Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (118)

Search Parameters:
Keywords = partial EMT

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 10345 KB  
Article
A Patient-Derived Scaffold-Based 3D Culture Platform for Head and Neck Cancer: Preserving Tumor Heterogeneity for Personalized Drug Testing
by Alinda Anameriç, Emilia Reszczyńska, Tomasz Stankiewicz, Adrian Andrzejczak, Andrzej Stepulak and Matthias Nees
Cells 2025, 14(19), 1543; https://doi.org/10.3390/cells14191543 - 2 Oct 2025
Viewed by 216
Abstract
Head and neck cancer (HNC) is highly heterogeneous and difficult to treat, underscoring the need for rapid, patient-specific models. Standard three-dimensional (3D) cultures often lose stromal partners that influence therapy response. We developed a patient-derived system maintaining tumor cells, cancer-associated fibroblasts (CAFs), and [...] Read more.
Head and neck cancer (HNC) is highly heterogeneous and difficult to treat, underscoring the need for rapid, patient-specific models. Standard three-dimensional (3D) cultures often lose stromal partners that influence therapy response. We developed a patient-derived system maintaining tumor cells, cancer-associated fibroblasts (CAFs), and cells undergoing partial epithelial–mesenchymal transition (pEMT) for drug sensitivity testing. Biopsies from four HNC patients were enzymatically dissociated. CAFs were directly cultured, and their conditioned medium (CAF-CM) was collected. Cryopreserved primary tumor cell suspensions were later revived, screened in five different growth media under 2D conditions, and the most heterogeneous cultures were re-embedded in 3D hydrogels with varied gel mixtures, media, and seeding geometries. Tumoroid morphology was quantified using a perimeter-based complexity index. Viability after treatment with cisplatin or Notch modulators (RIN-1, recombination signal-binding protein for immunoglobulin κ J region (RBPJ) inhibitor; FLI-06, inhibitor) was assessed by live imaging and the water-soluble tetrazolium-8 (WST-8) assay. Endothelial Cell Growth Medium 2 (ECM-2) medium alone produced compact CAF-free spheroids, whereas ECM-2 supplemented with CAF-CM generated invasive aggregates that deposited endogenous matrix. Matrigel with this medium and single-point seeding gave the highest complexity scores. Two of the three patient tumoroids were cisplatin-sensitive, and all showed significant growth inhibition with the FLI-06 Notch inhibitor, while the RBPJ inhibitor RIN-1 induced minimal change. The optimized scaffold retains tumor–stroma crosstalk and provides patient-specific drug response data within days after operation, supporting personalized treatment selection in HNC. Full article
(This article belongs to the Special Issue 3D Cultures and Organ-on-a-Chip in Cell and Tissue Cultures)
Show Figures

Figure 1

13 pages, 1249 KB  
Article
Dynamics of Telomerase-Based PD-L1 Circulating Tumor Cells as a Longitudinal Biomarker for Treatment Response Prediction in Patients with Non-Small Cell Lung Cancer
by Issei Sumiyoshi, Shinsaku Togo, Takahiro Okabe, Kanae Abe, Junko Watanabe, Yusuke Ochi, Kazuaki Hoshi, Shoko Saiwaki, Shuko Nojiri, Yuichi Fujimoto, Yukiko Namba, Yoko Tabe, Yasuo Urata and Kazuhisa Takahashi
Int. J. Mol. Sci. 2025, 26(19), 9583; https://doi.org/10.3390/ijms26199583 - 1 Oct 2025
Viewed by 241
Abstract
Noninvasive liquid biopsy for monitoring circulating tumor cells offers valuable insights for predicting therapeutic responses. We developed TelomeScan® (OBP-401), based on the detection of telomerase activity as a universal cancer cell marker and an indicator of the presence of viable circulating tumor [...] Read more.
Noninvasive liquid biopsy for monitoring circulating tumor cells offers valuable insights for predicting therapeutic responses. We developed TelomeScan® (OBP-401), based on the detection of telomerase activity as a universal cancer cell marker and an indicator of the presence of viable circulating tumor cells (CTCs) for patients with advanced non-small cell lung cancer (NSCLC). This system evaluated CTC subtypes characterized by programmed death ligand 1 (PD-L1), an immune checkpoint molecule, and vimentin, an epithelial–mesenchymal transition (EMT) marker, using a multi-fluorescent color microscope reader. The prognostic value and therapeutic responses were predicted by dynamically monitoring CTC counts in 79 patients with advanced NSCLC. The sensitivity and specificity values of TelomeScan® for PD-L1(+) cells (≥1 cell) were 75% and 100%, respectively, indicating high diagnostic accuracy. PD-L1(+) and EMT(+) in CTCs were detected in 75% and 12% of patients, respectively. Detection of PD-L1(+)CTCs and PD-L1(+)EMT(+) CTCs before treatment was associated with poor prognosis (p < 0.05). Monitoring of reducing and increasing PD-L1(+) CTC counts in two sequential samples (baseline, cycle 2 treatment) correlated significantly with partial response (p = 0.032) and progressive disease (p = 0.023), respectively. Monitoring PD-L1(+)CTCs by TelomeScan® will aid in anticipating responses or resistance to frontline treatments, optimizing precision medicine choices in patients with NSCLC. Full article
Show Figures

Figure 1

24 pages, 1263 KB  
Review
Shared and Context-Specific Mechanisms of EMT and Cellular Plasticity in Cancer and Fibrotic Diseases
by Victor Alexandre F. Bastos, Aline Gomes de Souza, Virginia C. Silvestrini Guedes and Thúlio M. Cunha
Int. J. Mol. Sci. 2025, 26(19), 9476; https://doi.org/10.3390/ijms26199476 - 27 Sep 2025
Viewed by 475
Abstract
Cellular plasticity enables cells to dynamically adapt their phenotype in response to environmental cues, a process central to development, tissue repair, and disease. Among the most studied plasticity programs is epithelial–mesenchymal transition (EMT), a transcriptionally controlled process by which epithelial cells acquire mesenchymal [...] Read more.
Cellular plasticity enables cells to dynamically adapt their phenotype in response to environmental cues, a process central to development, tissue repair, and disease. Among the most studied plasticity programs is epithelial–mesenchymal transition (EMT), a transcriptionally controlled process by which epithelial cells acquire mesenchymal traits. Originally described in embryogenesis, EMT is now recognized as a key driver in both tumor progression and fibrotic remodeling. In cancer, EMT and hybrid epithelial/mesenchymal (E/M) states promote invasion, metastasis, stemness, therapy resistance, and immune evasion. In fibrotic diseases, partial EMT (pEMT) contributes to fibroblast activation and excessive extracellular matrix deposition, sustaining organ dysfunction mainly in the kidney, liver, lung, and heart. This review integrates recent findings on the molecular regulation of EMT, including signaling pathways (TGF-β, WNT, NOTCH, HIPPO), transcription factors (SNAIL, ZEB, TWIST), and regulatory layers involving microRNAs and epigenetic modifications. Moreover, we discuss the emergence of pEMT states as drivers of phenotypic plasticity, functional heterogeneity, and poor prognosis. By comparing EMT in cancer and fibrosis, we reveal shared mechanisms and disease-specific features, emphasizing the translational relevance of targeting EMT plasticity. Finally, we explore how cutting-edge technologies, such as single-cell transcriptomics and lineage tracing, are reshaping our understanding of EMT across pathological contexts. Full article
(This article belongs to the Special Issue Cellular Plasticity and EMT in Cancer and Fibrotic Diseases)
Show Figures

Figure 1

23 pages, 3067 KB  
Article
Investigating the Relationship Between Long Non-Coding RNAs and miR-200 Family Expression in Clear Cell Renal Cell Carcinoma
by Tanja Čugura, Nina Hauptman, Jera Jeruc and Emanuela Boštjančič
Cancers 2025, 17(19), 3123; https://doi.org/10.3390/cancers17193123 - 25 Sep 2025
Viewed by 267
Abstract
Objectives: MicroRNAs of the miR-200 family are recognized as key inhibitors of epithelial-to-mesenchymal transition (EMT). However, there is limited data on the potential regulation of miR-200 family expression by long non-coding RNAs (lncRNAs) in RCC. Methods: We conducted a comprehensive literature and database [...] Read more.
Objectives: MicroRNAs of the miR-200 family are recognized as key inhibitors of epithelial-to-mesenchymal transition (EMT). However, there is limited data on the potential regulation of miR-200 family expression by long non-coding RNAs (lncRNAs) in RCC. Methods: We conducted a comprehensive literature and database search to identify lncRNAs that had been already functionally validated as regulators of any member of the miR-200 family. We analyzed the expression levels of the miR-200 family and the identified lncRNAs by qPCR. The study included 42 samples of carcinoma and non-carcinoma tissue from 25 RCC patients. In addition, we used RNA sequencing data from The Cancer Genome Atlas (TCGA), encompassing 511 kidney RCC (KIRC) samples, to further analyze the expression of miRNAs and lncRNAs. Results: We identified 127 lncRNAs with confirmed regulatory functions, 31 of which were validated in our samples. The majority of lncRNAs, along with all members of the miR-200 family, showed consistent downregulation in carcinoma tissues compared to non-carcinoma tissues. We observed a significant correlation between the expression of at least one member of the miR-200 family and 17 lncRNAs. In particular, three lncRNAs (MALAT1, OIP5-AS1, and LINC00467) showed a correlation with the expression of all members of the miR-200 family. Our results were at least partially confirmed in KIRC samples from the TCGA dataset. Conclusions: Our results suggest that the expression of the miR-200 family in RCC might be at least partially influenced by lncRNAs. Based on our cohort of samples, MALAT1, OIP5-AS1, and LINC00467 appear to be potentially important contributors to RCC development. Full article
Show Figures

Graphical abstract

28 pages, 50380 KB  
Review
Changes in Epithelial Cell Polarity and Adhesion Guide Human Endometrial Receptivity: How In Vitro Systems Help to Untangle Mechanistic Details
by Irmgard Classen-Linke, Volker U. Buck, Anna K. Sternberg, Matthias Kohlen, Liubov Izmaylova and Rudolf E. Leube
Biomolecules 2025, 15(8), 1057; https://doi.org/10.3390/biom15081057 - 22 Jul 2025
Viewed by 1160
Abstract
Tissue remodeling of human endometrium occurs during the menstrual cycle to prepare for embryo adhesion and invasion. The ovarian steroid hormones 17β-estradiol and progesterone control the menstrual cycle to achieve the receptive state during the “window of implantation” (WOI). Here, we focus on [...] Read more.
Tissue remodeling of human endometrium occurs during the menstrual cycle to prepare for embryo adhesion and invasion. The ovarian steroid hormones 17β-estradiol and progesterone control the menstrual cycle to achieve the receptive state during the “window of implantation” (WOI). Here, we focus on the human endometrial epithelium and its changes in polarity, adhesion, cytoskeletal organization and the underlying extracellular matrix enabling embryo implantation. The adhesion and invasion of the trophoblast via the apical plasma membrane of epithelial cells is a unique cell biological process, which is coupled to partial epithelial–mesenchymal transition (EMT). Given the fundamental species differences during implantation, we restrict the review mainly to the human situation and focus on cell culture systems to study the interaction between human trophoblast and endometrial cells. We summarize current knowledge based on the relatively scarce in vivo data and the steadily growing in vitro observations using various cell culture systems. Full article
Show Figures

Figure 1

51 pages, 1586 KB  
Review
ECM Mechanics Control Jamming-to-Unjamming Transition of Cancer Cells
by Claudia Tanja Mierke
Cells 2025, 14(13), 943; https://doi.org/10.3390/cells14130943 - 20 Jun 2025
Viewed by 2133
Abstract
Cancer metastasis constitutes a multifactorial phenomenon that continues to confound therapeutic strategies. The biochemical signals governing motile phenotypes have been extensively characterized, but mechanobiological interactions have only recently been integrated into cancer cell motility models and remain less well elucidated. The identification of [...] Read more.
Cancer metastasis constitutes a multifactorial phenomenon that continues to confound therapeutic strategies. The biochemical signals governing motile phenotypes have been extensively characterized, but mechanobiological interactions have only recently been integrated into cancer cell motility models and remain less well elucidated. The identification of the biochemically and mechanically controlled epithelial–mesenchymal transition (EMT) of cancer cells, which occurs either completely or partially, has led to a major breakthrough and a universal phenomenon in cancers. In addition, a relatively new theory based on mechanobiological aspects called “jamming-to-unjamming transition” is being proposed to explain the transition of cancer cells to an invasive phenotype. The latter transition may help to better understand the different types of 3D migration and invasion of cancer cells. Similarly to EMT, the transition from jamming to unjamming seems to be controlled by molecular and physical factors, including cell mechanics and mechanical cues from the extracellular matrix (ECM) of the tumor microenvironment (TME). It is challenging to grasp the distinctions between the transition from jamming to unjamming and EMT, as they appear to be the same at first glance. However, upon closer examination, the two transitions are quite separate. Moreover, it is still unclear whether both transitions may act synergistically. This review highlights the most important breakthroughs in the transition from jamming to unjamming, with a focus on mechanobiology and extracellular environmental aspects, and it compares them with those of EMT. In addition, the impact of the TME, such as ECM scaffold and cancer-associated fibroblasts (CAFs) on the jamming-to-unjamming transition is discussed. Finally, the research frontiers and future directions in the field of mechanobiological research in cancer metastasis are outlined. Full article
(This article belongs to the Special Issue Role of Extracellular Matrix in Cancer and Disease)
Show Figures

Figure 1

14 pages, 1255 KB  
Review
The Relationships Among Perineural Invasion, Tumor–Nerve Interaction and Immunosuppression in Cancer
by Jozsef Dudas, Rudolf Glueckert, Maria do Carmo Greier and Benedikt Gabriel Hofauer
Onco 2025, 5(2), 25; https://doi.org/10.3390/onco5020025 - 23 May 2025
Viewed by 2645
Abstract
Tumor cells and the tumor microenvironment (TME) produce factors, including neurotrophins, that induce axonogenesis and neurogenesis, and increase local nerve density. Proliferative growing cancer cell clusters and disseminated invasive tumor cells undergoing partial epithelial-to-mesenchymal transition (pEMT) can invade peripheral nerves. In the early [...] Read more.
Tumor cells and the tumor microenvironment (TME) produce factors, including neurotrophins, that induce axonogenesis and neurogenesis, and increase local nerve density. Proliferative growing cancer cell clusters and disseminated invasive tumor cells undergoing partial epithelial-to-mesenchymal transition (pEMT) can invade peripheral nerves. In the early stages of tumor–nerve interactions, Schwann cells (SCs) dedifferentiate, become activated and migrate to cancer cell nests; later, they induce pEMT in tumor cells and activate tumor cell migration along nerves. The SC–tumor–nerve interaction attracts myeloid-derived suppressor cells (MDSCs) and inflammatory monocytes, and the latter differentiate into macrophages. SCs and MDSCs are responsible for the activation of transforming growth factor-beta (TGF-beta) signaling. Intra-tumoral innervation is followed by perineural invasion (PNI), which has an unfavorable prognosis. What are the interventional options against PNI: local reduction in tumor nerves or inhibition of TGF-beta-related events, inhibition of downstream signaling of TGF-beta or immune activation, or intervention against immunosuppression? This systematic review is based on the Prisma 2009 search method and provides an overview of tumor–nerve interaction. Full article
Show Figures

Figure 1

17 pages, 11192 KB  
Article
Mechanism of Tumor Budding in Patient-Derived Metachronous Oral Primary Squamous Cell Carcinoma Cell Lines
by Takayuki Omae, Yuji Omori, Yuna Makihara, Koji Yamanegi, Soutaro Hanawa, Kyohei Yoshikawa, Kazuma Noguchi and Hiromitsu Kishimoto
Int. J. Mol. Sci. 2025, 26(7), 3347; https://doi.org/10.3390/ijms26073347 - 3 Apr 2025
Cited by 2 | Viewed by 864
Abstract
Tumor budding (TB) occurs at the deepest site of tumor invasion and is a significant prognostic indicator of cervical metastasis in oral squamous cell carcinoma (OSCC). The mechanism of TB, however, remains unclear. This study investigated the roles of the tumor microenvironment and [...] Read more.
Tumor budding (TB) occurs at the deepest site of tumor invasion and is a significant prognostic indicator of cervical metastasis in oral squamous cell carcinoma (OSCC). The mechanism of TB, however, remains unclear. This study investigated the roles of the tumor microenvironment and partial epithelial–mesenchymal transition (p-EMT) in TB expression using molecular and cellular physiological analyses. We established oral metachronous carcinoma cell lines (gingival carcinoma: 020, tongue carcinoma with high TB expression: 020G) from two cancers with pathologically different TB in the same patient and subjected them to exome analysis to detect gene mutations related to carcinogenesis and malignancy. Differences in EMT expression induced by transforming growth factor-β (TGF-β) between 020 and 020G were analyzed by Western blotting and reverse transcription polymerase chain reaction, and TGF-β-induced changes in cell morphology, proliferation, migration, and invasive ability were also examined. TGF-β expression was observed in the deepest tumor invasion microenvironment. TGF-β also induced the expression of several p-EMT markers and increased the migration and invasive abilities of 020G compared with 020 cells. In conclusion, TGF-β in the deep-tumor microenvironment can induce p-EMT in tumor cells, expressed as TB. Full article
Show Figures

Figure 1

16 pages, 3429 KB  
Article
PDLIM3 Regulates Migration and Invasion of Head and Neck Squamous Cell Carcinoma via YAP–Mediated Epithelial–Mesenchymal Transition
by Fan Yang, Ying Zhou, You Zhang, Weideng Wei, Fei Huang, Dan Yang, Yixin Zhang, Ruiyang Zhang, Xiaoqiang Xia, Qianming Chen, Yuchen Jiang and Xiaodong Feng
Int. J. Mol. Sci. 2025, 26(7), 3147; https://doi.org/10.3390/ijms26073147 - 28 Mar 2025
Cited by 1 | Viewed by 812
Abstract
Despite significant progress in characterizing the omics landscape of head and neck squamous cell carcinoma (HNSCC), the development of precision therapies remains limited. One key factor contributing to this challenge is the marked molecular heterogeneity of HNSCC. Further investigation of molecular profiles within [...] Read more.
Despite significant progress in characterizing the omics landscape of head and neck squamous cell carcinoma (HNSCC), the development of precision therapies remains limited. One key factor contributing to this challenge is the marked molecular heterogeneity of HNSCC. Further investigation of molecular profiles within HNSCC may facilitate the improvement in more effective precision treatments. Here, we focus on the dysregulation of PDZ and LIM domain protein 3 (PDLIM3) in HNSCC. The expression levels of PDLIM3 were analyzed using public datasets to assess its potential role in tumor progression. We found that PDLIM3 was downregulated in pan–cancer and HNSCC. The prognostic significance of PDLIM3 was evaluated through tissue microarray, and the downregulation of PDLIM3 was correlated with poor HNSCC prognosis. Investigating the implications of PDLIM3 for tumor metastatic ability in vitro, we found that PDLIM3 suppressed the migration and invasion of HNSCC, accompanied by partially impeding the process of epithelial–mesenchymal transition (EMT). Furthermore, PDLIM3 inhibited the transcriptional activity of Yes–associated protein (YAP), suggesting that YAP may be involved in the PDLIM3–mediated suppression of HNSCC metastatic ability. Our findings identify a potential signaling axis wherein PDLIM3 regulates YAP–EMT, thereby influencing tumor metastatic ability, and suggest the potential role of PDLIM3 as a tumor suppressor and prognostic biomarker for HNSCC. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

14 pages, 4134 KB  
Article
Patient-Derived Cancer-Associated Fibroblasts Support the Colonization of Tumor Cells in Head and Neck Squamous Cell Carcinoma
by Julia Federspiel, Teresa Bernadette Steinbichler, Samuel Moritz Vorbach, Marie Theres Eling, Wegene Borena, Christof Seifarth, Benedikt Gabriel Hofauer and Jozsef Dudas
Biomedicines 2025, 13(2), 358; https://doi.org/10.3390/biomedicines13020358 - 4 Feb 2025
Cited by 1 | Viewed by 1822
Abstract
Background: The crosstalk between cancer-associated fibroblasts (CAFs) and tumor cells promotes proliferation, tumor relapse, and the acquisition of a partial epithelial-to-mesenchymal (pEMT) phenotype in tumor cells. The aim of this study was to investigate the effects of patient-derived CAFs on tumor cell growth [...] Read more.
Background: The crosstalk between cancer-associated fibroblasts (CAFs) and tumor cells promotes proliferation, tumor relapse, and the acquisition of a partial epithelial-to-mesenchymal (pEMT) phenotype in tumor cells. The aim of this study was to investigate the effects of patient-derived CAFs on tumor cell growth and radioresistance in head and neck squamous cell carcinoma (HNSCC). Methods: CAFs were isolated and cultured in a three-dimensional spheroid formation. SCC-25 tumor cells educated by the CAFs (SCC25-E cells) were subjected to irradiation, and the response of the CAF-stimulated tumor cells to radiotherapy was determined using an MTT assay, a clonogenic assay, and Western blotting. Tumor cell morphological changes and growth dynamics were assessed using 3D holotomographic microscopy and a live video microscope. Results: Patient-derived CAFs significantly increased the growth rate of SCC-25 cells. CAFs drove fibrosis in the tumor microenvironment (TME), functioned as a physical barrier, temporarily stopped tumor growth, and induced the p38 mitogen-activated protein kinase (MAPK) signaling pathway. Viability after irradiation at 4–8 Gy was significantly higher in SCC25-E cells than in the controls (p = 8 × 10–4 or lower). Furthermore, irradiation triggered the pEMT profile in HNSCC cells. Conclusions: CAFs’ education of tumor cells and the induced p38 phosphorylation had no influence on irradiation sensitivity. SCC25-E cultures demonstrated increased tumor cell growth, viability, and stress-induced phospho-p38 activation. Full article
(This article belongs to the Special Issue Head and Neck Tumors, 4th Edition)
Show Figures

Figure 1

11 pages, 6125 KB  
Communication
Localized Effects in Graphene Oxide Systems: A Pathway to Hyperbolic Metamaterials
by Grazia Giuseppina Politano
Photonics 2025, 12(2), 121; https://doi.org/10.3390/photonics12020121 - 29 Jan 2025
Cited by 2 | Viewed by 1108
Abstract
Graphene oxide (GO) has emerged as a carbon-based nanomaterial providing a different pathway to graphene. One of its most notable features is the ability to partially reduce it, resulting in graphene-like sheets through the elimination of oxygen-including functional groups. In this paper, the [...] Read more.
Graphene oxide (GO) has emerged as a carbon-based nanomaterial providing a different pathway to graphene. One of its most notable features is the ability to partially reduce it, resulting in graphene-like sheets through the elimination of oxygen-including functional groups. In this paper, the effect of localized interactions in an Ag/GO/Au multilayer system was studied to explore its potential for photonic applications. GO was dip-coated onto magnetron-sputtered silver, followed by the deposition of a thin gold film to form an Ag/GO/Au structure. Micro-Raman Spectroscopy, SEM and Variable Angle Ellipsometry (VASE) measurements were performed on the Ag/GO/Au structure. An interesting behavior of the GO deposited on magnetron-sputtered silver with the formation of Ag nanostructures on top of the GO layer is reported. In addition to typical GO bands, Micro-Raman analysis reveals peaks such as the 1478 cm−1 band, indicating a transition from sp3 to sp2 hybridization, confirming the partial reduction of GO. Additionally, calculations based on effective medium theory (EMT) highlight the potential of Ag/GO structures in hyperbolic metamaterials for photonics. The medium exhibits dielectric behavior up to 323 nm, transitions to type I HMM between 323 and 400 nm and undergoes an Epsilon Near Zero and Pole (ENZP) transition at 400 nm, followed by type II HMM behavior. Full article
(This article belongs to the Special Issue Photonics Metamaterials: Processing and Applications)
Show Figures

Figure 1

21 pages, 609 KB  
Review
The Multifaceted Roles of MicroRNA-181 in Stem Cell Differentiation and Cancer Stem Cell Plasticity
by Chun Yang, Rui Wang and Pierre Hardy
Cells 2025, 14(2), 132; https://doi.org/10.3390/cells14020132 - 17 Jan 2025
Cited by 2 | Viewed by 1766
Abstract
Stem cells are undifferentiated or partially differentiated cells with an extraordinary ability to self-renew and differentiate into various cell types during growth and development. The epithelial–mesenchymal transition (EMT), a critical developmental process, enhances stem cell-like properties in cells, and is associated with both [...] Read more.
Stem cells are undifferentiated or partially differentiated cells with an extraordinary ability to self-renew and differentiate into various cell types during growth and development. The epithelial–mesenchymal transition (EMT), a critical developmental process, enhances stem cell-like properties in cells, and is associated with both normal stem cell function and the formation of cancer stem cells. Cell stemness and the EMT often coexist and are interconnected in various contexts. Cancer stem cells are a critical tumor cell population that drives tumorigenesis, cancer progression, drug resistance, and metastasis. Stem cell differentiation and the generation of cancer stem cells are regulated by numerous molecules, including microRNAs (miRNAs). These miRNAs, particularly through the modulation of EMT-associated factors, play major roles in controlling the stemness of cancer stem cells. This review presents an up-to-date summary of the regulatory roles of miR-181 in human stem cell differentiation and cancer cell stemness. We outline studies from the current literature and summarize the miR-181-controlled signaling pathways responsible for driving human stem cell differentiation or the emergence of cancer stem cells. Given its critical role in regulating cell stemness, miR-181 is a promising target for influencing human cell fate. Modulation of miR-181 expression has been found to be altered in cancer stem cells’ biological behaviors and to significantly improve cancer treatment outcomes. Additionally, we discuss challenges in miRNA-based therapies and targeted delivery with nanotechnology-based systems. Full article
(This article belongs to the Section Stem Cells)
Show Figures

Graphical abstract

18 pages, 4974 KB  
Article
MOMAST® Downregulates AQP3 Expression and Function in Human Colon Cells
by Ines Angelini, Mariangela Centrone, Giusy Rita Caponio, Annarita Di Mise, Andrea Gerbino, Marianna Ranieri, Giovanna Valenti and Grazia Tamma
Antioxidants 2025, 14(1), 26; https://doi.org/10.3390/antiox14010026 - 28 Dec 2024
Viewed by 1508
Abstract
The water channel AQP3 is an aquaglyceroporin expressed in villus epithelial cells, and it plays a role in water transport across human colonic surface cells. Beyond water, AQP3 can mediate glycerol and H2O2 transport. Abnormal expression and function of AQP3 [...] Read more.
The water channel AQP3 is an aquaglyceroporin expressed in villus epithelial cells, and it plays a role in water transport across human colonic surface cells. Beyond water, AQP3 can mediate glycerol and H2O2 transport. Abnormal expression and function of AQP3 have been found in various diseases often characterized by altered cell growth and proliferation. Here, the beneficial effects of MOMAST® have been evaluated. MOMAST® is an antioxidant-patented natural phenolic complex obtained from olive wastewater (OWW) of the Coratina cultivar. Treatment of human colon HCT8 cells with MOMAST® reduced cell viability. Confocal studies and Western Blotting analysis demonstrated that treatment with MOMAST® significantly decreased the staining and the expression of AQP3. Importantly, functional studies revealed that the reduction of AQP3 abundance correlates with a significant decrease in glycerol and H2O2 uptake. Indeed, the H2O2 transport was partially but significantly reduced in the presence of MOMAST® or DFP00173, a selective inhibitor of AQP3. In addition, the MOMAST®-induced AQP3 decrease was associated with reduced epithelial-mesenchymal transition (EMT)-related proteins such as vimentin and β-catenin. Together, these findings propose MOMAST® as a potential adjuvant in colon diseases associated with abnormal cell growth by targeting AQP3. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

25 pages, 8337 KB  
Article
Partial Inhibition of Epithelial-to-Mesenchymal Transition (EMT) Phenotypes by Placenta-Derived DBMSCs in Human Breast Cancer Cell Lines, In Vitro
by Yasser Basmaeil, Abdullah Al Subayyil, Haya Bin Kulayb, Altaf A. Kondkar, Maha Alrodayyan and Tanvir Khatlani
Cells 2024, 13(24), 2131; https://doi.org/10.3390/cells13242131 - 23 Dec 2024
Cited by 1 | Viewed by 1525
Abstract
Stem cell-based therapies hold significant potential for cancer treatment due to their unique properties, including migration toward tumor niche, secretion of bioactive molecules, and immunosuppression. Mesenchymal stem cells (MSCs) from adult tissues can inhibit tumor progression, angiogenesis, and apoptosis of cancer cells. We [...] Read more.
Stem cell-based therapies hold significant potential for cancer treatment due to their unique properties, including migration toward tumor niche, secretion of bioactive molecules, and immunosuppression. Mesenchymal stem cells (MSCs) from adult tissues can inhibit tumor progression, angiogenesis, and apoptosis of cancer cells. We have previously reported the isolation and characterization of placenta-derived decidua basalis mesenchymal stem cells (DBMSCs), which demonstrated higher levels of pro-migratory and anti-apoptotic genes, indicating potential anti-cancer effects. In this study, we analyzed the anti-cancer effects of DBMSCs on human breast cancer cell lines MDA231 and MCF7, with MCF 10A used as control. We also investigated how these cancer cells lines affect the functional competence of DBMSCs. By co-culturing DBMSCs with cancer cells, we analyzed changes in functions of both cell types, as well as alterations in their genomic and proteomic profile. Our results showed that treatment with DBMSCs significantly reduced the functionality of MDA231 and MCF7 cells, while MCF 10A cells remained unaffected. DBMSC treatment decreased epithelial-to-mesenchymal transition (EMT)-related protein levels in MDA231 cells and modulated expression of other cancer-related genes in MDA231 and MCF7 cells. Although cancer cells reduced DBMSC proliferation, they increased their expression of anti-apoptotic genes. These findings suggest that DBMSCs can inhibit EMT-related proteins and reduce the invasive characteristics of MDA231 and MCF7 breast cancer cells, highlighting their potential as candidates for cell-based cancer therapies. Full article
Show Figures

Figure 1

30 pages, 9646 KB  
Article
Plakin Expression in Serous Epithelial Ovarian Cancer Has the Potential to Impede Metastatic Spread and Epithelial–Mesenchymal Transition: A Comparative Expression Analysis of Immunohistochemical and In Silico Datasets
by Tamsin Wesley, Ruth M. Escalona, George Kannourakis and Nuzhat Ahmed
Cancers 2024, 16(23), 4087; https://doi.org/10.3390/cancers16234087 - 6 Dec 2024
Cited by 2 | Viewed by 1459
Abstract
Epithelial ovarian cancer is aggressive and causes high mortality among women worldwide. Members of the plakin family are essential to maintain cytoskeletal integrity and key cellular processes. In this study we characterised the expression of plakins, particularly plectin (PLEC), periplakin (PPL), envoplakin (EVPL), [...] Read more.
Epithelial ovarian cancer is aggressive and causes high mortality among women worldwide. Members of the plakin family are essential to maintain cytoskeletal integrity and key cellular processes. In this study we characterised the expression of plakins, particularly plectin (PLEC), periplakin (PPL), envoplakin (EVPL), and EMT-related proteins by immunohistochemistry in n = 48 patients’ samples to evaluate a potential correlation of plakin expression with EMT as EOC progresses. These tissue plakin and EMT expression analyses were further evaluated by in vitro cell line expression and correlated with the expression of these molecules using publicly available datasets such as Cancer Genome Atlas (TCGA) and Clinical Proteome Tumour Analysis Consortium (CPTAC) datasets. We demonstrate that the expression of PPL and PLEC plakins is decreased in high-grade compared to low-grade EOCs with mixed EMT marker protein expression. This is supported by the correlation of high PPL and PLEC expression with an epithelial rather than mesenchymal phenotype. Our data suggest a partial loss of plakin expression as EOC tumours progress. This may impact the connections of plakins with membrane-bound receptors, which impede the downstream signalling required for the initiation of EMT as the tumours progress. Full article
(This article belongs to the Section Cancer Metastasis)
Show Figures

Figure 1

Back to TopTop