Next Article in Journal
The Role of Akkermansia muciniphila on Improving Gut and Metabolic Health Modulation: A Meta-Analysis of Preclinical Mouse Model Studies
Previous Article in Journal
Impact of Human Milk Oligosaccharides and Probiotics on Gut Microbiome and Mood in Autism: A Case Report
Previous Article in Special Issue
Effect of Probiotic Bacteria on the Gut Microbiome of Mice with Lipopolysaccharide-Induced Inflammation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

The Anti-Constipation Effect of Bifidobacterium Longum W11 Is Likely Due to a Key Genetic Factor Governing Arabinan Utilization

by
Francesco Di Pierro
1,2,3,*,
Nicola Zerbinati
3,
Massimiliano Cazzaniga
2,
Alexander Bertuccioli
1,4,
Chiara Maria Palazzi
1,
Ilaria Cavecchia
1,5,
Mariarosaria Matera
1,6,
Edoardo Labrini
7,
Valeria Sagheddu
7 and
Sara Soldi
7
1
Microbiota International Clinical Society, 10123 Torino, Italy
2
Scientific & Research Department, Velleja Research, 20125 Milano, Italy
3
Department of Medicine and Technological Innovation, University of Insubria, 21100 Varese, Italy
4
Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122 Urbino, Italy
5
Microbiomic Department, Koelliker Hospital, 10134 Turin, Italy
6
Department of Paediatric Emergencies, Misericordia Hospital, 58100 Grosseto, Italy
7
AAT—Advanced Analytical Technologies, Fiorenzuola d’Arda, 29017 Piacenza, Italy
*
Author to whom correspondence should be addressed.
Microorganisms 2024, 12(8), 1626; https://doi.org/10.3390/microorganisms12081626
Submission received: 21 July 2024 / Revised: 7 August 2024 / Accepted: 7 August 2024 / Published: 9 August 2024
(This article belongs to the Special Issue Probiotics, Prebiotics, and Gut Microbes)

Abstract

:
Recent investigations have highlighted, both experimentally and clinically, that probiotic strains equipped with arabinofuranosidase, in particular abfA and abfB, favor regular intestinal motility, thus counteracting constipation. By analyzing the gene expression and the proliferative response in the presence of arabinan of the probiotic B. longum W11, a strain previously validated as an anti-constipation probiotic, we have speculated that its response mechanism to arabinan can effectively explain its clinical action. Our approach could be used in the future to select probiotics endowed with arabinofuranosidase-related anti-constipation effects.

1. Introduction

Orally administered probiotics are widely used to alleviate functional constipation, a disorder affecting about 14% of the adult population and 9.5% of children, and recent meta-analysis reported that probiotic use moderately increases stool frequency and reduces the whole-gut transit time [1,2,3,4]. Probiotic ingestion can modify the gut microbial fermentation leading to a short-chain fatty acids (SCFAs) profile that interacts differently with the host immune system and enteric nervous system, thus ameliorating gut motility [5]. However, the effect of probiotics in functional constipation seems to be strain dependent [6]. Recently, Zhang et al. showed that B. longum strains possessing the encoding arabinofuranosidases cluster abfA can ameliorate functional constipation through enhanced arabinan utilization in the gut in both mice and humans [7]. Their results would seem to suggest the existence of a potentially specific therapeutic tool (i.e., B. longum equipped with abfA, administered together with arabinan fiber) for functional constipation. The B. longum strain W11 (LMG P-21586), a probiotic presenting intrinsic and non-transferable resistance to rifaximin [8,9,10], has also been described as being effective in treating functional constipation [11,12,13]. We have, therefore, investigated whether the results describing the role of the strain W11 in constipation could be linked to the presence and functionality of the same gene cluster. Three genes putatively encoding arabinofuranosidases have been described (abfI, abfA, and abfB) [14]. However, we have focused only on abfA and abfB as abfI has been shown not to affect arabinofuranosyl residues. As shown by our methods and results, we have evaluated both the presence of two arabinofuranosidases clusters (abfA and abfB) and their action in metabolizing arabinan which also supports the in vitro growth of the probiotic strain. The detection of the genes in the strain genome, their overexpression in arabinan-enriched medium, and the strain condition-specific in vitro growth have provided a probable explanation for its effectiveness in constipation.

2. Materials and Methods

2.1. In Silico Evaluation of abfA and abfB Genes Presence in B. Longum W11 Genome

For the in silico evaluation of the presence of the genes abfA and, abfB, we conducted a search using BV-BRC ver 3.35.5 software employing the deposited sequence of the B. longum strain W11 (NCBI Accession n° PRJNA356203).

2.2. Viable Counts of B. Longum W11 in Different Sugar-Restricted Media

Growth assays of B. longum W11 were performed in sugar-restricted basal medium [15] alone (1.0% Bacto peptone, 0.5% Bacto yeast extract, 0.5% sodium acetate trihydrate, 0.2% diammonium hydrogen citrate, 0.08% L-cysteine hydrochloride monohydrate, 0.02% magnesium sulfate heptahydrate, 1.36% L-ascorbic acid, 0.44% sodium carbonate anhydrous; all reagents from Merck, Milan, Italy) or supplemented with 2.0% glucose or arabinan (from sugar beet; Megazyme, MI, USA) [16]. The first viable counts were performed at T0 and after 48 h of incubation at 37 °C under anaerobic conditions. We performed viable counts according to ISO 29981/IDF 220:2010, a method for the selective enumeration of presumptive bifidobacteria in milk products by using a colony count technique at 37 °C under anaerobic conditions [17].

2.3. RNA Extraction, Quantification and Retro Transcription for Downstream Analysis

An appropriate volume of the T0 and of the three tested conditions (no sugar, glucose and arabinan) at T16, T24 and T48, was collected for the total RNA extraction. The volume was chosen to reach the cell concentration of 1–2 × 109 bacteria. The cultures were centrifuged, supernatants discarded, and the pellets underwent a lysis phase with 10 mg/mL of lysozyme and 300 U/mL of mutanolysin for 1 h and 30 min at 37 °C (enzymes from Merck, Milan, Italy). The Aurum™ Total RNA Mini Kit (Bio-Rad, Hercules, CA, USA) was used for the isolation and purification of total RNA. The extracted RNA was quantified through Nanodrop and the Qubit RNA HS assay (Invitrogen, Segrate, Italy). DNA contamination was checked with the Qubit ds DNA HS assay (Invitrogen, Segrate, Italy). A treatment with DNase was performed to remove gDNA traces. RNA underwent the retro transcription using the iScript™ cDNA Synthesis Kit (Bio-Rad, Hercules, CA, USA) following manufacturer instructions.

2.4. qPCR Amplification

Quantitative PCR (qPCR) analysis was performed using StepOne Plus Instrument (Applied Biosystems, Waltham, MA, USA) with fluorescence signal detection (SYBR green) after each amplification cycle. PCRs were performed in a 25-μL reaction mixture: 12.5 μL of Bio-Rad SsoAdvanced Universal mix (Bio-Rad, Hercules, CA, USA), 0.1 (for 16S DNA) or 1 (target genes) ng of cDNA, each forward and reverse primer at the proper concentration and sufficient nuclease-free water to obtain a final volume of 25 μL. Negative controls for each primer set were included in each run. Primer sets (16S DNA housekeeping gene, abfA and abfB) and thermal protocols were assessed as previously described [14]. Gene expression data were analyzed through the ΔΔCt method by comparing the relative concentration of the target gene in the treated sample to that of the control one (B. longum W11 at T0). Tests were performed in triplicate.

2.5. Statistical Analysis

We performed statistical analysis using the Brown–Forsythe method (Prism- GraphPad, v8). Statistical significance was set for p value < 0.05.

3. Results

We performed an in silico analysis of the deposited genome sequence of B. longum W11 to find the abfA and abfB genes. As shown in Figure 1, the bioinformatic research highlighted the presence of the two genes. We then evaluated the B. longum W11 viable counts in different sugar-restricted media containing glucose or arabinan at 2% (w/v). As shown in Table 1, the total counts obtained for B. longum W11 in arabinan-supplemented medium, in comparison with others, demonstrate that the strain actively metabolized this sugar, producing a proliferative effect of more than 1 log. We then quantified the abfA and abfB expression in the presence of arabinan at different time points. The precise RNA and DNA quantifications are available in Supplementary Table S1. As shown in Figure 2, after 16 h of incubation, the two genes abfA and abfB were overexpressed in the sugar-restricted medium supplemented with 2% of arabinan. After 24 h (as after 48 h), the overexpression detected at 16 h was no longer observable. Indeed, as previously described [7], abfA and abfB seem to be expressed more in the first phase of bacterial growth in the presence of arabinan.

4. Discussion

As recently described, B. longum strains holding the abfA cluster can ameliorate functional constipation in animals and in humans through enhanced arabinan utilization, metabolically yielding acetate, butyrate, chenodeoxycholic acid, and uracil [7]. Acetate and butyrate are two SCFAs implicated in improving intestinal motility either by interacting with G-protein-coupled receptor 41 (GPR41) and G-protein-coupled receptor 43 (GPR43) or directly acting on colonic smooth muscle [18,19]. Acetate and butyrate producers are associated with the bile acid level (i.e., chenodeoxycholic acid) in the gut [20,21]. For patients with irritable bowel syndrome constipation-type (IBS-C), it is reported that treatment with chenodeoxycholic acid can increase stool frequency, improve stool consistency, and improve ease of stool passage by acting on the membrane-bound G protein-coupled bile-acid receptor (e.g., TGR5) on enterocytes [22,23,24]. Similarly, a has been established between increased microbiota-induced uracil levels and decreased IBS-C disease activity [20]. One can assume that, in the proximal part of the colon, the glucose supply is sufficient to guarantee the microbial production of SCFAs, bile acids, and uracil. Differently, in the distal colon, the glucose supply should be exhausted. In this circumstance, microorganisms equipped with abfA clusters could stably proliferate and produce the beneficial metabolites above described.
B. longum W11, previously reported to be clinically effective against constipation [10,11,12,13] possesses the abfA and abfB genes and increases the expression of both abfA and abfB genes in the presence of arabinan. Moreover, B. longum W11 proliferates in the presence of arabinan. The results seem to be quite specific since the same strain (B. longum W11) does not increase the amounts of these two genes and does not proliferate in the presence of glucose. Our results should seem then to indicate a possible mechanism of action by which the strain W11 counteracts constipation. Moreover, our study demonstrates that B. longum W11 can be considered a “precision probiotics” [25] since, on a genetic basis, it presents different and concomitant peculiarities (rifaximin resistant and anti-constipation). Of course, our results do not exclude that other possible mechanisms of action could take part in ameliorating such clinical outcome. Having established a certain relationship between the presence of arabinofuranosidases and the anti-constipation effect, however, does not mean that any bacterial strain, with reference to those with evident probiotic characteristics, equipped with the same genes, is capable of anti-constipation efficacy. For the effect to be clinically observed, it is necessary for the genes to be functional, that the bacterium equipped with them remains viable inside the host for a sufficiently long time, and that it finds, in the ecosystem in which it has been introduced, an appropriate quantity of specific fiber, arabinan, or its potential precursors such as pectin or arabinoxylans [26,27]. As amply demonstrated, the abfA and abfB genes are not present only in the species B. longum [7]. By in silico analysis, we have explored the presence of this gene cluster in the probiotic genera like Bacillus and Bifidobacterium and in what until recently was simply called Lactobacillus [28]. Our analyses have revealed that this gene cluster is quite present among probiotics, especially among bifidobacteria. Among the probably most studied and worldwide marketed probiotic strains, such as the Bacillus clausii ENT-pro®strains [29], B. animalis lactis BB-12® [30], and L. rhamnosus GG® [31], the latter, reviewed as ineffective in functional constipation [32,33], does not present arabinofuranosidase genes. In contrast, B. clausii strains and BB-12® have both been described to have clinical anti-constipation effects [34,35,36,37,38].

5. Conclusions

Although our work was limited mainly to the B. longum W11 strain, it lays the groundwork for identifying a rationale between the strain’s ability to metabolize arabinan and the consequent overexpression of the abfA and abfB genes. This finding could reveal the mechanism underlying the anti-constipation action exerted by the strain under investigation. As such, this approach might be used to select or better characterize other probiotics with the same function. Last but not least, to our knowledge, our work identifies for the first time the possibility for a “precision probiotic” to be characterized by two completely unrelated features.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms12081626/s1, Supplementary Table S1: RNA and DNA quantifications.

Author Contributions

Conceptualization, F.D.P. and V.S.; methodology, V.S., E.L. and S.S.; validation, F.D.P. and V.S.; formal analysis, F.D.P. and V.S.; investigation; V.S., E.L. and S.S.; resources, F.D.P. and V.S.; data curation, V.S., E.L. and S.S.; writing—original draft preparation; F.D.P. and V.S.; writing—review and editing, F.D.P. and V.S.; visualization, F.D.P. and V.S.; supervision, I.C., M.M., N.Z., A.B., C.M.P. and M.C.; project administration, F.D.P., V.S. and S.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The raw data supporting the conclusions of this article will be made available by the authors on request.

Conflicts of Interest

F.D.P. belongs to the Scientific Board of Pharmextracta (the company trading the strain tested); A.B. and M.C. are Pharmextracta consultants. The other authors declare no conflicts of interest and no financial relationships.

References

  1. Garzon Mora, N.; Jaramillo, A.P. Effectiveness of Probiotics in Patients with Constipation: A Systematic Review and Meta-Analysis. Cureus 2024, 16, e52013. [Google Scholar] [CrossRef] [PubMed]
  2. van der Schoot, A.; Helander, C.; Whelan, K.; Dimidi, E. Probiotics and synbiotics in chronic constipation in adults: A systematic review and meta-analysis of randomized controlled trials. Clin. Nutr. 2022, 41, 2759–2777. [Google Scholar] [CrossRef] [PubMed]
  3. Dong, M.; Wu, Y.; Zhang, M.; Chen, P.; Zhang, Z.; Wang, S. Effect of probiotics intake on constipation in children: An umbrella review. Front. Nutr. 2023, 10, 1218909. [Google Scholar] [CrossRef] [PubMed]
  4. Zhang, C.; Jiang, J.; Tian, F.; Zhao, J.; Zhang, H.; Zhai, Q.; Chen, W. Meta-analysis of randomized controlled trials of the effects of probiotics on functional constipation in adults. Clin. Nutr. 2020, 39, 2960–2969. [Google Scholar] [CrossRef] [PubMed]
  5. Dimidi, E.; Christodoulides, S.; Scott, S.M.; Whelan, K. Mechanisms of action of probiotics and the gastrointestinal microbiota on gut motility and constipation. Adv. Nutr. 2017, 8, 484–494. [Google Scholar] [CrossRef] [PubMed]
  6. Vriesman, M.H.; Koppen, I.J.N.; Camilleri, M.; Di Lorenzo, C.; Benninga, M.A. Management of functional constipation in children and adults. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 21–39. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, C.; Yu, L.; Ma, C.; Jiang, S.; Zhang, Y.; Wang, S.; Tian, F.; Xue, Y.; Zhao, J.; Zhang, H.; et al. A key genetic factor governing arabinan utilization in the gut microbiome alleviates constipation. Cell Host Microbe 2023, 31, 1989–2006.e8. [Google Scholar] [CrossRef] [PubMed]
  8. Graziano, T.; Amoruso, A.; Nicola, S.; Deidda, F.; Allesina, S.; Pane, M.; Piffanelli, P.; Strozzi, F.; Mogna, L.; Del Piano, M. The Possible Innovative Use of Bifidobacterium longum W11 in Association with Rifaximin: A New Horizon for Combined Approach? J. Clin. Gastroenterol. 2015, 50, S153–S156. [Google Scholar] [CrossRef] [PubMed]
  9. Di Pierro, F.; Bertuccioli, A.; Pane, M.; Ivaldi, L. Effects of rifaximin-resistant Bifidobacterium longum W11 in subjects with symptomatic uncomplicated diverticular disease treated with rifaximin. Minerva Gastroenterol. Dietol. 2019, 65, 259–264. [Google Scholar] [CrossRef]
  10. Di Pierro, F.; Pane, M. Bifidobacterium longum W11: Uniqueness and individual or combined clinical use in association with rifaximin. Clin. Nutr. ESPEN 2021, 42, 15–21. [Google Scholar] [CrossRef]
  11. Colecchia, A.; Vestito, A.; La Rocca, A.; Pasqui, F.; Nikiforaki, A.; Festi, D.; Symbiotic Study Group. Effect of a symbiotic preparation on the clinical manifestations of irritable bowel syndrome, constipation-variant. Results of an open, uncontrolled multicenter study. Minerva Gastroenterol. Dietol. 2006, 52, 349–358. [Google Scholar] [PubMed]
  12. Dughera, L.; Elia, C.; Navino, M.; Cisarò, F.; ARMONIA Study Group. Effects of symbiotic preparations on constipated irritable bowel syndrome symptoms. Acta Biomed. 2007, 78, 111–116. [Google Scholar] [PubMed]
  13. Amenta, M.; Cascio, M.T.; Di Fiore, P.; Venturini, I. Diet and chronic constipation. Benefits of oral supplementation with symbiotic zir fos (Bifidobacterium longum W11 + FOS Actilight). Acta Biomed. 2006, 77, 157–162. [Google Scholar] [PubMed]
  14. Savard, P.; Roy, D. Determination of Differentially Expressed Genes Involved in Arabinoxylan Degradation by Bifidobacterium longum NCC2705 Using Real-Time RT-PCR. Probiotics Antimicrob. Proteins 2009, 1, 121. [Google Scholar] [CrossRef]
  15. Komeno, M.; Hayamizu, H.; Fujita, K.; Ashida, H. Two Novel α-l-Arabinofuranosidases from Bifidobacterium longum subsp. longum Belonging to Glycoside Hydrolase Family 43 Cooperatively Degrade Arabinan. Appl. Environ. Microbiol. 2019, 85, e02582-18. [Google Scholar] [CrossRef] [PubMed]
  16. Moon, J.S.; Shin, S.Y.; Choi, H.S.; Joo, W.; Cho, S.K.; Li, L.; Kang, J.H.; Kim, T.J.; Han, N.S. In vitro digestion and fermentation properties of linear sugar-beet arabinan and its oligosaccharides. Carbohydr. Polym. 2015, 131, 50–56. [Google Scholar] [CrossRef]
  17. ISO 29981:2010|IDF; Milk Products-Enumeration of Presumptive Bifidobacteria-Colony Count Technique at 37 °C. ISO: Geneva, Switzerland, 2010. Available online: https://www.iso.org/standard/45765.html (accessed on 20 July 2024).
  18. Tazoe, H.; Otomo, Y.; Kaji, I.; Tanaka, R.; Karaki, S.I.; Kuwahara, A. Roles of short-chain fatty acids receptors, GPR41 and GPR43 on colonic functions. J. Physiol. Pharmacol. 2008, 59, 251–262. [Google Scholar]
  19. Soret, R.; Chevalier, J.; De Coppet, P.; Poupeau, G.; Derkinderen, P.; Segain, J.P.; Neunlist, M. Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. Gastroenterology 2010, 138, 1772–1782. [Google Scholar] [CrossRef]
  20. Mars, R.A.T.; Yang, Y.; Ward, T.; Houtti, M.; Priya, S.; Lekatz, H.R.; Tang, X.; Sun, Z.; Kalari, K.R.; Korem, T.; et al. Longitudinal multi-omics reveals subset-specific mechanisms underlying irritable bowel syndrome. Cell 2020, 182, 1460–1473.e17. [Google Scholar] [CrossRef]
  21. Lancaster, S.M.; Lee-McMullen, B.; Abbott, C.W.; Quijada, J.V.; Hornburg, D.; Park, H.; Perelman, D.; Peterson, D.J.; Tang, M.; Robinson, A.; et al. Global, distinctive, and personal changes in molecular and microbial profiles by specific fibers in humans. Cell Host Microbe 2022, 30, 848–862.e7. [Google Scholar] [CrossRef]
  22. Bazzoli, F.; Malavolti, M.; Petronelli, A.; Barbara, L.; Roda, E. Treatment of constipation with chenodeoxycholic acid. J. Int. Med. Res. 1983, 11, 120–123. [Google Scholar] [CrossRef]
  23. Rao, A.S.; Wong, B.S.; Camilleri, M.; Odunsi-Shiyanbade, S.T.; McKinzie, S.; Ryks, M.; Burton, D.; Carlson, P.; Lamsam, J.; Singh, R.; et al. Chenodeoxycholate in females with irritable bowel syndrome-constipation: A pharmacodynamic and pharmacogenetic analysis. Gastroenterology 2010, 139, 1549–1558. [Google Scholar] [CrossRef]
  24. Alemi, F.; Poole, D.P.; Chiu, J.; Schoonjans, K.; Cattaruzza, F.; Grider, J.R.; Bunnett, N.W.; Corvera, C.U. The receptor TGR5 mediates the prokinetic actions of intestinal bile acids and is required for normal defecation in mice. Gastroenterology 2013, 144, 145–154. [Google Scholar] [CrossRef] [PubMed]
  25. Veiga, P.; Suez, J.; Derrien, M.; Elinav, E. Moving from probiotics to precision probiotics. Nat. Microbiol. 2020, 5, 878–880. [Google Scholar] [CrossRef]
  26. Pascale, N.; Gu, F.; Larsen, N.; Jespersen, L.; Respondek, F. The Potential of Pectins to Modulate the Human Gut Microbiota Evaluated by In Vitro Fermentation: A Systematic Review. Nutrients 2022, 14, 3629. [Google Scholar] [CrossRef] [PubMed]
  27. Zannini, E.; Bravo Núñez, Á.; Sahin, A.W.; Arendt, E.K. Arabinoxylans as Functional Food Ingredients: A Review. Foods 2022, 11, 1026. [Google Scholar] [CrossRef]
  28. Zheng, J.; Wittouck, S.; Salvetti, E.; Franz, C.M.A.P.; Harris, H.M.B.; Mattarelli, P.; O’Toole, P.W.; Pot, B.; Vandamme, P.; Walter, J. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int. J. Syst. Evol. Microbiol. 2020, 70, 2782–2858. [Google Scholar] [CrossRef] [PubMed]
  29. Khatri, I.; Sharma, G.; Subramanian, S. Composite genome sequence of Bacillus clausii, a probiotic commercially available as Enterogermina®, and insights into its probiotic properties. BMC Microbiol. 2019, 19, 307. [Google Scholar] [CrossRef]
  30. Jungersen, M.; Wind, A.; Johansen, E.; Christensen, J.E.; Stuer-Lauridsen, B.; Eskesen, D. The Science behind the Probiotic Strain Bifidobacterium animalis subsp. lactis BB-12(®). Microorganisms 2014, 2, 92–110. [Google Scholar] [CrossRef]
  31. Boggio Marzet, C.; Burgos, F.; Del Compare, M.; Gerold, I.; Tabacco, O.; Vinderola, G. Approach to probiotics in pediatrics: The role of Lactobacillus rhamnosus GG. Arch. Argent. Pediatr. 2022, 120, e1–e7. [Google Scholar]
  32. Chmielewska, A.; Szajewska, H. Systematic review of randomised controlled trials: Probiotics for functional constipation. World J. Gastroenterol. 2010, 16, 69–75. [Google Scholar] [PubMed]
  33. Wegh, C.A.M.; Benninga, M.A.; Tabbers, M.M. Effectiveness of Probiotics in Children with Functional Abdominal Pain Disorders and Functional Constipation: A Systematic Review. J. Clin. Gastroenterol. 2017, 52 (Suppl. S1), S10–S26. [Google Scholar] [CrossRef] [PubMed]
  34. Lojanatorn, P.; Phrommas, J.; Tanpowpong, P.; Getsuwan, S.; Lertudomphonwanit, C.; Treepongkaruna, S. Efficacy of Bacillus clausii in Pediatric Functional Constipation: A Pilot of a Randomized, Double-Blind, Placebo-Controlled Trial. Indian Pediatr. 2023, 60, 453–458. [Google Scholar] [CrossRef] [PubMed]
  35. Giua, C.; Romano, F.; Keber, E.; Pellegrino, P.; Perez, M.; Uboldi, M.C.; SIFAC group of clinical pharmacists (SGCP). A Prospective Real-World Study of Bacillus clausii Evaluating Use, Treatment Habits and Patient Satisfaction in Italian Community Pharmacies: The PEGASO Study. Drugs–Real World Outcomes 2024, 11, 137–147. [Google Scholar] [CrossRef] [PubMed]
  36. Pitkala, K.H.; Strandberg, T.E.; Finne Soveri, U.H.; Ouwehand, A.C.; Poussa, T.; Salminen, S. Fermented cereal with specific bifidobacteria normalizes bowel movements in elderly nursing home residents. A randomized, controlled trial. J. Nutr. Health Aging 2007, 11, 305–311. [Google Scholar] [PubMed]
  37. Uchida, K.; Akashi, K.; Kusunoki, I.; Ikeda, T.; Katano, N.; Motoshima, H.; Benno, Y. Effect of fermented milk containing Bifidobacterium lactis BB-12® on stool frequency, defecation, fecal microbiota and safety of excessive ingestion in healthy female students. J. Nutr. Food 2005, 8, 39–51. [Google Scholar]
  38. Nishida, S.; Gotou, M.; Akutsu, S.; Ono, M.; Hitomi, Y.; Nakamura, T.; Iino, H. Effect of yogurt containing Bifidobacterium lactis BB-12 on improvement of defecation and fecal microflora of healthy female adults. Milk Sci. 2004, 53, 71–80. [Google Scholar]
Figure 1. In silico evaluation of the presence of abfA and abfB genes.
Figure 1. In silico evaluation of the presence of abfA and abfB genes.
Microorganisms 12 01626 g001
Figure 2. abfA and abfB genes’ expression at T0, T16 and T24 in the tested conditions. * p < 0.05; ** p < 0.01.
Figure 2. abfA and abfB genes’ expression at T0, T16 and T24 in the tested conditions. * p < 0.05; ** p < 0.01.
Microorganisms 12 01626 g002
Table 1. Viable counts at two timepoints (T0 and T24).
Table 1. Viable counts at two timepoints (T0 and T24).
Sugar restricted, T0Sugar restricted, T24
−5−6CFU/mLLog (CFUs)−5−6CFU/mLLog (CFUs)DL (CFUs)
BLW11187151.8 × 1077.3161231.67 × 1077.20.0
Sugar restricted + glucose, T0Sugar restricted + glucose, T24
−5−6CFU/mLLog (CFUs)−7−8CFU/mLLog (CFUs)DL (CFUs)
BLW11175141.7 × 1077.24464.55 × 1077.70.4
Sugar restricted + arabinan, T0Sugar restricted + arabinan, T24
−5−6CFU/mLLog (CFUs)−7−8CFU/mLLog (CFUs)DL (CFUs)
BLW11179181.8 × 1077.382118.45 × 1078.91.7
BLW11: B. longum W11; DL: Delta Log; CFU: Colony Forming Units.
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

Di Pierro, F.; Zerbinati, N.; Cazzaniga, M.; Bertuccioli, A.; Palazzi, C.M.; Cavecchia, I.; Matera, M.; Labrini, E.; Sagheddu, V.; Soldi, S. The Anti-Constipation Effect of Bifidobacterium Longum W11 Is Likely Due to a Key Genetic Factor Governing Arabinan Utilization. Microorganisms 2024, 12, 1626. https://doi.org/10.3390/microorganisms12081626

AMA Style

Di Pierro F, Zerbinati N, Cazzaniga M, Bertuccioli A, Palazzi CM, Cavecchia I, Matera M, Labrini E, Sagheddu V, Soldi S. The Anti-Constipation Effect of Bifidobacterium Longum W11 Is Likely Due to a Key Genetic Factor Governing Arabinan Utilization. Microorganisms. 2024; 12(8):1626. https://doi.org/10.3390/microorganisms12081626

Chicago/Turabian Style

Di Pierro, Francesco, Nicola Zerbinati, Massimiliano Cazzaniga, Alexander Bertuccioli, Chiara Maria Palazzi, Ilaria Cavecchia, Mariarosaria Matera, Edoardo Labrini, Valeria Sagheddu, and Sara Soldi. 2024. "The Anti-Constipation Effect of Bifidobacterium Longum W11 Is Likely Due to a Key Genetic Factor Governing Arabinan Utilization" Microorganisms 12, no. 8: 1626. https://doi.org/10.3390/microorganisms12081626

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop