Skip to main content

Profiling of the fecal microbiota and circulating microRNA-16 in IBS subjects with Blastocystis infection : a case–control study

Abstract

Irritable bowel syndrome (IBS) is a prevalent gastrointestinal (GI) tract disorder. Although the main reason for IBS is not clear, the interaction between intestinal microorganisms and the gut barrier seems to play an important role in pathogenesis of IBS. The current study aimed to investigate the effect of Blastocystis on the gut microbiota profile and the circulation levels of microRNA (mir)-16 of IBS patients compared to healthy subjects. Stool and blood samples were collected from 80 participants including 40 samples from each IBS and healthy group. Upon DNA extraction from stool samples, barcoding region and quantitative real-time PCR were analyzed to investigate Blastocystis and the microbiota profile, respectively. RNA was extracted from serum samples of included subjects and the expression of mir-16 was evaluated using stem-loop protocol and qreal-time PCR. Significant changes between IBS patients and healthy controls was observed in Firmicutes, Actinobacteria, Faecalibacterium, and Alistipes. In IBS patients, the relative abundance of Bifidobacteria was directly correlated with the presence of Blastocystis, while Alistipes was decreased with Blastocystis. Lactobacillus was significantly increased in Blastocystis carriers. In healthy subjects, the relative abundance of Bifidobacteria was decreased, but Alistipes was increased in Blastocystis carriers. The changes in the Firmicutes/Bacteroidetes ratio was not significant in different groups. The relative expression of mir-16 in Blastocystis-negative IBS patients and healthy carriers was significantly overexpressed compared to control group. The presence of Blastocystis, decreased the relative expression of mir-16 in IBS patients compared to Blastocystis-negative IBS patients. The present study revealed that Blastocystis has the ability to change the abundance of some phyla/genera of bacteria in IBS and healthy subjects. Moreover, Blastocystis seems to  modulate the relative expression of microRNAs  to control the gut atmosphere, apply its pathogenicity, and provide a favor niche for its colonization.

Introduction

Irritable bowel syndrome (IBS) is one of the most common functional disorders of the digestive system [1, 2]. The pathophysiological mechanisms behind the IBS have not been cleared yet; however, the main symptoms are abdominal bloating, visceral pain, and changes in the stool pattern [3]. IBS is a heterogeneous disorder with four main types: IBS-C with constipation stool type, IBS-D with diarrhea stool type, IBS-M with intermittent bowel pattern, and IBS-U with not-classified pattern of stool [4]. This disease affects about 10–20% of the world's population [5]. It was estimated that about 20% of the population and more than half of the visits to gastroenterology clinics in the UK complain from IBS symptoms, with a higher prevalence among women [6]. It is believed that the genetic, psychological and social factors, motility changes in the digestive system, increased visceral sensitivity, gut–brain axis disorders, activation of mast cells in the intestinal mucosa, and changes in serotonin metabolism play major role in the development and pathogenesis of IBS [7]. Furthermore, the interaction between microorganisms and the digestive system is thought to be involved in the progress of this disease [1]. There is evidence indicating significant role of gut microbiota disturbance in IBS patients [6]. Variation in the microbiota composition and metabolites compared to the healthy subjects highlights the role of gut microbiota in IBS [8, 9]. Therefore, the gut microbiota has emerged as a key issue in the investigation of gastrointestinal diseases such as IBS [4, 10].

The microbial community that lives in the human body (in the gut) is called the microbiota, which consists of a wide range of microorganisms including bacteria, viruses, fungi, and other eukaryotes [10]. Bacteria cover a large and eukaryotes comprise very small portion of the gut microbiota; nevertheless, protists such as Blastocystis may play important role in gut microbiota composition and richness [1, 11].

Blastocystis is a cosmopolitan protozoan in humans and animals [12], which is commonly reported from patients who suffer from IBS [13, 14]. Blastocystis is the most common parasite living in the human body, which affects about 5–30% of people in advanced and industrial societies and about 30–50% of people in non-industrial societies [15]. Although pathogenicity of Blastocystis is controversial, it was suggested that the protozoan can affect intestinal permeability [16, 17], and probably induces apoptosis throughout the gut [18].

There is a little data about the correlation of Blastocystis and the gut barrier functions. Recently, the role of Blastocystis on the regulation of micro RNAs (miRNAs/mir) and tight junction (TJ) proteins of the gut has been suggested [19]. It was shown that total antigen derived from Blastocystis subtype 3 induces the expression of mir-223 and mir-874, which both are associated with gut barrier dysfunctions [19]. MiRNAs are short non-coding, single-stranded RNAs that found in all eukaryotic and human cells, with an average length of 19–25 nucleotides, which control protein-coding gene expression at the post-transcriptional stage [20]. MiRNAs, for example mir-16, are considered as regulators in controlling of the expression of intestinal TJ proteins and the intestinal epithelial barrier [21]. A negative correlation was suggested between the expression levels of mir-16 and TJ proteins (cingulin and claudin-2) in IBS-D patients [21].

In the current study, we investigated the pattern of selected gut microbiota composition and mir-16 expression in IBS patients and healthy controls regarding the presence of Blastocystis.

Materials and methods

Ethics approval and consent to participate

This study received ethical approval from the Ethics Committee of the Islamic Azad University, Science and Research Branch, Tehran, Iran (IR.IAU.SRB.REC.1400.241).

Informed consent was verbally obtained from all participates and/or their legal guardian(s). For those patients with age ≤ 16, informed consent was obtained from their respective parent(s)/guardian(s) as well.

Sample collection and DNA extraction

Stool samples were collected from 80 participants including 40 samples from each IBS and healthy group. To analyze the effects of Blastocystis on the gut microbiota and mir-16 expression levels, four groups were considered for the study including 20 samples for each Blastocystis-positive IBS patients (BPI), Blastocystis-negative IBS patients (BNI), Blastocystis-positive healthy subjects (BPH), and Blastocystis-negative healthy subjects (BNH) (Fig. 1). All stool samples were examined for the presence of intestinal parasites using direct microscopy. The presence of Blastocystis in stool samples was confirmed using amplification of the “barcoding region” through the small subunit ribosomal RNA (SSU rRNA) gene [22]. The presence of any other cysts/oocysts/eggs in stool samples was considered as exclusion criteria. Healthy controls were collected from those subjects who either intend to participate in our study or referred to the laboratory for periodical checkup. Healthy controls did not complain any gastrointestinal disorders and those who suffer from any gastrointestinal problems were excluded from the study.

Fig. 1
figure 1

The flowchart describing allocation of samples in studied groups

Demographic data, drug consumption, stool appearance at the time of sampling, and symptoms during sampling were recorded. The presence of IBS was confirmed by gastroenterologists, and consumption of any antibiotics, immunosuppressant drugs, alcohol, following a specific diet, and the presence of any immunodeficiency were considered as exclusion criteria. In addition, in healthy controls, the presence of any gastrointestinal symptoms was considered as exclusion criterion.

To analyze the expression of mir-16, peripheral blood samples were collected from all participants, as well. To analyze the expression of circulating mir-16, serum was isolated from blood samples. All samples were transferred to the Parasitology lab in the Research Institute for Gastroenterology and Liver Diseases for further analyses. Stool and serum samples were kept out at −20 and −70 ˚C, respectively, until experiments.

To characterize the presence of Blastocystis and selected microbiota, total DNA was extracted from stool samples using stool DNA extraction kit (Yekta Tajhiz, Tehran, Iran). For this purpose, 200 mg of stool samples was washed three times with sterile phosphate buffer saline (PBS; pH = 7.5) and after discarding supernatant, DNA was extracted from remained pellet. Isolated DNA was stored at −20 °C until use.

Microbiota profiling

Quantitative real-time PCR was employed to investigate the microbiota profiling of a selection of phyla/classes/genera according to the literature review, including phyla Firmicutes, Bacteroidetes, Actinobacteria, and Fusobacteria, and genera Lactobacillus, Bifidobacteria, Streptococcus, Faecalibacterium, Blautia, Ruminococcus, Roseburia, Alistipes, Prevotella, and Methanobacteria. Amplification was performed using phyla- and genus-specific, and universal primers, which were mentioned (Table 1).

Table 1 Targeted microorganism and their primers

Quantitative real-time PCR was performed in total volume 15 μL using a Rotor-Gene Q (QIAGEN, Germany) real-time instrument based on the following conditions: 7.5 μL of 2X real-time PCR Master Mix (BioFACT™, Korea), 5 ρM of each primer, 3.5 μL of distilled water, and 3 μL of template DNA. The cycling profile was 95 °C for 10 min followed by 40 cycles: 95 °C for 25 s, 56 °C for 30 s and 72 °C for 20 s and ramping from 70 °C to 95 °C at 1 °Cs − 1. Positive control and sterile distillated water were run together with each sample set, as positive and negative controls, respectively. To exclude the probability of false amplification and primer-dimers, melting curve analysis was performed. In the cases of cycle of threshold (Ct) value more than 35, amplification plot and melting profile were considered to exclude negative samples. The relative abundance of bacterial taxonomic group was calculated according to the method described elsewhere [23, 24] based on the following formula:

$$X \, = \, \left( {{\text{Eff}}.{\text{Univ}}} \right)^{C_t {\text{ univ}}} / \, \left( {{\text{Eff}}.{\text{Spec}}} \right)^{C_t {\text{ spec}}} \times {1}00.$$

The Eff. Univ represents the calculated efficiency of the universal primers for Eubacteria (2 = 100% and 1 = 0%), the Eff. Spec indicates the efficiency of the taxon-specific primers. In addition, Ct univ and Ct spec refer to the released Ct by the thermocycler, and “X” is the percentage (%) of taxon-specific 16S rRNA gene copy numbers in single fecal sample.

RNA extraction, cDNA synthesis, and quantitative real-time PCR

The expression level of mir-16 was evaluated using stem-loop reverse transcriptase and real-time PCR using primers and protocols, which were previously explained [19]. For this purpose, RNA was extracted using Trizol extraction protocol (BioMix) from serum samples, which were collected from enrolled participants. Briefly, 750 µL of Trizol reagent was added to 250 µL of serum. After agitating for 10 s and cooling at − 20 ˚C for 10 min, 250 µL of chloroform was added and cooled again at – 20 ˚C for 7 min. Samples were vortexed at 4 ˚C for 20 min, centrifuged at 12000 × g, and supernatant was collected and mixed with isopropanol alcohol. After centrifuging at 12000 × g in 4 ˚C for 20 min, RNA was isolated from remained pellet. The concentration of extracted RNAs was determined by NanoDrop (NanoDrop Technologies, USA) apparatus, and RNA adjustment was performed before complementary DNA (cDNA) synthesis. cDNA was constructed for mir-16 and U6, as housekeeping gene, using cDNA synthesis kit (Pars Tous, Mashhad, Iran), as explained previously [19].

Relative expression of the mir-16 in serum samples of participants were evaluated by qreal-time PCR using Rotor-Gene Q (Qiagen, Germany) in a 20 µL reaction mixture containing 10 µL SYBR Green qPCR master mix 2X (Ampliqon, Denmark), 5 ρM of each primer, and 2 µL of constructed cDNA as template. The amplification condition for mi6-16 was adjusted with previously released protocol [19]. As mentioned above, to avoid from non-specific amplification, melting curves were analyzed for each run. The relative expression of mir-16 was adjusted to housekeeping gene (U6 snRNA) and calculated comparing to Blastocystis-negative healthy controls [19].

Statistical analysis

The descriptive statistics were presented as frequency and prevalence. The quantity of bacteria and levels of mir-16 were presented as mean and standard deviation (SD). The normality of distribution was tested by the Shapiro–Wilk test, and Student’s t test (independent and paired sample) was employed for the parametric data analysis. Mann–Whitney and Wilcoxon tests were used for non-parametric data analysis. In addition, the principal coordinate analysis (PCoA) was calculated using the same program based on the Bray–Curtis dissimilarity method. P value < 0.05 was considered statistically significant. REST, SPSS v.24, GraphPad Prism version 8.3.0.538, and the open-source statistical program R version 3.6.1 (R Core Team, Vienna, Austria) software were applied for data analysis. GraphPad Prism software version 8.3.0.538 was employed for visualizing data.

Results

Demographic data

Regardless the presence of Blastocystis, the mean of age ± SD in IBS patients and healthy subjects was 41.9 ± 13.02 and 40.33 ± 13.93, respectively. The gender distribution in all studied subjects was 39 (48.75%) and 41 (51.25%) for female and male, respectively (Table 2).

Table 2 Demographic data of participated subjects

Microbiota comparisons

Real-time PCR was performed on all included samples and results of analyses showed significant changes between IBS patients and healthy controls in four phyla and genera including, Firmicutes, Actinobacteria, Faecalibacterium, and Alistipes, respectively. Accordingly, the relative abundance of Firmicutes, Actinobacteria, and Faecalibacterium, in IBS patients was 14.52 ± 3.04, 7.73 ± 1.17, and 9.75 ± 1.41, respectively, which were higher than the relative abundance of mentioned taxa in healthy subjects. In contrast the relative abundance of Alistipes was increased from 2.06 ± 2.58 in IBS patients to 5.13 ± 5.79 in healthy subjects.

Regarding the presence of Blastocystis, the relative abundance of Bifidobacteria in IBS patients was directly associated with the presence of Blastocystis and was increased with the protist. In contrast, the relative abundance of Alistipes was conversely associated with Blastocystis. A statistically significant change in the relative abundance of taxa was seen in Lactobacillus, which was increased from 5.01 ± 1.46 in Blastocystis-negative IBS (BNI) subjects to 5.77 ± 0.93 in BPI patients (P value = 0.033). In non-IBS subjects, the presence of Blastocystis significantly affected the relative abundance of Bifidobacteria and Alistipes. Accordingly, the relative abundance of Bifidobacteria was conversely related with the Blastocystis, and was decreased from 8.12 ± 2.61 in Blastocystis-negative healthy BNH to 7.58 ± 1.84 in BPH (P value = 0.028). In contrast, the abundance of Alistipes was increased from 2.84 ± 3.42 in BNH to 4.35 ± 5.67 in BPH (P value = 0.000) (Table 3; Fig. 2).

Table 3 The relative abundance of the gut microbiota in four studied groups
Fig. 2
figure 2

The bar-plot analysis represents the relative abundance and distribution of each targeted bacteria in studied groups as mean ± SD

Firmicutes/Bacteroidetes ratio

To evaluate the hemostasis condition of the gut microbiota in studied groups, the Firmicutes/Bacteroidetes ratio was evaluated. The analysis showed a similar violin shape of the ratio in groups, while there were no significant differences in calculated ratio between IBS and healthy controls, regardless the presence of Blastocystis. The presence of Blastocystis changed the violin shape of Firmicutes/Bacteroidetes ratio in different groups, but the differences were not statistically significant (Fig. 3).

Fig. 3
figure 3

The Firmicutes to Bacteroidetes (F/B) ratio in fecal samples of A IBS patients and healthy controls, and B IBS patients and healthy controls based on the presence of Blastocystis. Statistical analyses based on Mann–Whitney test showed no significant association

Principal coordinate analysis

Diversity of studied microbiota was evaluated by the PCoA analysis. The PCoA suggested a similar diversity in investigated taxa, regardless the presence of Blastocystis. The comparison of groups to each other represented a similarity through the diversity of microbiota among three groups BNH, BNI patients, and BPI patients, while they were different from healthy carriers (Fig-PCoA) (Fig. 4A, B).

Fig. 4
figure 4

The PCoA analysis represented a comparison between the microbial communities from A IBS and healthy controls A without and B with considering the presence of Blastocystis. The comparison of groups showed a difference between healthy Blastocystis carriers with other three groups

Relative expression of mir-16

The relative expression of mir-16 revealed non-significant downregulation in BPI group (16.80 ± 59.70; P value = 0.191) compared to BNH. The relative expression of mir-16 in BNI patients and BPH was significantly overexpressed compared to control group (120.3 ± 163; P value = 0.0023) and (32.17 ± 54.13; P value = 0.014), respectively. In IBS patients, the presence of Blastocystis, significantly changed the relative expression of mir-16. Accordingly, the presence of Blastocystis, decreased the relative expression of mir-16 from 120.3 ± 163 to 16.80 ± 59.70 (P value = 0.0011) (Fig. 5).

Fig. 5
figure 5

Comparison of the relative expression of circulating mir-16 in studied groups compared with Blastocystis-negative healthy subjects. Independent t test was employed to analyze statistical association. The analysis suggests a modulatory role of Blastocystis on the expression levels of mir-16 in IBS patients. *P value < 0.05; **P value < 0.01

Discussion

Alongside with increasing studies on the distribution of Blastocystis, many reports have indicated a high prevalence of this protist in IBS patients. However, the association between the presence of IBS and carrying Blastocystis is still unclear. IBS is a multifactorial disease of the GI tract, which is associated with the gut microbiota disturbance [33, 34]. Moreover, correlation between colonization of Blastocystis and the richness and diversity of the gut microbiota has been evaluated [35,36,37]. On the other hand, correlation between Blastocystis and the gut permeability has complicated IBS-Blastocystis associations [19, 38,39,40]. In the current study, we investigated correlation between Blastocysts, with a couple of bacterial taxa and the serum levels of mir-16 in IBS patients compared to healthy controls.

As a finding, regardless the presence of Blastocystis, the relative abundance of Firmicutes, Actinobacteria, and Faecalibacterium in IBS patients was significantly higher than healthy controls, which was in contrast with Alistipes. The richness of Firmicutes in IBS patients compared to healthy controls is controversial. Although available evidence supports our results, which indicate higher abundance of Firmicutes in IBS patients [8], many studies have demonstrated lower richness of Firmicutes in IBS patients compared to control subjects [2, 41, 42]. This contentious trend was also observed for the abundance of Actinobacteria, and Faecalibacterium [2, 8, 43, 44]. However, this controversial results most probably backs to the method of investigations and IBS types [1]. For example, Zhuang et al. [34], demonstrated that the abundance of Firmicutes was decreased in IBS-D patients, while Bacteroidetes was increased. Nevertheless, in the line of previous studies [8, 45], the relative abundance of Alistipes in IBS patients was decreased in comparison to healthy subjects.

Alistipes is commonly isolated from healthy gut, however, there are contradicting reports of some species such as A. obesi and A. ihumii in morbidly obese and anorexia patients, respectively [46]. It was suggested that Alistipes is associated with short chain fatty acids (SCFAs) and could be an acetate and propionate producer [47, 48]. Moreover, SCFAs significantly contribute in pathophysiology and severity of IBS [49]. Butyrate, acetate, and propionate levels in stool are conversely related with IBS-C, while it is positively associated with IBS-D [50, 51]. In the current study, diarrhea was not seen in most of participated IBS patients, therefore, higher and lower abundance of Firmicutes and Alistipes in our samples compared to healthy controls could be rational. Splitting IBS and healthy groups based on Blastocystis, revealed a decreased abundance of Bifidobacterium and Alistipes in BPH and BNH controls, respectively. However, Lactobacillus and Bifidobacterium were enriched in BPI patients. Lactobacillus, Alistipes, and Bifidobacterium are SCFAs-producing bacterial genera [47, 52]. On the other hand, enrichment of SCFAs-producing bacteria was documented to be correlated with Blastocystis subtypes. For example, Deng et al. [53] demonstrated a positive association between Blastocystis subtype 4 with SCFAs-producing bacteria, while this correlation was converse for subtype 7. Controversial correlation between Blastocystis subtypes and enrichment of SCFAs-producing bacteria was supported by other studies [54, 55]. In the current study, Blastocystis -positive subjects were probably consisted of a diverse subtype pattern. Therefore, conflicting correlation between enrichment of SCFAs-producing bacteria and the presence of Blastocystis could be related to not only the gut conditions, like IBS, but also different subtypes.

The intestinal barrier plays an important role in maintaining and balancing intestinal homeostasis, and prevents the transfer of contents from the lumen to the lower layer and the circulatory system [56]. Therefore, the disturbance in the functions of the intestinal barrier can cause disturbances in the immune system of the GI tract [56, 57]. TJ proteins are responsible for the integrity of the intestinal barriers, and the presence of disorder in these proteins affects the intercellular permeability [56]. In addition, it is believed that the permeability of the intestinal barrier is regulated by miRNAs [58, 59]. Martinez et al. [21] demonstrated a significant reverse association between the mir-16 and TJ proteins, caludin-2 and cingulin, in IBS-D patients. They reported that the number of mast cells was negatively correlated with has-mir-16 and has-mir-125b, while it was positively associated with caludin-2 and cingulin, highlighting the significant role of miRNAs in expression of TJ proteins and the severity of clinical symptoms in IBS patients [21]. The protecting role of mir-16 in IBS-D patients was supported by Xi et al. [60], who showed that mir-16 may inhibit TLR4/NF-κB pathway signaling, leading to prevention from apoptosis and stimulation of enterocyte viability. In a single study, evaluating the effects of Blastocystis on the expression of miRNAs, Mohammad Rahimi et al. [19], suggested significant effects of Blastocystis soluble total antigen (B3STA), extracted from subtype 3, on the expression of a couple of miRNAs (mir-223, mir-874, and mir-29a) and claudin-7, which all are contributed to the gut barrier integrity. They showed that although B3STA decreased the expression of mir-29a, it induced the expression of mir-223, mir-874, which are all involved in the dysfunction of the gut barrier. In the current study, the circulating level of mir-16 in BPI patients was significantly lower than BNI group, and was similar to BNH subjects. Our results suggested that Blastocystis may induce inflammation and elevate the gut permeability via dysregulating miRNAs in IBS patients, while the association between Blastocystis and miRNAs could be different in healthy controls. However, colonization of Blastocystis in the gut may either directly or indirectly induce inflammation and affects the gut integrity via changing the microbiota, in a non-healthy condition, like IBS. Although there are studies describing the effects of Blastocystis on the gut microbiota, this is the first study analyzing the effects of this protist on not only the gut microbiota, but also the transcriptional expression of mir-16, which is involved in the gut permeability in IBS patients. However, the most important limitation of this study is the lack of metagenomics data, which can provide much more data about the microbiome of the gut in IBS patients. In addition, the lack of a microarray analysis to highlight a table of miRNAs, which are affected by Blastocystis, is another limitation, that are recommended to be considered in future studies.

Conclusion

The present study revealed that although the presence of Blastocystis was not significantly associated with the Firmicutes/Bacteroidetes ratio, this protozoan decreased the abundance of Bifidobacterium and Alistipes in healthy controls, while in IBS patients, Lactobacillus and Bifidobacterium were enriched in Blastocystis carriers. In addition, although in the line of previous studies mir-16 was strongly increased in IBS patients, the presence of Blastocystis can modulated circulating levels of mir-16. Taken together, it seems that Blastocystis may induce inflammation, disrupt the gut integrity, and modulate the gut environment to provide a stable condition for its colonization. However, due to the lack of metagenomics data to exclude the probable role of other variables, the role of Blastocystis in modulating the gut microbiota and the expression of miRNAs should be carefully interpreted.

Availability of data and materials

All generated data from the current study are included in the article.

References

  1. Olyaiee A, Sadeghi A, Yadegar A, Mirsamadi ES, Mirjalali H. Gut microbiota shifting in irritable bowel syndrome: the mysterious role of Blastocystis sp. Front Med (Lausanne). 2022;9: 890127.

    Article  PubMed  Google Scholar 

  2. Mei L, Zhou J, Su Y, Mao K, Wu J, Zhu C, He L, Cui Y. Gut microbiota composition and functional prediction in diarrhea-predominant irritable bowel syndrome. BMC Gastroenterol. 2021;21:105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Johnsen PH, Hilpüsch F, Valle PC, Goll R. The effect of fecal microbiota transplantation on IBS related quality of life and fatigue in moderate to severe non-constipated irritable bowel: Secondary endpoints of a double blind, randomized, placebo-controlled trial. EBioMedicine. 2020;51: 102562.

    Article  PubMed  Google Scholar 

  4. Nagel R, Traub RJ, Allcock RJ, Kwan MM, Bielefeldt-Ohmann H. Comparison of faecal microbiota in Blastocystis-positive and Blastocystis-negative irritable bowel syndrome patients. Microbiome. 2016;4:47.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Dale HF, Lied GA. Gut microbiota and therapeutic approaches for dysbiosis in irritable bowel syndrome: recent developments and future perspectives. Turk J Med Sci. 2020;50:1632–41.

    Article  CAS  PubMed  Google Scholar 

  6. Zhao L, Yang W, Chen Y, Huang F, Lu L, Lin C, Huang T, Ning Z, Zhai L, Zhong LL, et al. A Clostridia-rich microbiota enhances bile acid excretion in diarrhea-predominant irritable bowel syndrome. J Clin Invest. 2020;130:438–50.

    Article  CAS  PubMed  Google Scholar 

  7. Gasbarrini A, Lauritano EC, Garcovich M, Sparano L, Gasbarrini G. New insights into the pathophysiology of IBS: intestinal microflora, gas production and gut motility. Eur Rev Med Pharmacol Sci. 2008;12(Suppl 1):111–7.

    PubMed  Google Scholar 

  8. Rajilić-Stojanović M, Biagi E, Heilig HG, Kajander K, Kekkonen RA, Tims S, de Vos WM. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology. 2011;141:1792–801.

    Article  PubMed  Google Scholar 

  9. Shankar V, Reo NV, Paliy O. Simultaneous fecal microbial and metabolite profiling enables accurate classification of pediatric irritable bowel syndrome. Microbiome. 2015;3:73.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Fagoonee S, Pellicano R. Does the microbiota play a pivotal role in the pathogenesis of irritable bowel syndrome? J Clin Med. 2019;8:1808.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Nieves-Ramírez M, Partida-Rodríguez O, Laforest-Lapointe I, Reynolds L, Brown E, Valdez-Salazar A, Morán-Silva P, Rojas-Velázquez L, Morien E, Parfrey L. Asymptomatic intestinal colonization with protist Blastocystis is strongly associated with distinct microbiome ecological patterns. Msystems. 2018;3:e00007-00018.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Nemati S, Zali MR, Johnson P, Mirjalali H, Karanis P. Molecular prevalence and subtype distribution of Blastocystis sp. in Asia and in Australia. J Water Health. 2021;19:687–704.

    Article  PubMed  Google Scholar 

  13. Cifre S, Gozalbo M, Ortiz V, Soriano JM, Merino JF, Trelis M. Blastocystis subtypes and their association with Irritable Bowel Syndrome. Med Hypotheses. 2018;116:4–9.

    Article  PubMed  Google Scholar 

  14. Dogruman-Al F, Simsek Z, Boorom K, Ekici E, Sahin M, Tuncer C, Kustimur S, Altinbas A. Comparison of methods for detection of Blastocystis infection in routinely submitted stool samples, and also in IBS/IBD Patients in Ankara. Turkey PLoS One. 2010;5: e15484.

    Article  PubMed  Google Scholar 

  15. Beghini F, Pasolli E, Truong TD, Putignani L, Cacciò SM, Segata N. Large-scale comparative metagenomics of Blastocystis, a common member of the human gut microbiome. ISME J. 2017;11:2848–63.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Asnicar F, Berry SE, Valdes AM, Nguyen LH, Piccinno G, Drew DA, Leeming E, Gibson R, Le Roy C, Khatib HA. Microbiome connections with host metabolism and habitual diet from 1,098 deeply phenotyped individuals. Nat Med. 2021;27:321–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Karamati SA, Mirjalali H, Niyyati M, Yadegar A, Asadzadeh Aghdaei H, Haghighi A, Seyyed Tabaei SJ. Association of Blastocystis ST6 with higher protease activity among symptomatic subjects. BMC Microbiol. 2021;21:285.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mirza H, Wu Z, Teo JD, Tan KS. Statin pleiotropy prevents rho kinase-mediated intestinal epithelial barrier compromise induced by Blastocystis cysteine proteases. Cell Microbiol. 2012;14:1474–84.

    Article  CAS  PubMed  Google Scholar 

  19. Mohammad Rahimi H, Yadegar A, Asadzadeh Aghdaei H, Mirjalali H, Zali MR. Modulation of microRNAs and claudin-7 in Caco-2 cell line treated with Blastocystis sp., subtype 3 soluble total antigen. BMC Microbiol. 2022;22:111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chatterjee B, Sarkar M, Bose S, Alam MT, Chaudhary AA, Dixit AK, Tripathi PP, Srivastava AK: MicroRNAs: Key modulators of inflammation-associated diseases. In: Seminars in Cell & Developmental Biology. Elsevier; 2023. https://doi.org/10.1016/j.semcdb.2023.01.009

  21. Martínez C, Rodiño-Janeiro BK, Lobo B, Stanifer ML, Klaus B, Granzow M, González-Castro AM, Salvo-Romero E, Alonso-Cotoner C, Pigrau M. miR-16 and miR-125b are involved in barrier function dysregulation through the modulation of claudin-2 and cingulin expression in the jejunum in IBS with diarrhoea. Gut. 2017;66:1537–8.

    Article  PubMed  Google Scholar 

  22. Norouzi M, Pirestani M, Arefian E, Dalimi A, Sadraei J, Mirjalali H. Exosomes secreted by Blastocystis subtypes affect the expression of proinflammatory and anti-inflammatory cytokines (TNFα, IL-6, IL-10, IL-4). Front Med (Lausanne). 2022;9: 940332.

    Article  PubMed  Google Scholar 

  23. Gholam-Mostafaei FS, Azimirad M, Naseri K, Nabavi-Rad A, Asadzadeh Aghdaei H, Shahrokh S, Ebrahimi Daryani N, Yadegar A, Zali MR. Intestinal microbiota changes pre-and post-fecal microbiota transplantation for treatment of recurrent Clostridioides difficile infection among Iranian patients with concurrent inflammatory bowel disease. Front Microbiol. 2023;14:499.

    Article  Google Scholar 

  24. De Gregoris TB, Aldred N, Clare AS, Burgess JG. Improvement of phylum-and class-specific primers for real-time PCR quantification of bacterial taxa. J Microbiol Method. 2011;86:351–6.

    Article  Google Scholar 

  25. Moszak M, Szulińska M, Bogdański P. You are what you eat—the relationship between diet, microbiota, and metabolic disorders—a review. Nutrients. 2020;12:1096.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Thursby E, Juge N. Introduction fo the human gut flora. Biochem J. 2017;474:1823–36.

    Article  CAS  PubMed  Google Scholar 

  27. Hermann-Bank ML, Skovgaard K, Stockmarr A, Larsen N, Mølbak L. The gut microbiotassay: a high-throughput qPCR approach combinable with next generation sequencing to study gut microbial diversity. BMC Genomic. 2013;14:1–14.

    Article  Google Scholar 

  28. Springham N, Robert G. Experience based co-design reduces formal complaints on an acute mental health ward. BMJ Open Qual. 2015;4(u209153): w203970.

    Google Scholar 

  29. Keshavarz Azizi Raftar S, Ashrafian F, Yadegar A, Lari A, Moradi HR, Shahriary A, Azimirad M, Alavifard H, Mohsenifar Z, Davari M. The protective effects of live and pasteurized Akkermansia muciniphila and its extracellular vesicles against HFD/CCl4-induced liver injury. Microbiol Spect. 2021;9:e00484-e1421.

    Article  Google Scholar 

  30. Wang I-K, Lai H-C, Yu C-J, Liang C-C, Chang C-T, Kuo H-L, Yang Y-F, Lin C-C, Lin H-H, Liu Y-L. Real-time PCR analysis of the intestinal microbiotas in peritoneal dialysis patients. Appl Environ Microbiol. 2012;78:1107–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Quigley EM. Gut bacteria in health and disease. Gastroenterol Hepatol. 2013;9:560.

    Google Scholar 

  32. Vigsnæs LK, Brynskov J, Steenholdt C, Wilcks A, Licht TR. Gram-negative bacteria account for main differences between faecal microbiota from patients with ulcerative colitis and healthy controls. Beneficial Microb. 2012;3:287–97.

    Article  Google Scholar 

  33. Tap J, Derrien M, Törnblom H, Brazeilles R, Cools-Portier S, Doré J, Störsrud S, Le Nevé B, Öhman L, Simrén M. Identification of an intestinal microbiota signature associated with severity of irritable bowel syndrome. Gastroenterology. 2017;152:111-123.e118.

    Article  PubMed  Google Scholar 

  34. Zhuang X, Tian Z, Li L, Zeng Z, Chen M, Xiong L. Fecal microbiota alterations associated with diarrhea-predominant irritable bowel syndrome. Front Microbiol. 2018;9:1600.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Deng L, Wojciech L, Gascoigne NR, Peng G, Tan KS. New insights into the interactions between Blastocystis, the gut microbiota, and host immunity. PLoS Pathog. 2021;17: e1009253.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kim MJ, Lee YJ, Kim TJ, Won EJ. Gut microbiome profiles in colonizations with the enteric protozoa Blastocystis in Korean populations. Microorganisms. 2021;10:34.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Stensvold CR, Sørland BA, Berg R, Andersen LO, van der Giezen M, Bowtell JL, El-Badry AA, Belkessa S, Kurt Ö, Nielsen HV. Stool microbiota diversity analysis of Blastocystis-positive and Blastocystis-negative individuals. Microorganisms. 2022;10:326.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Olivo-Diaz A, Romero-Valdovinos M, Gudino-Ramirez A, Reyes-Gordillo J, Jimenez-Gonzalez DE, Ramirez-Miranda ME, Martinez-Flores WA, Martinez-Hernandez F, Flisser A, Maravilla P. Findings related to IL-8 and IL-10 gene polymorphisms in a Mexican patient population with irritable bowel syndrome infected with Blastocystis. Parasitol Res. 2012;111:487–91.

    Article  PubMed  Google Scholar 

  39. Ismail MH, Molan AL, Abbas SK. Serological levels of cytokines in irritable bowel syndrome (IBS) patients and non-IBS subjects with and without Blastocystis spp. infection. Ann Parasitol. 2022;68:77–85.

    PubMed  Google Scholar 

  40. Nagel R, Traub RJ, Kwan MM, Bielefeldt-Ohmann H. Blastocystis specific serum immunoglobulin in patients with irritable bowel syndrome (IBS) versus healthy controls. Parasit Vectors. 2015;8:453.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Chen H, Ou R, Tang N, Su W, Yang R, Yu X, Zhang G, Jiao J, Zhou X. Alternation of the gut microbiota in irritable bowel syndrome: an integrated analysis based on multicenter amplicon sequencing data. J Transl Med. 2023;21:117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lee SM, Kim N, Yoon H, Kim YS, Choi SI, Park JH, Lee DH. Compositional and functional changes in the gut microbiota in irritable bowel syndrome patients. Gut Liver. 2021;15:253–61.

    Article  PubMed  Google Scholar 

  43. Lopetuso LR, Petito V, Graziani C, Schiavoni E, Paroni Sterbini F, Poscia A, Gaetani E, Franceschi F, Cammarota G, Sanguinetti M, et al. Gut microbiota in health, diverticular disease, irritable bowel syndrome, and inflammatory bowel diseases: time for microbial marker of gastrointestinal disorders. Dig Dis. 2018;36:56–65.

    Article  PubMed  Google Scholar 

  44. Barandouzi ZA, Lee J, Maas K, Starkweather AR, Cong XS. Altered gut microbiota in irritable bowel syndrome and its association with food components. J Pers Med. 2021;11:35.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Carroll IM, Ringel-Kulka T, Keku TO, Chang YH, Packey CD, Sartor RB, Ringel Y. Molecular analysis of the luminal- and mucosal-associated intestinal microbiota in diarrhea-predominant irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol. 2011;301:G799-807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Parker BJ, Wearsch PA, Veloo ACM, Rodriguez-Palacios A. The genus alistipes: gut bacteria with emerging implications to inflammation, cancer, and mental health. Front Immunol. 2020;11:906.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. O’Riordan KJ, Collins MK, Moloney GM, Knox EG, Aburto MR, Fülling C, Morley SJ, Clarke G, Schellekens H, Cryan JF. Short chain fatty acids: microbial metabolites for gut-brain axis signalling. Mol Cell Endocrinol. 2022;546: 111572.

    Article  CAS  PubMed  Google Scholar 

  48. Oliphant K, Allen-Vercoe E. Macronutrient metabolism by the human gut microbiome: major fermentation by-products and their impact on host health. Microbiome. 2019;7:91.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Jiang W, Wu J, Zhu S, Xin L, Yu C, Shen Z. The role of short chain fatty acids in irritable bowel syndrome. J Neurogastroenterol Motil. 2022;28:540–8.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Sun Q, Jia Q, Song L, Duan L. Alterations in fecal short-chain fatty acids in patients with irritable bowel syndrome: A systematic review and meta-analysis. Medicine (Baltimore). 2019;98: e14513.

    Article  CAS  PubMed  Google Scholar 

  51. Gargari G, Taverniti V, Gardana C, Cremon C, Canducci F, Pagano I, Barbaro MR, Bellacosa L, Castellazzi AM, Valsecchi C, et al. Fecal clostridiales distribution and short-chain fatty acids reflect bowel habits in irritable bowel syndrome. Environ Microbiol. 2018;20:3201–13.

    Article  CAS  PubMed  Google Scholar 

  52. Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat Rev Gastroenterol Hepatol. 2019;16:461–78.

    Article  PubMed  Google Scholar 

  53. Deng L, Wojciech L, Png CW, Kioh DYQ, Gu Y, Aung TT, Malleret B, Chan ECY, Peng G, Zhang Y, et al. Colonization with two different Blastocystis subtypes in DSS-induced colitis mice is associated with strikingly different microbiome and pathological features. Theranostics. 2023;13:1165–79.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Deng L, Wojciech L, Png CW, Koh EY, Aung TT, Kioh DYQ, Chan ECY, Malleret B, Zhang Y, Peng G, et al. Experimental colonization with Blastocystis ST4 is associated with protective immune responses and modulation of gut microbiome in a DSS-induced colitis mouse model. Cell Mol Life Sci. 2022;79:245.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Mayneris-Perxachs J, Arnoriaga-Rodríguez M, Garre-Olmo J, Puig J, Ramos R, Trelis M, Burokas A, Coll C, Zapata-Tona C, Pedraza S, et al. Presence of Blastocystis in gut microbiota is associated with cognitive traits and decreased executive function. Isme J. 2022;16:2181–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Schoultz I, Keita ÅV. The intestinal barrier and current techniques for the assessment of gut permeability. Cells. 1909;2020:9.

    Google Scholar 

  57. Lee SH. Intestinal permeability regulation by tight junction: implication on inflammatory bowel diseases. Intestin Res. 2015;13:11.

    Article  Google Scholar 

  58. Kozieł MJ, Ziaja M, Piastowska-Ciesielska AW. Intestinal barrier, claudins and mycotoxins. Toxins. 2021;13:758.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Zhao X, Zeng H, Lei L, Tong X, Yang L, Yang Y, Li S, Zhou Y, Luo L, Huang J. Tight junctions and their regulation by non-coding RNAs. Int J Biol Sci. 2021;17:712.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Xi M, Zhao P, Li F, Bao H, Ding S, Ji L, Yan J. MicroRNA-16 inhibits the TLR4/NF-κB pathway and maintains tight junction integrity in irritable bowel syndrome with diarrhea. J Biol Chem. 2022;298(11): 102461.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank all members of the Foodborne and Waterborne Diseases Research Center for their supports. We would like to sincerely thank Dr. Sara Mohammad Ali Gol for her assistance in collecting samples from IBS patients.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

Conceived and designed: HM; sample collecting: AO, AS; laboratory experiments: AO, AY, ESM, HM; writing the manuscript: AO, HM; reviewing and editing the manuscript: HM. All authors read and approved the final version of the manuscript.

Corresponding author

Correspondence to Hamed Mirjalali.

Ethics declarations

Ethics approval and consent to participate

All experimental protocols were in accordance to the ethical principles and the national norms and standards for conducting Medical Research in Iran. This study was approved by the ethical standards (IR.IAU.SRB.REC.1400.241) released by Ethical Review Committees of Islamic Azad University- Science and Research Branch, Tehran, Iran.

Consent for publication

All authors declare that they have seen and approved the submitted version of this manuscript.

Competing interests

The authors declare that they have no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Olyaiee, A., Yadegar, A., Mirsamadi, E.S. et al. Profiling of the fecal microbiota and circulating microRNA-16 in IBS subjects with Blastocystis infection : a case–control study. Eur J Med Res 28, 483 (2023). https://doi.org/10.1186/s40001-023-01441-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-023-01441-8

Keywords