Buscar en
Annals of Hepatology
Toda la web
Inicio Annals of Hepatology Oral 24-week probiotics supplementation did not decrease cardiovascular risk mar...
Journal Information
Vol. 28. Issue 1.
(January - February 2023)
Share
Share
Download PDF
More article options
Visits
744
Vol. 28. Issue 1.
(January - February 2023)
Original article
Open Access
Oral 24-week probiotics supplementation did not decrease cardiovascular risk markers in patients with biopsy proven NASH: A double-blind placebo-controlled randomized study
Visits
744
Samantha Thifani Alrutz Barcelosa, Amanda Souza Silva-Sperba, Helena Abadie Moraesa, Larisse Longoa,b, Bruna Concheski de Mouraa, Matheus Truccolo Michalczuka,c, Carolina Uribe-Cruza,b, Carlos Thadeu Schmidt Cerskia,d, Themis Reverbel da Silveiraa, Valesca Dall'Albaa,b,e, Mário Reis Álvares-da-Silvaa,b,c,
Corresponding author
marioreis@live.com

Corresponding author: Mario Reis Alvares-da-Silva, GI/Liver Division, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2350 sala 2033, 90035-903 Porto Alegre, Rio Grande do Sul, Brazil
a Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Rio Grande do Sul, Brazil
b Experimental Laboratory of Hepatology and Gastroenterology, Center for Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, Rio Grande do Sul, Brazil
c Division of Gastroenterology, HCPA, Porto Alegre 90035-903, Rio Grande do Sul, Brazil
d Unit of Surgical Pathology, HCPA, Porto Alegre 90035-903, Rio Grande do Sul, Brazil
e Division of Nutrition, HCPA, Porto Alegre 90035-903, Rio Grande do Sul, Brazil
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (1)
Additional material (1)
Abstract
Introduction and Objectives

Cardiovascular disease (CVD) is the major cause of death in non-alcoholic fatty liver disease (NAFLD), a clinical condition without any approved pharmacological therapy. Probiotics are often indicated for the disease, but their results are controversial in part due to the poor quality of studies. Thus, we investigated the impact of 24-week probiotics supplementation on cardiovascular risk (CVR) in biopsy-proven non-alcoholic steatohepatitis (NASH) patients.

Patients and Methods

Double-blind, placebo-controlled, single-center study (NCT03467282), adult NASH, randomized for 24 weeks daily sachets of probiotic mix (109CFU of Lactobacillus acidophilus, Lactobacillus rhamnosus, Lactobacillus paracasei and Bifidobacterium lactis) or placebo. Clinical scores (atherogenic indexes, atherosclerotic cardiovascular disease-ASCVD and systematic coronary risk evaluation-SCORE), biochemistry, miR-122, miR-33a, plasminogen activator inhibitor-1 (PAI-1), intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), were determined before and after the intervention.

Results

Forty-six patients were enrolled (23 received probiotics and 23 placebo), with a mean age of 51.7 years, most of them females and whites. Clinical and demographic features were similar between the groups at the baseline. The Median NAFLD activity score was 4.13 in both groups. Fibrosis was mild in most patients (15.2% and 65.2% F0 and F1, respectively). Treatment did not promote any clinically significant changes in body mass index or laboratory, including lipid and glucose profile. High CVR patients through atherogenic indexes decreased from baseline in both groups, as well as PAI-1 and miR-122 levels, although there was no difference between probiotics and placebo.

Conclusions

A 24-week probiotic mix administration was not superior to placebo in reducing CVR markers in patients with NASH.

Keywords:
non-alcoholic fatty liver disease
probiotics
microRNAs
cardiovascular risk
clinical trial
Abbreviations:
AC
ALT
APRI
AST
ASCVD
BMI
ASV
CRI
CVD
CVR
ELSA
GM
HDL
HOMA-IR
IPAQ
LDL
MAFLD
MS
NAFLD
NFS
NASH
PAI-1
SCORE
TC
T2DM
VCAM-1
WC
Full Text
1Introduction

Non-alcoholic fatty liver disease (NAFLD), recently renamed metabolic-associated fatty liver disease (MAFLD) [1], is the leading cause of chronic liver disease worldwide [2], with an estimated prevalence between 30-40% in the general population [3, 4]. The increase in the prevalence of this disease has caused a great impact on the clinical and economic burden on society, such that nearly 1 billion people globally are affected [5]. The term NAFLD comprises liver conditions varying in the severity of the injury as hepatic steatosis and non-alcoholic steatohepatitis (NASH). The more advanced the disease (NASH, especially with fibrosis), the greater the cardiovascular risk (CVR) [6].

Cardiovascular disease (CVD) is the major cause of death in NAFLD patients [7], independently of other traditional CVR factors or metabolic syndrome (MS) [4, 8]. Abnormalities in microRNAs, inflammatory and endothelial injury markers, as well as gut dysbiosis can alter NAFLD and CVR [9–12]. Inflammation has been linked to coronary syndrome [13]. Soluble forms of vascular adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) are over-regulated in patients with higher hepatic fat content, suggesting greater endothelial dysfunction and vascular injury [14], as well as endothelial markers such as plasminogen activator inhibitor-1 (PAI-1) [15] and pro-coagulation factors such as fibrinogen [15]. NAFLD is also associated with the dysregulation of many microRNAs, responsible for the regulation of gene expression at the post-transcriptional level and also the synthesis of pro-inflammatory cytokines, culminating in the influx of inflammatory cells into the liver and activation of stellate cells, mechanisms linked to the progression of the clinical picture [16–18]. miR-122 is a liver-characteristic miRNA that composes about 70% of the total miRNAs found in normal hepatocytes, most probably due to the fact that it positively regulates the accumulation of cholesterol and triglycerides and the metabolism of fatty acids [19]. Additionally, miR-33a inhibits genes involved in HDL synthesis and reverse cholesterol transport, which may contribute to the development of NAFLD-related metabolic disorders and CVD [16].

Probiotics are living microorganisms that provide benefits to the host [20]. They can modulate the pathways of liver inflammation and improve lipid and glycemic profiles [21]. Despite their importance, CVD and CVR are outcomes rarely evaluated in NASH studies [22–25]. The effect of probiotics on human CVR is still conflicting [10, 26-28] and studies focusing CVR effects of probiotics in NAFLD patients are lacking. This study aims to evaluate the impact of probiotics supplementation on CVR markers in patients with biopsy-proven NASH.

2Patients and methods

Triple-blind, randomized, placebo-controlled, single-center study carried out in a university hospital in southern Brazil - NCT03467282 with the aim of evaluating CVR as the primary outcome through clinical scores, microRNAs, inflammatory and adherence molecules. Adult patients (>18 years) presenting with NASH (liver biopsy less than one year before inclusion) were enrolled from January to June 2018. Patients with cirrhosis and those infected with human immunodeficiency, hepatitis B or hepatitis C virus, with significant alcohol intake (> 15g ethanol/day) were excluded, as well as pregnant women, transplant recipients, patients using immunosuppressant, corticosteroids, valproic acid, tetracycline and amiodarone, and those carriers of other chronic inflammatory diseases and history of diarrhea. Patients using antibiotics were also excluded or included after three months of withdrawal. Potentially eligible patients were identified at the NAFLD outpatient clinic. The patient's eligibility was confirmed by the responsible researcher. As there are no previous studies considering CVR in NASH patients receiving probiotics, a convenience sample was used. This study is registered at ClinicalTrials.gov as NCT0346782.

2.1Randomization and intervention

Patients were allocated through a randomization list made by an online program (randomization.com website) being divided into two groups: intervention (probiotics) and control (placebo). Patients allocated to the intervention group received mixed probiotics supplementation, which consists of a 1g sachet containing Lactobacillus acidophilus NCFM (1 × 109 CFU) + Lactobacillus rhamnosus HN001 (1 × 109 CFU) + Lactobacillus paracasei LPC-37 (1 × 109 CFU) + Bifidobacterium lactis HN019 (1 × 109 CFU) while those allocated to the placebo group received a 1g sachet with an identical appearance (physical and organoleptic) containing polydextrose/maltodextrin as the placebo. Patients were instructed to ingest two sachets daily with water at room temperature for a period of 24 weeks. All patients received diet and physical activity general guidelines. Every 45 days, patients were instructed to return for a follow-up appointment, carrying notes on a standard spreadsheet to check adherence to the supplement, possible side effects, or even unusual medications (the participants were instructed to advise the research team about the need to use any other non-routine medications and to inform the team when they use a product that contains probiotics).

Patients and the researchers administering the study did not know the composition of each sachet of supplements and the participant's allocation treatment. An external researcher was unblinded. Researchers will know which supplements each participant received only at the end of the study. The external researcher was informed about the composition of each supplement if needed.

2.2Diagnosis and liver histology

Liver biopsy was done in the Hospital setting and reviewed for histological examination by a blinded pathologist, graded through NASH-Clinical Research Network, NAFLD activity score (NAS) and steatosis-activity-fibrosis (SAF) [29, 30].

2.3Clinical and physical evaluation

A medical doctor carried out the evaluations. Clinical evaluation included data on age, sex and ethnicity. The presence of T2DM, hypertension, hypothyroidism, dyslipidemia with or without treatment and current or previous smoking was seen before the interventions started and after the 24-week follow-up. Hypertension, dyslipidemia and T2DM diagnosis followed the 2018, 2016 and 2013 European Society of Cardiology guidelines, respectively [31–33]. All medications taken by the patient were analyzed. Family history of coronary artery disease in a first-degree relative [34] was asked. The presence of acute myocardial infarction, coronary syndrome, previous arterial revascularization and stroke were analyzed before the study. During the same, the presence of coronary syndrome or the presence of cardiovascular events was seen.

Anthropometric variables (body mass index [BMI] and waist circumference [WC]) were evaluated before and after the follow-up period by experienced nutritionists. Blood pressure was always checked by the same researcher. For the diagnosis of MS, the presence of ≥ 3 of the criteria presented in the study conducted by Alberti et al. [35], and for the purpose of WC the database of the ELSA study (Longitudinal Study of Adult Health) was used (men ≥ 92 cm, women ≥ 86 cm) [36].

2.4Physical-activity assessment

The International Physical Activity Questionnaire – Short Form (IPAQ) was applied to evaluate the weekly time spent in physical exercise before the interventions start and after de 24-week follow-up period [37].

2.5Laboratory assessment

We conducted blood laboratory analysis before and after the intervention, consisting of complete blood count, liver enzymes, factor VII, fibrinogen, C-reactive protein (CRP) lipid and glucose profile, and these evaluations were performed using the Labmax 560 equipment. Insulin resistance was determined by the homeostasis model assessment for insulin resistance (HOMA-IR) [38].

2.6Cardiovascular risk scores

The CVR scores were assessed before and after the intervention. The American College of Cardiology Atherosclerotic Cardiovascular Disease (ASCVD) score was used. Patients were classified as low CVR (<5%), borderline (5-7.4%), intermediate (7.5 - 19.9%) ​​and high CVR (≥ 20%) [39]. Framingham score was also calculated for all patients, classifying them in low CVR (≤ 10%), intermediate (10-20%), or high CVR (≥ 20%) [40]. Additionally, the European Society of Cardiology (SCORE) was performed, categorizing patients into different grades: very high ≥ 10%, high ≥ 5% and <10%, moderate ≥ 1 and < 5%, and low CVR < 1% [41]. The presence of altered high-density lipoprotein (HDL) and elevated WC was also investigated; the latter was seen using the usual patterns [35] and the ELSA study [36].

Atherogenic ratios were calculated using the results of the lipid profile to predict CVR. Lipid profile included low-density lipoprotein (LDL), HDL and total cholesterol (TC). Such ratios were calculated in the following ways: Castelli's Risk Index (CRI)-I = TC/HDL, CRI-II= LDL/HDL and Atherogenic Coefficient (AC) = (TC–HDL)/HDL [42]. The cut-off values for atherogenic indices were obtained from previous studies and stratified by sex [43, 44]. Low risk was considered if CRI-I > 3.5 for men and > 3.0 for women; CRI-II and AC values were considered low risk when less than 3.0 and 2.0, respectively, for both sexes [45].

2.7Analysis of markers of endothelial dysfunction

To detect serum changes before and after the intervention in the endothelial dysfunction markers, we analyzed ICAM-1 and vascular cell adhesion molecule-1 (VCAM-1) using the multiplex assay of the Luminex platform (Millipore, Germany). The results were expressed in ng/mL. Serum evaluation of the PAI-1 was performed using the ELISA kit (Invitrogen, USA). The absorbance was measured in a spectrophotometer at a wavelength of 450 nm (Zenyth 200 rt) and the results were expressed in pg/mL. All processes were performed according to the manufacturer's instructions and analyzes were performed in duplicate.

2.8Analysis of the circulating microRNAs

Total RNA was extracted from serum using miRNeasy serum/plasma kit (Qiagen, USA) to analyze the circulating microRNAs before and after the intervention. Then, cel-miR-39 (1.6 × 108 copies) spike in control (Qiagen, USA) was added to provide an internal reference. cDNA conversion was performed from 10ng of total RNA using the TaqMan microRNA Reverse Transcription kit (Applied Biosystems, USA). Analysis of the gene expression of miR-122 and miR-33a, together with the cell-miR-39 normalizer, was performed by RT-qPCR using TaqMan assay (Applied Biosystems, USA). The sequences and codes of the assessed microRNAs are described in Supplementary Table 1. Values were calculated by formula 2 −(ΔΔCt)[46].

2.9Sample size and statistical analysis

Sample size estimation was performed using the WINPEPI 11.20 program (Brixton Health, Israel), based on a published study that found a mean reduction in fibrosis score of 9.36 ± 1.9 to 6.38 ± 1.5 in NAFLD patients taking symbiotic supplementation (p < 0.001, compared to placebo) [47]. Thus, considering a power of 90% and a significance of 5%, adding 10% to compensate for eventual losses, it will be necessary to include 46 patients with NAFLD in the present study.

Normality was verified for all variables using the Shapiro-Wilk test and histograms. Quantitative variables were described as mean and standard deviation / standard error and categorical variables as absolute and relative frequencies. Comparisons over time were evaluated using the Generalized Estimation Equations model complemented by the Least Significant Difference test. The comparison of means was performed by the t-student test and the comparison of proportions by the chi-square test or Fisher's exact test. The level of significance adopted was 5% (p <0.05) and the analyzes were performed using the SPSS version 21.0 program.

2.10Ethical statement

Written informed consent was obtained from each patient included in the study and the study protocol conforms to the ethical guidelines of the 1975 Declaration of Helsinki as reflected in a priori approval by the Hospital de Clínicas de Porto Alegre Ethics Committee (CAAE 86120718.6.0000.5327 and CAAE 97777318.2.0000.5327).

3Results

During the study period, 239 patients were evaluated, of which 46 met the inclusion criteria, were randomized and effectively underwent the intervention (Fig. 1). During the study, one patient was lost to follow-up in each group.

Fig. 1.

Consort diagram. E, exclusion; HCV, hepatitis C virus, I, inclusion, NASH, non-alcoholic steatohepatitis

(0.4MB).
3.1Baseline Profile and Safety

The clinical and demographic baseline data of the patients under study are shown in Table 1. The study sample had a mean age of 51.7 years and a predominance of females and white ethnicity in both groups. As shown, there was no statistically significant difference between the placebo and the probiotics group regarding BMI, WC, smoking, cardiovascular disease, MS features and exercise, although patients in the placebo group presented a higher percentage of T2DM and MS than those in the probiotics group. There was also a statistical difference concerning the presence of previous cardiovascular disease, with predominance in the placebo group, and although the randomization could not separate those patients as equal, we adjusted the cardiovascular variables for these features.

Table 1.

Clinical and demographic data of patients under study

Variables#  Placebo (n = 23)  Probiotic (n = 23)  p 
Age (years)  51.7 ± 11.9  51.7 ± 11.4  0.990 
Gender       
Female  12 (52.2)  15 (65.2)  0.549
Male  11 (47.8)  8 (34.8) 
Ethnicity       
White  19 (82.6)  20 (87.0)  1.000
Non-white  4 (17.4)  3 (13.0) 
Menopause  9 (64.3)  9 (60.0)  1.000 
Mean WC  104.2 ± 11  105.143 ± 13.1  0.801 
Mean BMI  32.3 ± 5.6  32.852 ± 7.1  0.767 
Smoking       
Current  1 (4.3)  1 (4.3)  1.000 
Previous  4 (17.4)  9 (39.1)  0.190 
Previous cardiovascular disease  6 (26.1)  1 (4.3)  0.029 
Atherosclerosis*  3 (13)  4 (17.4)  1.000 
Family history⁎⁎  6 (26.1)  7 (30.4)  1.000 
Hypothyroidism  3 (13)  6 (26.1)  0.457 
Dyslipidemia  11 (47.8)  11 (47.8)  1.000 
Hypertension  14 (60.9)  16 (69.6)  0.757 
Diabetes  13 (56.5)  9 (39.1)  0.376 
Metabolic syndrome  19 (82.6)  16 (69.6)  0.489 
IPAQ       
Low  9 (39.1)  9 (39.1)  0.909
Moderate  10 (43.5)  11 (47.8) 
High  4 (17.4)  3 (13.0) 
#

Variables described as mean ± standard deviation or frequency (%).

Atherosclerosis defined by abdominal ultrasound before the study

⁎⁎

Family history of cardiovascular disease defined by first-degree relative male <55 years old or female <65 years old. BMI, body mass index, IPAQ, International Physical Activity Questionnaire, WC, waist circumference

Baseline liver biopsy findings are shown in Table 2. There was no significant difference between the two groups. All included patients had NASH (NAS 4.13 ± 0.87 - placebo group; and 4.13 ± 0.87 - probiotics group; p = 1.000). Most cases were of absent (F0) or mild (F1) fibrosis (19 - placebo group; 18 - probiotics group; p = 0.730). Only five patients taking placebo and four receiving probiotics had intermediate (F2) or advanced (F3) fibrosis (p = 0.730). Twenty-three patients received placebo and twenty-three patients received probiotics. At the end of 24 weeks, 44 patients completed the study. Antibiotics were used by six patients in the placebo group (26.1%) and four (17.4%) in the probiotics group (p = 0.721). No patient has any adverse event.

Table 2.

Histological findings assessed according to NAS and SAF

Variables#  Placebo (n = 23)  Probiotic (n = 23)  p 
NAS  4.13 ± 0.87  4.13 ± 0.87  1.000 
Steatosis       
Mild  9 (39.1)  9 (39.1)  0.654
Moderate  10 (43.5)  12 (52.2) 
Severe  4 (17.4)  2 (8.7) 
Lobular inflammation       
Mild  17 (73.9)  20 (87.0)  0.459
Moderate  6 (26.1)  3 (13.0) 
Ballooning       
Mild  21 (91.3)  16 (69.6)  0.135
Moderate  2 (8.7)  7 (30.4) 
SAF score       
S - Steatosis       
9 (39.1)  11 (47.8)  0.799
11 (47.8)  10 (43.5) 
3 (13.0)  2 (8.7) 
A - Inflammation activity       
4 (17.4)  3 (13)  0.841
11 (47.8)  14 (60.9) 
7 (30.4)  5 (21.7) 
1 (4.3)  1 (4.3) 
F - Fibrosis       
4 (17.4)  3 (13.0)  0.730
14 (60.9)  16 (69.6) 
1 (4.3)  2 (8.7) 
4 (17.4)  2 (8.7) 
#

Variables described as mean ± standard deviation or frequency (%). Mild lobular inflammation: < 2 foci in a 20-x field; moderate lobular inflammation: 2-4 foci in a 20-x field. A, inflammation activity, F, fibrosis; NAS, NAFLD activity score; S, steatosis; SAF, steatosis-activity-fibrosis

3.2Comparison between Baseline and End of Study Results

The mean weight, BMI and WC were not statistically different between placebo and probiotics before and after the intervention, as well as mean systolic and diastolic blood pressure and IPAQ (Supplementary Table 2 and Supplementary Fig. 1). The baseline and final laboratory are shown in Table 3 and Supplementary Table 3. As for the findings referring to CRP, creatine kinase, fibrinogen and factor VII, there was no difference after intervention in both groups (Supplementary Table 4). Combining high WC and low HDL (according to the International Diabetes Federation or ELSA Study recommendations), there were no differences between the two groups either in the baseline or after intervention (Supplementary Table 5).

Table 3.

Comparison of laboratory tests before and after intervention

Variables#  Placebo (n = 23)  Probiotic (n = 23)  p 
AST (U/L)       
Before  39.3 ± 4.80  38.2 ± 5.34  0.880 
After  37.6 ± 5.65  36.6 ± 4.18  0.887 
Difference % (CI 95%)  -1.66 (-7.41 to 4.07)  -1.57 (-10.2 to 7.12)  0.994 
p  0.570  0.722   
ALT (U/L)       
Before  50.8 ± 5,24  49.6 ± 8.13  0.896 
After  50.0 ± 6,99  49.4 ± 7.91  0.959 
Difference % (CI 95%)  -0.87 (-12.0 to 10.3)  -0.15 (-12.1 to 11.8)  0.933 
P  0.879  0.980   
GGT (U/L)       
Before  101.1 ± 46.7  81.8 ± 19.6  0.704 
After  114.8 ± 61.0  98.7 ± 32.1  0.816 
Difference % (CI 95%)  13.6 (-16.1 to 43.4)  16.9 (-17.3 to 51.1)  0.735 
p  0.369  0.333   
Fasting glucose (mg/dL)       
Before  124.8 ± 9.32  103.8 ± 6.34  0.063 
After  120.8 ± 6.45  110.7 ± 7.50  0.310 
Difference % (CI 95%)  -4.00 (-17.1 to 9.09)  6.90 (-3.45 to 17.2)  0.200 
p  0.549  0.191   
Insulin (lU/L)       
Before  17.1 ± 1.94  22.6 ± 3.99  0.213 
After  17.6 ± 2.19  19.46 ± 2.96  0.619 
Difference % (CI 95%)  0.464 (-2.42 to 3.35)  -3.22 (-7.61 to 1.16)  0.162 
p  0.753  0.150   
HbA1C (%)       
Before  6.38 ± 0.29  6.10 ± 0.24  0.473 
After  6.44 ± 0.26  6.44 ± 0.32  0.991 
Difference % (CI 95%)  0.05 (-0.36 to 0.47)  0.33 (0.01 to 0.65)  0.292 
p  0.801  0.037   
HOMA IR       
Before  5.52 ± 0.94  5.65 ± 0.83  0.919 
After  5.30 ± 0.80  5.70 ± 1.24  0.791 
Difference % (CI 95%)  -0.22 (-1.87 to 1.43)  0.04 (-1.43 to 1.52)  0.811 
p  0.792  0.954   
Total cholesterol (mg/dL)       
Before  173.0 ± 8.38  178.6 ± 6.46  0.593 
After  171.7 ± 9.17  184.7 ± 8.65  0.304 
Difference% (CI 95%)  -1.22 (-11.4 to 8.96)  6.07 (-3.09 to 15.2)  0.297 
p  0.813  0.194   
HDL (mg/dL)       
Before  44.1 ± 2.60  46.7 ± 2.67  0.492 
After  45.5 ± 2.98  46.9 ± 2.38  0.722 
Difference % (CI 95%)  1.37 (-1.98 to 4.72)  0.17 (-2.26 to 2.60)  0.570 
p  0.423  0.891   
LDL (mg/dL)       
Before  97.9 ± 7.50  98.9 ± 6.26  0.920 
After  95.5 ± 7.72  103.1 ± 7.71  0.483 
Difference % (CI 95%)  -2.48 (-10.5 to 5.60)  4.20 (-4.73 to 13.1)  0.277 
Triglycerides (mg/dL)       
Before  169.7 ± 17.5  179.0 ± 18.8  0.719 
After  153.6 ± 16.2  170.0 ± 14.7  0.436 
Difference % (CI 95%)  -16.0 (-42.3 to 10.2)  -8.22 (-42.9 to 26.5)  0.688 
p  0.232  0.643   
#

Variables described as mean ± standard deviation and confidence interval (CI 95%). ALT, alanine aminotransferase, AST, aspartate aminotransferase, GGT, gamma-glutamyl transferase, HbA1C, glycosylated hemoglobin, HDL, high-density lipoprotein, HOMA-IR, homeostasis model assessment for insulin resistance, LDL, low-density lipoprotein

The evaluation of the CVR by clinical scores in qualitative (considering only high-risk cases) form before and after the 24 weeks of intervention can be seen in Table 4. There was a significant decrease in CRI and AC in the placebo group (p = 0.045 and 0.048, respectively) and a smaller decrease in the probiotics group (p = 0.058 for both). There were no differences in the absolute value of CRI-I, CRI-II and AC between the groups (Supplementary Table 6). Applying ASCVD, SCORE and Framingham, there was no difference between groups (Supplementary Fig. 2). Endothelial lesion markers and microRNAs before and after intervention are shown in Table 5. There was no significant difference between the two groups. However, from the baseline to the end of the study, PAI-1 levels significantly (p < 0.001) decreased in both groups, as well as miR-122. After the intervention, the placebo and probiotic groups had no alteration of the CVR by ASCVD and SCORE. The variation of miR-122 and PAI-1 resulted in a decrease in CVR by 95% and 72.7%, respectively, in the probiotic group, without significance. However, in the placebo group, there was a decrease in CVR in 82.4% and 95.5% of patients with a variation of miR-122 and PAI-1, respectively, without significance.

Table 4.

Evaluation of the cardiovascular risk by clinical scores before and after intervention considering high-risk patients

Variables#  Placebo (n = 23)  Probiotic (n = 23)  p 
CRI-I: high risk       
Before  18 (78.3)  22 (95.7)  0.070 
After  14 (63.6)  18 (81.8)  0.167 
Difference % (CI 95%)  -14.6 (-28.9 to 0.00)  -13.8 (-28.2 to 0.00)  0.377 
p  0.045  0.058   
CRI-II: high risk       
Before  4 (17.4)  0 (0.0)  0.028 
After  7 (31.8)  3 (13.6)  0.140 
Difference % (CI 95%)  14.4 (-4.5 to 33.3)  13.6 (-0.7 to 27.9)  1.000 
p  0.134  0.062   
AC: high risk       
Before  19 (82.6)  22 (95.7)  0.146 
After  15 (68.2)  18 (81.8)  0.290 
Difference % (CI 95%)  -14.4 (-28.7 to -0.00)  -13.8 (-28.2 to 0.00)  0.428 
p  0.048  0.058   
#

Variables described as mean ± standard deviation or frequency (%) and confidence interval (CI 95%). AC, Atherogenic Coefficient and CRI, Castelli's Risk Index

Table 5.

Endothelial lesion markers and microRNAs before and after intervention

Variables#  Placebo (n = 23)  Probiotic (n = 23)  p 
PAI-1 (pg/ml)       
Before  5379.5 ± 108.1  5457.8 ± 86.4  0.572 
After  4617.2 ± 230.4  4500.5 ± 179.3  0.689 
Difference % (CI 95%)  -762.3 (-1196.4 to -327.7)  -957.3 (-1229.4 to -685.2)  0.456 
p  0.001  <0.001   
VCAM-1 (ng/ml)       
Before  10.0 ± 0.54  9.73 ± 0.72  0.706 
After  15.7 ± 1.05  15.8 ± 1.47  0.977 
Difference % (CI 95%)  5.67 (3.86 to 7.48)  6.06 (3.99 to 8.13)  0.779 
p  <0.001  <0.001   
ICAM-1 (ng/ml)       
Before  0.59 ± 0.28  0.66 ± 0.39  0.174 
After  0.80 ± 0.21  0.77 ± 0.03  0.434 
Difference % (CI 95%)  0.21 (0.13 to 0.28)  0.11 (0.005 to 0.21)  0.126 
p  <0.001  0.038   
miR-122       
Before  2.43 ± 0.92  1.69 ± 0.63  0.509 
After  0.35 ± 0.99  0.38 ± 0.12  0.838 
Difference % (CI 95%)  -2.08 (-3.93 to -0.22)  -1.30 (-2.56 to -0.04)  0.515 
p  0.028  0.042   
miR-33a       
Before  1.53 ± 0.48  1.73 ± 0.43  0.759 
After  2.09 ± 0.50  2.84 ± 0.59  0.342 
Difference % (CI 95%)  0.56 (-1.03 to 2.16)  1.10 (-0.55 to 2.76)  0.306 
p  0.488  0.191   
#

Variables described as mean ± standard deviation and confidence interval (CI 95%). ICAM-1, intercellular adhesion molecule-1; PAI-1, plasminogen activator inhibitor-1; VCAM-1, vascular cell adhesion molecule-1

During the study, one patient in the placebo group had an acute myocardial infarction, one had stable angina and one patient had subepicardial ischemia (p = 0.223). No cardiovascular events were observed in the probiotics group.

4Discussion

In this study, oral supplementation with probiotics for a 24-week period did not promote any significant changes in CVR markers in comparison to placebo. Indeed, PAI-1 and miR-122 decreased after the intervention, but this difference occurred in both groups, not only in those who received probiotics. To the best of our knowledge, the present study is the first triple-blinded random control trial with probiotics in biopsy-proven NASH patients intended to evaluate CVR. This study fulfills the expectation regarding random sequence generation (selection bias), allocation concealment (selection bias), blinding of participants and personnel (performance bias), and blinding of outcome assessment (detection bias). Thus, it seems to have internal validity and, since it included common patients in clinical practice, external validity.

Probiotics are promising for NAFLD therapy due to their relatively easy availability, low cost, and absence of serious side effects [48], but their use is still quite controversial. Recently, a randomized, double-blind, placebo-controlled study evaluated whether probiotic supplementation for six months is able to improve hepatic steatosis, fibrosis, and other clinical markers in NAFLD patients [49]. This study demonstrated that the use of probiotics did not promote significant clinical improvement in the patient with NAFLD; however, at the microenvironment level, their use was effective in controlling the increase in intestinal permeability [49]. In fact, the use of probiotics, prebiotics and symbiotics has been considered a potential and promising strategy to regulate the intestinal microbiota. However, they are not able to play a healing role. Its use can be an adjuvant therapy in pathological processes involving NAFLD and its spectra, either by improving the intestinal barrier or preventing the formation of metabolites toxic to the liver, or acting on the immune system [50]. More studies with larger sample sizes, longer duration and different strains are needed to assess the real benefit of probiotics in NAFLD, as their therapeutic use is not supported by high-quality clinical studies to date [51]. There are previous random control trials using different strains and doses of probiotics that demonstrated some interesting effects on blood glucose, insulin resistance, lipid profile and MS [52, 53]. However, even meta-analysis differs on the usefulness of probiotics in patients with NAFLD [21, 53, 54].

Regarding demographic and clinical variables, such as age, gender and comorbidities, the population included was quite consistent with previously published studies [3, 55-58]. Except for the history of CVD and the presence of T2DM, which were higher in placebo, all other baseline variables were similar between the groups. All included patients had biopsy-proven NASH (NAS ≥ 4). This is important not only to verify the liver injury but also because CVR seems to strongly depend on the presence of more advanced fatty liver disease [59]. However, most of the patients in both groups presented grade 1 fibrosis, and this mild disease could probably exert an influence on the CVR.

Our research group has been developing studies with the objective of evaluating the cardiovascular manifestations associated with NAFLD since CVD is the main cause of death and is an outcome that has been poorly evaluated [16, 45]. In fact, CVD associated with NAFLD takes a long time to cause clinical consequences, so we chose, in this study, to carry out CVR assessments through molecular analysis and scores applied in clinical practice. As there is a lack of studies that assess CVR as the main outcome in patients with NASH who underwent probiotic supplementation, and considering that CVR is related to the presence of fibrosis, the sample size calculation was based on the reduction of hepatic fibrosis. Inflammation and oxidative stress seem to be involved in the onset and progression of NAFLD [88] and the production of PAI-1 may be over-regulated by inflammatory factors [60]. These mechanisms and mutual interactions seem to explain the association between NAFLD and CVD [61]. Increased levels of PAI-1 are predictors of future cardiovascular events and have been reported in patients with coronary artery disease [62]. Our study shows an improvement in PAI-1 in both groups, more pronounced in the probiotics group. Our finding may show an improvement in CVR based on the study by Jung et al., where PAI-1 levels were higher in a patient who had major cardiovascular events [60]. Besides that, Tofler et al. concluded that the analyses of PAI-1 are predictive of CVD after considering the established risk factors [63]. Although the patients in this study had a diagnosis of NASH, most had mild or absent fibrosis, preventing the performance of a stratified analysis of the impact of probiotic supplementation on CVR according to the NAS score as would be advisable [64].

Due to the role of microRNAs in regulating metabolic pathways (like lipogenesis, glycolysis, gluconeogenesis) and also the association of microRNAs with oxidative stress, they are considered biomarkers and potential therapeutic targets for NAFLD [9]. Especially, miR-122 is quite important in liver diseases, including NAFLD [65]. In our study, it decreased after intervention in both groups. This finding is consistent with the results we observed in PAI-1 since miR-122 has also been considered a potential biomarker for the diagnosis and prognosis of CVD, mainly with the presence and severity of coronary artery disease, independent of other CVR factors [66]. No deaths occurred during the study, perhaps due to the reduced number of participants and the length of clinical follow-up. However, during the study in the placebo group, we can observe the appearance of some cardiovascular events, highlighting the high CVR these patients often present.

In this randomized clinical trial, there was no benefit from the use of probiotics over CVR. Studies with a number of patients and time similar to ours have shown discrepant results, some showing improvement and another worsening of profiles related to CVR and use of probiotics. Duseja et al. [26] conducted a randomized, double-blind, multicenter study with several strains of probiotics for 12 months in 39 Indian patients with NAFLD without T2DM. Patients who used probiotics had a significantly greater reduction in ALT and inflammatory cytokines compared to placebo. In the study by Wong et al. [67], biopsy-proven NASH patients were randomized to receive probiotics (multiple strains of 20 × 106 CFU; 10 patients) or no medication (10 patients) for six months. Metabolic parameters were evaluated but without any other evaluation of the RCV. The use of probiotics was not associated with changes in BMI, waist circumference, glycemic and lipid profile. Therefore, this relationship between probiotics with RCV is still open.

This study has some strengths, such as its triple blinding, the strict monitoring of patients, the high adherence index, the inclusion of only biopsy-proven NASH, and the investigative approach regarding different possibilities of CVR assessment, such as inflammatory markers, microRNAs, and also the evaluation through questionnaires and physical examination. However, it does present some limitations, like being a single-center study, the number of patients, the short period of treatment, and the low severity of NASH patients included. Furthermore, this study does not apply to the suggested concept of MAFLD, meaning that there were excluded patients with other causes of liver diseases, such as viruses or alcohol, among others [45].

5Conclusions

In summary, in this double-blind placebo-controlled randomized clinical trial, probiotics supplementation was not able to significantly decrease CVR markers in comparison to placebo in NASH patients.

Author contributions

Barcelos STA, Silva-Sperb A, Moraes HA, Longo L, Moura BC, Uribe-Cruz C, Silveira TR, Dall'Alba V and Álvares-da-Silva MR performed the conceptualization, methodology, formal analysis, investigation, data curation, writing of the original draft, writing-review, and editing; Michalczuk MT and Cerski CT performed the methodology and formal analysis; writing review and editing.

Declaration of Interest

None.

Funding

This study is financed by the Research and Events Fund from the Hospital de Clínicas de Porto Alegre (FIPE), Coordination for the Improvement of Higher Education Personnel (CAPES/PROAP), National Council for Scientific and Technological Development – Brazil (CNPq, Universal 1/2016). We thank the Farmoquimica Company for the probiotics donated. No funding sources are involved in the study design, data collection, analysis, and interpretation of data.

References
[1]
M Eslam, PN Newsome, SK Sarin, QM Anstee, G Targher, M Romero-Gomez, et al.
A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement.
J Hepatol, 73 (2020), pp. 202-209
[2]
Z Younossi, F Tacke, M Arrese, CB Sharma, I Mostafa, E Bugianesi, et al.
Global perspectives on nonalcoholic fatty liver disease and nonalcoholic steatohepatitis.
Hepatology, 69 (2019), pp. 2672-2682
[3]
ZM Younossi, AB Koenig, D Abdelatif, Y Fazel, L Henry, M. Wymer.
Global epidemiology of non-alcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes.
Hepatology, 64 (2016), pp. 73-84
[4]
R Patil, GK. Sood.
Non-alcoholic fatty liver disease and cardiovascular risk.
World J Gastrointest Pathophysiol, 8 (2017), pp. 51-58
[5]
M Eslam, AJ Sanyal, J George, IC. Panel.
MAFLD: a consensus-driven proposed nomenclature for metabolic associated fatty liver disease.
Gastroenterology, 158 (2020), pp. 1999-2014
[6]
R Aller, C Fernández-Rodríguez, O Lo Iacono, R Bañares, J Abad, JA Carrión, et al.
Consensus document. Management of non-alcoholic fatty liver disease (NAFLD). Clinical practice guideline.
Gastroenterol Hepatol, 41 (2018), pp. 328-349
[7]
AC Sheka, O Adeyi, J Thompson, B Hameed, PA Crawford, S. Ikramuddin.
Nonalcoholic steatohepatitis: a review.
JAMA, 323 (2020), pp. 1175-1183
[8]
LS Bhatia, NP Curzen, PC Calder, CD. Byrne.
Non-alcoholic fatty liver disease: a new and important cardiovascular risk factor?.
Eur Heart J, 33 (2012), pp. 1190-1200
[9]
M Gjorgjieva, C Sobolewski, D Dolicka, M Correia de Sousa, M. Foti.
miRNAs and NAFLD: from pathophysiology to therapy.
[10]
A Dixon, K Robertson, A Yung, M Que, H Randall, D Wellalagodage, et al.
Efficacy of probiotics in patients of cardiovascular disease risk: a systematic review and meta-analysis.
Curr Hypertens Rep, 22 (2020), pp. 74-101
[11]
D Ferro, F Baratta, D Pastori, N Cocomello, A Colantoni, F Angelico, et al.
New insights into the pathogenesis of non-alcoholic fatty liver disease: gut-derived lipopolysaccharides and oxidative stress.
Nutrients, 12 (2020), pp. 2762-2776
[12]
ML Thakur, S Sharma, A Kumar, SP Bhatt, K Luthra, R Guleria, et al.
Non-alcoholic fatty liver disease is associated with subclinical atherosclerosis independent of obesity and metabolic syndrome in Asian Indians.
Atherosclerosis, 223 (2012), pp. 507-511
[13]
ET Yeh, JT. Willerson.
Coming of age of C-reactive protein: using inflammation markers in cardiology.
Circulation, 107 (2003), pp. 370-371
[14]
R Lautamäki, R Borra, P Iozzo, M Komu, T Lehtimäki, M Salmi, et al.
Liver steatosis coexists with myocardial insulin resistance and coronary dysfunction in patients with type 2 diabetes.
Am J Physiol Endocrinol Metab, 291 (2006), pp. 282-290
[15]
G Targher, M Chonchol, L Miele, G Zoppini, I Pichiri, M. Muggeo.
Nonalcoholic fatty liver disease as a contributor to hypercoagulation and thrombophilia in the metabolic syndrome.
Semin Thromb Hemost, 35 (2009), pp. 277-287
[16]
L Longo, PH Rampelotto, E Filippi-Chiela, VEG de Souza, F Salvati, CT Cerski, et al.
Gut dysbiosis and systemic inflammation promote cardiomyocyte abnormalities in an experimental model of steatohepatitis.
World J Hepatol, 13 (2021), pp. 2052-2070
[17]
G. Baffy.
MicroRNAs in nonalcoholic fatty liver disease.
J Clin Med, 4 (2015), pp. 1977-1988
[18]
M Eslam, L Valenti, S. Romeo.
Genetics and epigenetics of NAFLD and NASH: clinical impact.
J Hepatol, 68 (2018), pp. 268-279
[19]
CJ Pirola, T Fernández Gianotti, GO Castaño, P Mallardi, J San Martino, M Mora Gonzalez Lopez Ledesma, et al.
Circulating microRNA signature in non-alcoholic fatty liver disease: from serum non-coding RNAs to liver histology and disease pathogenesis.
[20]
C Hill, F Guarner, G Reid, GR Gibson, DJ Merenstein, B Pot, et al.
Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic.
Nat Rev Gastroenterol Hepatol, 11 (2014), pp. 506-514
[21]
Y Tang, J Huang, WY Zhang, S Qin, YX Yang, H Ren, et al.
Effects of probiotics on non-alcoholic fatty liver disease: a systematic review and meta-analysis.
Therap Adv Gastroenterol, 12 (2019), pp. 1-23
[22]
MJ Westerouen Van Meeteren, JPH Drenth, ETTL. Tjwa.
Elafibranor: a potential drug for the treatment of non-alcoholic steatohepatitis (NASH).
Expert Opin Investig Drugs, 29 (2020), pp. 117-123
[23]
S Traussnigg, JM Schattenberg, M Demir, J Wiegand, A Geier, G Teuber, et al.
Norursodeoxycholic acid versus placebo in the treatment of non-alcoholic fatty liver disease: a double-blind, randomised, placebo-controlled, phase 2 dose-finding trial.
Lancet Gastroenterol Hepatol, 4 (2019), pp. 781-793
[24]
ZM Younossi, V Ratziu, R Loomba, M Rinella, QM Anstee, Z Goodman, et al.
Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial.
Lancet, 394 (2019), pp. 2184-2196
[25]
CP Oliveira, HP Cotrim, JT Stefano, ACG Siqueira, ALA Salgado, ER. Parise.
N-acetylcysteine and/or ursodeoxycholic acid associated with metformin in non-alcoholic steatohepatitis: an open-label multicenter randomized controlled trial.
Arq Gastroenterol, 56 (2019), pp. 184-190
[26]
A Duseja, SK Acharya, M Mehta, S Chhabra, S Rana, A Das, et al.
High potency multistrain probiotic improves liver histology in non-alcoholic fatty liver disease (NAFLD): a randomised, double-blind, proof of concept study.
BMJ Open Gastroenterol, 6 (2019), pp. 1-9
[27]
MY Khan, AB Mihali, MS Rawala, A Aslam, WJ. Siddiqui.
The promising role of probiotic and synbiotic therapy in aminotransferase levels and inflammatory markers in patients with non-alcoholic fatty liver disease - a systematic review and meta-analysis.
Eur J Gastroenterol Hepatol, 31 (2019), pp. 703-715
[28]
PM Bock, GH Telo, R Ramalho, M Sbaraini, G Leivas, AF Martins, et al.
The effect of probiotics, prebiotics or synbiotics on metabolic outcomes in individuals with diabetes: a systematic review and meta-analysis.
Diabetologia, 64 (2021), pp. 26-41
[29]
P Bedossa, C Poitou, N Veyrie, JL Bouillot, A Basdevant, V Paradis, et al.
Histopathological algorithm and scoring system for evaluation of liver lesions in morbidly obese patients.
Hepatology, 56 (2012), pp. 1751-1759
[30]
EM Brunt, DE Kleiner, LA Wilson, P Belt, BA Neuschwander-Tetri.
CRN) NCRN. Non-alcoholic fatty liver disease (NAFLD) activity score and the histopathologic diagnosis in NAFLD: distinct clinicopathologic meanings.
Hepatology, 53 (2011), pp. 810-820
[31]
B Williams, G Mancia, W Spiering, EA Rosei, M Azizi, M Burnier, et al.
2018 ESC/ESH Guidelines for the management of arterial hypertension. The Task Force for the management of arterial hypertension of the European Society of Cardiology (ESC) and the European Society of Hypertension (ESH).
European Heart Journal, (2018), pp. 3021-3104
[32]
AL Catapano, I Graham, G De Backer, O Wiklund, MJ Chapman, H Drexel, et al.
2016 ESC/EAS Guidelines for the Management of Dyslipidaemias.
Rev Esp Cardiol (Engl Ed), 70 (2017), pp. 105-114
[33]
L Rydén, PJ Grant, SD Anker, C Berne, F Cosentino, N Danchin, et al.
ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD: the Task Force on diabetes, pre-diabetes, and cardiovascular diseases of the European Society of Cardiology (ESC) and developed in collaboration with the European Association for the Study of Diabetes (EASD).
Eur Heart J, 34 (2013), pp. 3035-3087
[34]
H Saghafi, MJ Mahmoodi, H Fakhrzadeh, R Heshmat, A Shafaee, B. Larijani.
Cardiovascular risk factors in first-degree relatives of patients with premature coronary artery disease.
Acta Cardiol, 61 (2006), pp. 607-613
[35]
KG Alberti, RH Eckel, SM Grundy, PZ Zimmet, JI Cleeman, KA Donato, et al.
Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity.
Circulation, 120 (2009), pp. 1640-1645
[36]
TR Cardinal, A Vigo, BB Duncan, SMA Matos, MJM da Fonseca, SM Barreto, et al.
Optimal cut-off points for waist circumference in the definition of metabolic syndrome in Brazilian adults: baseline analyses of the Longitudinal Study of Adult Health (ELSA-Brasil).
Diabetol Metab Syndr, 10 (2018), pp. 49-58
[37]
PH Lee, DJ Macfarlane, TH Lam, SM. Stewart.
Validity of the International Physical Activity Questionnaire Short Form (IPAQ-SF): a systematic review.
Int J Behav Nutr Phys Act, 8 (2011), pp. 115-126
[38]
S Chitturi, S Abeygunasekera, GC Farrell, J Holmes-Walker, JM Hui, C Fung, et al.
NASH and insulin resistance: Insulin hypersecretion and specific association with the insulin resistance syndrome.
Hepatology, 35 (2002), pp. 373-379
[39]
DC Goff, DM Lloyd-Jones, G Bennett, S Coady, RB D'Agostino, R Gibbons, et al.
2013 ACC/AHA guideline on the assessment of cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines.
Circulation, 129 (2014), pp. 49-73
[40]
RB D'Agostino, RS Vasan, MJ Pencina, PA Wolf, M Cobain, JM Massaro, et al.
General cardiovascular risk profile for use in primary care: the Framingham Heart Study.
Circulation, 117 (2008), pp. 743-753
[41]
MF Piepoli, AW Hoes, S Agewall, C Albus, C Brotons, AL Catapano, et al.
European Guidelines on cardiovascular disease prevention in clinical practice: The Sixth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of 10 societies and by invited experts): Developed with the special contribution of the European Association for Cardiovascular Prevention & Rehabilitation (EACPR).
Eur J Prev Cardiol 2016, 23 (2016), pp. 1-96
[42]
R Sujatha, S. Kavitha.
Atherogenic indices in stroke patients: a retrospective study.
Iran J Neurol, 16 (2017), pp. 78-82
[43]
S Bhardwaj, J Bhattacharjee, MK Bhatnagar, S. Tyagi.
Atherogenic index of plasma, castelli risk index and atherogenic coefficient- new parameters in assessing cardiovascular risk.
Int J Pharm Biol Sci, 3 (2013), pp. 359-364
[44]
MA Olamoyegun, R Oluyombo, SO. Asaolu.
Evaluation of dyslipidemia, lipid ratios, and atherogenic index as cardiovascular risk factors among semi-urban dwellers in Nigeria.
Ann Afr Med, 15 (2016), pp. 194-199
[45]
GTS Guerreiro, L Longo, MA Fonseca, VEG de Souza, MR. Álvares-da-Silva.
Does the risk of cardiovascular events differ between biopsy-proven NAFLD and MAFLD?.
Hepatol Int, 15 (2021), pp. 380-391
[46]
KJ Livak, TD. Schmittgen.
Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method.
Methods, 25 (2001), pp. 402-408
[47]
T Eslamparast, H Poustchi, F Zamani, M Sharafkhah, R Malekzadeh, A. Hekmatdoost.
Synbiotic supplementation in non-alcoholic fatty liver disease: a randomized, double-blind, placebo-controlled pilot study.
Am J Clin Nutr, 99 (2014), pp. 535-542
[48]
AS Lavekar, DV Raje, T Manohar, AA. Lavekar.
Role of probiotics in the treatment of nonalcoholic fatty liver disease: a meta-analysis.
Euroasian J Hepatogastroenterol, 7 (2017), pp. 130-137
[49]
MH Mohamad Nor, N Ayob, NM Mokhtar, RA Raja Ali, GC Tan, Z Wong, et al.
The effect of probiotics (MCP ® BCMC ® strains) on hepatic steatosis, small intestinal mucosal immune function, and intestinal barrier in patients with non-alcoholic fatty liver disease.
Nutrients, 13 (2021), pp. 3192
[50]
RZ Carpi, SM Barbalho, KP Sloan, LF Laurindo, HF Gonzaga, PC Grippa, et al.
The effects of probiotics, prebiotics and synbiotics in non-alcoholic fat liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH): a systematic review.
Int J Mol Sci, 23 (2022), pp. 8805
[51]
G Tarantino, C. Finelli.
Systematic review on intervention with prebiotics/probiotics in patients with obesity-related non-alcoholic fatty liver disease.
Future Microbiol, 10 (2015), pp. 889-902
[52]
A Sepideh, P Karim, A Hossein, R Leila, M Hamdollah, EG Mohammad, et al.
Effects of multistrain probiotic supplementation on glycemic and inflammatory indices in patients with nonalcoholic fatty liver disease: a double-blind randomized clinical trial.
J Am Coll Nutr, 35 (2016), pp. 500-505
[53]
J Sun, N. Buys.
Effects of probiotics consumption on lowering lipids and CVD risk factors: a systematic review and meta-analysis of randomized controlled trials.
Ann Med, 47 (2015), pp. 430-440
[54]
Barengolts E.Gut Microbiota.
Prebiotics, probiotics, and synbiotics in management of obesity and prediabetes: review of randomized controlled trials.
Endocr Pract, 22 (2016), pp. 1224-1234
[55]
AM Allen, TM Therneau, JJ Larson, A Coward, VK Somers, PS. Kamath.
Non-alcoholic fatty liver disease incidence and impact on metabolic burden and death: A 20 year-community study.
Hepatology, 67 (2018), pp. 1726-1736
[56]
A Duseja, N. Chalasani.
Epidemiology and risk factors of non-alcoholic fatty liver disease (NAFLD).
Hepatol Int, 7 (2013), pp. 755-764
[57]
N Chalasani, Z Younossi, JE Lavine, M Charlton, K Cusi, M Rinella, et al.
The diagnosis and management of non-alcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases.
Hepatology, 67 (2018), pp. 328-357
[58]
BLCA Oliveira, RR. Luiz.
Racial density and the socioeconomic, demographic and health context in Brazilian cities in 2000 and 2010.
Rev Bras Epidemiol, 22 (2019), pp. 1-12
[59]
G Targher, CD Byrne, H. Tilg.
NAFLD and increased risk of cardiovascular disease: clinical associations, pathophysiological mechanisms and pharmacological implications.
Gut, 69 (2020), pp. 1691-1705
[60]
RG Jung, P Motazedian, FD Ramirez, T Simard, P Di Santo, S Visintini, et al.
Association between plasminogen activator inhibitor-1 and cardiovascular events: a systematic review and meta-analysis.
[61]
C Pisetta, C Chillè, G Pelizzari, MG Pigozzi, M Salvetti, A Paini, et al.
Evaluation of cardiovascular risk in patient with primary non-alcoholic fatty liver disease.
High Blood Press Cardiovasc Prev, 27 (2020), pp. 321-330
[62]
V Xanthakis, DM Enserro, JM Murabito, JF Polak, KC Wollert, JL Januzzi, et al.
Ideal cardiovascular health: associations with biomarkers and subclinical disease and impact on incidence of cardiovascular disease in the Framingham Offspring Study.
Circulation, 130 (2014), pp. 1676-1683
[63]
GH Tofler, J Massaro, CJ O'Donnell, PWF Wilson, RS Vasan, PA Sutherland, et al.
Plasminogen activator inhibitor and the risk of cardiovascular disease: The Framingham Heart Study.
Thromb Res, 140 (2016), pp. 30-35
[64]
M Ekstedt, H Hagström, P Nasr, M Fredrikson, P Stål, S Kechagias, et al.
Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up.
Hepatology, 61 (2015), pp. 1547-1554
[65]
NC Salvoza, DC Klinzing, J Gopez-Cervantes, MO. Baclig.
Association of Circulating Serum miR-34a and miR-122 with dyslipidemia among patients with non-alcoholic fatty liver disease.
[66]
W Gao, HW He, ZM Wang, H Zhao, XQ Lian, YS Wang, et al.
Plasma levels of lipometabolism-related miR-122 and miR-370 are increased in patients with hyperlipidemia and associated with coronary artery disease.
Lipids Health Dis, 11 (2012), pp. 55-63
[67]
VW Wong, GL Won, AM Chim, WC Chu, DK Yeung, KC Li, et al.
Treatment of non-alcoholic steatohepatitis with probiotics. A proof-of-concept study.
Ann Hepatol, 12 (2013), pp. 256-262
Article options
Tools
Supplemental materials
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos