Next Article in Journal
Genotype Combinations Drive Variability in the Microbiome Configuration of the Rhizosphere of Maize/Bean Intercropping System
Next Article in Special Issue
Mucosal Genes Encoding Clock, Inflammation and Their Mutual Regulators Are Disrupted in Pediatric Patients with Active Ulcerative Colitis
Previous Article in Journal
Buffering Mitigates Chondrocyte Oxidative Stress, Metabolic Dysfunction, and Death Induced by Normal Saline: Formulation of a Novel Arthroscopic Irrigant
Previous Article in Special Issue
Pro-Inflammatory Cytokines Enhanced In Vitro Cytotoxic Activity of Clostridioides difficile Toxin B in Enteric Glial Cells: The Achilles Heel of Clostridioides difficile Infection?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue: “Inflammatory Signaling Pathways Involved in Gastrointestinal Diseases”

by
Marianna Lauricella
* and
Diana Di Liberto
*
Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(2), 1287; https://doi.org/10.3390/ijms25021287
Submission received: 16 January 2024 / Accepted: 17 January 2024 / Published: 20 January 2024
(This article belongs to the Special Issue Inflammatory Signaling Pathways Involved in Gastrointestinal Diseases)
Inflammation is a defensive response of the innate and adaptive immune systems against injury and/or harmful microorganisms to restore homeostasis. The activation of inflammatory signaling pathways has been recognized in several gastrointestinal diseases, and signaling molecules that are implicated in these pathways are thought to be promising targets for new therapeutic strategies.
In the light of these considerations, we have organized this Special Issue, titled “Inflammatory Signaling Pathways Involved in Gastrointestinal Diseases”, containing six papers (two research articles and four reviews).
Chronic inflammation of the gastrointestinal tract (GT) is a typical feature of Inflammatory Bowel Disease (IBD), a chronic idiopathic disease including Ulcerative Colitis (UC) and Crohn’s disease (CD) [1]. IBD’s etiopathogenesis involves many different factors, such as genetic predisposition, epigenetic variations, environmental factors, alterations in the intestinal microbiota, deregulation of the immune response, and chronic inflammation [2,3,4]. It is now well known that persistent oxidative stress and inflammatory responses leading to dysbiosis of the gut microbiota are associated with the onset and the progression of IBD [5]. Oxidative stress and gut dysbiosis, consisting of a decreased gut microbial diversity [6] with a higher presence of bacterial pathogenic species than beneficial species, are often induced by external stimuli, including a high-fat diet, smoking, disturbed circadian rhythms, and drug interventions. Over time, this leads to intestinal epithelium damage and increased intestinal permeability [7], with a consequent release into the blood of toxins, proinflammatory chemokines, and cytokines, such as tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), IL-6, and interferon-γ (IFN-γ) [5], as well as the activation of proinflammatory enzymes (e.g., iNOS, COX-2, and NOX) [8] and Mitogen-Activated Protein Kinases (MAPKs) signaling pathways [9]. As a result, a further increase in ROS levels in the GT [10] and a remodeling of the gut microbiota occur in a vicious cycle, in which dysbiosis and inflammation support each other, eventually causing a worsening of the inflammatory process that characterizes IBD patients [11].
Given the key role that is exerted by inflammation in the pathogenesis of IBD, the identification of new markers that are related to it represents a crucial aim in IBD research. In this scenario, the study by Scalia et al. [12] suggests a correlation between heat shock proteins (HSPs) and IBDs. HSPs are proteins that are expressed by the cells in stress conditions, whose genetic or acquired dysregulation has been connected to several pathological conditions called chaperonopathies [13,14]. The review highlighted that genetic and epigenetic deregulation of HSPs have been reported in IBD. However, the studies are controversial, because some of these deregulations may act as factors of susceptibility to the development of IBD, while others may act as protective factors assisting in the maintenance of the healthy/not inflamed intestinal tissue. Understanding these mechanisms is of fundamental importance to developing increasingly targeted therapies that consider molecular chaperones.
Another contribution, provided by Dvornikova et al. [15], outlines the main functions of histamine and its receptors (HRs) in the healthy and inflamed gut. Histamine sustains normal gut functioning and mediates pathogenic conditions by directly or indirectly modulating immune responses. The role of histamine and its signaling pathways in the inflammatory response makes it one of the main actors in the pathogenesis of IBD. In particular, the stimulation of H1R and H4R seems to be associated with proinflammatory effects, while H2R seems to mediate an anti-inflammatory effect in active IBD disease. However, the actual molecular mechanisms that underlie histamine/HRs-associated signaling pathways and are responsible for inducing inflammation in IBD are still unclear [16,17]. The authors also evaluated the experimental data from the literature from recent years, presenting a schematic overview of the main potential molecular signaling pathways that are associated with histamine receptors in IBD. The aim is that future research in this field could lead to the development of novel therapeutic approaches for IBD treatment based on agonists/antagonists of HRs or components of their signaling pathways.
The main goal of clinical treatment of IBD is to minimize the inflammatory response and heal the intestinal mucosal ulcers that characterize affected patients [18]. Currently, IBD treatment consists of immunosuppressants, aminosalicylic acid drugs, antibiotics, and monoclonal antibodies, such as anti-TNFα, with the aim of turning off the inflammation in the acute phase of the disease and/or extend remission periods [19]. However, long-term use of these drugs can lead to serious adverse reactions, poor clinical efficacy, and high recurrence rates and costs [20]. Collectively, novel therapeutic strategies for IBD treatment that are able to reduce oxidative stress and inflammation are urgently required.
Polyphenols are secondary metabolites of plants that are largely present in vegetables, fruits, nuts, and beverages such as coffee, tea, wine, and beer [21,22]. Due to their chemical structure, which is composed of multiple phenolic units, they can efficiently scavenge ROS, thus exerting an important antioxidant and anti-inflammatory activity both in vitro and in vivo [23,24,25,26]. A protective role for polyphenols against inflammation is making their use desirable in the treatment of IBDs [27]. The study by Pratelli and colleagues [28] highlights how polyphenols can restore gut dysbiosis and intestinal epithelium damage and reduce inflammation in IBD patients. Molecular mechanisms that are modulated by dietary polyphenols in IBD include the inhibition of NF-κB and the activation of Nrf2 transcriptional activity [29,30]. Furthermore, Pratelli et al. suggest that an “omics” approach could be useful for evaluating the individual response to nutrients and polyphenols that are introduced with diet, leading to a so-called “precision nutrition” research initiative that is associated with IBD management and clinical endpoint improvement.
A new therapeutic strategy for IBD was also investigated by Scalavino and colleagues [31] in their article, where they analyzed the effect of miR-369-3p administration in IBD mice models. The authors demonstrated that miR-369-3p can reduce the expression of the immunoproteasome PSMB9 subunit and consequently modulate its catalytic activity, which is frequently increased in macrophages following their activation during immune responses to inflammatory and stress stimuli [32,33]. The immunoproteasome is a multi-catalytic protein complex that is expressed in all hematopoietic cells. An increased expression of immuno-subunits, followed by increased proteasome activities in activated macrophages, is frequently found in IBD patients, demonstrating its potential role in IBD’s pathogenesis [34,35,36]. Therefore, the identification of molecules that are able to inhibit the activities of this complex is widely attempted, as it is a future alternative therapeutic approach for the treatment of inflammatory diseases, such as IBD [37,38,39].
Disorders involving gut–brain interactions (DGBIs), formerly called functional gastrointestinal disorders (FGIDs), include irritable bowel syndrome (IBS) and functional dyspepsia (FD). The pathophysiology of DGBIs is complex and includes a combination of motility disturbance, visceral hypersensitivity, altered mucosal and immune function, altered gut microbiota, and altered central nervous system processing. Immune system deregulation and inflammation and a compromised barrier function have been correlated with DGBIs and represent risk factors for the development of IBD [40,41,42]. Due to the complex etiology of the problem, the available treatment methods do not completely cure the disease. The study by Szadkowska et al. [43] discusses the beneficial effect of Cirsium palustre on gut disorders. Cirsium palustre is an herbaceous plant belonging to the Asteraceae family, which displays anti-inflammatory and antimicrobial effects due to the presence of flavonoids. The authors demonstrated that selected preparations and flavonoids from C. palustre have a pronounced motility-regulating effect on the swine colon, as well as proven beneficial antioxidant and antibacterial effects in this ex vivo system, suggesting the need for further investigations into their use in disorders involving DGBI patients’ treatment [44,45,46].
Long-standing intestinal inflammation in the GT, leading to the disruption of the normal intestinal structure and to the dysregulation of the intestinal mucosal immune system [47], can also predispose to cancer.
Barrett’s esophagus (BE) is a premalignant lesion involving the development of intestinal metaplasia in the distal esophagus. This represents an adaptational mechanism in the distal esophagus, which is caused predominantly by a long-term exposure to bile reflux. Some recent investigations by Maslenkina et al. [48] described the role of NOTCH, hedgehog (Hh), NF-κB, and IL6/STAT3 signaling in the pathogenesis of Barrett’s esophagus (BE). The study also focused on catastrophic genetic events due to impaired DNA reparation [49,50,51], the rapid activation and amplification of oncogenes such as MYC and MDM2 oncogenes, and the inactivation of tumor suppressor genes [52] following repeated reflux exposure and leading to the formation of dysplasia and esophageal adenocarcinoma (EAC) [53,54,55,56]. Their data provide evidence that genomic catastrophe is important in malignant transformations in BE, thus serving as an alternative mechanism of carcinogenesis and a “point of no return” after which malignant transformation becomes inevitable [57].
We would like to share our gratitude to all the authors who submitted their outstanding research to this Special Issue. Their manuscripts highlighted some of the molecular mechanisms underlying inflammation that characterize different gastrointestinal diseases and compounds with potential antioxidant action for application as adjuvant molecules in the treatment of these diseases.

Author Contributions

Conceptualization, M.L. and D.D.L.; writing—original draft preparation, M.L and D.D.L.;—review and editing M.L. and D.D.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ng, S.C.; Shi, H.Y.; Hamidi, N.; Underwood, F.E.; Tang, W.; Benchimol, E.I.; Panaccione, R.; Ghosh, S.; Wu, J.C.Y.; Chan, F.K.L.; et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: A systematic review of population-based studies. Lancet 2017, 390, 2769–2778. [Google Scholar] [CrossRef] [PubMed]
  2. Kobayashi, T.; Siegmund, B.; Le Berre, C.; Wei, S.C.; Ferrante, M.; Shen, B.; Bernstein, C.N.; Danese, S.; Peyrin-Biroulet, L.; Hibi, T. Ulcerative colitis. Nat. Rev. Dis. Primers 2020, 6, 74. [Google Scholar] [CrossRef] [PubMed]
  3. Petronis, A.; Petroniene, R. Epigenetics of inflammatory bowel disease. Gut 2000, 47, 302–306. [Google Scholar] [CrossRef] [PubMed]
  4. Ji, Y.; Yang, Y.; Sun, S.; Dai, Z.; Ren, F.; Wu, Z. Insights into diet-associated oxidative pathomechanisms in inflammatory bowel disease and protective effects of functional amino acids. Nutr. Rev. 2022, 81, 95–113. [Google Scholar] [CrossRef] [PubMed]
  5. Weiss, G.A.; Hennet, T. Mechanisms and consequences of intestinal dysbiosis. Cell. Mol. Life Sci. 2017, 74, 2959–2977. [Google Scholar] [CrossRef] [PubMed]
  6. Li, H.; Christman, L.M.; Li, R.; Gu, L. Synergic interactions between polyphenols and gut microbiota in mitigating inflammatory bowel diseases. Food Funct. 2020, 11, 4878–4891. [Google Scholar] [CrossRef] [PubMed]
  7. Camilleri, M. Leaky gut: Mechanisms, measurement and clinical implications in humans. Gut 2019, 68, 1516–1526. [Google Scholar] [CrossRef]
  8. Chervy, M.; Sivignon, A.; Dambrine, F.; Buisson, A.; Sauvanet, P.; Godfraind, C.; Allez, M.; Le Bourhis, L.; The Remind, G.; Barnich, N.; et al. Epigenetic master regulators HDAC1 and HDAC5 control pathobiont Enterobacteria colonization in ileal mucosa of Crohn’s disease patients. Gut Microbes 2022, 14, 2127444. [Google Scholar] [CrossRef]
  9. Yao, L.; Gu, Y.; Jiang, T.; Che, H. Inhibition effect of PPAR-gamma signaling on mast cell-mediated allergic inflammation through down-regulation of PAK1/ NF-kappaB activation. Int. Immunopharmacol. 2022, 108, 108692. [Google Scholar] [CrossRef]
  10. Wang, L.; Hu, Y.; Song, B.; Xiong, Y.; Wang, J.; Chen, D. Targeting JAK/STAT signaling pathways in treatment of inflammatory bowel disease. Inflamm. Res. 2021, 70, 753–764. [Google Scholar] [CrossRef]
  11. Nguepi Tsopmejio, I.S.; Yuan, J.; Diao, Z.; Fan, W.; Wei, J.; Zhao, C.; Li, Y.; Song, H. Auricularia polytricha and Flammulina velutipes reduce liver injury in DSS-induced Inflammatory Bowel Disease by improving inflammation, oxidative stress, and apoptosis through the regulation of TLR4/NF-kappaB signaling pathways. J. Nutr. Biochem. 2023, 111, 109190. [Google Scholar] [CrossRef] [PubMed]
  12. Scalia, F.; Carini, F.; David, S.; Giammanco, M.; Mazzola, M.; Rappa, F.; Bressan, N.I.; Maida, G.; Tomasello, G. Inflammatory Bowel Diseases: An Updated Overview on the Heat Shock Protein Involvement. Int. J. Mol. Sci. 2023, 24, 12129. [Google Scholar] [CrossRef]
  13. Macario, A.J.L.; Conway de Macario, E.; Cappello, F. The Chaperonopathies: Diseases with Defective Molecular Chaperones; Springer: Berlin/Heidelberg, Germany, 2013. [Google Scholar]
  14. Scalia, F.; Vitale, A.M.; Santonocito, R.; Conway de Macario, E.; Macario, A.J.L.; Cappello, F. The Neurochaperonopathies: Anomalies of the Chaperone System with Pathogenic Effects in Neurodegenerative and Neuromuscular Disorders. Appl. Sci. 2021, 11, 898. [Google Scholar] [CrossRef]
  15. Dvornikova, K.A.; Platonova, O.N.; Bystrova, E.Y. Inflammatory Bowel Disease: Crosstalk between Histamine, Immunity, and Disease. Int. J. Mol. Sci. 2023, 24, 9937. [Google Scholar] [CrossRef] [PubMed]
  16. Smolinska, S.; Groeger, D.; Perez, N.R.; Schiavi, E.; Ferstl, R.; Frei, R.; Konieczna, P.; Akdis, C.A.; Jutel, M.; O’Mahony, L. Histamine Receptor 2 Is Required to Suppress Innate Immune Responses to Bacterial Ligands in Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2016, 22, 1575–1586. [Google Scholar] [CrossRef]
  17. Schirmer, B.; Neumann, D. The Function of the Histamine H4 Receptor in Inflammatory and Inflammation-Associated Diseases of the Gut. Int. J. Mol. Sci. 2021, 22, 6116. [Google Scholar] [CrossRef] [PubMed]
  18. Elhag, D.A.; Kumar, M.; Saadaoui, M.; Akobeng, A.K.; Al-Mudahka, F.; Elawad, M.; Al Khodor, S. Inflammatory Bowel Disease Treatments and Predictive Biomarkers of Therapeutic Response. Int. J. Mol. Sci. 2022, 23, 6966. [Google Scholar] [CrossRef] [PubMed]
  19. Cai, Z.; Wang, S.; Li, J. Treatment of Inflammatory Bowel Disease: A Comprehensive Review. Front. Med. 2021, 8, 765474. [Google Scholar] [CrossRef]
  20. Park, K.T.; Bass, D. Inflammatory bowel disease-attributable costs and cost-effective strategies in the United States: A review. Inflamm. Bowel Dis. 2011, 17, 1603–1609. [Google Scholar] [CrossRef]
  21. Tsao, R. Chemistry and biochemistry of dietary polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef]
  22. Rudrapal, M.; Khairnar, S.J.; Khan, J.; Dukhyil, A.B.; Ansari, M.A.; Alomary, M.N.; Alshabrmi, F.M.; Palai, S.; Deb, P.K.; Devi, R. Dietary Polyphenols and Their Role in Oxidative Stress-Induced Human Diseases: Insights into Protective Effects, Antioxidant Potentials and Mechanism(s) of Action. Front. Pharmacol. 2022, 13, 806470. [Google Scholar] [CrossRef] [PubMed]
  23. Di Liberto, D.; Iacuzzi, N.; Pratelli, G.; Porrello, A.; Maggio, A.; La Bella, S.; De Blasio, A.; Notaro, A.; D’Anneo, A.; Emanuele, S.; et al. Cytotoxic Effect Induced by Sicilian Oregano Essential Oil in Human Breast Cancer Cells. Cells 2023, 12, 2733. [Google Scholar] [CrossRef] [PubMed]
  24. Pratelli, G.; Di Liberto, D.; Carlisi, D.; Emanuele, S.; Giuliano, M.; Notaro, A.; De Blasio, A.; Calvaruso, G.; D’Anneo, A.; Lauricella, M. Hypertrophy and ER Stress Induced by Palmitate Are Counteracted by Mango Peel and Seed Extracts in 3T3-L1 Adipocytes. Int. J. Mol. Sci. 2023, 24, 5419. [Google Scholar] [CrossRef] [PubMed]
  25. Pratelli, G.; Carlisi, D.; D’Anneo, A.; Maggio, A.; Emanuele, S.; Palumbo Piccionello, A.; Giuliano, M.; De Blasio, A.; Calvaruso, G.; Lauricella, M. Bio-Waste Products of Mangifera indica L. Reduce Adipogenesis and Exert Antioxidant Effects on 3T3-L1 Cells. Antioxidants 2022, 11, 363. [Google Scholar] [CrossRef] [PubMed]
  26. Niwano, Y.; Kohzaki, H.; Shirato, M.; Shishido, S.; Nakamura, K. Metabolic Fate of Orally Ingested Proanthocyanidins through the Digestive Tract. Antioxidants 2022, 12, 17. [Google Scholar] [CrossRef]
  27. Lu, Y.; Zamora-Ros, R.; Chan, S.; Cross, A.J.; Ward, H.; Jakszyn, P.; Luben, R.; Opstelten, J.L.; Oldenburg, B.; Hallmans, G.; et al. Dietary Polyphenols in the Aetiology of Crohn’s Disease and Ulcerative Colitis-A Multicenter European Prospective Cohort Study (EPIC). Inflamm. Bowel Dis. 2017, 23, 2072–2082. [Google Scholar] [CrossRef]
  28. Pratelli, G.; Tamburini, B.; Carlisi, D.; De Blasio, A.; D’Anneo, A.; Emanuele, S.; Notaro, A.; Affranchi, F.; Giuliano, M.; Seidita, A.; et al. Foodomics-Based Approaches Shed Light on the Potential Protective Effects of Polyphenols in Inflammatory Bowel Disease. Int. J. Mol. Sci. 2023, 24, 14619. [Google Scholar] [CrossRef]
  29. Hamalainen, M.; Nieminen, R.; Vuorela, P.; Heinonen, M.; Moilanen, E. Anti-inflammatory effects of flavonoids: Genistein, kaempferol, quercetin, and daidzein inhibit STAT-1 and NF-kappaB activations, whereas flavone, isorhamnetin, naringenin, and pelargonidin inhibit only NF-kappaB activation along with their inhibitory effect on iNOS expression and NO production in activated macrophages. Mediat. Inflamm. 2007, 2007, 45673. [Google Scholar]
  30. Qin, S.; Hou, D.X. Multiple regulations of Keap1/Nrf2 system by dietary phytochemicals. Mol. Nutr. Food Res. 2016, 60, 1731–1755. [Google Scholar] [CrossRef]
  31. Scalavino, V.; Piccinno, E.; Valentini, A.M.; Mastronardi, M.; Armentano, R.; Giannelli, G.; Serino, G. A Novel Mechanism of Immunoproteasome Regulation via miR-369-3p in Intestinal Inflammatory Response. Int. J. Mol. Sci. 2022, 23, 13771. [Google Scholar] [CrossRef]
  32. Qureshi, N.; Vogel, S.N.; Van Way, C.; Papasian, C.J.; Qureshi, A.A.; Morrison, D.C. The proteasome: A central regulator of inflammation and macrophage function. Immunol. Res. 2005, 31, 243–260. [Google Scholar] [CrossRef] [PubMed]
  33. Kimura, H.; Caturegli, P.; Takahashi, M.; Suzuki, K. New Insights into the Function of the Immunoproteasome in Immune and Nonimmune Cells. J. Immunol. Res. 2015, 2015, 541984. [Google Scholar] [CrossRef] [PubMed]
  34. Fitzpatrick, L.R.; Small, J.S.; Poritz, L.S.; McKenna, K.J.; Koltun, W.A. Enhanced Intestinal Expression of the Proteasome Subunit Low Molecular Mass Polypeptide 2 in Patients with Inflammatory Bowel Disease. Dis. Colon. Rectum 2007, 50, 337–350. [Google Scholar] [CrossRef] [PubMed]
  35. Visekruna, A.; Slavova, N.; Dullat, S.; Gröne, J.; Kroesen, A.-J.; Ritz, J.-P.; Buhr, H.-J.; Steinhoff, U. Expression of catalytic proteasome subunits in the gut of patients with Crohn’s disease. Int. J. Color. Dis. 2009, 24, 1133–1139. [Google Scholar] [CrossRef] [PubMed]
  36. Visekruna, A.; Joeris, T.; Schmidt, N.; Lawrenz, M.; Ritz, J.-P.; Buhr, H.J.; Steinhoff, U. Comparative expression analysis and characterization of 20S proteasomes in human intestinal tissues. Inflamm. Bowel. Dis. 2009, 15, 526–533. [Google Scholar] [CrossRef] [PubMed]
  37. Schmidt, N.; Gonzalez, E.; Visekruna, A.; Kuhl, A.A.; Loddenkemper, C.; Mollenkopf, H.; Kaufmann, S.H.E.; Steinhoff, U.; Joeris, T. Targeting the proteasome: Partial inhibition of the proteasome by bortezomib or deletion of the immunosubunit LMP7 attenuates experimental colitis. Gut 2010, 59, 896–906. [Google Scholar] [CrossRef] [PubMed]
  38. Scalavino, V.; Liso, M.; Serino, G. Role of microRNAs in the Regulation of Dendritic Cell Generation and Function. Int. J. Mol. Sci. 2020, 21, 1319. [Google Scholar] [CrossRef]
  39. Miller, Z.; Ao, L.; Bo Kim, K.; Lee, W. Inhibitors of the Immunoproteasome: Current Status and Future Directions. Curr. Pharm. Des. 2013, 19, 4140–4151. [Google Scholar] [CrossRef]
  40. Drossman, D.A. Functional Gastrointestinal Disorders: History, Pathophysiology, Clinical Features and Rome IV. Gastroenterology 2016, 150, 1262–1279.e2. [Google Scholar] [CrossRef]
  41. Gubert, C.; Gasparotto, J.; Morais, L.H. Convergent pathways of the gut microbiota-brain axis and neurodegenerative disorders. Gastroenterol. Rep. 2022, 10, goac017. [Google Scholar] [CrossRef]
  42. Shivaji, U.N.; Ford, A.C. Prevalence of functional gastrointestinal disorders among consecutive new patient referrals to a gastroenterology clinic. Frontline Gastroenterol. 2014, 5, 266–271. [Google Scholar] [CrossRef] [PubMed]
  43. Szadkowska, D.; Chłopecka, M.; Strawa, J.V.; Jakimiuk, K.; Augustynowicz, D.; Tomczyk, M.; Mendel, M. Effects of Cirsium palustre Extracts and Their Main Flavonoids on Colon Motility—An Ex Vivo Study. Int. J. Mol. Sci. 2023, 24, 17283. [Google Scholar] [CrossRef] [PubMed]
  44. Azab, A.; Nassar, A.; Azab, A.N. Anti-Inflammatory Activity of Natural Products. Molecules 2016, 21, 1321. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, S.; Wang, R.; Li, D.; Zhao, L.; Zhu, L. Role of gut microbiota in functional constipation. Gastroenterol. Rep. 2021, 9, 392–401. [Google Scholar] [CrossRef] [PubMed]
  46. Pusceddu, M.M.; Gareau, M.G. Visceral pain: Gut microbiota, a new hope? J. Biomed. Sci. 2018, 25, 73. [Google Scholar] [CrossRef] [PubMed]
  47. Hnatyszyn, A.; Hryhorowicz, S.; Kaczmarek-Rys, M.; Lis, E.; Slomski, R.; Scott, R.J.; Plawski, A. Colorectal carcinoma in the course of inflammatory bowel diseases. Hered. Cancer Clin. Pract. 2019, 17, 18. [Google Scholar] [CrossRef] [PubMed]
  48. Maslenkina, K.; Mikhaleva, L.; Naumenko, M.; Vandysheva, R.; Gushchin, M.; Atiakshin, D.; Buchwalow, I.; Tiemann, M. Signaling Pathways in the Pathogenesis of Barrett’s Esophagus and Esophageal Adenocarcinoma. Int. J. Mol. Sci. 2023, 24, 9304. [Google Scholar] [CrossRef]
  49. Contino, G.; Vaughan, T.L.; Whiteman, D.; Fitzgerald, R.C. The Evolving Genomic Landscape of Barrett’s Esophagus and Esophageal Adenocarcinoma. Gastroenterology 2017, 153, 657–673.e1. [Google Scholar] [CrossRef]
  50. Nones, K.; Waddell, N.; Wayte, N.; Patch, A.M.; Bailey, P.; Newell, F.; Holmes, O.; Fink, J.L.; Quinn, M.C.J.; Tang, Y.H.; et al. Genomic catastrophes frequently arise in esophageal adenocarcinoma and drive tumorigenesis. Nat. Commun. 2014, 5, 5224. [Google Scholar] [CrossRef]
  51. Secrier, M.; Li, X.; de Silva, N.; Eldridge, M.D.; Contino, G.; Bornschein, J.; MacRae, S.; Grehan, N.; O’Donovan, M.; Miremadi, A.; et al. Mutational signatures in esophageal adenocarcinoma define etiologically distinct subgroups with therapeutic relevance. Nat. Genet. 2016, 48, 1131–1141. [Google Scholar] [CrossRef]
  52. Stephens, P.J.; Greenman, C.D.; Fu, B.; Yang, F.; Bignell, G.R.; Mudie, L.J.; Pleasance, E.D.; Lau, K.W.; Beare, D.; Stebbings, L.A.; et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 2011, 144, 27–40. [Google Scholar] [CrossRef] [PubMed]
  53. Evans, J.A.; Carlotti, E.; Lin, M.L.; Hackett, R.J.; Haughey, M.J.; Passman, A.M.; Dunn, L.; Elia, G.; Porter, R.J.; McLean, M.H.; et al. Clonal Transitions and Phenotypic Evolution in Barrett’s Esophagus. Gastroenterology 2022, 162, 1197–1209.e13. [Google Scholar] [CrossRef] [PubMed]
  54. Stachler, M.D.; Taylor-Weiner, A.; Peng, S.; McKenna, A.; Agoston, A.T.; Odze, R.D.; Davison, J.M.; Nason, K.S.; Loda, M.; Leshchiner, I.; et al. Paired exome analysis of Barrett’s esophagus and adenocarcinoma. Nat. Genet. 2015, 47, 1047–1055. [Google Scholar] [CrossRef] [PubMed]
  55. Stachler, M.D.; Camarda, N.D.; Deitrick, C.; Kim, A.; Agoston, A.T.; Odze, R.D.; Hornick, J.L.; Nag, A.; Thorner, A.R.; Ducar, M.; et al. Detection of Mutations in Barrett’s Esophagus Before Progression to High-Grade Dysplasia or Adenocarcinoma. Gastroenterology 2018, 155, 156–167. [Google Scholar] [CrossRef]
  56. Peters, Y.; Al-Kaabi, A.; Shaheen, N.J.; Chak, A.; Blum, A.; Souza, R.F.; Di Pietro, M.; Iyer, P.G.; Pech, O.; Fitzgerald, R.C.; et al. Barrett oesophagus. Nat. Rev. Dis. Primers 2019, 5, 35. [Google Scholar] [CrossRef]
  57. Panda, A.; Shin, M.R.; Cheng, C.; Bajpai, M. Barrett’s Epithelium to Esophageal Adenocarcinoma: Is There a “Point of No Return”? Front. Genet. 2021, 12, 706706. [Google Scholar] [CrossRef]
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

Lauricella, M.; Di Liberto, D. Special Issue: “Inflammatory Signaling Pathways Involved in Gastrointestinal Diseases”. Int. J. Mol. Sci. 2024, 25, 1287. https://doi.org/10.3390/ijms25021287

AMA Style

Lauricella M, Di Liberto D. Special Issue: “Inflammatory Signaling Pathways Involved in Gastrointestinal Diseases”. International Journal of Molecular Sciences. 2024; 25(2):1287. https://doi.org/10.3390/ijms25021287

Chicago/Turabian Style

Lauricella, Marianna, and Diana Di Liberto. 2024. "Special Issue: “Inflammatory Signaling Pathways Involved in Gastrointestinal Diseases”" International Journal of Molecular Sciences 25, no. 2: 1287. https://doi.org/10.3390/ijms25021287

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