Next Article in Journal
Endoscopic Management of Gastric Disruptions
Previous Article in Journal
In the Driving Seat with High-Resolution Ano-Rectal Manometry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Probiotic Interventions in Coeliac Disease: A Systematic Review with a Focus on Cardiovascular Risk

1
Centre for Integrated Research in Life and Health Sciences, University of Roehampton, London SW15 4JD, UK
2
Department of Medicine, Surgery and Dentistry Scuola Medica Salernitana, University of Salerno, 84084 Fisciano, SA, Italy
3
OptiBiotix Health PLC, York YO10 5DG, UK
*
Author to whom correspondence should be addressed.
Gastrointest. Disord. 2024, 6(1), 114-130; https://doi.org/10.3390/gidisord6010008
Submission received: 29 November 2023 / Revised: 29 December 2023 / Accepted: 3 January 2024 / Published: 10 January 2024

Abstract

:
People with coeliac disease (CD) have a higher risk of developing cardiovascular disease (CVD), potentially due to inflammation. Probiotics can influence CVD risk through several mechanisms including modifying inflammation. We performed a systematic review of probiotic interventions in people with CD. In total, 4 databases were systematically searched for studies published up to March 2023. All outcomes, inclusive of any cardiovascular risk factors, were collated and reported. We screened 8084 articles and 11 publications reporting on 7 RCTs and 2 non-RCTs met the inclusion criteria for qualitative analysis. In total, 1 RCT and both non-RCTs were considered to have a high risk of bias. There was large heterogeneity between the studies and adherence to a gluten-free diet was only measured in two studies. No specific outcomes related to cardiovascular risk were reported. Two studies reported a significant reduction on serum TNF-α in children over time after probiotic supplementation. One study reported no significant change in intestinal permeability over a 3-week intervention. Currently there is insufficient evidence to advocate a positive impact of probiotics on inflammation in CD, due, in part, to the limited data on adherence to the gluten-free diet and active disease.

1. Introduction

Coeliac disease (CD) is a chronic inflammatory bowel condition, triggered by an autoimmune response which is characterized by a permanent sensitivity to gluten in genetically predisposed people [1]. The small intestines are primarily affected at the onset and duration of the disease, however, there are broad clinical manifestations usually presenting as both intestinal and extraintestinal symptoms. Gluten is the term used for storage proteins derived from cereal grains barley, rye, and wheat. The prevalence of CD is steadily increasing with approximately 1% of people affected in the world’s population [2]. With no cure, lifelong adherence to a gluten-free diet (GFD) (a combination of naturally gluten-free and processed foods comprising less than 20 ppm gluten) is recognized as the only effective treatment for CD [3]. Adhering to a GFD can be very challenging; it requires knowledge, skills and modified behaviours to undertake the substantial changes to dietary habits [4].
The gut microbiota, which is a complex community of microorganisms, within people with CD differs compared with controls, whether this CD-associated dysbiosis is the consequence of the inflammation or a concurring causative factor remains to be established [5]. Intestinal dysbiosis persists on the GFD, which itself can contribute due to the reduced fibre content [6,7]. Dysbiosis has been associated with cardiovascular disease (CVD), through several mechanisms including microbiota-derived metabolites [8,9]. People with CD are more likely to develop atherosclerosis, likely as a result of systemic inflammation and low high-density lipoprotein levels that may contribute to atherosclerosis [10,11]. It has been reported that a third of patients with CD adhering to a GFD had concurrent non-alcoholic fatty liver disease (NAFLD), accounting for a three-fold increased risk compared with the general population [12]. Conroy et al. (2023) report, analysis from UK Biobank data, people with CD have a higher risk of developing cardiovascular disease than did people with no coeliac disease [13].
Studies have suggested a GFD itself may also contribute to metabolic syndrome and hyperlipidaemia observed in people with CD [14,15,16,17,18], with significantly more saturated fat in GF compared with gluten containing counter parts [19]. Based on findings from a large epidemiological study, Lebwohl et al. (2017) suggest the avoidance of gluten may result in reduced consumption of beneficial whole grains, which may affect cardiovascular risk [20]. A systematic review of studies up to 2021, indicated there was insufficient evidence to link a GFD with CVD, however, there was considerable heterogeneity within the studies [21].
The gut microbiota has an essential role in regulating lipid absorption in the human intestine [9]. Two studies reported participants consuming Lactobacillus plantarum ECGC 13110402 (LPLDL®) supplement, exhibited reductions in their LDL-cholesterol and increased HDL Cholesterol [22,23]. Derosa et al. (2020) reported significant reductions were seen in blood pressure and level of LDL cholesterol after three months of LPLDL® [24]. They concluded that major risk factors for cardiovascular events such as high blood pressure and hypercholesterolemia could be managed with a probiotic [24].
Previous literature reviews on probiotics in coeliac disease have reported their potential role in inflammation and gastrointestinal symptoms [25,26]. We performed a systematic review of the literature to evaluate the effects of probiotic interventions in people with CD to collate knowledge from all outcomes inclusive of any cardiovascular risk factors.

2. Results

Records identified through initial literature search were 9672 in total. This number was reduced to 8084 after duplicates were removed. As shown in the flow chart (Figure 1) we reviewed thirty-one full-text studies. Upon further review, we excluded twenty studies, and the remaining eleven publications met the inclusion criteria and were eligible for qualitative synthesis which is summarised in Table 1.

2.1. Characteristics of Studies

All studies included were in English and did not require translation. We performed analysis on 11 reports [27,28,29,30,31,32,33,34,35,36,37,38,39,40,41] including 7 RCTS with a total of 618 participants. In total, 3 articles reported the findings from 1 RCT [29,31,34]. All studies included were published from 2013 to 2022. The studies were conducted in medical centres with 2 studies from Argentina, 1 from Spain, 1 from Australia,1 from Brazil, 1 from Italy, 1 from Pakistan and 3 from Slovenia. The 3 Slovenian reports had a common intervention cohort from which three publications reported outcomes [29,31,34]. The majority of all participants from the studies were female, 6 studies included adults whereas 3 studies (5 publications) included only children.
Four studies reported only faecal analysis from their participants [35,37,39,41], whilst one reported blood analyses only [29]. Five studies conducted both faecal and blood analyses [27,28,30,34,36]. One study reported a duodenal biopsy analysis only [32]. The probiotic strains and dosages were the same for the three publications of the Slovenia cohort as well as the two Argentina studies but were different in all other studies. Six studies monitored adverse events [27,28,29,30,35,36] comparing probiotics to placebo no differences in the number of adverse events were reported. All except one RCT used either a placebo capsule or sachet with the one RCT [37] did not use a placebo for their controlled study. Every study excluded participants who were on any medications including antibiotics and immunosuppressants as they could have had effects on the outcome. Studies also excluded participants who had severe malnutrition, organic disorders, or suspicion of cancer.
Most reports, except four [27,28,32,37], participants had been diagnosed of coeliac disease by serologic testing and had been on a GFD for over 6 months before starting in the study. Persistent symptoms in participants were reported in three studies while on a GFD [30,35,36]. Francavilla et al., reported participants had an additional diagnosis of irritable bowel syndrome [35]. The studies undertaken by Ali et al. and Olivares et al. included newly diagnosed children who started a GFD for the study [28,37] and one study included newly diagnosed adults who were on a GFD with no other comorbidities [27]. Only two studies monitored the adherence to a GFD [28,30] even though for all other studies it was a pre-requisite to be on GFD in order to be included in the study. Olivares et al. used the 72 h food diary whilst Harnett et al. used the 3-day food diary.

2.2. Risk of Bias Assessment

Five RCTs were considered to have a low risk of bias, three with some concerns and one with high risk of bias; details for each domain can be found within Table 1. Full details of subcategories within Supplementary Materials in Tables S1 and S2. Both non-RCTs were considered to have a high risk of bias; details for each domain can be found in Table 2. The domain ‘measurement of outcomes’ was analysed for two outcomes: For gastrointestinal symptoms, four reports were judged as LOW risk of bias [27,28,35,36] whilst one had a HIGH risk of bias [37]. For inflammatory markers, three studies measured this outcome and were judged as LOW risk of bias [27,29,30,31,32,33].
Table 1. Risk of bias for Randomised Controlled Trials.
Table 1. Risk of bias for Randomised Controlled Trials.
Randomisation ProcessDeviations from Intended InterventionMissing Outcome DataMeasurement of Outcome Gastro SymptomsMeasurement of Outcome
Inflammation
Selection of Reported ResultsOverall
Smecuol et al., 2013 [27] Some concerns
Olivares et al., 2014 [28] Some Concerns
Klemenak et al., 2015 [29] Not measured Low risk of bias
Harnett et al., 2016 [30] Not measuredNot measured Low risk of bias
Quagliariello et al., 2016 [31] Not measuredNot measured Low risk of bias
Primec et al., 2019 [34] Not measuredNot measured Low risk of bias
Francavilla et al., 2019 [35] Not measured Low risk of bias
Smecuol et al., 2020 [36] Not measured Some concerns
Ali et al., 2022
[37]
Not measured High risk of bias
Green: Low risk of bias; Orange: Some concerns; Red: High risk of bias. Full details of subcategories within supplementary information.
Table 2. Risk of bias for Non-Randomised Controlled Trials.
Table 2. Risk of bias for Non-Randomised Controlled Trials.
Non RCTsBias Due to ConfoundingBias in Selection of Participants into the StudyBias in Classification of InterventionsBias Due to Deviations from Intended InterventionsBias Due to Missing DataBias in Measurement of OutcomesBias in Selection of the Reported ResultOverall
Pinto-Sanchez et al., 2016 [32] High risk of bias
Martinello et al., 2017 [33] High risk of bias
Green: Low risk of bias; Orange: Some concerns; Red: High risk of bias. Full details of subcategories within supplementary information.

2.3. Impact of Probiotic Intervention on Outcomes

The Effects of Probiotics on Gut Microbiota: Eight studies conducted faecal analysis but only four measured the microbiota composition and reported data for absolute abundance inclusive of bifidobacterial and lactobacilli species [28,30,31,35]. One of the four studies reported significant increases in bifidobacteria species after probiotic intervention when compared to placebo and no significant increase was observed in the lactobacilli species. Only two studies out of the four had lactobacilli as part of their formulations, however, the counts of the lactobacilli species were not high.
The Effect of Probiotics on Cardiovascular Risk Factors: None of the studies reported outcomes on cardiovascular risk factors (Table 3 and Table 4). Inflammatory markers: Two studies assessed the effects of probiotics on serum TNF-α in children (Table 3 and Table 4). In the Klemenak et al. study, participants had been on a GFD for at least 6 months, they reported a significant reduction over time [29]. Whereas patients in Olivares et al. study patients started the GFD simultaneously with probiotics, a potential trend towards a reduction in TNF-α was observed [28]. One study reported intestinal permeability with the use of lactulose/mannitol ratio intestinal permeability test [27]. No change in intestinal permeability was observed over the 3-week intervention.
The Effect of Probiotics on the Quality of Life (QoL): Only two studies reported outcomes for QoL (Table 3 and Table 4). Francavilla et al. used the validated irritable bowel syndrome-QoL questionnaire tool (IBS-QoL) and Harnett et al. used the validated coeliac disease questionnaire tool (CDQ). In both studies, QoL scores after treatment did not differ between probiotic and placebo treatments [30,35].
The Effect of Probiotics on GI Symptoms: Five studies assessed the impact of probiotics on GI symptoms (Table 3 and Table 4). Four studies reported improvements in GI symptoms after comparing probiotic to placebo [27,28,35,37]. Smecuol et al. reported improvements in bloating, diarrhoea, abdominal pains, and abdominal distension from self-reported records [27]. Olivares et al. also reported improvements in abdominal pains, vomiting, constipation and diarrhoea from self-reported records [28]. Ali et al. reported significant improvements in stool frequency [37], though the study had a high risk of bias (Table 1). Francavilla et al. reported no difference in diarrhoea when using the Bristol Stool Form Scale and was considered a low risk of bias (Table 2) [35]. Smecuol et al. and Francavilla et al. [27,35] used gastrointestinal symptom rating scale (GSRS) questionnaires and reported a significant reduction in GSRS scores compared with baseline values. Smecuol et al. [36] used the coeliac symptoms index (CSI) while Harnett et al. [30] used CDQ-GI to assess GI symptoms of participants. Probiotics when compared to placebo did not show significant changes in CSI scores.
Table 3. Characteristics of studies and reported outcomes for RCTs.
Table 3. Characteristics of studies and reported outcomes for RCTs.
Author
Year Country
Study Design
Sample SizePopulation CharacteristicsInterventions (Probiotic and Placebo)Follow Up DurationOutcomes
Smecuol et al., 2013 [27]
 
Argentina
 
DBRCT
Probiotic: 12
Placebo: 10
M/F: 4/18
Probiotic: Age 46 (29–62) years
Placebo: Age 40 (20–71) years
Dx: active CD, on gluten containing diet before GFD.
No other active chronic gastrointestinal pathologies
No additional diagnosis.
Blood and urine sample analysed.
B. infantis NLS super strain (2 × 109 Colony-Forming Units) vs. Placebo, 3 times per day, 15 min before meals
5 g of lactulose and 2 g of Mannitol in 450 mL of water for fractional excretion ratio
2-week run -in, 3-week treatment, 50-day f/u to initiate GFDQuality of life:
None reported.
Gastro Symptoms: bloating and abdominal distention (20/22), abdominal pain (19/22), diarrhoea (11/22) for both groups
Markers of active disease:
Probiotics: Reduction in serum antibody concentrations (10% for IgA tTG and IgA DGP antibodies)
Placebo: Increased antibody serum concentrations (IgA tTG, 7% and IgA DGP, 10%) at the end of the trial.
Biochemical and microbial data:
Primary endpoint:
Non-significant increase of mean lactulose/mannitol ratio from baseline for the probiotic (p = 0.064) and placebo (p = 0.342) and
Secondary endpoint:
Probiotic: significant reduction in indigestion (p = 0.0035) and constipation (p = 0.0483) symptoms, however, borderline for reflux symptoms (p = 0.0586).
Placebo: No significant changes in any syndrome (indigestion, diarrhoea, constipation, abdominal pain). Significant improvement in diarrhoea symptoms
Outcome of inflammatory mediators:
Probiotic and placebo: No significant changes in Th1 serum cytokines and serum chemokines.
Significant increase in high baseline serum concentration of MIP-1β, (p < 0.04), but not in the placebo group.
Cardiovascular risk factors: None reported.
Olivares et al., 2014
[32]
Spain
 
DBRCT
Probiotics: 17
Placebo: 16
Probiotic: Age 6.8 ± 0.9 years
M/F (7/10) Placebo: Age 8.5 ± 1.2 years
M/F (8/8)
Dx: biopsy newly diagnosed CD.
Adherence to GFD after diagnosis confirmation. Food records for 72 h (2 weekdays, 1 weekend day) before start and after 3 mnths intervention.
No additional diagnosis.
 
Blood and faecal sample analysed; DNA sequencing with QIAamp DNA stool Mini Kit and Real time PCR to quantify content of bacterial groups.
Bifidobacterium longum CECT 7347 (109 CFU) vs. Placebo At lunch time3 monthsQuality of life:
Not reported.
Gastro Symptoms: Self-reported improvements in diarrhoea, constipation, abdominal pain, and vomiting. No significant changes in both groups for energy intake other specific nutrients (p > 0.05). GFD adherence led to slight decrease in dietary fibre.
Probiotic: higher mean increment in weight and height percentiles than in the placebo group.
Markers of active disease:
Probiotic: Decreases in CD3+ (p = 0.013) and HLA-DR+ (p = 0.029) T cell populations but no significant changes in placebo.
No significant differences seen in T lymphocytes HLA-DR+, CD4+, CD4+Foxp3+ and CD8+ for both groups (p = 0.328, p = 0.970, p = 0.504 and p = 0.376).
Biochemical and microbial data:
Slight reduction in serum TNF-α for probiotic group (p = 0.067); but difference between both groups was not significant (p = 0.085).
No significant differences found in both groups for serum IL-10, interferon-y, IL-13, transforming growth factor-β1, IgG1 and IgG4.
Placebo: Significant increase in gene copy Bacteroides fragilis group (p = 0.013), Enterobacteriaceae (p = 0.038), non-significant decrease in Bifidobacterium spp. (p = 0.151).
Probiotic: No significant differences in Lactobacillus group, Bifidobacterium spp./B. fragilis group and Enterobacteriaceae. Significant decrease in faecal sIgA concentration
Cardiovascular risk factors: None reported.
Slovenia RCT 3 publications
Klemenak et al., 2015
[33]
Slovenia
 
DBRCT
49 patients Placebo: 24 Probiotic: 22 HC: 18Placebo: Age 10.81 ± 5.0 years, M/F (10/14)
Time on GFD 7.1 ± 5.5 years,
Compliance on GFD (80%)
Probiotic: Age 10.4 ± 4.2 years M/F (6/16)
Time on GFD 5.6 ± 3.7 years,
HC: Age 8.8 ± 6.0 years M/F (7/11)
Dx: biopsy On GFD from 0.5 to 15 years.
No additional diagnosis.
Compliance on GFD (91%) in probiotic group and 80% in placebo
Blood sample analysed.
Lyophilized 50% B. breve BR03 (109 CFU) and 50% B. breve B632 (109 CFU) vs. Placebo Daily at breakfast3 months; 3 months f/uQuality of life:
None reported.
Gastro Symptoms:
None reported.
Markers of active disease:
None reported.
Biochemical and microbial data:
TNF-α in serum at baseline, significantly higher (p = 0.015) in the probiotic group (14.78 ± 6.43) than placebo group (10.58 ± 3.57).
TNF-α in serum at the end of the study, significant decrease (p = 0.020) in the probiotic group (11.97 ± 3.58) from baseline levels.
TNF-α in serum on f/u, significantly higher in the probiotic group
No correlation between positive serologic markers of CD and levels of TNF-α and IL-10 in individual patient with CD.
Values for cytokine IL-10 in serum, below assay detection limit (5 pg/mL) and so not analysed.
Cardiovascular risk factors: None reported.
Quagliariello et al., 2016
[35]
Slovenia
 
DBRCT
Probiotic: 20 Placebo: 20 HC: 16Age 1–19 years
Dx: biopsy On GFD for at least 3 months.
No additional diagnosis.
Faecal sample analysed; DNA extraction and sequencing with QIAamp DNA stool Mini Kit and molecular analyses; absolute quantification using Quantitative PCR.
B. breve BR03 (DSM 16604) (109 CFU) and B. breve B632 (DSM 24706) (1:1) (109 CFU) vs. Placebo daily at breakfast3 monthsQuality of life:
None reported.
Gastro Symptoms:
None reported.
Markers of active disease:
None reported.
Biochemical and microbial data:
Metagenomic analysis; 6 phyla revealed after sequencing (5 Bacteria and 1 Archaea). Firmicutes, high in control group (60–70%) and 50–60% in probiotic group. Bacteriodetes 20–40% in CD group, 10–20% in control group. Proteobacteria and Verrucomicrobia high in the placebo group. Statistically, Actinobacteria was low in CD group but increased after probiotic intake. Euryarchaeota, the only Archaea was found predominately in the control group.
Cardiovascular risk factors: None reported.
Primec et al., 2019
[38]
Slovenia
 
DBRCT
Probiotic: 20 Placebo: 19 HC: 14Probiotic: Age 9.2 ± 4.4 yeras
Male 20% Placebo: Age 10.5 ± 5.1 years
Male 31%
HC: Age 10.1 ± 6.0 years
Male 36%
Dx: biopsy On GFD from 6 months to 15 years.
No additional diagnosis.
Blood and faecal sample analysed; DNA extraction and sequencing with QIAamp DNA stool Mini Kit and molecular analyses; absolute quantification using Quantitative PCR.
B. breve 72 BR03 (DSM 16604) (2 × 109 CFU) and B. breve B632 (DSM 24706) (2 × 109 CFU) vs. Placebo Daily at breakfast3 months; 3 months follow-upQuality of life:
None reported.
Gastro Symptoms:
None reported.
Markers of active disease:
None reported.
Biochemical and microbial data:
In the probiotic and placebo group, TNF-α had a similar level as the healthy children. In CD patients, TNF-α had positive correlation with Verrucomicrobia and negative one with Parcubacteria. There was a high statistical significance between TNF-α and unclassified Bacteria group and positive correlation with TNF- alpha and unclassified Archaea.
In CD patients, Proteobacteria correlated positively with acetic and propionic acid (p = 0.452, p = 0.004 and p = 0.331, p = 0.045, respectively), which led to Proteobacteria and total SCFAs (p = 0.380. p = 0.017). Euryarchaeota phylum had a positive correlation (p = 0.351, p = 0.029) to acetic acid.
In Healthy Children, TNF-alpha had a negative association with Firmicutes (p = 0.660, p = 0.010) and negative correlation to Euryarchaeota (p = 0.654, p = 0.011).
Cardiovascular risk factors: None reported.
Harnett et al., 2016
[34]
Australia
 
DBRCT
Probiotics: 21
Placebo: 21
Probiotic: Age 47.1 ± 16.1 years
M/F (3/18)
Placebo: Age
47.5 ± 12.9 years
M/F (4/17)
Dx: biopsy On GFD for at least 12 months and with persistent symptoms
GFD Adherence: the three-day diet diary
Compliance was 95.2%
No additional diagnosis.
Duodenal biopsy, blood and faecal samples analysed; DNA and PCR analysis.
VSL #3 blend of probiotic bacteria vs. Placebo sachet orally with water/juice at breakfast and super12-week treatmentQuality of life:
Assessed with the CDQ. 2 participants on the placebo and 2 participants on probiotic reported mild bloating. Partial improvement due to GFD, 100% of participants.
However, residual gastrointestinal symptoms (mild to moderate) and fatigue were reported by all.
Gastro Symptoms:
None reported.
Markers of active disease:
1 probiotic and 1 placebo participant had persistent villous atrophy.
Biochemical and microbial data:
Descriptive statistics for the predominant bacteria showed that only Streptomyces spp. (p = 0.058) was different between the two groups at baseline and 12 weeks and Mycoplasma sp. (p = 0.026), 12 weeks only.
Bifidobacteria (p = 0.001) reduced significantly, and Escherichia coli (p = 0.005) increased over time.
Cardiovascular risk factors: None reported.
Francavilla et al., 2019
[39]
Italy
 
DBRCT
Probiotics: 54
Placebo: 55
Probiotic: Age 43.3 (18.8–62.2) years Male 11%
Placebo: Age: 44.6 (19.3–63.4) years Male: 16%
Dx: biopsy on GFD > 2 years Persistent symptoms [IBS Severity Score System >75]
Duration of GFD: Probiotic, 6.8 (2.6–16.7) years
Placebo, 7.4 (3.5–17.5) years
Additional Diagnosis: IBS
Faecal sample analysed; DNA extraction with FastDNA Spin kit for soil, RNA extraction with stool total RNA purification kit.
Mixture of: Lactobacillus casei LMG 101/37 P-17504 (5 × 109 CFU), Lactobacillus plantarum CECT 4528 (5 × 109 CFU), Bifidobacterium animalis subsp. lactis Bi1 LMG P-17502 (10 × 109 CFU), Bifidobacterium breve Bbr8 LMG P-17501 (10 × 109 CFU), B. breve Bl10 LMG P-17500 (10 × 109 CFU) vs. Placebo sachet Daily2-week run-in; 6-week treatment; 6-week follow-upQuality of life:
Irritable bowel syndrome quality of life scores was not different in both probiotic and placebo groups.
Gastro Symptoms:
GI symptoms reduced. Gastrointestinal symptom rating scale: Probiotic, 12.2 ± 5.5 and Placebo, 16.7 ± 6.7, reduced significantly for both groups from baseline.
At the end of follow up Probiotic, 10.1 ± 4.1 Placebo 9.6 ± 4.2.
Markers of active disease:
TTG-IgA (IU/mL): Probiotic, 0.8 (0–1.2); Placebo, 0.5 (0.2.1)
Biochemical and microbial data:
Probiotics: significant increased levels of presumptive lactic acid bacteria (Lactobacillus, Lactococcus, and Streptococcus) Bifidobacterium spp. and Staphylococcus spp. Higher levels of Bifidobacterium spp. after 6 weeks.
Placebo: No statical differences found in cultivable microbes. No statistical differences found between both groups for total bacterial community richness.
Cardiovascular risk factors:
None reported.
Smecuol et al., 2020
[40]
Argentina
 
DBRCT
Probiotic: 7 Placebo: 5Probiotic: Male 0%
Placebo: Male 20%
Age 53 (43–57) years
Dx: biopsy On GFD for >2 years
No additional diagnosis.
Blood and faecal sample analysed; Faecal total DNA extracted and sequenced.
B. infantis NLS-SS (4 × 109 CFU) vs. Placebo DailyOne week run in 3 weeks treatment 2 weeks washout 3 weeks switched treatmentQuality of life:
None reported.
Gastro Symptoms:
Two patients in the probiotic group had gluten indigestion.
Markers of active disease:
No significant changes in the coeliac symptom index when comparing probiotic and placebo groups.
Biochemical and microbial data:
Significant improvement in CD symptom in probiotic group compared to highly symptomatic placebo group, p = 0.046.
No difference between both groups with positive and negative serology.
No significant differences in both groups for gluten immunogenic peptide excretion.
Probiotic group had decreased levels of Ruminococcus spp. and Bifidobacterium adolescentis.
Cardiovascular risk factors:
None reported.
Ali et al., 2022
[41]
Pakistan Randomized clinical trial, descriptive cross-sectional study.
Probiotic: 85
No medication: 85
Age 8–10 years
Dx: CD from intestinal biopsy.
No additional diagnosis.
Stool frequency analysed.
Clostridium butyricum and Bifidobacterium spp. in 75–100 mL of boiled water twice daily.28 daysQuality of life:
None reported.
Gastro Symptoms:
Significant reduction in frequency of stools per day.
Markers of active disease:
None reported.
Biochemical and microbial data:
None reported.
Cardiovascular risk factors:
None reported.
CD: Coeliac Disease, DBRCT: Double Blind Randomized Controlled Trial, GFD: Gluten-free Diet, M/F: Male/Female, HC: Healthy Control, Dx: Diagnosis, F/u: Follow-up, IBS: Irritable bowel syndrome.
Table 4. Characteristics of studies and reported outcomes for non-RCTs.
Table 4. Characteristics of studies and reported outcomes for non-RCTs.
Author
Year Country Study Design
Sample SizePopulation CharacteristicsInterventions (Probiotic and Placebo)Follow Up DurationOutcomes
Pinto-Sanchez et al., 2016
[36]
Argentina
 
Clinical trial with external controls
Active CD with probiotic: 12
Active CD without probiotic: 24.
Control CD on 1 y GFD: 5
CD Active on probiotics: Age 41 (22–53) years
Female n (%): 8 (67)
CD Active no treatment: Age 40 (29–54) years
Female n (%): 23 (95.8)
CD on GFD: Age 35 (31–45) years
Female n (%): 4 (80)
Dx: biopsy Active cases
No additional diagnosis.
Duodenal biopsy analysed.
Bifdobacterium infantis NSL-SS3 weeksQuality of life:
None reported.
Gastro Symptoms:
None reported.
Markers of active disease:
None reported.
Biochemical and microbial data:
The probiotic reduced the Paneth cells (PC) counts without the GFD. However, decreased macrophage counts (p = 0.02) were seen after 1 y GFD as well as further decreased in patients treated with the probiotic only.
Similarly, the expression of mucosal HD-5 was significantly decreased in the probiotic group but not in 1 y GFD (p < 0.001).
IgA TG: Active CD without probiotic 129 (104–200), Active CD with probiotic 200 (192–216), CD on 1 y GFD 8 (4–17)
IgA DGP: Active CD without probiotic 200 (120–300), Active CD with probiotic 152 (64–300), CD on 1 y GFD 7 (4–18)
Cardiovascular risk factors:
None reported.
Martinello et al., 2017
[37]
Brazil
 
Non-randomized clinical trial
CD: 14
HC: 17
CD: Age 18–60 years 10/4 (F/M)
HC: Age 18–85 years years 10/7 (F/M)
Dx: biopsy On GFD and asymptomatic
No additional diagnosis. Faecal sample analysed; for bifidobacteria content and faecal pH.
108 CFU of Lactobacillus acidophilus and Bifdobacterium lactis as 100 g of yogurt per day. the average concentration of bifidobacteria was 6.67 × 108 ± 10.3 × 108 CFU/g of yoghurt.30 daysQuality of life:
None reported.
Gastro Symptoms:
None reported.
Markers of active disease:
None reported.
Biochemical and microbial data:
Healthy individuals presented a significantly higher concentration of bifidobacteria (2.3 × 108 ± 6.3 × 107 CFU/g) before the probiotic-containing yogurt intake when compared to the celiac group (1.0 × 107 ± 1.7 × 107 CFU/g) Celiac patients presented, in average, 83% less bifidobacteria than healthy individuals. Still, celiac faecal pH (7.19 ± 0.521) was not significantly different from the faecal pH of the control group (7.18 ± 0.522).
Healthy individuals presented a significantly higher bifidobacteria concentration (14.7 × 108 ± 0.2 × 108 CFU/g) than celiac patients (0.76 × 108 ± 0.1 × 108 CFU/g). However, faecal pH of celiac patients (7.28 ± 0.518) did not show significant difference from the faecal pH of healthy individuals (7.07 ± 0.570) after the yogurt intake.
Cardiovascular risk factors:
None reported.
CD: Coeliac Disease, GFD: Gluten-free Diet, M/F: Male/Female, HC: Healthy Control, Dx: Diagnosis, F/u: Follow-up.

3. Discussion

Data on the effect of probiotics in coeliac disease related gastrointestinal symptoms are limited, and data on CVD risks is even less studied. CVD is associated with a chronic state of low-grade inflammation, whereby the microbiota has an impact on maintaining the gut barrier function, critical for reducing the amount of pro-inflammatory bacterial byproducts that can cross into the bloodstream [8]. Notably, the two studies that evaluated the effects of probiotics on serum TNF-α reported a reduction in levels [28,29], agrees with a meta-analysis of 12 RCTs in people with diabetes or prediabetes [8]. Naseri et al. suggests the modulating effects of probiotics on TNF-α levels (and CRP levels) were more pronounced in patients with heightened inflammation [8]. The two studies included European children with CD only, thus, it is clear further studies exploring the impact of probiotics on cardiovascular risk and inflammation are needed. Smecuol et al. [27] reported that a probiotic intervention had no significant impact on intestinal permeability, however, the intervention was only 3 weeks, and it is well known that it may take some time before mucosal integrity returns to baseline [38]. Patients with CD continue to have gluten exposures which may account for persistence of histologic inflammation and residual symptoms. If patients are able to completely exclude all gluten, then is remains debated whether the inflammation remains [39].
Four studies reported improvements in GI symptoms after comparing probiotic to placebo [27,28,35,37], although there was a high risk of bias in the outcome measurement for Ali et al., study. Since completing the systematic literature search, Khorzoghi et al. reported improved fatigue scores with probiotic supplementation in a small cohort of adults with CD from Iran [40]. A meta-analysis by Seiler et al. [25] concluded probiotics may improve gastrointestinal symptoms, however, the quality of evidence was low or very low; we confirm the recent publications have not been able to overcome this criticism.
A strength of our systematic review is collating all reported outcomes from RCTs and non RCTs relating to the impact of probiotic supplementation in adults and children with coeliac disease. We also bring attention to the reader that three publications were from the same intervention study in Slovenia [29,31,34], thus the body of evidence for probiotics in CD is small. This systematic review was conducted rigorously with pre-defined outcomes and a pre-registered protocol. We highlight that only two studies reported how they determined adherence to a GFD, it is well recognised that adhering to the gluten-free diet is challenging and many patients do not achieve full adherence [4]. We recommend all future studies investigating probiotics in CD include an assessment for dietary adherence to the GFD as consumption of gluten would have a substantial impact on study outcomes such as intestinal permeability and inflammatory markers.
Four studies reported the composition of the microbiota with an increase in bifidobacterial evident after the probiotic interventions as would be expected, Mozafarybazargany et al. [26] reported similar findings from pooled analysis from a broad range of studies including people with CD and potential CD. The dysbiosis in treated CD maybe mainly food-induced, but in a small sample of children there is a demonstration of a constant alteration of some species that clustered in CD and not in healthy controls [41]. The gut microbiota differs from person to person geographically; thus, the effectiveness of a probiotic may vary for persons in specific geographic areas [42]. The review comprised of studies conducted in Europe (Italy, Spain and Slovenia), Australia, South America (Brazil and Argentina) and South Asia (Pakistan) with none from Africa, East Asia and North America; therefore, the results cannot be easily generalized. Knowing the impact of probiotics on the microbial ecosystem is essential; however, it must be emphasized that not all probiotics contain the same bacterial strains and can have different beneficial effects.
To conclude, our systematic review reveals there remains very sparce data on outcomes related to cardiovascular risk, and presently our knowledge of the area relies on data from other disease-based populations. There is emerging evidence of an impact on inflammation and gastrointestinal symptoms in adults and children with CD.

4. Methods

The systematic review was undertaken and reported in line with the guidelines of preferred reporting items for systematic reviews and meta-analyses [43]. A population, intervention, comparison and outcomes (PICO) framework and literature search strategy criteria were developed—including the PICO table. This review has been registered on Prospero, the international prospective register of systematic reviews (CRD42023380433)

4.1. Inclusion and Exclusion

We adapted the PICO table strategy for the inclusion and exclusion criteria. Participants/persons, i.e., children and adults diagnosed with coeliac disease using standard procedures including specific serologic tests (anti-tissue transglutaminase, anti-deamidated gliadin peptide, or anti-endomysia antibodies) and confirmed by duodenal biopsy (Marsh score 3 or higher; or equivalent). Interventions included (any dietary intervention using probiotics with a duration of at least 21 days. For comparison, dietary interventions compared to placebo, or any or any other therapy were considered.
Outcomes assessed were all reported outcomes, this included quality of life, gastro symptoms, biochemical and microbial data, inflammatory markers and any cardiovascular risk factors. Assessment of stool frequency, stool consistency, bloating, flatulence, abdominal pain prior during and after the dietary intervention. Markers of active disease (total immunological immunoglobin A (IgA) and IgA tissue transglutaminase; IgA endomysial antibodies (EMA), IgG deamidated gliadin peptide (DGP) or IgG tTG). Studies that did not meet the criteria were excluded.

4.2. Type of Studies

The systematic review included randomized controlled trials, experimental studies, and non-controlled trials, which were human trials only in this study. There were no restrictions on study size. Observational studies, case reports, and case series were excluded. Duplicate studies and studies without validated diagnoses of CD were also excluded. We conducted a comprehensive search in the following databases: CINAHL, Web of Science, CENTRAL, PubMed, Australian and New Zealand Clinical Trials Register and ClinicalTrials.gov from inception up to March 2023.

4.3. Search Terms

The search strategy was based on Medical Subject Headings (MeSH) and other corresponding words. The terms include “Probiotics”, “Bifdobacterium”, “Lactobacillus”, “Coeliac Disease”, “Celiac disease”. We used the logical operators “OR” or “AND” to connect the terms above. The search syntax: “Coeliac disease” OR “Celiac disease” AND probiotics OR “probiotic supplementation” was used on CINAHL, CENTRAL and Web of Science Core Selection. The syntax: (“Celiac Disease” [MeSH Terms] OR “Celiac Disease” [Text Word] OR “Coeliac Disease” [Title/Abstract] OR “CD” [All Fields]) AND (“Probiotics” [MeSH Terms] OR “probiotic*” [Text Word] OR “probiotic supplement” [Title/Abstract] OR “Probiotic supplementation” [Title/Abstract]) was used on PubMed Search. The total search results from PubMed Search were 665 results, Web of Science had 6264 results, CENTRAL produced 67 results and CINAHL, 2676 results.

4.4. Selection of Studies

Two independent authors (LF and CS), screened titles and abstracts and cleared duplicates as well. Full-text screening was performed by the same reviewers independently. All data were collated in the AI software tool Rayyan and after screening, selected articles were collected into a Rayyan file. For any disagreement case, with the help of an independent third author (YJ), a resolution was reached.
Two independent reviewers (LF and YJ) extracted data and information on authors, publication type (abstract/full text), publication year, geographic region (country), language, follow-up duration, age, gender, ethnicity, method of diagnosis (biopsy/serology/HLA DQ2/8 status), adherence to a gluten-free diet, additional diagnosis such as IBS, Probiotic strain/combination and type of delivery, dosage, frequency (timing), duration of intervention (length). Comparator population characteristics and dosage, frequency, duration of intervention placebo type, GI symptom scores, QOL scores, adverse events, markers of active disease such as serum TNF-α, intestinal permeability.

4.5. Data Synthesis Strategy

Our data synthesis was conducted based on data availability and it comprised studies grouped under all reported outcomes and study details (e.g., number of participants, setting, etc.). Individual study data including specific study outcomes, methods of analysis and results were reported in tables. A narrative synthesis was then carried out by tabulation and narrative summary of populations, interventions, and outcomes. Meta-analysis was not undertaken due to the large heterogeneity between the studies.

4.6. Risk of Bias Assessment

Selected articles were reviewed for risk of bias using Cochrane risk of bias in RCTs (ROB-2 tool) and non RCTs (ROBINS-1 tool) [44,45]. The Cochrane risk of bias assessment tools were used to assess randomization; concealment and blinding of participants, outcome assessors; deviations from intended interventions, missing outcome data, measurement of outcomes and selection of reported results. The first reviewer independently (LF) assessed the quality of selected articles for all domains of the tool so did a second reviewer (YJ). A table summarising the findings was created to display the quality of each study.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/gidisord6010008/s1.

Author Contributions

Conceptualization, Y.J., A.C., C.C. and S.K.; methodology, Y.J. and L.O.F.; formal analysis, L.O.F., Y.J. and C.S.; writing—original draft preparation, L.O.F. and Y.J.; writing—review and editing, All authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

S.K. is employed by OptiBiotix Health PLC; A.C. has received funding from OptiBiotix Health PLC.

References

  1. Ludvigsson, J.F.; Leffler, D.A.; Bai, J.C.; Biagi, F.; Fasano, A.; Green, P.H.R.; Hadjivassiliou, M.; Kaukinen, K.; Kelly, C.P.; Leonard, J.N.; et al. The Oslo definitions for coeliac disease and related terms. Gut 2013, 62, 43–52. [Google Scholar] [CrossRef] [PubMed]
  2. Singh, P.; Arora, A.; Strand, T.A.; Leffler, D.A.; Catassi, C.; Green, P.H.; Kelly, C.P.; Ahuja, V.; Makharia, G.K. Global prevalence of celiac disease: Systematic review and meta-analysis. Clin. Gastroenterol. Hepatol. 2018, 16, 823–836. [Google Scholar] [CrossRef] [PubMed]
  3. National Institute for Health and Care Excellence (NICE). Coeliac Disease Quality Standard. 2016. Available online: www.Nice.Org.Uk/guidance/qs134 (accessed on 1 October 2023).
  4. Muhammad, H.; Reeves, S.; Jeanes, Y.M. Identifying and improving adherence to the gluten-free diet in people with coeliac disease. Proc. Nut. Soc. 2019, 78, 418–425. [Google Scholar] [CrossRef] [PubMed]
  5. Sacchetti, L.; Nardelli, C. Gut microbiome investigation in celiac disease: From methods to its pathogenetic role. Clin. Chem. Lab. Med. 2020, 58, 340–349. [Google Scholar] [CrossRef] [PubMed]
  6. De Palma, G.; Nadal, I.; Collado, M.C.; Sanz, Y. Effects of a gluten-free diet on gut microbiota and immune function in healthy adult human subjects. Br. J. Nutr. 2009, 102, 1154–1160. [Google Scholar] [CrossRef] [PubMed]
  7. Cristofori, F.; Indrio, F.; Miniello, V.L.; Angelis, M.D.; Francavilla, R. Probiotics in celiac disease. Nutrients 2018, 10, 1824. [Google Scholar] [CrossRef]
  8. Naseri, K.; Saadati, S.; Ghaemi, F.; Ashtary-Larky, D.; Asbaghi, O.; Sadeghi, A.; Afrisham, R.; de Courten, B. The effects of probiotic and synbiotic supplementation on inflammation, oxidative stress, and circulating adiponectin and leptin concentration in subjects with prediabetes and type 2 diabetes mellitus: A GRADE-assessed systematic review, meta-analysis, and meta-regression of randomized clinical trials. Eur. J. Nutr. 2023, 62, 543–561. [Google Scholar]
  9. Yntema, T.; Koonen, D.P.Y.; Kuipers, F. Emerging Roles of Gut Microbial Modulation of Bile Acid Composition in the Etiology of Cardiovascular Diseases. Nutrients 2023, 15, 1850. [Google Scholar] [CrossRef]
  10. Naaraayan, A.; Nimkar, A.; Jesmajian, S.; Gitler, B.; Acharya, P. Atherosclerotic cardiovascular disease prevalence among patients with celiac disease in the United States. Mayo Clin. Proc. 2021, 96, 666–676. [Google Scholar] [CrossRef]
  11. Wang, Y.; Chen, B.; Ciaccio, E.J.; Jneid, H.; Virani, S.S.; Lavie, C.J.; Lebovits, J.; Green, P.H.R.; Krittanawong, C. Celiac Disease and the Risk of Cardiovascular Diseases. Int. J. Mol. Sci. 2023, 24, 9974. [Google Scholar] [CrossRef]
  12. Tovoli, F.; Negrini, G.; Farì, R.; Guidetti, E.; Faggiano, C.; Napoli, L.; Bolondi, L.; Granito, A. Increased risk of nonalcoholic fatty liver disease in patients with coeliac disease on a gluten-free diet: Beyond traditional metabolic factors. Aliment Pharmacol. Ther. 2018, 48, 538–546. [Google Scholar] [CrossRef] [PubMed]
  13. Conroy, M.; Allen, N.; Lacey, B.; Soilleux, E.; Littlejohns, T. Association between coeliac disease and cardiovascular disease: Prospective analysis of UK Biobank data. BMJ Med. 2023, 2, e000371. [Google Scholar] [CrossRef]
  14. Zanini, B.; Mazzoncini, E.; Lanzarotto, F.; Ricci, C.; Cecana, B.; Villanacci, V.; Lanzini, A. Impact of gluten-free diet on cardiovascular risk factors. A retrospective analysis in a large cohort of coeliac patients. Dig. Liver Dis. 2013, 45, 810–815. [Google Scholar] [CrossRef] [PubMed]
  15. Tortora, R.; Capone, P.; De Stefano, G.; Imperatore, N.; Gerbino, N.; Donetto, S.; Monaco, V.; Caporaso, N.; Rispo, A. Metabolic syndrome in patients with coeliac disease on a gluten-free diet. Aliment Pharmacol. Ther. 2015, 41, 352–359. [Google Scholar] [CrossRef]
  16. Assa, A.; Frenkel-Nir, Y.; Tzur, D.; Katz, L.H.; Shamir, R. Cardiovascular risk factors in adolescents with celiac disease: A cross-sectional population-based study. J. Pediatr. Gastroenterol. Nutr. 2017, 65, 190–194. [Google Scholar] [CrossRef]
  17. Tetzlaff, W.F.; Merono, T.; Menafra, M.; Martin, M.; Botta, E.; Matoso, M.D.; Sorroche, P.; DePaula, J.A.; Boero, L.E.; Brites, F. Markers of inflammation and cardiovascular disease in recently diagnosed celiac disease patients. World J. Cardiol. 2017, 9, 448–456. [Google Scholar] [CrossRef] [PubMed]
  18. Agarwal, A.; Singh, A.; Mehtab, W.; Gupta, V.; Chauhan, A.; Rajput, M.S.; Singh, N.; Ahuja, V.; Makharia, G. Patients with celiac disease are at high risk of developing metabolic syndrome and fatty liver. Intest. Res. 2021, 19, 106–114. [Google Scholar] [CrossRef]
  19. Fry, L.; Madden, A.M.; Fallaize, R. An investigation into the nutritional composition and cost of gluten free versus regular food products in the UK. J. Hum. Nutr. Diet. 2018, 31, 108–120. [Google Scholar] [CrossRef]
  20. Lebwohl, B.; Cao, Y.; Zong, G.; Hu, F.B.; Green, P.H.R.; Neugut, A.I.; Rimm, E.B.; Sampson, L.; Dougherty, L.W.; Giovannucci, E.; et al. Long term gluten consumption in adults without celiac disease and risk of coronary heart disease: Prospective cohort study. BMJ 2017, 357, j1892. [Google Scholar] [CrossRef]
  21. Schmucker, C.; Eisele-Metzger, A.; Meerpohl, J.J.; Lehane, C.; Kuellenberg de Gaudry, D.; Lohner, S.; Schwingshackl, L. Effects of a gluten-reduced or gluten-free diet for the primary prevention of cardiovascular disease. Cochrane Database Syst. Rev. 2022, CD013556. [Google Scholar] [CrossRef]
  22. Costabile, A.; Buttarazzi, I.; Kolida, S.; Quercia, S.; Baldini, J.; Swann, J.R.; Brigidi, P.; Gibson, G.R. An in vivo assessment of the cholesterol-lowering efficacy of Lactobacillus plantarum ECGC 13110402 in normal to mildly hypercholesterolaemic adults. PLoS ONE 2017, 12, e0187964. [Google Scholar] [CrossRef]
  23. Keleszade, E.; Kolida, S.; Costabile, A. The cholesterol lowering efficacy of Lactobacillus plantarum ECGC 13110402 in hypercholesterolemic adults: A double-blind, randomized, placebo controlled, pilot human intervention study. J. Func. Foods 2022, 89, 1756–4646. [Google Scholar] [CrossRef]
  24. Derosa, G.; Gaudio, G.; D’Angelo, A.; Maffioli, P. Evaluation in Patients with High NORMAL Blood Pressure of a Supplement Containing Arginine, Lactobacillus Plantarum Lp-LDL, Coenzime Q10 and Vitamin B1: A PILOT Study. J. Food Nutr. Res. 2020, 8, 273–278. [Google Scholar] [CrossRef]
  25. Seiler, C.; Kiflen, M.; Stefanolo, J.P.; Bai, J.C.; Bercik, P.; Kelly, C.P.; Verdu, E.F.; Moayyedi, P.; Pinto-Sanchez, M.I. Probiotics for Celiac Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Am. J. Gastroenterol. 2020, 115, 1584–1595. [Google Scholar] [CrossRef]
  26. Mozafarybazargany, M.; Khonsari, M.; Sokoty, L.; Ejtahed, H.S.; Qorbani, M. The effects of probiotics on gastrointestinal symptoms and microbiota in patients with celiac disease: A systematic review and meta-analysis on clinical trials. Clin. Exp. Med. 2023, 23, 2773–2788. [Google Scholar] [CrossRef]
  27. Smecuol, E.; Hwang, H.J.; Sugai, E.; Corso, L.; Chernavsky, A.C.; Bellavite, F.P.; Gonzalez, A.; Vodanovich, F.; Moreno, M.L.; Vazquez, H.; et al. Exploratory, randomized, doubleblind, placebo-controlled study on the effects of Bifidobacterium infantis natren life start strain super strain in active celiac disease. J. Clin. Gastroenterol. 2013, 47, 139–147. [Google Scholar] [CrossRef]
  28. Olivares, M.; Castillejo, G.; Varea, V.; Sanz, Y. Double-blind, randomised, placebo-controlled intervention trial to evaluate the effects of Bifidobacterium longum CECT 7347 in children with newly diagnosed coeliac disease. Br. J. Nutr. 2014, 112, 30–40. [Google Scholar] [CrossRef] [PubMed]
  29. Klemenak, M.; Dolinsek, J.; Langerholc, T.; Gioia, D.D.; Micetic-Turk, D. Administration of Bifidobacterium breve decreases the production of TNF- α in children with celiac disease. Dig. Dis. Sci. 2015, 60, 3386–3392. [Google Scholar] [CrossRef]
  30. Harnett, J.; Myers, S.; Rolfe, M. Probiotics and the microbiome in celiac disease: A randomised controlled trial. Evid. Based Complement Altern Med. 2016, 2016, 9048574. [Google Scholar] [CrossRef]
  31. Quagliariello, A.; Aloisio, I.; Cionci, N.B.; Luiselli, D.; D’Auria, G.; Martinez-Priego, L.; Perez-Villarroya, D.; Langerholc, T.; Primec, M.; Micetic-Turk, D.; et al. Effect of Bifidobacterium breve on the intestinal microbiota of coeliac children on a gluten free diet: A pilot study. Nutrients 2016, 8, 660. [Google Scholar] [CrossRef]
  32. Pinto-Sanchez, M.I.; Smecuol, E.C.; Temprano, M.P.; Sugai, E.; Gonzalez, A.; Moreno, M.L.; Huang, X.; Bercik, P.; Cabanne, A.; Vazquez, H.; et al. Bifidobacterium infantis NLS super strain reduces the expression of α-defensin-5, a marker of innate immunity, in the mucosa of active celiac disease patients. J. Clin. Gastroenterol. 2016, 51, 814–817. [Google Scholar] [CrossRef] [PubMed]
  33. Martinello, F.; Roman, C.F.; Alves de Souza, P. Effects of probiotic intake on intestinal bifidobacteria of celiac patients. Arq. Gastroenterol. 2017, 54, 85–90. [Google Scholar] [CrossRef]
  34. Primec, M.; Klemenak, M.; Gioia, D.D.; Aloisio, I.; Cionci, N.B.; Quagliariello, A.; Gorenjak, M.; Micetic-Turk, D.; Langerholc, T. Clinical intervention using Bifidobacterium strains in celiac disease children reveals novel microbial modulators of TNF- α and short-chain fatty acids. Clin. Nutr. 2019, 38, 1373–1381. [Google Scholar] [CrossRef]
  35. Francavilla, R.; Piccolo, M.; Francavilla, A.; Polimeno, L.; Semeraro, F.; Cristofori, F.; Castellaneta, S.; Barone, M.; Indrio, F.; Gobbetti, M.; et al. Clinical and microbiological effect of a multispecies probiotic supplementation in celiac patients with persistent IBS-type symptoms: A randomized, double-blind, placebo-controlled, multicenter trial. J. Clin. Gastroenterol. 2019, 53, e117. [Google Scholar] [CrossRef]
  36. Smecuol, E.; Constante, M.; Temprano, M.P.; Costa, A.F.; Moreno, M.L.; Pinto-Sanchez, M.I.; Vazquez, H.; Stefanolo, J.P.; Gonzalez, A.F.; D’Adamo, C.R.; et al. Effect of Bifidobacterium infantis NSL super strain in symptomatic coeliac disease patients on long-term gluten-free diet: An exploratroy study. Benef Microbes. 2020, 11, 527–534. [Google Scholar] [CrossRef] [PubMed]
  37. Ali, B.; Khan, A.R. Efficacy of probiotics in management of celiac disease. Cureus 2022, 14, e22031. [Google Scholar] [CrossRef] [PubMed]
  38. Gan, J.; Nazarian, S.; Teare, J.; Darzi, A.; Ashrafian, H.; Thompson, A.J. A case for improved assessment of gut permeability: A meta-analysis quantifying the lactulose:mannitol ratio in coeliac and Crohn’s disease. BMC Gastroenterol. 2022, 22, 16. [Google Scholar] [CrossRef]
  39. Syage, J.A.; Mäki, M.; Leffler, D.A.; Silvester, J.A.; Sealey-Voyksner, J.A.; Wu, T.T.; Murray, J.A. A Composite Morphometric Duodenal Biopsy Mucosal Scale for Celiac Disease Encompassing both Morphology and Inflammation. Clin. Gastroenterol. Hepatol. 2023, 10, S1542–S3565. [Google Scholar] [CrossRef]
  40. Khorzoghi, M.S.; Rostami-Nejad, M.; Yadegar, A.; Dabiri, H.; Hadadi, A.; Rodrigo, L. Impact of probiotics on gut microbiota composition and clinical symptoms of coeliac disease patients following gluten-free diet. Contemp. Clin. Trials Commun. 2023, 35, 101201. [Google Scholar] [CrossRef]
  41. Zafeiropoulou, K.; Nichols, B.; Mackinder, M.; Biskou, O.; Rizou, E.; Karanikolou, A.; Clark, C.; Buchanan, E.; Cardigan, T.; Duncan, H.; et al. Alterations in Intestinal Microbiota of Children With Celiac Disease at the Time of Diagnosis and on a Gluten-free Diet. Gastroenterology 2020, 159, 2039–2051. [Google Scholar] [CrossRef]
  42. He, Y.; Wu, W.; Zheng, H.M.; Li, P.; McDonald, D.; Sheng, H.F.; Chen, M.X.; Chen, Z.H.; Ji, G.Y.; Zheng, Z.D.X.; et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat. Med. 2018, 24, 1532–1535. [Google Scholar] [CrossRef]
  43. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  44. Sterne, J.A.C.; Savović, J.; Page, M.J.; Elbers, R.G.; Blencowe, N.S.; Boutron, I.; Cates, C.J.; Cheng, H.Y.; Corbett, M.S.; Eldridge, S.M.; et al. RoB 2: A revised tool for assessing risk of bias in randomised trials. BMJ 2019, 366, l4898. [Google Scholar] [CrossRef] [PubMed]
  45. Sterne, J.A.C.; Hernán, M.A.; Reeves, B.C.; Savović, J.; Berkman, N.D.; Viswanathan, M.; Henry, D.; Altman, D.G.; Ansari, M.T.; Boutron, I.; et al. ROBINS-I: A tool for assessing risk of bias in non-randomised studies of interventions. BMJ 2016, 355, i4919. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow Chart of Study Selection.
Figure 1. Flow Chart of Study Selection.
Gastrointestdisord 06 00008 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fagbemi, L.O.; Soldaini, C.; Costabile, A.; Kolida, S.; Ciacci, C.; Jeanes, Y. Probiotic Interventions in Coeliac Disease: A Systematic Review with a Focus on Cardiovascular Risk. Gastrointest. Disord. 2024, 6, 114-130. https://doi.org/10.3390/gidisord6010008

AMA Style

Fagbemi LO, Soldaini C, Costabile A, Kolida S, Ciacci C, Jeanes Y. Probiotic Interventions in Coeliac Disease: A Systematic Review with a Focus on Cardiovascular Risk. Gastrointestinal Disorders. 2024; 6(1):114-130. https://doi.org/10.3390/gidisord6010008

Chicago/Turabian Style

Fagbemi, Lorretta Olu, Carlo Soldaini, Adele Costabile, Sofia Kolida, Carolina Ciacci, and Yvonne Jeanes. 2024. "Probiotic Interventions in Coeliac Disease: A Systematic Review with a Focus on Cardiovascular Risk" Gastrointestinal Disorders 6, no. 1: 114-130. https://doi.org/10.3390/gidisord6010008

Article Metrics

Back to TopTop