic blood pressure (SBP), percentage of DM and HTN, and serum C-reactive protein (CRP) and TMAO levels. Patients with CKD in the PAS group had a lower estimated glomerular filtration rate (eGFR) compared to those in the control group (p = 0.001); however, when classifying patients into CKD stages, no significant difference was observed between those in the PAS and control groups (p = 0.019). Viewed more closely, a trend could be observed showing a higher percentage of CKD stages 4 and 5 in patients belonging to the PAS group compared to the control group (chi-squared test for trend, p = 0.044, data not shown).
We applied multivariate logistic regression analysis on age, HTN, DM, SBP, fasting glucose, eGFR, CRP, and TMAO to analyze possible factors indicative of PAS occurrence. It showed that patients with CKD who were older had higher SBP and serum TMAO, with a 1.081-fold (95% confidence interval [CI] = 1.043–1.121, p < 0.001), 1.042-fold (95% CI = 1.004–1.081, p = 0.031), and 1.016-fold (95% CI = 1.002–1.029, p = 0.021) increased risk of PAS occurrence (Table 2).
To determine the serum TMAO level relative to PAS occurrence, a receiver operating characteristic curve analysis was applied, which showed an optimal serum cut-off level of 46.78 μg/L with 39.71% and 84.27% in sensitivity and specificity, individually. The area under the curve was 0.629 (95% CI = 0.548–0.705, p = 0.0052, Figure 1).
Table 3 shows the correlation between clinical variables and baPWV [left and right] and serum TMAO by Spearman correlation analysis. First, left baPWV was strongly correlated with right baPWV (r = 0.913, p < 0.001). Second, both left and right baPWV were significantly positively correlated with aging, SBP, and serum TMAO. Third, in addition to the positive association with baPWV, serum TMAO level was found to be positively correlated with aging (r = 0.219, p = 0.006) and CRP (r = 0.261, p = 0.001), but negatively associated with eGFR (r = −0.442, p < 0.001).
To examine the potential risk of TAMO related with PAS in CKD patients, a subgroup analysis was performed by dividing patients into groups with or without DM, HTN, and hyperlipidemia (Table 4). The analysis revealed that a higher TMAO value significantly correlated with the occurrence of PAS specifically in CKD patients who were without DM, and those with or without HTN or hyperlipidemia. TMAO in CKD stage 4 and 5 patients had an increasingly independently 1.025-fold (p = 0.046), and 1.036-fold (p = 0.021) higher risk of developing PAS, respectively. It indicated that there was increased possibility of TMAO in relation to the occurrence of PAS in advanced CKD.

3. Discussion

In this study, TMAO associated with aging and high SBP could be a useful marker for the occurrence of PAS in patients with advanced CKD. Furthermore, serum TMAO increased as patients got older, exhibited a greater inflammatory status, or exhibited a decline of renal function.
CVD has been known to be the leading cause of long-term morbidity or mortality in patients with dialytic or non-dialytic CKD [18]. In this study, aging and high SBP were found to be associated with PAS, as indicated by baPWV. As is already known, many traditional and uremia-specific risk factors could lead to irreversible vascular damage with results in high vascular smooth muscle tone and BP, as manifested in arterial stiffness [19,20]. Previous studies had shown that old age and high BP contributed to AS [25,26], which could, in turn, promote further CVD development, renal dysfunction, and even mortality [19,20]. Furthermore, a recent study showed that with old age and SBP, TMAO was also associated with increased PWV in mice as well as in healthy young- to old-aged adults [27].
In addition to the traditional risk factors, such as old age or high BP (SBP or DBP), gut-derived uremic toxins, such as indoxyl sulfate, p-cresol sulfate, and TMAO, were demonstrated to play important roles in CVD pathogenesis [2,3,4]. Through oxidative stress modulation and inflammatory and cytokine responses, which then induced endothelium and smooth muscle cell dysfunctions, these gut-derived uremic toxins were found to have detrimental effects on vascular health [28,29]. Our previous studies also showed that indoxyl sulfate and p-cresol sulfate were related to the occurrence of arterial stiffness in patients with CKD, which highlighted the important pathogenetic role of gut-derived uremic toxins on AS [22,23,24]. Furthermore, TMA, produced from dietary choline, phosphatidylcholine, L-carnitine, and betaine by intestinal bacteria, and then converted to TMAO in the liver, was accumulated in patients with CKD [1]. Besides, serum TMAO was significantly elevated in association with DM, age, and BMI [6], and was positively dose-dependently associated with HTN, as increased levels of per-5 mμmol/L or 10-mμmol/L would increase the risk of HTN by 9% and 20%, respectively [30]. In animal studies, increased dietary TMAO would induce tubule-interstitial fibrosis, collagen deposition, and Smad3 phosphorylation, and then renal function indicated increased serum cystatin C [9]. Another animal study also showed that TMAO-activated inflammasomes release interleukin-1 beta and interleukin-18 to accelerate renal inflammation and fibrosis [10]. Conversely, after targeting TAMO inhibition, it attenuated serum cystatin C levels as well as renal tubule-interstitial fibrosis severity and collagen deposition [11]. In a meta-analysis, patients with advanced CKD had a 67.9 mμmol/L increase in TMAO concentration, whereas patients with CKD had the highest TMAO level of 12.9 mmol/L decreases in eGFR [12]. In regard to renal function, elevated TMAO could only return to normal range after receiving renal transplantation without receiving dialysis [31]. Farhangi et al. found a positively dose-dependent nonlinear relationship between serum TMAO and CRP (the highest category had a 0.27 mg/L increase in CRP compared to the lowest) [8]. Because TMAO was a metabolite derived from gut microbial metabolism and dietary intake, Mishima et al. found that TMAO could originate from both microbiota and dietary components [32]; Costabile et al. found that marine derived diets induce higher TMAO in a healthy population [33]; in a randomized crossover designed study comparing plant-based alternative meat to animal meat, a significant lower level of TMAO was observed in a healthy population on plant-based alternative meat [34]; another study showed a 2-month vegan diet could significantly reduce plasma TMAO in obese or hyperglycemic adults [35]; but a meta-analysis comparing microbiota-driven therapy, including prebiotics, probiotics or synbiotics, with placebo did not reveal a reduction in circulating TMAO [36]. Although we did not record the dietary contents of CKD patients in this study, we still found that, as already shown, there was an inverse relationship between serum TMAO and kidney function and a positive correlation between TMAO and CRP.
Aside from the harmful effects on renal function, many studies reported the TMAO effects on CV and long-term adverse outcomes. Evidence had shown an association between TMAO and severity and mortality in patients with heart failure [5]. Tarng et al. conducted a cohort study on patients with stage 3 to 5 CKD, with a prognostic value of TMAO in predicting 5-year mortality risk [9]. Several meta-analyses showed that TMAO had a dose-dependent effect relationship with increased risk of HTN (the highest category had a 2.36 mmHg increase in SBP compared to the lowest) [14], an increased risk of all-cause mortality and CV events in the general and elderly population [15]. In patients with CKD with the highest TMAO group, a 1.29-fold and 1.45-fold increased risk was observed of all-cause and CV mortality; besides, a 3% higher all-cause mortality per 1 unit increases the TMAO level [17]. Studies have proven that TMAO plays a role in the atherosclerosis process through mediating immune response by recruiting macrophages to aortic lesions with increased CD36, tumor necrotic factor α, and interleukin-6 expressions through the p38 MAPK and JNK 1/2 pathways [37], or activation of NLRP3 inflammasome to cause endothelial dysfunction and inflammation through redox regulation and lysosomal dysfunction [38]. Furthermore, endothelial and smooth muscle cells exposed to TMAO enhanced the recruitment of leukocytes and induced various inflammatory cytokines by activating nuclear factor-kappa β [39]. In 5/6 nephrectomy rats, TMAO with superoxide and proinflammatory cytokines were found to be significantly elevated but with reduced production of endothelial nitric oxide, which together contributed to endothelial dysfunction [13]. Most importantly, TMAO was recently found to modulate the vascular calcification process [40]. Zhang et al. found that TMAO could upregulate osteoblast-specific protein expression, such as Runt-related transcription factor 2 and bone morphogenetic protein-2, to induce calcification by activating nuclear factor-kappa β and NLRP3 inflammasomes [40]. Brunt et al. conducted a study and showed plasma TMAO with aging positively correlated with aortic PWV and SBP in humans and mice; in vitro study additionally revealed the adverse effects of TMAO on intrinsic mechanical stiffness by modulating the formation of advanced glycation end-products and reactive oxygen species [27]. Additionally, this study showed interesting findings that TMAO levels for those CKD patients who did not have DM, regardless of HTN or hyperlipidemia, showed a significantly higher risk for the occurrence of PAS. However, the TMAO of diabetic CKD patients did not correlate with the occurrence of PAS by multivariate logistic regression analysis. This result showed the possible role of DM for influencing the role of TMAO on PAS. Taken together, we reported a positive correlation between serum TMAO level and baPWV in patients with advanced CKD, with a possible mechanism from inflammatory activations and DM influencing the pathogenesis of TMAO on PAS.
This study had several limitations. First, it was a cross-sectional and single-center design with a limited sample size of patients with CKD. Furthermore, patients with stage 1–2 CKD and on dialysis were excluded. Therefore, the results could not be extrapolated to other populations, except for advanced CKD. Second, gut microbiota was not assessed in this study, and the mechanism by which TMAO affects PAS remains unclear. Third, the association between TMAO and PAS (baPWV) was not stronger than age and SBP. Nevertheless, we thought that there could be a role of TMAO in the development of PAS, probably through the activation of inflammatory responses. Fourth, we did not record the dietary contents of these CKD patients. Indeed, plasma TMAO still presents in animals without microbiota [32], and the level directly correlates with the intake of fish, vegetables, and whole-grain products, but not meat, processed meat, and dairy products in humans with high cardiometabolic risk [33]. Therefore, further longitudinal studies with large samples and cells as well as animal studies are needed to clarify these findings.

4. Conclusions

In addition to older age and elevated SBP, increased serum TMAO was independently indicative of PAS in patients with stage 3–5 CKD. Besides the positive correlation with baPWV, serum TMAO was also positively associated with serum CRP levels. These findings suggested that TMAO may be an important upstream modulator of the PAS development process among patients with advanced CKD who are diagnosed with stage 3–5 CKD, but the detailed mechanism remains to be investigated.

5. Materials and Methods

5.1. Patients

From January to December 2016, patients with CKD who were diagnosed with stage 3 to 5 CKD and were old than 18 years with regular follow-up were recruited. CKD was defined using the eGFR by the Chronic Kidney Disease Epidemiology Collaboration equation [18]. Patients were separated into stages 3, 4, and 5 based on their calculated eGFRs of 30–59 mL/min/1.73 m2, 15–29 mL/min/1.73 m2, and <15 mL/min/1.73 m2 [18]. Patients with acute kidney injury, malignancies, acute infectious diseases, congestive heart failure at the time of blood sampling, or who refused to participate in this study were excluded. This study was approved by the Research Ethics Committee of Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation (IRB108-96-B).
The demographic and baseline characteristics regarding the smoking status, DM, HTN, and use of medications were recorded by medical record review.

5.2. Anthropometric and Biochemical Examination

Body height and weight were examined to the nearest 0.5 cm and 0.5 kg and BMI was calculated as weight (kg) divided by height squared (m2). The serum fasting glucose, blood urea nitrogen, creatinine, albumin, total cholesterol, triglyceride, low-density lipoprotein cholesterol, blood urea nitrogen, creatinine, and CRP levels were determined using an autoanalyzer (Siemens Advia 1800; Siemens Healthcare GmbH, Henkestr, Erlangen, Germany) after overnight fasting for at least 8 h.

5.3. Determination of Serum TMAO Levels by High-Performance Liquid Chromatography–Mass Spectrometry

We used a Waters e2695 high-performance liquid chromatography system containing a mass spectrometer (ACQUITY QDa, Waters Corporation, Milford, MA, USA) to determine serum TMAO levels according to a previous study [41]. Mass spectrometry was performed in full scan ranges of 50–450 m/z for positive-ion modes and 100–350 m/z for negative-ion modes, respectively, to monitor the participants’ compounds (TMAO: 76.0 m/z; d9-TMAO: 85.1 m/z). The retention time for TMAO and d9-TMAO was 2.54 min. The procurement and analysis of all examinations were performed using the Empower® 3.0 software (New York, NY, USA).

5.4. Examination of Left and Right Brachial-Ankle PWV

Using this apparatus for volume plethysmography (VaSera VS-1000, Fukuda Denshi Co. Ltd., Tokyo, Japan), patients lay in a supine position with four pneumatic cuffs wrapped around both upper arms and ankles, connected to both plethysmographic and oscillometric sensors to assess baPWV after blood sampling, and rested for 10 min [25,26]. We used baPWV of >18 m/s (either left or right) as a cut-off value to diagnose PAS [25,26].

5.5. Statistical Analysis

The Kolmogorov–Smirnov test was used to examine the normality of continuous variables. The results indicated that these continuous data were expressed as the mean ± standard deviation or median with an interquartile range. To compare the difference of continuous variables between the control and PAS groups, the Student’s independent t-test or Mann–Whitney μ-test were used for the Kolmogorov–Smirnov test, as appropriate. Numbers with percentages and compared between categorical variables were calculated using the chi-squared test. Factors showing significant differences in Table 1 were adopted for multivariate logistic regression analysis, including DM, HTN, age, SBP, fasting glucose, eGFR, CRP, and TMAO, to determine possible factors associated with the occurrence of PAS (which is defined as baPWV as >18.0 m/s). A receiver operating characteristic curve analysis was used to determine the optimal serum TMAO levels indicative of PAS occurrence. The Spearman rank correlation coefficient was used to analyze the correlation between clinical variables and baPWV (left and right) and TMAO. Subgroup analysis by univariate logistic regression analysis was applied to analyze the relation between the occurrence of PAS and risk factors of CKD, including CKD stage, and underlying diseases such as DM, HTN, and hyperlipidemia. The SPSS software for Windows (version 19.0; SPSS, Chicago, IL, USA) was used for all statistical analyses. A p < 0.05 was indicative of statistical significance.

Author Contributions

Conceptualization, B.-G.H.; methodology, J.-P.T. and B.-G.H.; formal analysis, J.-P.T. and B.-G.H.; investigation, C.-H.W., Y.-L.L., Y.-H.L. and B.-G.H.; data curation, C.-H.W., Y.-L.L., Y.-H.L. and B.-G.H.; funding acquisition, B.-G.H.; writing—original draft preparation, J.-P.T.; writing—review and editing, J.-P.T. and B.-G.H.; supervision, B.-G.H. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by grants from Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taiwan, under Grant TCRD108-70.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Research Ethics Committee, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation (IRB108-96-B) on 3 June 2019.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tomlinson, J.A.P.; Wheeler, D.C. The role of trimethylamine N-oxide as a mediator of cardiovascular complications in chronic kidney disease. Kidney Int. 2017, 92, 809–815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Wu, I.W.; Hsu, K.H.; Lee, C.C.; Sun, C.Y.; Hsu, H.J.; Tsai, C.J.; Tzen, C.Y.; Wang, Y.C.; Lin, C.Y.; Wu, M.S. p-Cresyl sulphate and indoxyl sulphate predict progression of chronic kidney disease. Nephrol. Dial. Transpl. 2011, 26, 938–947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lin, C.J.; Liu, H.L.; Pan, C.F.; Chuang, C.K.; Jayakumar, T.; Wang, T.J.; Chen, H.H.; Wu, C.J. Indoxyl sulfate predicts cardiovascular disease and renal function deterioration in advanced chronic kidney disease. Arch. Med. Res. 2012, 43, 451–456. [Google Scholar] [CrossRef]
  4. Barreto, F.C.; Barreto, D.V.; Liabeuf, S.; Meert, N.; Glorieux, G.; Temmar, M.; Choukroun, G.; Vanholder, R.; Massy, Z.A.; European Uremic Toxin Work, G. Serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin. J. Am. Soc. Nephrol. 2009, 4, 1551–1558. [Google Scholar] [CrossRef] [Green Version]
  5. Troseid, M.; Ueland, T.; Hov, J.R.; Svardal, A.; Gregersen, I.; Dahl, C.P.; Aakhus, S.; Gude, E.; Bjorndal, B.; Halvorsen, B.; et al. Microbiota-dependent metabolite trimethylamine-N-oxide is associated with disease severity and survival of patients with chronic heart failure. J. Int. Med. 2015, 277, 717–726. [Google Scholar] [CrossRef] [PubMed]
  6. Dambrova, M.; Latkovskis, G.; Kuka, J.; Strele, I.; Konrade, I.; Grinberga, S.; Hartmane, D.; Pugovics, O.; Erglis, A.; Liepinsh, E. Diabetes is associated with higher trimethylamine N-oxide plasma levels. Exp. Clin. Endocrinol. Diabetes 2016, 124, 251–256. [Google Scholar] [CrossRef] [Green Version]
  7. Rhee, E.P.; Clish, C.B.; Ghorbani, A.; Larson, M.G.; Elmariah, S.; McCabe, E.; Yang, Q.; Cheng, S.; Pierce, K.; Deik, A.; et al. A combined epidemiologic and metabolomic approach improves CKD prediction. J. Am. Soc. Nephrol. 2013, 24, 1330–1338. [Google Scholar] [CrossRef] [Green Version]
  8. Farhangi, M.A.; Vajdi, M. Novel findings of the association between gut microbiota-derived metabolite trimethylamine N-oxide and inflammation: Results from a systematic review and dose-response meta-analysis. Crit. Rev. Food Sci. Nutr. 2020, 60, 2801–2823. [Google Scholar] [CrossRef]
  9. 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] [Green Version]
  10. Fang, Q.; Zheng, B.; Liu, N.; Liu, J.; Liu, W.; Huang, X.; Zeng, X.; Chen, L.; Li, Z.; Ouyang, D. Trimethylamine N-oxide exacerbates renal inflammation and fibrosis in rats with diabetic kidney disease. Front. Physiol. 2021, 12, 682482. [Google Scholar] [CrossRef] [PubMed]
  11. Gupta, N.; Buffa, J.A.; Roberts, A.B.; Sangwan, N.; Skye, S.M.; Li, L.; Ho, K.J.; Varga, J.; DiDonato, J.A.; Tang, W.H.W.; et al. Targeted inhibition of gut microbial trimethylamine N-oxide production reduces renal tubulointerstitial fibrosis and functional impairment in a murine model of chronic kidney disease. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1239–1255. [Google Scholar] [CrossRef]
  12. Zeng, Y.; Guo, M.; Fang, X.; Teng, F.; Tan, X.; Li, X.; Wang, M.; Long, Y.; Xu, Y. Gut microbiota-derived trimethylamine N-oxide and kidney function: A systematic review and meta-analysis. Adv. Nutr. 2021, 12, 1286–1304. [Google Scholar] [CrossRef] [PubMed]
  13. Li, T.; Gua, C.; Wu, B.; Chen, Y. Increased circulating trimethylamine N-oxide contributes to endothelial dysfunction in a rat model of chronic kidney disease. Biochem. Biophys. Res. Commun. 2018, 495, 2071–2077. [Google Scholar] [CrossRef] [PubMed]
  14. Abbasalizad Farhangi, M.; Vajdi, M. Gut microbiota-associated trimethylamine N-oxide and increased cardiometabolic risk in adults: A systematic review and dose-response meta-analysis. Nutr. Rev. 2021, 79, 1022–1042. [Google Scholar] [CrossRef] [PubMed]
  15. Farhangi, M.A. Gut microbiota-dependent trimethylamine N-oxide and all-cause mortality: Findings from an updated systematic review and meta-analysis. Nutrition 2020, 78, 110856. [Google Scholar] [CrossRef] [PubMed]
  16. Chen, G.; He, L.; Dou, X.; Liu, T. Association of trimethylamine-N-oxide levels with risk of cardiovascular disease and mortality among elderly subjects: A systematic review and meta-analysis. Cardiorenal Med. 2022, 12, 39–54. [Google Scholar] [CrossRef]
  17. Zhou, Z.; Jin, H.; Ju, H.; Sun, M.; Chen, H.; Li, L. Circulating trimethylamine-N-oxide and risk of all-cause and cardiovascular mortality in patients with chronic kidney disease: A systematic review and meta-analysis. Front. Med. 2022, 9, 828343. [Google Scholar] [CrossRef]
  18. Stevens, P.E.; Levin, A.; Kidney Disease: Improving Global Outcomes Chronic Kidney Disease Guideline Development Work Group Members. Evaluation and management of chronic kidney disease: Synopsis of the kidney disease: Improving global outcomes 2012 clinical practice guideline. Ann. Intern. Med. 2013, 158, 825–830. [Google Scholar] [CrossRef] [Green Version]
  19. Vlachopoulos, C.; Aznaouridis, K.; Stefanadis, C. Prediction of cardiovascular events and all-cause mortality with arterial stiffness: A systematic review and meta-analysis. J. Am. Coll. Cardiol. 2010, 55, 1318–1327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Karras, A.; Haymann, J.P.; Bozec, E.; Metzger, M.; Jacquot, C.; Maruani, G.; Houillier, P.; Froissart, M.; Stengel, B.; Guardiola, P.; et al. Large artery stiffening and remodeling are independently associated with all-cause mortality and cardiovascular events in chronic kidney disease. Hypertension 2012, 60, 1451–1457. [Google Scholar] [CrossRef] [Green Version]
  21. Chen, S.C.; Chang, J.M.; Liu, W.C.; Tsai, Y.C.; Tsai, J.C.; Hsu, P.C.; Lin, T.H.; Lin, M.Y.; Su, H.M.; Hwang, S.J.; et al. Brachial-ankle pulse wave velocity and rate of renal function decline and mortality in chronic kidney disease. Clin. J. Am. Soc. Nephrol. 2011, 6, 724–732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wang, S.C.; Lai, Y.H.; Liu, C.H.; Wang, C.H.; Hsu, B.G.; Tsai, J.P. Association between serum indoxyl sulfate levels with carotid-femoral pulse wave velocity in patients with chronic kidney disease. Ren. Fail. 2021, 43, 796–802. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, C.H.; Lai, Y.H.; Kuo, C.H.; Lin, Y.L.; Tsai, J.P.; Hsu, B.G. Association between serum indoxyl sulfate levels and endothelial function in non-dialysis chronic kidney disease. Toxins 2019, 11, 589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Lai, Y.H.; Wang, C.H.; Kuo, C.H.; Lin, Y.L.; Tsai, J.P.; Hsu, B.G. Serum p-cresyl sulfate is a predictor of central arterial stiffness in patients on maintenance hemodialysis. Toxins 2019, 12, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Tomiyama, H.; Shiina, K. State of the art review: Brachial-ankle PWV. J. Atheroscler. Thromb. 2020, 27, 621–636. [Google Scholar] [CrossRef] [PubMed]
  26. Tanaka, A.; Tomiyama, H.; Maruhashi, T.; Matsuzawa, Y.; Miyoshi, T.; Kabutoya, T.; Kario, K.; Sugiyama, S.; Munakata, M.; Ito, H.; et al. Physiological diagnostic criteria for vascular failure. Hypertension 2018, 72, 1060–1071. [Google Scholar] [CrossRef] [PubMed]
  27. Brunt, V.E.; Casso, A.G.; Gioscia-Ryan, R.A.; Sapinsley, Z.J.; Ziemba, B.P.; Clayton, Z.S.; Bazzoni, A.E.; VanDongen, N.S.; Richey, J.J.; Hutton, D.A.; et al. Gut microbiome-derived metabolite trimethylamine N-oxide induces aortic stiffening and increases systolic blood pressure with aging in mice and humans. Hypertension 2021, 78, 499–511. [Google Scholar] [CrossRef]
  28. Dou, L.; Jourde-Chiche, N.; Faure, V.; Cerini, C.; Berland, Y.; Dignat-George, F.; Brunet, P. The uremic solute indoxyl sulfate induces oxidative stress in endothelial cells. J. Thromb. Haemos. 2007, 5, 1302–1308. [Google Scholar] [CrossRef]
  29. Sun, C.Y.; Chang, S.C.; Wu, M.S. Uremic toxins induce kidney fibrosis by activating intrarenal renin-angiotensin-aldosterone system associated epithelial-to-mesenchymal transition. PLoS ONE 2012, 7, e34026. [Google Scholar] [CrossRef] [Green Version]
  30. Ge, X.; Zheng, L.; Zhuang, R.; Yu, P.; Xu, Z.; Liu, G.; Xi, X.; Zhou, X.; Fan, H. The gut microbial metabolite trimethylamine N-oxide and hypertension risk: A systematic review and dose-response meta-analysis. Adv. Nutr. 2020, 11, 66–76. [Google Scholar] [CrossRef]
  31. Missailidis, C.; Hallqvist, J.; Qureshi, A.R.; Barany, P.; Heimburger, 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] [PubMed] [Green Version]
  32. Mishima, E.; Fukuda, S.; Mukawa, C.; Yuri, A.; Kanemitsu, Y.; Matsumoto, Y.; Akiyama, Y.; Fukuda, N.N.; Tsukamoto, H.; Asaji, K.; et al. Evaluation of the impact of gut microbiota on uremic solute accumulation by a CE-TOFMS-based metabolomics approach. Kidney Int. 2017, 92, 634–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Costabile, G.; Vetrani, C.; Bozzetto, L.; Giacco, R.; Bresciani, L.; Del Rio, D.; Vitale, M.; Della Pepa, G.; Brighenti, F.; Riccardi, G.; et al. Plasma TMAO increase after healthy diets: Results from 2 randomized controlled trials with dietary fish, polyphenols, and whole-grain cereals. Am. J. Clin. Nutr. 2021, 114, 1342–1350. [Google Scholar] [CrossRef] [PubMed]
  34. Crimarco, A.; Springfield, S.; Petlura, C.; Streaty, T.; Cunanan, K.; Lee, J.; Fielding-Singh, P.; Carter, M.M.; Topf, M.A.; Wastyk, H.C.; et al. A randomized crossover trial on the effect of plant-based compared with animal-based meat on trimethylamine-N-oxide and cardiovascular disease risk factors in generally healthy adults: Study with appetizing plantfood-meat eating alternative trial (SWAP-MEAT). Am. J. Clin. Nutr. 2020, 112, 1188–1199. [Google Scholar]
  35. Argyridou, S.; Davies, M.J.; Biddle, G.J.H.; Bernieh, D.; Suzuki, T.; Dawkins, N.P.; Rowlands, A.V.; Khunti, K.; Smith, A.C.; Yates, T. Evaluation of an 8-week vegan diet on plasma trimethylamine-N-oxide and postchallenge glucose in adults with dysglycemia or obesity. J. Nutr. 2021, 151, 1844–1853. [Google Scholar] [CrossRef] [PubMed]
  36. Miao, L.; Du, J.; Chen, Z.; Shi, D.; Qu, H. Effects of microbiota-driven therapy on circulating trimethylamine-N-oxide metabolism: A Systematic review and meta-analysis. Front. Cardiovasc. Med. 2021, 8, 710567. [Google Scholar] [CrossRef] [PubMed]
  37. Geng, J.; Yang, C.; Wang, B.; Zhang, X.; Hu, T.; Gu, Y.; Li, J. Trimethylamine N-oxide promotes atherosclerosis via CD36-dependent MAPK/JNK pathway. Biomed. Pharmacother. 2018, 97, 941–947. [Google Scholar] [CrossRef]
  38. Boini, K.M.; Hussain, T.; Li, P.L.; Koka, S. Trimethylamine-N-oxide instigates NLRP3 inflammasome activation and endothelial dysfunction. Cell. Physiol. Biochem. 2017, 44, 152–162. [Google Scholar] [CrossRef]
  39. Seldin, M.M.; Meng, Y.; Qi, H.; Zhu, W.; Wang, Z.; Hazen, S.L.; Lusis, A.J.; Shih, D.M. Trimethylamine N-oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear factor-kappaB. J. Am. Heart Assoc. 2016, 5, e002767. [Google Scholar] [CrossRef] [Green Version]
  40. Zhang, X.; Li, Y.; Yang, P.; Liu, X.; Lu, L.; Chen, Y.; Zhong, X.; Li, Z.; Liu, H.; Ou, C.; et al. Trimethylamine-N-Oxide Promotes Vascular Calcification Through Activation of NLRP3 (Nucleotide-Binding Domain, Leucine-Rich-Containing Family, Pyrin Domain-Containing-3) Inflammasome and NF-kappaB (Nuclear Factor kappaB) Signals. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 751–765. [Google Scholar] [CrossRef]
  41. Yang, M.; Zhang, R.; Zhuang, C.; Wu, Y.; Yang, Q.; Yu, Z.; Liu, J.; Zha, B.; Gong, Q.; Yang, B.; et al. Serum trimethylamine N-oxide and the diversity of the intestinal microbial flora in type 2 diabetes complicated by diabetic kidney disease. Clin. Lab. 2022, 68, 210836. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Levels of TMAO relative to the occurrence of peripheral arterial stiffness in chronic kidney disease patients by receiver operating characteristic curve analysis.
Figure 1. Levels of TMAO relative to the occurrence of peripheral arterial stiffness in chronic kidney disease patients by receiver operating characteristic curve analysis.
Toxins 14 00526 g001
Table 1. Baseline characteristics of the 157 chronic kidney disease patients in the control and peripheral arterial stiffness group.
Table 1. Baseline characteristics of the 157 chronic kidney disease patients in the control and peripheral arterial stiffness group.
CharacteristicsAll Patients
(n = 157)
Control group
(n = 89)
PAS Group
(n = 68)
p Value
Age (years)67.90 ± 13.0063.35 ± 12.0473.85 ± 11.81<0.001 *
Body mass index (kg/m2)26.12 ± 3.9526.27 ± 4.3925.92 ± 3.300.587
Female, n (%)76 (48.4)41 (46.1)35 (51.5)0.502
TMAO (μg/L)27.43 (16.68–49.54)25.28 (15.56–42.47)35.83 (17.95–77.79)0.006 *
Left baPWV (m/s)17.30 ± 3.4614.93 ± 1.5420.40 ± 2.74<0.001 *
Right baPWV (m/s)17.26 ± 3.3914.97 ± 1.8020.26 ± 2.54<0.001 *
Diabetes mellitus, n (%)52 (33.1)23 (25.8)29 (42.6)0.027 *
Hypertension, n (%)97 (61.8)49 (55.1)48 (70.6)0.047 *
Hyperlipidemia, n (%)93 (59.2)55 (61.8)38 (55.9)0.555
Smoking, n (%)16 (10.2)10 (11.2)6 (8.8)0.244
SBP (mmHg)144.31 ± 17.84140.53 ± 19.49149.25 ± 14.090.002 *
DBP (mmHg)82.14 ± 10.2182.06 ± 10.3182.25 ± 10.150.907
Total cholesterol (mg/dL)163.01 ± 43.75165.22 ± 47.37160.12 ± 38.640.470
Triglyceride (mg/dL)121.0 (87.5–165.0)116.0 (88.5–166.5)128.50 (86.25–163.0)0.430
LDL–C (mg/dL)92.63 ± 36.2895.93 ± 39.4188.31 ± 31.500.193
Fasting glucose (mg/dL)104.0 (98.0–134.0)100.0 (97.0–118.5)110.0 (100.0–141.75)0.010 *
Blood urea nitrogen (mg/dL)33.00 (24.00–45.00)31.00 (24.50–44.00)35.50 (24.00–49.50)0.562
Creatinine (mg/dL)1.90 (1.40–2.70)1.80 (1.40–2.60)2.05 (1.60–3.00)0.163
eGFR (mL/min)31.37 ± 15.3334.06 ± 15.4227.84 ± 14.580.011 *
CKD stage 3, n (%)79 (50.3)51 (57.3)28 (41.2)0.119
CKD stage 4, n (%)45 (28.7)23 (25.8)22 (32.4)
CKD stage 5, n (%)33 (21.0)15 (16.9)18 (26.4)
Albumin (g/dL)4.07 ± 0.324.10 ± 0.334.04 ± 0.290.256
Hemoglobin (g/dL)11.68 ± 2.5811.71 ± 2.0311.64 ± 3.170.870
CRP (mg/dL)0.13 (0.05–1.20)0.06 (0.05–1.12)0.34 (0.08–163)0.012 *
ARB use, n (%)88 (56.1)46 (51.7)42 (61.8)0.207
β–blocker use, n (%)50 (31.8)25 (28.1)25 (36.8)0.248
CCB use, n (%)60 (38.2)31 (34.8)29 (42.6)0.318
α–adrenergic blocker use, n (%)21 (13.4)10 (11.2)11 (16.2)0.368
Statin use, n (%)73 (46.5)41 (46.1)32 (47.1)0.902
Fibrate use, n (%)25 (15.9)15 (16.9)10 (14.7)0.715
Values for continuous variables are given as mean ± standard deviation or median and interquartile range and tested by Student’s t-test or Mann–Whitney U test according to normal distribution; values are presented as number (%) and analysis was done using the chi-square test. PAS, peripheral arterial stiffness; baPWV, brachial-ankle pulse wave velocity; SBP, systolic blood pressure; DBP, diastolic blood pressure; LDL-C, low-density lipoprotein cholesterol; eGFR, estimated glomerular filtration rate; TMAO, Trimethylamine N-oxide; CRP, C-reactive protein; ARB, angiotensin-receptor blocker; CCB, calcium-channel blocker; CKD, chronic kidney disease. * p < 0.05 was considered statistically significant.
Table 2. Factors correlated with peripheral arterial stiffness of chronic kidney disease patients.
Table 2. Factors correlated with peripheral arterial stiffness of chronic kidney disease patients.
VariablesOdds Ratio95% CIp Value
Trimethylamine N-oxide, 1 μg/L1.0161.002–1.0290.021 *
Age, 1 year1.0811.043–1.121<0.001 *
systolic blood pressure, 1 mmHg1.0421.004–1.0810.031 *
Hypertension, present0.5960.171–2.0720.415
Diabetes mellitus, present3.0620.970–9.6600.056
Fasting glucose, 1 mg/dL0.9980.987–1.0090.710
Estimated glomerular filtration rate, 1 mL/min0.9910.962–1.0200.538
C-reactive protein, 1 mg/dL1.0850.889–1.3250.422
Data analysis was performed using multivariate logistic regression analysis (adopted factors: diabetes mellitus, hypertension, age, systolic blood pressure, fasting glucose, estimated glomerular filtration rate, C-reactive protein and trimethylamine N-oxide). CI, confidence interval. * p < 0.05 was considered statistically significant.
Table 3. Correlation between TMAO, left baPWV, and right baPWV, with clinical variables of chronic kidney disease stage 3–5 patients.
Table 3. Correlation between TMAO, left baPWV, and right baPWV, with clinical variables of chronic kidney disease stage 3–5 patients.
VariablesTMAOLeft PWVRight PWVAgeBMIeGFRCRPGlucoseLDL–CTGSBPDBP
TMAOr
p
0.256
0.001 *
0.267
0.001 *
0.219
0.006 *
−0.098
0.222
−0.442
<0.001 *
0.261
0.001 *
−0.015
0.856
−0.082
0.305
−0.079
0.325
0.029
0.716
−0.049
0.546
Left PWVr
p
0.256
0.001 *
0.913
<0.001 *
0.453
<0.001 *
−0.013
0.870
−0.147
0.066
0.182
0.022 *
0.132
0.100
0.038
0.637
0.035
0.666
0.328
<0.001 *
0.103
0.200
Right PWVr
p
0.267
0.001 *
0.913
<0.001 *
0.411
<0.001 *
−0.036
0.650
−0.151
0.060
0.157
0.050
0.126
0.117
0.039
0.629
0.059
0.459
0.291
< 0.001 *
0.084
0.294
Ager
p
0.219
0.006 *
0.453
<0.001 *
0.411
<0.001 *
−0.006
0.941
−0.110
0.171
0.147
0.066
0.013
0.867
−0.024
0.767
−0.129
0.109
0.041
0.611
−0.346
<0.001 *
BMIr
p
−0.098
0.222
−0.013
0.870
−0.036
0.650
−0.006
0.941
0.166
0.038
−0.072
0.373
0.125
0.118
0.028
0.724
0.128
0.109
0.221
0.006 *
0.127
0.113
eGFRr
p
−0.442
<0.001 *
−0.147
0.066
−0.151
0.060
−0.110
0.171
0.166
0.038 *
−0.185
0.021 *
0.126
0.116
0.088
0.270
−0.033
0.680
−0.184
0.021 *
−0.058
0.472
CRPr
p
0.261
0.001 *
0.182
0.022 *
0.157
0.050
0.147
0.066
−0.072
0.373
−0.185
0.021 *
−0.034
0.673
−0.088
0.272
−0.006
0.937
0.050
0.537
−0.072
0.370
Glucoser
p
−0.015
0.856
0.132
0.100
0.126
0.117
0.013
0.867
0.125
0.118
0.126
0.116
−0.034
0.673
−0.106
0.185
0.167
0.036 *
0.028
0.729
0.031
0.697
LDL–Cr
p
−0.082
0.305
0.038
0.637
0.039
0.629
−0.024
0.767
0.028
0.724
0.088
0.270
−0.088
0.272
−0.106
0.185
0.196
0.014 *
−0.028
0.730
0.120
0.133
TGr
p
−0.079
0.325
0.035
0.666
0.059
0.459
−0.129
0.109
0.128
0.109
−0.033
0.680
−0.006
0.937
0.167
0.036 *
0.196
0.014 *
−0.075
0.352
0.043
0.592
SBPr
p
0.029
0.716
0.328
<0.001 *
0.291
<0.001 *
0.041
0.611
0.221
0.006 *
−0.184
0.021 *
0.050
0.537
0.028
0.729
−0.028
0.730
−0.075
0.352
0.566
<0.001 *
DBPr
p
−0.049
0.546
0.103
0.200
0.084
0.294
−0.346
<0.001 *
0.127
0.113
−0.058
0.472
−0.072
0.370
0.031
0.697
0.120
0.133
0.043
0.592
0.566
<0.001 *
Analysis of data was performed using Spearman correlation analysis. BMI, body mass index; eGFR, estimated glomerular filtration rate; CRP, C-reactive protein; TG, triglyceride; DBP, diastolic blood pressure; SBP, systolic blood pressure; LDL-C, low-density lipoprotein cholesterol; PWV, pulse wave velocity; TMAO, trimethylamine N-oxide. * p < 0.05 was considered statistically significant.
Table 4. Correlation between peripheral arterial stiffness and TMAO of CKD patients by subgroup analysis.
Table 4. Correlation between peripheral arterial stiffness and TMAO of CKD patients by subgroup analysis.
Peripheral arterial stiffnessOdds ratio95% CIp Value
Diabetes mellitus, yes1.0070.995–1.0200.249
Diabetes mellitus, no1.0351.016–1.054<0.001 *
Hypertension, yes1.0161.004–1.0280.011 *
Hypertension, no1.0321.008–1.0560.008 *
Hyperlipidemia, yes1.0141.003–1.0250.017 *
Hyperlipidemia, no1.0381.011–1.0660.006 *
Chronic kidney disease
Stage 31.0060.992–1.0190.432
Stage 41.0251.004–1.0500.046 *
Stage 51.0361.006–1.0680.021 *
Analysis data was done using the univariate logistic regression analysis. CI, confidence interval. * p < 0.05 was considered statistically significant.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hsu, B.-G.; Wang, C.-H.; Lin, Y.-L.; Lai, Y.-H.; Tsai, J.-P. Serum Trimethylamine N-Oxide Level Is Associated with Peripheral Arterial Stiffness in Advanced Non-Dialysis Chronic Kidney Disease Patients. Toxins 2022, 14, 526. https://doi.org/10.3390/toxins14080526

AMA Style

Hsu B-G, Wang C-H, Lin Y-L, Lai Y-H, Tsai J-P. Serum Trimethylamine N-Oxide Level Is Associated with Peripheral Arterial Stiffness in Advanced Non-Dialysis Chronic Kidney Disease Patients. Toxins. 2022; 14(8):526. https://doi.org/10.3390/toxins14080526

Chicago/Turabian Style

Hsu, Bang-Gee, Chih-Hsien Wang, Yu-Li Lin, Yu-Hsien Lai, and Jen-Pi Tsai. 2022. "Serum Trimethylamine N-Oxide Level Is Associated with Peripheral Arterial Stiffness in Advanced Non-Dialysis Chronic Kidney Disease Patients" Toxins 14, no. 8: 526. https://doi.org/10.3390/toxins14080526

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