ijms-logo

Journal Browser

Journal Browser

Molecular Mechanism in Epithelial-Mesenchymal Transition (EMT) and Fibrosis: 2nd Edition

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Biology".

Deadline for manuscript submissions: 20 December 2024 | Viewed by 3240

Special Issue Editors


E-Mail Website
Guest Editor
Department of Basic Medical Sciences, Neurosciences and Sensory Organs (SMBNOS), Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, Bari, Italy
Interests: autoimmunity; Sjogren’s syndrome; inflammation; salivary gland dysfunction; epithelial–mesenchymal transition; fibrosis
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari Medical School, Bari, Italy
Interests: pathophysiology and molecular immunology applied to immunological research lines; molecular processes underlying the interaction between receptors of the immune response, inflammation and characterization of new anti-inflammatory molecules; novel therapies for Sjögren’s syndrome autoimmune disease; evaluation of the molecular mechanisms linking chronic inflammation to fibrosis in Sjögren’s syndrome and others autoimmune diseases
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The process known as epithelial–mesenchymal transition (EMT), essential for accurate development during embryogenesis, is involved in many pathological processes, such as degenerative fibrosis and cancer. EMT is a biological process that allows epithelial cells to assume a mesenchymal phenotype with enhanced migratory capacity, invasiveness, elevated resistance to apoptosis, stem-like features, and the increased production of ECM components.

During EMT, distinct molecular processes are activated: the loss of junctions and apical–basal polarity by epithelial cells, the activation of transcription factors, the downregulation of epithelial cell-surface proteins and upregulation of mesenchymal markers, and the reorganization and expression of cytoskeletal proteins.

EMT is linked to wound healing, tissue regeneration, and organ fibrosis. During the course of fibrosis, EMT can lead to organ failure following the persistent release of a variety of inflammatory signals.

EMT is now considered to be a converging point among inflammation, fibrotic diseases, cancer, and pathologies characterized by chronic inflammation, such as autoimmune diseases. However, despite intense investigation in recent years, relatively little is known about the mechanisms of fibrosis pathogenesis and how all of these events are integrated and participate in the same process, and how the mesenchymal state is maintained. Deep knowledge of these aspects will help to exploit the plasticity of this process to reverse the metastatic phenotype of many cancers and design potential therapeutic approaches.

Papers related to any aspect of EMT and fibrosis will be considered for publication within this Special Issue.

Dr. Sabrina Lisi
Dr. Margherita Sisto
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • inflammation
  • cancer
  • EMT
  • autoimmunity
  • fibrosis
  • salivary glands

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Published Papers (3 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

13 pages, 2606 KiB  
Article
Assessment of Imatinib Anti-Remodeling Activity on a Human Precision Cut Lung Slices Model
by Sara Bozzini, Eleonora Bozza, Cecilia Bagnera, Patrizia Morbini, Sara Lettieri, Matteo Della Zoppa, Giulio Melloni, Laura Saracino, Mirko Belliato and Federica Meloni
Int. J. Mol. Sci. 2024, 25(15), 8186; https://doi.org/10.3390/ijms25158186 - 26 Jul 2024
Viewed by 374
Abstract
Recent studies have emphasized the critical role of alteration in cellular plasticity in the development of fibrotic disorders, particularly pulmonary fibrosis, prompting further investigation into molecular mechanisms and therapeutic approaches. In this context, Precision Cut Lung Slices (PCLSs) emerge as a valuable ex [...] Read more.
Recent studies have emphasized the critical role of alteration in cellular plasticity in the development of fibrotic disorders, particularly pulmonary fibrosis, prompting further investigation into molecular mechanisms and therapeutic approaches. In this context, Precision Cut Lung Slices (PCLSs) emerge as a valuable ex vivo research tool. The process of PCLSs generation preserves most features of the naïve lung tissue, such as its architecture and complex cellular composition. We previously stimulated normal lung PCLSs with two different stimuli (fibrotic cocktail, composed by platelet lysate and TGFβ, or neutrophil extracellular traps) and we observed a significant elevation of Epithelial–Mesenchymal Transition (EMT) markers from 24 h to 72 h of culture. The aim of our work was to exploit this PCLSs based ex vivo model of EMT, to evaluate the effect of imatinib, an old tyrosine kinase inhibitor with reported anti-remodeling activities in vitro and in animal models. Imatinib treatment significantly decreased α-SMA and collagen expression already starting from 24 h on stimulated PCLS. Imatinib showed a significant toxicity on unstimulated cells (3-fold increase in ACTA2 expression levels at 24 h, 1.5-fold increase in COL1A1 expression levels at 24 h, 2-fold increase in COL3A1 expression levels at 72 h). Further evaluations on specific cell lines pointed out that drug effects were mainly directed towards A549 and LFs. In conclusion, our model confirms the anti-remodeling activity of imatinib but suggests that its direct delivery to alveolar epithelial cells as recently attempted by inhalatory preparation of the drug might be associated with a non-negligible epithelial cell toxicity. Full article
Show Figures

Figure 1

12 pages, 4390 KiB  
Article
Semaglutide May Ameliorate Fibrosis and Inhibit Epithelial–Mesenchymal Transition in Intrauterine Adhesion Models
by Luming Wu, Yue Zhan and Yiqing Wang
Int. J. Mol. Sci. 2024, 25(11), 6196; https://doi.org/10.3390/ijms25116196 - 4 Jun 2024
Viewed by 839
Abstract
The purpose of this study was to explore the effect of Semaglutide on intrauterine adhesions and discover new drugs for such adhesions. In this study, the cell model was simulated by TGF-β1-induced human endometrial epithelial cells, and the animal model was established through [...] Read more.
The purpose of this study was to explore the effect of Semaglutide on intrauterine adhesions and discover new drugs for such adhesions. In this study, the cell model was simulated by TGF-β1-induced human endometrial epithelial cells, and the animal model was established through mechanical curettage and inflammatory stimulation. After co-culturing with TGF-β1 with or without different concentrations of Semaglutide for 48 h, cells were collected for RT-qPCR and Western blotting analyses. Three doses were subcutaneously injected into experimental mice once a day for two weeks, while the control group received sterile ddH2O. The serum and uterine tissues of the mice were collected. HE and Masson staining were used for the uterine histomorphological and pathological analyses. RT-qPCR and Western blotting were used for mRNA and protein expression analyses. Serum indicators were detected using ELISA kits. The results showed that Semaglutide significantly reduced the mRNA levels of fibrosis indicators ACTA2, COL1A1, and FN and inflammatory indicators TNF-α, IL-6, and NF-κB in the two models. Semaglutide improved endometrium morphology, increased the number of endometrial glands, and reduced collagen deposition in IUA mice. The results also showed that Semaglutide could inhibit vimentin, E-Cadherin, and N-Cadherin in the two models. In summary, Semaglutide can ameliorate fibrosis and inflammation of intrauterine adhesions as well as inhibit epithelial–mesenchymal transition in IUA models. Full article
Show Figures

Figure 1

Review

Jump to: Research

13 pages, 2239 KiB  
Review
Exploring Salivary Epithelial Dysfunction in Sjögren’s Disease
by Braxton Noll, Micaela Beckman, Farah Bahrani Mougeot and Jean-Luc Mougeot
Int. J. Mol. Sci. 2024, 25(9), 4973; https://doi.org/10.3390/ijms25094973 - 2 May 2024
Cited by 1 | Viewed by 885
Abstract
Sjögren’s Disease (SjD) is an autoimmune disease of the exocrine tissues. Etiological events result in the loss of epithelial homeostasis alongside extracellular matrix (ECM) destruction within the salivary and lacrimal glands, followed by immune cell infiltration. In this review, we have assessed the [...] Read more.
Sjögren’s Disease (SjD) is an autoimmune disease of the exocrine tissues. Etiological events result in the loss of epithelial homeostasis alongside extracellular matrix (ECM) destruction within the salivary and lacrimal glands, followed by immune cell infiltration. In this review, we have assessed the current understanding of epithelial–mesenchymal transition (EMT)-associated changes within the salivary epithelium potentially involved in salivary dysfunction and SjD pathogenesis. We performed a PubMed literature review pertaining to the determination of pathogenic events that lead to EMT-related epithelial dysfunction and signaling in SjD. Molecular patterns of epithelial dysfunction in SjD salivary glands share commonalities with EMT mediating wound healing. Pathological changes altering salivary gland integrity and function may precede direct immune involvement while perpetuating MMP9-mediated ECM destruction, inflammatory mediator expression, and eventual immune cell infiltration. Dysregulation of EMT-associated factors is present in the salivary epithelium of SjD and may be significant in initiating and perpetuating the disease. In this review, we further highlight the gap regarding mechanisms that drive epithelial dysfunction in salivary glands in the early or subclinical pre-lymphocytic infiltration stages of SjD. Full article
Show Figures

Figure 1

Back to TopTop