IMR Press / FBL / Volume 28 / Issue 11 / DOI: 10.31083/j.fbl2811318
Open Access Original Research
MicroRNA-599-Regulated Susceptibility to Acute Kidney Injury in Patients with Cirrhosis is Mediated by the Sirtuin 1 (SIRT1) rs4746720 Single Nucleotide Polymorphism
Show Less
1 Department of Nephrology, Ningbo No.2 Hospital, 315010 Ningbo, Zhejiang, China
*Correspondence: nbeyluoqun@126.com (Qun Luo)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(11), 318; https://doi.org/10.31083/j.fbl2811318
Submitted: 7 March 2023 | Revised: 14 May 2023 | Accepted: 1 June 2023 | Published: 29 November 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Objective: The aim of this case-control study was to analyze the association between sirtuin 1 (SIRT1) single nucleotide polymorphism (SNP) and the risk of acute kidney injury (AKI) in Han Chinese patients with cirrhosis and to explore its potential mechanism. Methods: Twenty-nine AKI patients with cirrhosis (AKI group) and 87 non-AKI patients with cirrhosis (control group) were recruited from a Han Chinese population. SNaPshot sequencing technology was used for the detection of SNPs. Dual luciferase reporter vectors were constructed and co-transfected into HK-2 human proximal tubular epithelial cells. SIRT1-overexpressing recombinant plasmids were constructed and co-transfected into HK-2 cells. The expression of microRNA-599 (miR-599) and SIRT1/peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α)/nuclear respiratory factor 1 (NRF1)/mitochondrial transcription factor A (TFAM) was detected by the quantitative polymerase chain reaction, and the expression of the corresponding proteins was detected by Western blotting. Results: There were no statistically significant between-group differences in the genotype and allele frequencies of SIRT1 rs4746720. In the subgroup of patients with hepatic encephalopathy, the SIRT1 rs4746720 SNP was significantly associated with the development of AKI, and the risk of AKI in patients with the T allele was six times higher than in those with the C allele. The results of the in vitro experiments demonstrated that the T allele of SIRT1 rs4746720 increased the binding of miR-599 to the rs4746720 locus within the 3-UTR of SIRT1 (p < 0.001). The results of the SIRT1-overexpressing recombinant plasmid experiments confirmed that the T allele of SIRT1 rs4746720 mediated the binding of miR-599, leading to decreased SIRT1 and PGC-1α, NRF1, and TFAM (p < 0.05). Conclusions: The SIRT1 rs4746720 SNP might be linked with AKI in cirrhotic patients, and the T allele increased the risk of AKI in those with hepatic encephalopathy. The rs4746720 SNP in the SIRT1 3-UTR is linked to the development of AKI in cirrhotic patients with hepatic encephalopathy, potentially by mediating the binding of miR-599.

Keywords
acute kidney injury
cirrhosis
sirtuin 1
single-nucleotide SNP
microRNA-599
1. Introduction

Acute kidney injury (AKI) is a common complication in patients with cirrhosis. In fact, in hospitalized patients with cirrhosis, the incidence of AKI can be as high as 20–40% [1, 2, 3]. The occurrence of AKI in patients with cirrhosis prolongs the hospital stay and increases their risk of multiple organ failure, such that their 30-day and 1-year mortality rates are 10- and 8-fold higher, respectively, than those of cirrhotic patients without AKI [4, 5, 6]. Thus, early detection of and interventions for AKI are important measures to improve patient outcomes [3, 7]. There are several risk factors for AKI patients with cirrhosis, including advanced age, diabetes, and infection [8, 9, 10, 11]. Moreover, candidate gene studies have suggested that some patients have a genetic predisposition to developing AKI in cirrhosis, such as the endothelial nitric oxide synthase gene (eNOS) G894T single nucleotide polymorphism (SNP), the vasopressin 1a receptor gene (AVPR1A) promoter region rs113481894 SNP, or an angiotensin-converting enzyme gene insertion or deletion, as these are associated with the risk of hepatorenal syndrome [12, 13, 14].

Silent mating type information regulator 2 homolog 1 (SIRT1) is a highly conserved nicotinamide adenine dinucleotide-dependent protein deacetylase [15], which has been shown to have nephroprotective effects in AKI models induced by multiple etiologies such as ischemia-reperfusion injury, sepsis, and nephrotoxic drugs [16, 17, 18, 19, 20]. For example, we showed that in the early stages of a rat model of AKI in cirrhosis, the expression of SIRT1 and peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) in the renal tissues of rats was significantly reduced, suggesting that the SIRT1/PGC-1α signaling pathway may be involved in the mechanism of cirrhosis.

Currently, the role of SIRT1 SNPs in AKI patients with cirrhosis remains unknown. Accordingly, in this study, we examined the function of SIRT1, as it is a candidate susceptibility gene for AKI in cirrhosis. Our aim was to develop a sound theoretical basis for the pathogenesis of AKI in cirrhosis that can be used for early diagnosis and the identification of therapeutic targets.

2. Materials and Methods
2.1 Participants

This study enrolled patients with liver cirrhosis who were admitted to the Department of Hepatology, Ningbo No.2 Hospital (Zhejiang, China) from October 2020 to January 2021. The inclusion criteria were being Han Chinese, aged 18 years old, and being hospitalized for a diagnosis of liver cirrhosis. The exclusion criteria had organic kidney disease, having received renal replacement therapy, intending to undergo or have undergone kidney transplantation or liver transplantation, having liver cancer and other malignant tumors, having a confirmed pregnancy, or having a life expectancy <3 days. Those meeting the inclusion criteria were divided into an AKI group and a non-AKI group. The research protocol was approved by the Medical Ethics Committee of the hospital, and all of the participants provided signed informed consent. The diagnostic criteria for AKI were based on the 2015 International Ascites Club guidelines: a serum creatinine (SCr) concentration >0.3 mg/dL (26.5 µmol/L) from baseline within 48 h of admission, or >50% from baseline within 7 days of admission.

2.2 Clinical Data Collection

The data collected on the general condition of the participants comprised their age, sex, primary disease, comorbidities, medical history, family history, having complications of cirrhosis (infection/spontaneous peritonitis, gastrointestinal (GI) bleeding, ascites, and hepatic encephalopathy), body mass index (BMI), systolic blood pressure, and diastolic blood pressure. The laboratory indicators of the participants that were recorded included SCr concentration (baseline SCr concentration, i.e., at admission, and highest SCr concentration), blood urea nitrogen (BUN) concentration, albumin concentration, total bilirubin concentration, alanine aminotransferase (ALT) concentration, aspartate aminotransferase (AST) concentration, glucose concentration, total cholesterol concentration, low-density lipoprotein concentration, triglyceride concentration, blood sodium concentration, C-reactive protein (CRP) concentration, hemoglobin concentration, platelet count, prothrombin time, and international normalized ratio. The simplified Modification of Diet in Renal Disease formula was used to determine the estimated glomerular filtration rate (eGFR).

2.3 Specimen Collection and Genomic DNA Extraction

A sample (0.5 mL) of peripheral venous blood was collected from the patients in the morning while they were at rest and had an empty stomachs. The sample was treated with an anticoagulant (ethylenediaminetetraacetic acid) and then stored at –80 °C. After all of the blood samples had been collected, their DNA was extracted using a blood genomic DNA extraction kit and then stored at –20 °C for later use.

2.4 Primer Design and Synthesis

The SNP locus information of SIRT1 was obtained, revealing that the functional SNP site of SIRT1 was rs4746720. Thus, the primer sequence was designed as shown in Table 1.

Table 1.Primer sequence of SIRT1 SNP.
SNP site Polymorphism Sequence 5-3
rs4746720 T/C F: CCAAAGAATGGTATTTTCACTT
R: AAGTTAGCTGCCACAGTT

Note: F, Forward primer; R, Reverse primer.

2.5 Cell Culture

The kidney proximal tubular epithelial cell line Human Kidney 2 (HK2, cat. ZB188) was purchased from the Shanghai Zhibei Biotechnology Co., Ltd., which has been validated by short tandem repeat profiling and was negative for mycoplasma contamination. Dulbecco’s modified Eagle’s medium/F-12 modification containing 10% fetal bovine serum was added to recovered human proximal tubular epithelial (HK-2) cells, which were then cultured in an incubator at 37 °C under an atmosphere of 5% CO2. Cells were passaged at a density of 5 × 105 cells/mL.

2.6 Dual Luciferase Reporter Gene Assay

LipofectamineTM3000 liposomal transfection reagent was used to co-transfect the pmir-GLO-SIRT1-3-UTR-T and pmir-GLO-SIRT1-3-UTR-C luciferase reporter plasmids with an miR-599 mimic and negative control (NC), respectively, into HK-2 cells. A luciferase kit was used to detect the relative luciferase activity of cells after 72 h.

2.7 Quantitative Polymerase Chain Reaction (qPCR)

Overexpressed recombinant plasmids pcDNA3.1-SIRT1-T and pcDNA3.1-SIRT1-C bearing different alleles of SIRT1 rs4746720 were constructed, and HK-2 cells were co-transfected with an miR-599 mimic, an miR-599 inhibitor, or an NC, respectively. Total cellular RNA was extracted using TRIzol reagent, and the concentration and purity of the extracted RNA were determined. RNA was reverse transcribed into complementary DNA (cDNA), which was detected by qPCR using an miR-599 upstream primer (5-GCACGGCAGTGTGTGTCAGTGTTTA-3) and downstream primer (5-TATGGTTCTTCACACGACTCCTTCAC-3). The upstream primer of the internal reference U6 was forward 5-CTCGCTTCGGCAGCACA-3, and the downstream primer was 5-AACGCTTCACGAATTTGCG-3. The PCR reaction system comprised 2× SYBR Color qPCR Master Mix (10 µL), upstream and downstream primers (0.6 µL), cDNA (8.8 µL), and double-distilled H2O (20 µL). The reaction conditions were 40 cycles comprising pre-denaturation at 95 °C for 30 s, denaturation at 95 °C for 10 s, and annealing at 60 °C for 30 s. The relative gene expression was calculated using the 2-ΔΔCT method.

2.8 Western Blot Analysis

Phenylmethylsulfonyl fluoride was added to lyse and extract total proteins, and the bicinchoninic acid assay was used to determine the total protein concentration. Subsequently, proteins were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and then electrotransferred to a polyvinylidene difluoride membrane. Then the membrane was blocked in blocking solution for 1 h, followed by incubation overnight at 4 °C with primary antibodies against SIRT1, PGC-1α, nuclear respiratory factor 1 (NRF1), mitochondrial transcription factor A (TFAM) (1:1000), and glyceraldehyde 3-phosphate dehydrogenase (1:5000). Next, the membrane was washed and then incubated for 1 h at room temperature with secondary antibody (1:5000). Finally, enhanced chemiluminescence was used for protein detection, and ImageJ software (v1.42; National Institutes of Health, Bethesda, MD, USA) was used to quantify the bands.

2.9 Statistical Analysis

Statistical Package for Social Sciences (SPSS) version 26.0 (IBM SPSS Statistics, Armonk, NY, USA) was used for the data analysis. The normally distributed measurements are expressed as the means ± standard deviations, the t-test was used for comparisons between groups, and analysis of variance was used for comparisons between multiple groups. The non-normally distributed measures are expressed as medians (interquartile intervals), and the Mann–Whitney U test was used for comparisons between groups. Counts are expressed as cases and percentages, and comparisons between groups were made using a goodness-of-fit chi-square (χ2) Hardy–Weinberg equilibrium (HWE) test. Logistic regression was used to analyze the degree of association in terms of an odds ratio (OR) with a 95% confidence interval (CI). p < 0.05 was considered statistically significant.

3. Results
3.1 Comparison of the General Data of Study Participants

Of the 116 participants, 29 (20 males) were assigned to the AKI group and had an average age of 59.0 ± 14.8 years, and 87 (58 males) were assigned to the control group and had an average age of 60.1 ± 11.2 years. There were no significant between-group differences in age, sex, BMI, mean arterial pressure, diabetes mellitus, hypertension, ascites, hepatic encephalopathy, GI bleeding, albumin concentration, blood sodium concentration, coagulation function, or baseline SCr concentration (p > 0.05). The infection rate, total bilirubin concentration, ALT concentration, AST concentration, CRP concentration, and model for end-stage liver disease (MELD) scores of the AKI group were significantly higher than those of the control group (p < 0.05). The eGFR of the AKI group was significantly lower than that of the control group (p < 0.001; Table 2).

Table 2.Clinical data of study population.
AKI group (n = 29) Control group (n = 87) p
Age (year) 59.0 ± 14.8 60.1 ± 11.2 0.677
Male (n, %) 20 (69.0) 58 (66.7) 0.819
BMI (Kg/m2) 23.7 ± 3.7 23.9 ± 4.0 0.805
Mean arterial pressure (mmHg) 87.9 ± 17.0 89.9 ± 12.9 0.577
Diabetes (n, %) 5 (17.2) 29 (33.3) 0.099
Hypertension (n, %) 6 (20.7) 20 (23.0) 0.797
Infection (n, %) 18 (62.1) 23 (26.4) 0.001*
Ascites (n, %) 22 (75.9) 50 (57.5) 0.077
Hepatic Encephalopathy (n, %) 9 (31.0) 21 (24.1) 0.463
Gastrointestinal Bleeding (n, %) 9 (31.0) 19 (21.8) 0.316
Albumin (g/L) 28.8 ± 5.5 30.2 ± 6.7 0.297
Total bilirubin (µmol/L) 50.8 (30.1, 168.9) 37.0 (21.9, 72.1) 0.037*
ALT (U/L) 43.0 (23.0, 103.0) 32.0 (19.0, 46.0) 0.038*
AST (U/L) 79.0 (48.5, 204.5) 56.0 (32.0, 81.0) 0.004*
Serum sodium (mmol/L) 136.6 ± 5.4 137.2 ± 5.2 0.614
CRP (mg/L) 18.1 (7.5, 45.1) 4.8 (1.9, 17.5) <0.001*
PT (s) 17.9 (15.9, 21.4) 17.1 (14.6, 19.6) 0.152
INR 1.5 (1.4, 2.0) 1.5 (1.3, 1.7) 0.118
Baseline SCr (µmol/L) 66 (52.3, 80.7) 60.8 (52.5, 75.0) 0.296
eGFR (mL/min/1.73 m2) 56.0 ± 23.1 101.9 ± 28.9 <0.001*
MELD score 20.8 ± 9.3 11.9 ± 6.1 <0.001*

Note: BMI, Body Mass Index; ALT, alanine aminotransferase; AST, aspartate aminotransferase; CRP, C-reactive protein; PT, prothrombin time; INR, International Normalized Ratio; SCr, serum creatinine; eGFR, estimation of glomerular filtration rate; MELD, Model of end-stage liver disease; AKI, acute kidney injury; *p < 0.05.

3.2 SIRT1 Genotyping

SNaPshot gene sequencing technology was used for SIRT1 genotyping. This revealed that SIRT1 rs4746720 sites exhibited CC, CT, or TT genotypes (Fig. 1).

Fig. 1.

The genotyping results of SIRT1 rs4746720 polymorphism in the AKI group and the control group. (A) CC homozygous. (B) CT heterozygotes. (C) TT homozygous. Note: SIRT1 rs4746720 extends forward, and the product is consistent with polymorphism.

3.3 Correlation Analysis of SIRT1 Gene SNPs and AKI in Cirrhosis
3.3.1 HWE Test

The goodness-of-fit χ2 test showed that the frequency distribution of SIRT1 rs4746720 and rs2273773 genotypes in the control group was consistent with the HWE (pHWE > 0.05), indicating that the participants were representative of the population (Table 3).

Table 3.HWE test of SIRT1 SNP genotype.
SNP site Genotype Observed value Expected value χ2 pHWE
rs4746720 TT 30 (34.5) 27 (31.0) 1.66 0.437
CT 37 (42.5) 17 (19.5)
CC 20 (23.0) 43 (49.4)

Note: pHWE: the control group tested the p-value.

3.3.2 Association Analysis of AKI in Cirrhosis

There was no significant between-group difference in the frequency distribution of the SIRT1 rs4746720 TT, CT, and CC genotypes (χ2 = 0.448, p = 0.799). The frequency of the T and C alleles was 35 (60.3%) and 23 (39.7%) in the AKI group and 97 (55.7%) and 77 (44.3%) in the control group, respectively, indicating that there was no significant between-group difference in the frequency distribution of these alleles (χ2 = 0.37, p = 0.540). Logistic regression analysis showed that after adjustment for age and sex, the SIRT1 rs4746720 genetic models (co-dominant: TT vs. CC, CT vs. CC; isotopic: T vs. C; dominant: TT+CT vs. CC; recessive: TT vs. CC+CT; hypersensitive: TT+CC vs. CT) were not significantly correlated with AKI susceptibility (p > 0.05; Table 4).

Table 4.Association analysis between SIRT1 rs4746720 polymorphism and AKI risk in cirrhosis.
Heredity model Genotype/allele AKI group (n = 29) Control group (n = 87) OR (95% CI) p
Co-dominant TT 12 (41.4) 30 (34.5) 1.34 (0.43–4.21) 0.611
CT 11 (37.9) 37 (42.5) 0.98 (0.32–3.07) 0.978
CC 6 (20.7) 20 (23.0) 1 (reference)
Allele T 35 (60.3) 97 (55.7) 1.21 (0.66–2.23) 0.534
C 23 (39.7) 77 (44.3) 1 (reference)
Dominant genes TT+ CT 23 (79.3) 67 (77.0) 1.14 (0.41–3.21) 0.802
CC 6 (20.7) 20 (23.0) 1 (reference)
Recessive genes TT 12 (41.4) 30 (34.5) 1.36 (0.57–3.24) 0.490
CC+ CT 17 (58.6) 57 (65.5) 1 (reference)
Hyperdominant genes TT+CC 18 (62.1) 50 (57.5) 1.22 (0.52–2.91) 0.647
CT 11 (37.9) 37 (42.5) 1 (reference)

Note: OR and p are parameters after adjusting age and gender.

3.3.3 Stratified Analyses

Stratified analyses based on age, sex, and complications of cirrhosis (infection, ascites, GI bleeding, and hepatic encephalopathy) were conducted to study the effect of genetic factors on the risk of AKI in patients with cirrhosis. In the subgroup with hepatic encephalopathy, SIRT1 rs4746720 SNPs were significantly linked with the development of AKI, and patients with the T allele had a six times greater risk of AKI than those with the C allele (OR = 6.00, 95% CI = 1.22–29.48; p = 0.027). However, in subgroups based on other factors, SIRT1 rs4746720 SNPs were not significantly associated with AKI in cirrhosis (p > 0.05; Table 5).

Table 5.Stratified analysis of SIRT1 rs4746720 polymorphism with AKI risk in cirrhosis.
Variable Genotype/allele AKI group (n = 29) Control group (n = 87) OR (95% CI) p
Age (years) <60 TT vs. CC 8/3 13/12 2.46 (0.53–11.50) 0.252
CT vs. CC 5/3 20/12 1.00 (0.20–4.96) 1.000
T vs. C 21/11 46/44 1.83 (0.79–4.22) 0.156
60 TT vs. CC 4/3 17/8 0.63 (0.11–3.49) 0.595
CT vs. CC 6/3 17/8 0.94 (0.19–4.76) 0.942
T vs. C 14/12 51/33 0.76 (0.31–1.83) 0.534
Gender male TT vs. CC 8/4 17/15 1.77 (0.44–7.06) 0.422
CT vs. CC 8/4 26/15 1.15 (0.30–4.49) 0.836
T vs. C 24/16 60/56 1.40 (0.68–2.91) 0.366
female TT vs. CC 4/2 13/5 0.77 (0.11–5.61) 0.796
CT vs. CC 3/2 11/5 0.68 (0.09–5.45) 0.718
T vs. C 11/7 37/21 0.89 (0.30–2.65) 0.837
Infection no TT vs. CC 4/3 20/15 1.00 (0.19–5.15) 1.000
CT vs. CC 4/3 29/15 0.69 (0.14–3.49) 0.653
T vs. C 12/10 69/59 1.03 (0.41–2.55) 0.956
yes TT vs. CC 8/3 10/5 1.33 (0.24–7.35) 0.741
CT vs. CC 7/3 8/5 1.46 (0.25–8.43) 0.673
T vs. C 23/13 28/18 1.14 (0.46–2.80) 0.780
Ascites no TT vs. CC 3/1 15/9 1.80 (0.16–20.03) 0.633
CT vs. CC 3/1 13/9 2.08 (0.19–23.30) 0.553
T vs. C 9/5 43/31 1.30 (0.40–4.25) 0.666
yes TT vs. CC 9/5 15/11 1.32 (0.35–5.05) 0.685
CT vs. CC 8/5 24/11 0.73 (0.20–2.76) 0.647
T vs. C 26/18 54/46 1.23 (0.60–2.52) 0.571
Hepatic encephalopathy no TT vs. CC 5/6 22/15 0.57 (0.15–2.21) 0.414
CT vs. CC 9/6 29/15 0.78 (0.23–2.59) 0.680
T vs. C 19/21 73/59 0.73 (0.36–1.49) 0.387
yes TT vs. CC 7/0 8/5 - -
CT vs. CC 2/0 8/5 - -
T vs. C 16/2 24/18 6.00 (1.22–29.48) 0.027*
Gastrointestinal bleeding no TT vs. CC 9/4 26/13 1.13 (0.29–4.35) 0.865
CT vs. CC 7/4 29/13 0.78 (0.20–3.16) 0.732
T vs. C 25/15 81/55 1.13 (0.55–2.34) 0.738
yes TT vs. CC 3/2 4/7 2.63 (0.30–23.00) 0.383
CT vs. CC 4/2 8/7 1.75 (0.24–12.64) 0.579
T vs. C 10/8 16/22 1.72 (0.56–5.33) 0.348

Note: *p < 0.05.

3.3.4 Relationship Between Genotype Distribution and Liver and Kidney Function

The relationship between genotype and liver and kidney function indexes was analyzed in each group. In the AKI group, patients with the SIRT1 rs4746720 TT genotype had significantly higher SCr and BUN concentrations and significantly lower eGFRs than those with the CC+CT genotypes (p <0.05). However, there were no significant between-genotype differences in other indexes (p > 0.05). In the control group, there was no significant between-genotype difference in liver or kidney function (p > 0.05; Table 6).

Table 6.Liver and kidney function level in different genotypes of SIRT1 rs4746720.
Variable AKI group p Control group p
TT CC+CT TT CC+CT
SCr (µmol/L) 171.5 ± 88.1 111.8 ± 48.4 0.026* 68.0 ± 16.3 72.0 ± 15.1 0.255
eGFR (mL/min/1.73 m2) 43.1 ± 20.7 65.1 ± 20.5 0.009* 104.4 ± 33.8 100.6 ± 26.2 0.564
BUN (mmol/L) 15.7 ± 6.8 9.5 ± 5.9 0.015* 5.2 ± 2.3 6.0 ± 3.7 0.256
Albumin (g/L) 27.6 ± 5.6 29.6 ± 5.4 0.350 30.5 ± 5.8 30.0 ± 7.1 0.748
Total bilirubin (µmol/L) 138.7 ± 191.3 117.7 ± 124.2 0.723 75.2 ± 85.3 56.0 ± 60.9 0.229
ALT (U/L) 48.7 ± 40.8 88.8 ± 97.2 0.191 59.6 ± 83.5 47.5 ± 64.2 0.454
AST (U/L) 79.0 ± 53.5 225.1 ± 309.7 0.073 84.9 ± 84.0 65.0 ± 55.0 0.189
MELD Score 23.5 ± 12.0 18.8 ± 6.6 0.193 12.2 ± 6.1 11.7 ± 6.1 0.685

Note: SCr, serum creatinine; eGFR, estimation of glomerular filtration rate; BUN, urea nitrogen; ALT, alanine aminotransferase; AST, aspartate aminotransferase; MELD, Model of end-stage liver disease; *p < 0.05.

3.4 SIRT1 rs4746720 Mediates the Effects of miR-599 on the Double Luciferase Reporter Gene

The bioinformatics software programs TargetScan and miRanda were used to predict the target gene of microRNA-599 (miRNA-599). To verify the effect of SIRT1 rs4746720 SNP binding with miR-599, whole-gene SIRT1–3-UTR-T/C sequences were synthesized, and the pmir-GLO-SIRT1–3-UTR-T/C double luciferase reporter gene plasmid was constructed. This was used to co-transfect HK-2 cells with NC and miR-599 mimics, respectively, and the changes in luciferase activity were compared. The results showed that the luciferase activity in the SIRT1-3-UTR-T+miR-599 mimic group was significantly lower than that in the SIRT1-3-UTR-T+NC group and the SIRT1-3-UTR-C+miR-599 mimic group (p < 0.001), whereas there was no significant difference in luciferase activity between the SIRT1-3-UTR-C+miR-599 mimic group and the SIRT1-3-UTR-C+NC group (p > 0.05). The above-described results showed that the expression of the T allele of SIRT1 rs4746720 was regulated by miR-599, resulting in significant inhibition of the expression of luciferase by miR-599, whereas the C allele of SIRT1 rs4746720 was not regulated by miR-599, resulting in no inhibition of the expression of luciferase by miR-599 (Fig. 2).

Fig. 2.

Results of dual luciferase reporter genes interaction between SIRT1 rs4746720 and miR-599 (***p < 0.001).

3.5 SIRT1 rs4746720 Mediates the Effect of miR-599 on SIRT1 Expression

To further study the effect of the SIRT1 rs4746720-mediated effect of miR-599 on SIRT1 expression, overexpression plasmids pcDNA3.1-SIRT1-T/C, which contained different alleles (i.e., T or C) of SIRT1 rs4746720, were constructed and verified by gene sequencing. Next, pcDNA3.1-SIRT1-T/C plasmids and pcDNA3.1 (empty plasmid) were used to transfect HK-2 cells with NC, an miR-599 mimic, or an miR-599 inhibitor, respectively. Subsequently, the relative expression of miR-599, SIRT1 mRNA, and protein in cells was determined by qPCR and Western blotting, respectively. The results were as follows: (1) Compared with transfection with NC, transfection with the miR-599 mimic caused the expression of miR-599 in the SIRT1-T overexpression group and the empty plasmid group to increase by 41% and 58%, respectively (p < 0.01), whereas transfection with the miR-599 inhibitor caused the expression of miR-599 in the SIRT1-T overexpression group, SIRT1-C overexpression group, and empty plasmid group to decrease by 39%, 34%, and 39%, respectively (p < 0.05). These results confirmed the effectiveness of transfection with the miR-599 mimic and miR-599 inhibitor, respectively (Fig. 3A). (2) Compared with the empty plasmid+NC group, in the SIRT1-T overexpression+NC group and the SIRT1-C overexpression+NC group, the mRNA and protein expression of SIRT1 was significantly upregulated (p < 0.05). This confirmed the functioning of the SIRT1-T/C overexpression plasmids. (3) Compared with the NC group, in the SIRT1-T overexpression group, co-transfection with the miR-599 mimic significantly reduced the expression of SIRT1 mRNA (p < 0.01). Protein expression of SIRT1 decreased while with no significance (p > 0.05). Co-transfection with the miR-599 inhibitor increased the expression of SIRT1 mRNA and SIRT1, but this difference was not statistically significant. (4) In the SIRT1-C overexpression group and empty plasmid groups, SIRT1 mRNA and SIRT1 expression were not significantly changed by transfection with the miR-599 mimic or the miR-599 inhibitor (p > 0.05). Taken together, these results indicate that the T allele of SIRT1 rs4746720 may mediate the interaction of miR-599 with SIRT1, resulting in significant inhibition of the mRNA expression of SIRT1, leading to a downward trend in the expression of SIRT1 (Fig. 3B,C).

Fig. 3.

Expression of miR-599 and SIRT1 in HK-2 cells after co-transfection with overexpression plasmid and miR-599. (A) Relative expression of miR-599. (B) Relative expression of SIRT1 mRNA. (C) Relative expression of SIRT1; *p <0.05, **p < 0.01, compared with the NC group; #p < 0.05, ##p < 0.01, ###p < 0.001, compared with the empty plasmid group. (D) SIRT1 protein detected by western blot.

3.6 SIRT1 rs4746720 Mediates the Effect of miR-599 on Downstream Pathways

The effect of the SIRT1 rs4746720 SNP-mediated interaction of miR-599 with SIRT1 on the PGC-1α/NRF1/TFAM signaling pathway downstream of SIRT1 was further investigated by determining the relative mRNA and protein expression of PGC-1α, NRF1, and TFAM with qPCR and Western blot analysis, respectively. The following results were obtained: (1) Compared with the empty plasmid+NC group; there was a significantly higher abundance of PGC-1α, NRF1, and TFAM mRNA and protein in the SIRT1-T overexpression+NC group and SIRT1-C overexpression+NC group (p < 0.001). (2) Compared with the NC group, in the SIRT1-T overexpression group, transfection with the miR-599 mimic caused a significant reduction in the mRNA and protein expression of PGC-1α, NRF1, and TFAM (p < 0.01), whereas co-transfection with an miR-599 inhibitor caused a significant increase in their levels (p < 0.05). (3) In the SIRT1-C overexpression group, co-transfection with the miR-599 inhibitor caused a significant reduction in the mRNA expression of TFAM (p < 0.001) but not in the protein expression of TFAM (p > 0.05), as well as a significant reduction in the mRNA levels of NRF1 and TFAM (p < 0.001) but not in the protein expression of NRF1 and TFAM (p > 0.05). (4) In the empty plasmid group, co-transfection with the miR-599 mimic and the miR-599 inhibitor, respectively, did not cause significant changes in the mRNA or protein levels of PGC-1α, NRF1, and TFAM (p > 0.05). Taken together, these results suggest that the T allele of SIRT1 rs4746720 may mediate the ability of miR-599 to significantly inhibit the activity of the PGC-1α/NRF1/TFAM pathway (Fig. 4).

Fig. 4.

Expression of PGC-1α/NRF1/TFAM in HK-2 cells after co-transfection with overexpression plasmid and miR-599. (A–C) PGC-1α/NRF1/TFAM mRNA relative expression. (D–G) PGC-1α/NRF1/TFAM protein relative expression; *p < 0.05, **p < 0.01, ***p < 0.001, compared with NC; ###p < 0.001, compared to empty plasmid.

4. Discussion

The rapid development of genomics technology has enabled research revealing the important role of genetic factors in the occurrence and development of AKI [21, 22, 23, 24]. However, there have been few studies on patients with cirrhosis and genetic susceptibility to develop AKI. Seckin et al. [12] reported that the GT/TT genotypes and T mutant allele of the eNOs G894T SNP might be risk factors for the development of hepatorenal syndrome, as patients with the GT or TT genotype had a 5.0 or 5.8 times greater risk, respectively, of developing hepatorenal syndrome than those with the GG genotype. Wang et al. [13] studied 60 patients with cirrhosis and found that those with the T allele of the promoter of AVPR1A rs113481894 had a 2.23 times higher risk of developing type I hepatorenal syndrome than those with the C allele of the promoter of AVPR1A rs113481894. These candidate genes code for proteins involved in classical splanchnic-vasodilation-related processes. In recent years, further research has increased our understanding of the pathogenesis of AKI in cirrhosis [25, 26]. In addition to traditional hemodynamic mechanisms, non-hemodynamic microvascular toxicities (caused by, for example, endotoxins or bile acids) may be directly related to the development of AKI [10, 27]. The synergistic effect of toxic factors and microvascular dysfunction increases damage to proximal tubular epithelial cells, mediates the downregulation of mitochondrial function, and triggers intrarenal activation of the renin-angiotensin-aldosterone system, resulting in a decreased GFR [28].

4.1 Association of SIRT1 SNPs with AKI in Cirrhosis

SIRT1 is a highly conserved protein deacetylase that is widely expressed in renal tissue. In ischemia-reperfusion injury, sepsis, and AKI caused by cisplatin, SIRT1 acts on many transcription factors to regulate members of downstream pathways, such as PGC-1α, forkhead box O, nuclear factor kappa B, and p53, resulting in nephroprotective effects [15, 16, 17]. Specifically, SIRT1 mediates the deacetylation of PGC-1α, activates NRF1 and TFAM, promotes mitochondrial biosynthesis and respiratory recovery, and drives proximal tubule repair, which may be a key pathway for renal protection [15, 29, 30]. In our early research using a rat model of AKI in cirrhosis, we found that the expression of SIRT1 and PGC-1α in the renal tissue of rats decreased in the early stages of AKI, and the SIRT1/PGC-1α signaling pathway may be involved in the mechanism of AKI in cirrhotic patients. Chou et al. [31] showed that knockout of SIRT1 increased tumor necrosis factor alpha-mediated renal insufficiency. Thus, SIRT1 and the downstream pathways that it affects play an important role in protecting the kidney against damage with various etiologies. SIRT1 SNPs are associated with aging, cardiovascular disease, alcoholic fatty liver disease, diabetic nephropathy, cardiorenal syndrome type 1, and many other diseases [32, 33, 34, 35]. The current study represents the first examination of the association between SIRT1 SNPs and AKI in cirrhosis. Our data analysis using Haploview 4.2 software (Broad Institute, Cambridge, MA,USA) and functional annotation screened out one functional SNP locus: the rs4746720 locus in the 3-UTR region of SIRT1.

4.2 Association of SIRT1 rs4746720 with AKI in Cirrhosis

SIRT1 rs4746720 is associated with aging, type 2 diabetes, diabetic nephropathy, and other diseases [36, 37, 38, 39]. In the current study, we found that the TT genotype and T allele frequencies of the SIRT1 rs4746720 locus in the AKI group were higher than those in the control group (41.4% vs. 34.5%, 60.3% vs. 55.7%), and the risk of AKI in cirrhosis was increased in patients with the T allele; however, the latter difference did not reach statistical significance (p = 0.534). Stratified analyses revealed that the T allele led to a greater risk of developing AKI in cirrhosis with hepatic encephalopathy. Analyses of the liver and kidney function indicators of the AKI group with respect to genotype demonstrated that the SCr and BUN concentrations of patients with the TT genotype were significantly higher, and their eGFRs were significantly lower than those with the CC+CT genotype (p < 0.05), indicating that among AKI patients with cirrhosis, those who had the TT genotype had significantly worse renal function than those who had the CC+CT genotype. Similarly, Tang et al. [36] found that compared with other genotypes, the SIRT1 rs4746720 TT genotype led to more severe diabetic nephropathy in Han Chinese patients with diabetes. Sarumaru et al. [40] showed that in Japanese patients with autoimmune thyroid disease, the thyroid autoantibody titer in patients with the SIRT1 rs4746720 T allele (i.e., the TT+TC genotype) was significantly higher than that in patients with the CC genotype, which may affect the expression of SIRT1 in inflammatory lesions of target tissues and increase susceptibility to disease. Zhang et al. [41] studied the relationship between SIRT1 functional loci and healthy aging in Han Chinese older adults and found that the participants exhibited a significantly higher frequency of the SIRT1 rs4746720 C allele than of the SIRT1 rs4746720 T allele, a higher frequency of the CC genotype than of the CT+TT genotypes and that those with the C allele may have a higher risk of unhealthy aging than those with the T allele. In addition, in the current study, SIRT1 rs4746720 was found to be most associated with AKI susceptibility in the hepatic encephalopathy subgroup, suggesting that gene-risk factors may jointly increase susceptibility to disease. MELD score, serum sodium concentrations, and hepatic encephalopathy are independent predictors of the risk of renal failure in cirrhosis cohorts, and hepatic encephalopathy is significantly associated with prognosis in patients with cirrhosis complicated by renal failure [42]. However, due to the small sample size of this study, further research is needed to confirm these findings.

4.3 SIRT1 rs4746720 may Mediate the Binding of miR-599 to SIRT1 to Affect the SIRT1/PGC-1α Pathway

Bioinformatics software was used to predict the miRNAs that may bind to rs4746720 in the 3-UTR region of SIRT1, and this revealed that miR-599 should be capable of performing this binding. Given that the site bound by miR-599 is in the 3-UTR, it is speculated that when the site has a T allele, it can be bound by miR-599, resulting in the inhibition of SIRT1 expression, whereas when the site has a C allele (due to a T-to-C mutation), it cannot be bound by miR-599, resulting in no effect on SIRT1 expression. The construction and use of a double luciferase reporter plasmid revealed that miR-599 could significantly inhibit the expression of luciferase by the T allele of rs4746720 but could not inhibit the expression of luciferase by the C allele of rs4746720, confirming that the polymorphic site of SIRT1 rs4746720 was likely to be the site where miR-599 acts. The recombinant overexpression plasmid of SIRT1 rs4746720 was constructed, and its use in experiments showed that the T allele of rs4746720 may mediate miR-599’s inhibition of the expression of SIRT1 mRNA and its downstream effectors PGC-1α, NRF1, and TFAM.

The above-described results, combined with those from other studies, indicate that the T allele of SIRT1 rs4746720 may mediate the ability of miR-599 to target and inhibit the expression of SIRT1 and its downstream PGC-1α/NRF1/TFAM signaling pathway, and may be involved in the pathogenesis of AKI in cirrhosis. The pathogenesis of AKI in cirrhosis involves the regulation of multiple genes, proteins, and signaling pathways. This is the first study to show the regulatory relationship between SIRT1 rs4746720 and miR-599. Current research on miR-599 has mainly focused on its involvement in the pathogenesis of lung cancer, liver cancer, kidney cancer, and other cancer-related pathogenesis, as miR-599 plays different roles in different tumor tissues. For example, in liver cancer, the expression of miR-599 is downregulated; this is manifested by changes in the abundance of tumor suppressor factors, which affect tumor production. By contrast, in lung cancer, the expression of miR-599 is upregulated, which promotes proliferation and invasion [43, 44]. However, the role of miR-599 in AKI has not been fully described, so further research is needed.

5. Limitations

This was a single-center retrospective case-control study with a small sample size that only included a Han Chinese population with cirrhosis. No relationship between SIRT1 rs4746720 and the development of AKI was observed in cirrhotic patients. Thus, our conclusions need to be validated in future centralized studies with larger sample sizes. Moreover, in the current study, the serum levels of SIRT1 and miR-599 in patients in the AKI and control groups were considered to fully represent the effect of SIRT1rs4746720 SNP-mediated miR-599 on AKI patients with cirrhosis at the population level. Furthermore, no in vivo functional verification experiments were performed in this study. Therefore, clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 technology could be used to construct a knock-in SIRT1 rs4746720 mutant mouse model of AKI in cirrhosis. This would enable verification of the mechanism of the susceptibility of SIRT1 and its downstream pathways to SIRT1 rs4746720 SNP-mediated miR-599 control.

6. Conclusions

The SIRT1 rs4746720 SNP might be linked with AKI in cirrhotic patients, and the T allele increased the risk of AKI in those with hepatic encephalopathy. In AKI patients with cirrhosis, the renal function of patients with the TT genotype was significantly lower than that of patients with the CC+CT genotype. The SIRT1 rs4746720 SNP mediated miR-599 binding to SIRT1, thereby affecting the expression of SIRT1 and its downstream effectors, PGC-1α/NRF1/TFAM, and may be involved in the mechanism of AKI in cirrhotic patients.

Availability of Data and Materials

The datasets generated during and analysed during the current study are available from the corresponding author on reasonable request.

Author Contributions

All authors contributed to the study conception and design. Material preparation, data collection and analysis were performed by FZ, YC, YX, and QL. The first draft of the manuscript was written by FZ and YC, and it was revised by QL. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

The research protocol was approved by the Medical Ethics Committee of Ningbo NO.2 hospital (YJ-KYSB-NBEY-2018-002-01), and all of the participants provided signed informed consent.

Acknowledgment

Not applicable.

Funding

This study was funded by the Project of NINGBO Leading Medical & Health Discipline (Project No. 2022-S03), China, and Medical Scientific Research Foundation of Zhejiang Province (Project No. 2019KY181).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Maiwall R, Pasupuleti SSR, Bihari C, Rastogi A, Singh PK, Naik V, et al. Incidence, Risk Factors, and Outcomes of Transition of Acute Kidney Injury to Chronic Kidney Disease in Cirrhosis: A Prospective Cohort Study. Hepatology. 2020; 71: 1009–1022.
[2]
Francoz C. Acute kidney injury in cirrhosis: An immediate threat but also a ticking time bomb. Journal of Hepatology. 2020; 72: 1043–1045.
[3]
Angeli P, Ginès P, Wong F, Bernardi M, Boyer TD, Gerbes A, et al. Diagnosis and management of acute kidney injury in patients with cirrhosis: revised consensus recommendations of the International Club of Ascites. Journal of Hepatology. 2015; 62: 968–974.
[4]
Desai AP, Knapp SM, Orman ES, Ghabril MS, Nephew LD, Anderson M, et al. Changing epidemiology and outcomes of acute kidney injury in hospitalized patients with cirrhosis - a US population-based study. Journal of Hepatology. 2020; 73: 1092–1099.
[5]
Kumar U, Kumar R, Jha SK, Jha AK, Dayal VM, Kumar A. Short-term mortality in patients with cirrhosis of the liver and acute kidney injury: A prospective observational study. Indian Journal of Gastroenterology. 2020; 39: 457–464.
[6]
Tandon P, James MT, Abraldes JG, Karvellas CJ, Ye F, Pannu N. Relevance of New Definitions to Incidence and Prognosis of Acute Kidney Injury in Hospitalized Patients with Cirrhosis: A Retrospective Population-Based Cohort Study. PLoS ONE. 2016; 11: e0160394.
[7]
European Association for the Study of the Liver. EASL Clinical Practice Guidelines for the management of patients with decompensated cirrhosis. Journal of Hepatology. 2018; 69: 406–460.
[8]
Kogiso T, Ogasawara Y, Sagawa T, Taniai M, Tokushige K. Risk and protective factors of acute kidney injury in decompensated cirrhotic patients with ascites on tolvaptan. JGH Open. 2021; 5: 1298–1305.
[9]
Shin YJ, Heo CM, Kim KM, Shim SG, Sinn DH. Prevalence, risk factors, and short-term outcomes of postparacentesis acute kidney injury using revised criteria of the international club of ascites. Medicine. 2021; 100: e27431.
[10]
Velez JCQ, Therapondos G, Juncos LA. Reappraising the spectrum of AKI and hepatorenal syndrome in patients with cirrhosis. Nature Reviews. Nephrology. 2020; 16: 137–155.
[11]
Mantle D, Turton N, Hargreaves IP. Depletion and Supplementation of Coenzyme Q10 in Secondary Deficiency Disorders. Frontiers in Bioscience-Landmark. 2022; 27: 322.
[12]
Seckin Y, Yigit A, Yesilada E, Gulbay G, Cagin YF, Gozukara H, et al. Association of eNOS Gene Polymorphisms G894T and T-786C with Risk of Hepatorenal Syndrome. Gastroenterology Research and Practice. 2016; 2016: 2579626.
[13]
Wang C, Yu Z, Luo X, Ye J, Liu S, Miu L, et al. Association of arginine vasopressin receptor 1a gene polymorphisms with hepatorenal syndrome. JPMA. The Journal of the Pakistan Medical Association. 2017; 67: 577–579.
[14]
Giani JF, Veiras LC, Shen JZY, Bernstein EA, Cao D, Okwan-Duodu D, et al. Novel roles of the renal angiotensin-converting enzyme. Molecular and Cellular Endocrinology. 2021; 529: 111257.
[15]
Morigi M, Perico L, Benigni A. Sirtuins in Renal Health and Disease. Journal of the American Society of Nephrology. 2018; 29: 1799–1809.
[16]
Sabet N, Soltani Z, Khaksari M. The effects of exercise on kidney injury: the role of SIRT1. Molecular Biology Reports. 2022; 49: 4025–4038.
[17]
Peasley K, Chiba T, Goetzman E, Sims-Lucas S. Sirtuins play critical and diverse roles in acute kidney injury. Pediatric Nephrology. 2021; 36: 3539–3546.
[18]
Gong L, He J, Sun X, Li L, Zhang X, Gan H. Activation of sirtuin1 protects against ischemia/reperfusion-induced acute kidney injury. Biomedicine & Pharmacotherapy. 2020; 125: 110021.
[19]
Xu S, Gao Y, Zhang Q, Wei S, Chen Z, Dai X, et al. SIRT1/3 Activation by Resveratrol Attenuates Acute Kidney Injury in a Septic Rat Model. Oxidative Medicine and Cellular Longevity. 2016; 2016: 7296092.
[20]
Kim JY, Jo J, Kim K, An HJ, Gwon MG, Gu H, et al. Pharmacological Activation of SIRT1 Ameliorates Cisplatin-Induced Acute Kidney Injury by Suppressing Apoptosis, Oxidative Stress, and Inflammation in Mice. Antioxidants. 2019; 8: 322.
[21]
Larach DB, Engoren MC, Schmidt EM, Heung M. Genetic variants and acute kidney injury: A review of the literature. Journal of Critical Care. 2018; 44: 203–211.
[22]
Ortega-Loubon C, Martínez-Paz P, García-Morán E, Tamayo-Velasco Á, López-Hernández FJ, Jorge-Monjas P, et al. Genetic Susceptibility to Acute Kidney Injury. Journal of Clinical Medicine. 2021; 10: 3039.
[23]
Ding H, Zhang L, Yang Q, Zhang X, Li X. Epigenetics in kidney diseases. Advances in Clinical Chemistry. 2021; 104: 233–297.
[24]
Vilander LM, Kaunisto MA, Pettilä V. Genetic predisposition to acute kidney injury–a systematic review. BMC Nephrology. 2015; 16: 197.
[25]
Chen B, Liu J, Li Y, He X, Zhou C, Chen Y, et al. Elevated D-Dimer levels correlate with the development of hepatorenal syndrome and a poor outcome in patients with cirrhosis. Scandinavian Journal of Gastroenterology. 2022; 57: 1486–1493.
[26]
Cullaro G, Kanduri SR, Velez JCQ. Acute Kidney Injury in Patients with Liver Disease. Clinical Journal of the American Society of Nephrology. 2022; 17: 1674–1684.
[27]
Sorribas M, Jakob MO, Yilmaz B, Li H, Stutz D, Noser Y, et al. FXR modulates the gut-vascular barrier by regulating the entry sites for bacterial translocation in experimental cirrhosis. Journal of Hepatology. 2019; 71: 1126–1140.
[28]
de Seigneux S, Martin PY. Preventing the Progression of AKI to CKD: The Role of Mitochondria. Journal of the American Society of Nephrology. 2017; 28: 1327–1329.
[29]
Son SH, Lee SM, Lee MH, Son YK, Kim SE, An WS. Omega-3 Fatty Acids Upregulate SIRT1/3, Activate PGC-1α via Deacetylation, and Induce NRF1 Production in 5/6 Nephrectomy Rat Model. Marine Drugs. 2021; 19: 182.
[30]
Collier JB, Schnellmann RG. Extracellular signal-regulated kinase 1/2 regulates NAD metabolism during acute kidney injury through microRNA-34a-mediated NAMPT expression. Cellular and Molecular Life Sciences. 2020; 77: 3643–3655.
[31]
Chou YT, Liu TT, Yang UC, Huang CC, Liu CW, Huang SF, et al. Intestinal SIRT1 Deficiency-Related Intestinal Inflammation and Dysbiosis Aggravate TNFα-Mediated Renal Dysfunction in Cirrhotic Ascitic Mice. International Journal of Molecular Sciences. 2021; 22: 1233.
[32]
Hou Y, Su B, Chen P, Niu H, Zhao S, Wang R, et al. Association of SIRT1 gene polymorphism and its expression for the risk of alcoholic fatty liver disease in the Han population. Hepatology International. 2018; 12: 56–66.
[33]
Hou J, Xie X, Tu Q, Li J, Ding J, Shao G, et al. SIRT1 gene polymorphisms are associated with nondiabetic type 1 cardiorenal syndrome. Annals of Human Genetics. 2019; 83: 445–453.
[34]
Zhao Y, Wei J, Hou X, Liu H, Guo F, Zhou Y, et al. SIRT1 rs10823108 and FOXO1 rs17446614 responsible for genetic susceptibility to diabetic nephropathy. Scientific Reports. 2017; 7: 10285.
[35]
Lin R, Yan D, Zhang Y, Liao X, Gong G, Hu J, et al. Common variants in SIRT1 and human longevity in a Chinese population. BMC Medical Genetics. 2016; 17: 31.
[36]
Tang K, Sun M, Shen J, Zhou B. Transcriptional Coactivator p300 and Silent Information Regulator 1 (SIRT1) Gene Polymorphism Associated with Diabetic Kidney Disease in a Chinese Cohort. Experimental and Clinical Endocrinology & Diabetes. 2017; 125: 530–537.
[37]
Cai MY, Cheng J, Zhou MY, Liang LL, Lian SM, Xie XS, et al. The association between pre-miR-27a rs895819 polymorphism and myocardial infarction risk in a Chinese Han population. Lipids in Health and Disease. 2018; 17: 7.
[38]
Yang JF, Xiong XF, Xiao Y, Wei L, Li L, Yang M, et al. The single nucleotide polymorphism rs11643718 in SLC12A3 is associated with the development of diabetic kidney disease in Chinese people with type 2 diabetes. Diabetic Medicine. 2020; 37: 1879–1889.
[39]
Lee KW, Shin D. Interactions between Bitter Taste Receptor Gene Variants and Dietary Intake Are Associated with the Incidence of Type 2 Diabetes Mellitus in Middle-Aged and Older Korean Adults. International Journal of Molecular Sciences. 2023; 24: 2199.
[40]
Sarumaru M, Watanabe M, Inoue N, Hisamoto Y, Morita E, Arakawa Y, et al. Association between functional SIRT1 polymorphisms and the clinical characteristics of patients with autoimmune thyroid disease. Autoimmunity. 2016; 49: 329–337.
[41]
Zhang WG, Bai XJ, Chen XM. SIRT1 variants are associated with aging in a healthy Han Chinese population. Clinica Chimica Acta; International Journal of Clinical Chemistry. 2010; 411: 1679–1683.
[42]
Matchett CL, Simonetto DA, Kamath PS. Renal Insufficiency in Patients with Cirrhosis. Clinics in Liver Disease. 2023; 27: 57–70.
[43]
Tian J, Hu X, Gao W, Zhang J, Chen M, Zhang X, et al. Identification a novel tumor-suppressive hsa-miR-599 regulates cells proliferation, migration and invasion by targeting oncogenic MYC in hepatocellular carcinoma. American Journal of Translational Research. 2016; 8: 2575–2584.
[44]
Zhao H, Zhao H, Xia X, Liu X. MicroRNA-599 targets high-mobility group AT-hook 2 to inhibit cell proliferation and invasion in clear cell renal carcinoma. Molecular Medicine Reports. 2018; 17: 7451–7459.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top