Next Article in Journal
Application of Direct Thermal Desorption–Gas Chromatography–Mass Spectrometry for Determination of Volatile and Semi-Volatile Organosulfur Compounds in Onions: A Novel Analytical Approach
Previous Article in Journal
The Autoinducer N-Octanoyl-L-Homoserine Lactone (C8-HSL) as a Potential Adjuvant in Vaccine Formulations
Previous Article in Special Issue
The Probiotic Strains Bifidοbacterium lactis, Lactobacillus acidophilus, Lactiplantibacillus plantarum and Saccharomyces boulardii Regulate Wound Healing and Chemokine Responses in Human Intestinal Subepithelial Myofibroblasts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue “Gut Microbiota, Inflammatory Bowel Diseases, and Therapeutic Targets”

Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(5), 714; https://doi.org/10.3390/ph16050714
Submission received: 25 April 2023 / Accepted: 4 May 2023 / Published: 8 May 2023
(This article belongs to the Special Issue Gut Microbiota, Inflammatory Bowel Diseases, and Therapeutic Targets)
The gut microbiota and its overall genetic composition, the microbiome, have been the subject of extensive research over the last decade within the fields of genomics, transcriptomics and metabolomics, and their role in various other targeted approaches and advanced technologies has been explored [1]. Various studies have demonstrated that the composition of the gut microbiota differs in patients with Inflammatory Bowel Disease (IBD) compared to the eubiotic microbiota of healthy controls [2]. Additionally, gut microbiota is different between patients with Ulcerative Colitis (UC) and Crohn’s Disease (CD) or among subtypes of CD [3]. This dysbiotic composition involves an increase or decrease in specific intestinal bacterial species in patients with IBD, and more specifically, a decrease in the abundance and diversity of specific genera has been suggested to play a key role in the pathogenetic mechanisms of intestinal inflammation [4]. Having underlined the significant role of gut microbiota in IBD, this Special Issue was mainly focused on its involvement in health and IBD pathogenesis, its interactions and effects on the host, and on new possible therapeutic strategies involving the use of probiotics.
Most studies in IBD have been focused on microbiome disturbances in the lower gastrointestinal tract, where bacteria are more abundant, and a dysbiosis of gut microbiota has been documented, whereas similar studies in the duodenum are rare, mainly in animal models [5,6]. In this Special Issue, Schmidt et al. studied the composition of the bacterial flora, attached to the duodenal mucosa, in children with CD, since the prevalence of CD is more common in children [7]. They examined bacterial relative abundance, alpha and beta composition, and diversity, analyzing the 16S ribosomal RNA bacterial gene in biopsies from duodenum and terminal ileum from treatment-naïve CD children and age- and sex-matched controls. They demonstrated that the duodenal microbiome in children is distinct from the terminal ileum and is characterized by an increased abundance of Bacteroidales in active CD children and by an increased abundance of Pseudomonodales and Spirochetes in healthy controls.
In addition to changes in microbial diversity in IBD, distinct microbial signatures have been reported in subtypes of the disease or in cases of colectomy or complications [8]. Dovrolis et al. have shown distinct microbiome signatures in complicated Crohn’s disease subphenotypes, with the bacterial diversity of the inflamed group found to be closer to that of healthy controls than to that of structuring and penetrating groups [3]. In this Special Issue, Bálint et al. examined the possible effect of anatomical variations, after colectomy, on the diversity of gut microbiota, using a clinical model of two separate bowel conditions: UC patients after ileal pouch-anal anastomosis (IPAA) surgery, compared with UC patients, familial adenomatous polyposis (FAP) patients after IPAA surgery and healthy controls [9]. They found that the microbiota diversity of the non-IBD colectomized group was closer to that of the colectomized UC patients than the healthy controls, suggesting that the anatomical status construct has an influence on microbiota composition, in addition to intestinal inflammation.
The microbiota metabolism ends in the production of several metabolites that participate in interactions with the host and ultimately regulate mucosal homeostasis in gut and influence the pathophysiology of various disorders [10,11]. In addition, the interaction between the intestinal microbiota and the host at the metabolite receptor level appears to be involved in intestinal inflammation and IBD [12]. Ιn this Special Issue, three reviews summarize the recent knowledge and discuss in detail the role of the microbial metabolites in IBD. A comprehensive review by Aldars-García et al. studies the implication of microbial metabolites in the pathogenetic mechanisms of IBD. Furthermore, it examines the changes in the metabolome related to disease subphenotype and response to treatment, as well as the possibility that these changes may be useful biomarkers in the management of patients with IBD [10]. Another review, by Dowdell and Colgan, gathers the current knowledge on metabolic host–microbiota interactions in the pathogenesis of IBD, focusing mainly on their implication in autophagy and their role in gut inflammation [13]. Finally, Bunt et al. comprehensively examine various metabolites produced through the fermentation and metabolism of various dietary sources by gut microbiota and report data showing anti-inflammatory and antioxidant properties of these metabolites in in vitro and in vivo models of IBD [14]. They also discuss the potential therapeutic effects of these compounds. Existing studies have revealed a critical role played by microbial metabolites as effectors for productive innate immunity and maintenance of homeostasis, and their changes are associated with mechanisms of intestinal inflammation. However, at present, there is insufficient evidence to determine the role of the microbial metabolite–host receptor interactions in pathogenesis and/or the potential for translating it into clinical practice.
The central role of the gut microbiota in regulating health and disease, mentioned earlier, has received prominent emphasis in the development of potential therapeutic strategies. In the regulation of gut microbiota and treatment of dysbiosis, various methods such as fecal transplantation and probiotic, prebiotic and postbiotic approaches have been used to influence the host microbiota and treat IBD [15,16,17,18]. It appears that effective modulation of the microbiota composition may be a therapeutic approach or may enhance the effectiveness of an existing treatment [19]. In this Special Issue, Filidou & Kolios present and discuss the current knowledge of the mechanisms underlying the healing process in the intestinal mucosa and examine the role of the gut microbiota in healing pathophysiological mechanisms in gut homeostasis and intestinal inflammation [20]. They also discuss the steps in these host–microbe interactions that result in mucosal healing, highlighting possible therapeutic targets, presenting current data on the effect of probiotics on intestinal healing in animal models of colitis, and indicating that probiotics can even lead to the amelioration of intestinal inflammation and improvement in mucosal wound healing.
Tarapatzi et al. in vitro examined the role of certain probiotics strains in wound healing, using cultures of subepithelial gut myofibroblasts; in this Special Issue, the authors demonstrated that the mix of Bifidobacterium lactis, Lactobacillus acidophilus, Lactiplantibacillus plantarum and Saccharomyces boulardii induced a moderate, but statistically significant, increase in the mRNA of certain healing factors, such as collagen type I and III, fibronectin and tissue factor and myofibroblasts migration, thus indicating a positive effect in wound healing [21]. In addition, they found that this probiotic combination induced a mild increase in the mRNA of specific chemokines, and they suggested that this was due rather to a possible immune alertness than a pathological inflammatory response, contributing in this way to the host’s defenses. The use of multiple probiotic strains was found to have a better effect on mucosal immunity than the use of single probiotics.
It has also been suggested that the regulation of microbiota composition may enhance the effectiveness of an existing treatment [19]. In this Special Issue, Saber et al. examined a possible synergistic effect of rosuvastatin and Lactobacillus in an experimental model of colitis [22]. Previous studies have shown a protective effect of rosuvastatin in animal models of colitis, and it has been suggested as a potential therapeutic agent in IBD [23,24]. In this study, the combination of rosuvastatin/Lactobacillus was found to be safe and decreased the Disease Activity Index of colitic animals via an anti-inflammatory effect, thus indicating a possible role in the management of IBD.
In conclusion, this Special Issue has once again highlighted that in IBD, the gut microbiome is distinct and varies from healthy controls, and that the anatomical variations in the intestine can significantly influence its composition. Apart from the microbiome itself, it is also underlined that a dysbiotic microbial metabolome may also contribute to the IBD pathogenesis, while some may exert positive effects on the host’s immune functions. Finally, aiming in this direction, this Special Issue highlights the potentially beneficial role of probiotics, not only in mucosal healing, but also in triggering and maintaining a mild immunological alertness in the host’s cell populations.

Author Contributions

Conceptualization, E.F. and G.K.; writing—original draft preparation, E.F. and G.K.; writing—review and editing, G.K.; All authors have read and agreed to the published version of the manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dovrolis, N.; Filidou, E.; Kolios, G. Systems biology in inflammatory bowel diseases: On the way to precision medicine. Ann. Gastroenterol. 2019, 32, 233–246. [Google Scholar] [CrossRef] [PubMed]
  2. Glassner, K.L.; Abraham, B.P.; Quigley, E.M. The microbiome and inflammatory bowel disease. J. Allergy Clin. Immunol. 2020, 145, 16–27. [Google Scholar] [CrossRef] [PubMed]
  3. Dovrolis, N.; Drygiannakis, I.; Filidou, E.; Kandilogiannakis, L.; Arvanitidis, K.; Tentes, I.; Kolios, G.; Valatas, V. Gut Microbial Signatures Underline Complicated Crohn’s Disease but Vary Between Cohorts; An In Silico Approach. Inflamm. Bowel Dis. 2018, 25, 217–225. [Google Scholar] [CrossRef] [PubMed]
  4. Haneishi, Y.; Furuya, Y.; Hasegawa, M.; Picarelli, A.; Rossi, M.; Miyamoto, J. Inflammatory Bowel Diseases and Gut Microbiota. Int. J. Mol. Sci. 2023, 24, 3817. [Google Scholar] [CrossRef] [PubMed]
  5. Suchodolski, J.S.; Dowd, S.; Wilke, V.; Steiner, J.M.; Jergens, A.E. 16S rRNA Gene Pyrosequencing Reveals Bacterial Dysbiosis in the Duodenum of Dogs with Idiopathic Inflammatory Bowel Disease. PLoS ONE 2012, 7, e39333. [Google Scholar] [CrossRef]
  6. Santana, P.T.; Rosas, S.L.B.; Ribeiro, B.E.; Marinho, Y.; de Souza, H.S.P. Dysbiosis in Inflammatory Bowel Disease: Pathogenic Role and Potential Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 3464. [Google Scholar] [CrossRef]
  7. Schmidt, K.; Noel-MacDonnell, J.; Vyhlidal, C.; Heruth, D.P.; Singh, V.; Ahmed, A.A.; Hudson, T.; Williams, V.; Shakhnovich, V. Characterization of the Mucosally-Adherent Duodenal Microbiome in Children with and without Crohn’s Disease. Pharmaceuticals 2022, 15, 850. [Google Scholar] [CrossRef]
  8. Halfvarson, J.; Brislawn, C.J.; Lamendella, R.; Vázquez-Baeza, Y.; Walters, W.A.; Bramer, L.M.; D’Amato, M.; Bonfiglio, F.; McDonald, D.; Gonzalez, A.; et al. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat. Microbiol. 2017, 2, 17004. [Google Scholar] [CrossRef]
  9. Bálint, A.; Farkas, K.; Méhi, O.; Kintses, B.; Vásárhelyi, B.M.; Ari, E.; Pál, C.; Madácsy, T.; Maléth, J.; Szántó, K.J.; et al. Functional Anatomical Changes in Ulcerative Colitis Patients Determine Their Gut Microbiota Composition and Consequently the Possible Treatment Outcome. Pharmaceuticals 2020, 13, 346. [Google Scholar] [CrossRef]
  10. Aldars-García, L.; Gisbert, J.P.; Chaparro, M. Metabolomics Insights into Inflammatory Bowel Disease: A Comprehensive Review. Pharmaceuticals 2021, 14, 1190. [Google Scholar] [CrossRef]
  11. Neuffer, J.; González-Domínguez, R.; Lefèvre-Arbogast, S.; Low, D.Y.; Driollet, B.; Helmer, C.; Du Preez, A.; de Lucia, C.; Ruigrok, S.R.; Altendorfer, B.; et al. Exploration of the Gut-Brain Axis through Metabolomics Identifies Serum Propionic Acid Associated with Higher Cognitive Decline in Older Persons. Nutrients 2022, 14, 4688. [Google Scholar] [CrossRef]
  12. Melhem, H.; Kaya, B.; Ayata, C.K.; Hruz, P.; Niess, J.H. Metabolite-Sensing G Protein-Coupled Receptors Connect the Diet-Microbiota-Metabolites Axis to Inflammatory Bowel Disease. Cells 2019, 8, 450. [Google Scholar] [CrossRef]
  13. Dowdell, A.S.; Colgan, S.P. Metabolic Host–Microbiota Interactions in Autophagy and the Pathogenesis of Inflammatory Bowel Disease (IBD). Pharmaceuticals 2021, 14, 708. [Google Scholar] [CrossRef]
  14. Bunt, D.V.; Minnaard, A.J.; El Aidy, S. Potential Modulatory Microbiome Therapies for Prevention or Treatment of Inflammatory Bowel Diseases. Pharmaceuticals 2021, 14, 506. [Google Scholar] [CrossRef]
  15. Tkach, S.; Dorofeyev, A.; Kuzenko, I.; Falalyeyeva, T.; Tsyryuk, O.; Kovalchuk, O.; Kobyliak, N.; Abenavoli, L.; Boccuto, L. Efficacy and safety of fecal microbiota transplantation via colonoscopy as add-on therapy in patients with mild-to-moderate ulcerative colitis: A randomized clinical trial. Front. Med. 2023, 9, 1049849. [Google Scholar] [CrossRef]
  16. Lê, A.; Mantel, M.; Marchix, J.; Bodinier, M.; Jan, G.; Rolli-Derkinderen, M. Inflammatory bowel disease therapeutic strategies by modulation of the microbiota: How and when to introduce pre-, pro-, syn-, or postbiotics? Am. J. Physiol. Liver Physiol. 2022, 323, G523–G553. [Google Scholar] [CrossRef]
  17. Zhou, J.; Li, M.; Chen, Q.; Li, X.; Chen, L.; Dong, Z.; Zhu, W.; Yang, Y.; Liu, Z.; Chen, Q. Programmable probiotics modulate inflammation and gut microbiota for inflammatory bowel disease treatment after effective oral delivery. Nat. Commun. 2022, 13, 3432. [Google Scholar] [CrossRef]
  18. Iyer, N.; Williams, M.A.; O’callaghan, A.A.; Dempsey, E.; Cabrera-Rubio, R.; Raverdeau, M.; Crispie, F.; Cotter, P.D.; Corr, S.C. Lactobacillus salivarius UCC118™ Dampens Inflammation and Promotes Microbiota Recovery to Provide Therapeutic Benefit in a DSS-Induced Colitis Model. Microorganisms 2022, 10, 1383. [Google Scholar] [CrossRef]
  19. Gowen, R.; Gamal, A.; Di Martino, L.; McCormick, T.S.; Ghannoum, M.A. Modulating the Microbiome for Crohn’s Disease Treatment. Gastroenterology 2023, 164, 828–840. [Google Scholar] [CrossRef]
  20. Filidou, E.; Kolios, G. Probiotics in Intestinal Mucosal Healing: A New Therapy or an Old Friend? Pharmaceuticals 2021, 14, 1181. [Google Scholar] [CrossRef]
  21. Tarapatzi, G.; Filidou, E.; Kandilogiannakis, L.; Spathakis, M.; Gaitanidou, M.; Arvanitidis, K.; Drygiannakis, I.; Valatas, V.; Kotzampassi, K.; Manolopoulos, V.G.; et al. The Probiotic Strains Bifidοbacterium lactis, Lactobacillus acidophilus, Lactiplantibacillus plantarum and Saccharomyces boulardii Regulate Wound Healing and Chemokine Responses in Human Intestinal Subepithelial Myofibroblasts. Pharmaceuticals 2022, 15, 1293. [Google Scholar] [CrossRef] [PubMed]
  22. Saber, S.; El-Fattah, E.E.A.; Yahya, G.; Gobba, N.A.; Maghmomeh, A.O.; Khodir, A.E.; Mourad, A.A.E.; Saad, A.S.; Mohammed, H.G.; Nouh, N.A.; et al. A Novel Combination Therapy Using Rosuvastatin and Lactobacillus Combats Dextran Sodium Sulfate-Induced Colitis in High-Fat Diet-Fed Rats by Targeting the TXNIP/NLRP3 Interaction and Influencing Gut Microbiome Composition. Pharmaceuticals 2021, 14, 341. [Google Scholar] [CrossRef] [PubMed]
  23. Maheshwari, R.A.; Balaraman, R.; Sailor, G.U.; Sen, D.B. Protective effect of simvastatin and rosuvastatin on trinitrobenzene sulfonic acid-induced colitis in rats. Indian J. Pharmacol. 2015, 47, 17–21. [Google Scholar] [CrossRef] [PubMed]
  24. Shin, S.K.; Cho, J.H.; Kim, E.J.; Kim, E.-K.; Park, D.K.; Kwon, K.A.; Chung, J.-W.; Kim, K.O.; Kim, Y.J. Anti-inflammatory and anti-apoptotic effects of rosuvastatin by regulation of oxidative stress in a dextran sulfate sodium-induced colitis model. World J. Gastroenterol. 2017, 23, 4559–4568. [Google Scholar] [CrossRef] [PubMed]
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

Filidou, E.; Kolios, G. Special Issue “Gut Microbiota, Inflammatory Bowel Diseases, and Therapeutic Targets”. Pharmaceuticals 2023, 16, 714. https://doi.org/10.3390/ph16050714

AMA Style

Filidou E, Kolios G. Special Issue “Gut Microbiota, Inflammatory Bowel Diseases, and Therapeutic Targets”. Pharmaceuticals. 2023; 16(5):714. https://doi.org/10.3390/ph16050714

Chicago/Turabian Style

Filidou, Eirini, and George Kolios. 2023. "Special Issue “Gut Microbiota, Inflammatory Bowel Diseases, and Therapeutic Targets”" Pharmaceuticals 16, no. 5: 714. https://doi.org/10.3390/ph16050714

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