Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (104)

Search Parameters:
Keywords = cancer-associated fibroblast features

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 9009 KiB  
Article
Cancer-Associated Fibroblasts Establish Spatially Distinct Prognostic Niches in Subcutaneous Colorectal Cancer Mouse Model
by Zhixian Lin, Jinmeng Wang, Yixin Ma, Yanan Zhu, Yuhan Li, Zhengtao Xiao and Wei Zhao
Cancers 2025, 17(14), 2402; https://doi.org/10.3390/cancers17142402 - 19 Jul 2025
Viewed by 502
Abstract
Background/Objectives: Subcutaneous tumor models are widely used in colorectal cancer (CRC) research due to their experimental accessibility; however, the spatial organization and regulatory mechanisms of their tumor microenvironment remain poorly understood. Methods: Here, we applied spatial transcriptomics to systematically characterize spatial heterogeneity within [...] Read more.
Background/Objectives: Subcutaneous tumor models are widely used in colorectal cancer (CRC) research due to their experimental accessibility; however, the spatial organization and regulatory mechanisms of their tumor microenvironment remain poorly understood. Methods: Here, we applied spatial transcriptomics to systematically characterize spatial heterogeneity within MC38 subcutaneous tumors in a syngeneic mouse model. Results: We identified two spatially distinct tumor zones, partitioned by cancer-associated fibroblasts (CAFs), that differ markedly in cellular composition, oncogenic signaling, immune infiltration, and metabolic states. One zone exhibited features of TGF-β-driven extracellular matrix remodeling, immune exclusion, and hyperproliferative metabolism, while the other was enriched for immunosuppressive macrophages, metabolic reprogramming via PPAR and AMPK pathways, and high-risk cell populations. Spatially resolved cell–cell communication networks further revealed zone-specific ligand–receptor interactions—such as ANGPTL4–SDC2 and PROS1–AXL—that underpin stromal remodeling and immune evasion and are associated with patient prognosis. Conclusions: Collectively, our study uncovers how region-specific cellular ecosystems and intercellular crosstalk establish prognostically divergent niches within subcutaneous CRC tumors, offering insights into spatially guided therapeutic strategies. Full article
(This article belongs to the Section Tumor Microenvironment)
Show Figures

Figure 1

19 pages, 5038 KiB  
Article
A Novel Hypoxia-Immune Signature for Gastric Cancer Prognosis and Immunotherapy: Insights from Bulk and Single-Cell RNA-Seq
by Mai Hanh Nguyen, Hoang Dang Khoa Ta, Doan Phuong Quy Nguyen, Viet Huan Le and Nguyen Quoc Khanh Le
Curr. Issues Mol. Biol. 2025, 47(7), 552; https://doi.org/10.3390/cimb47070552 - 16 Jul 2025
Viewed by 376
Abstract
Background: Hypoxia and immune components significantly shape the tumor microenvironment and influence prognosis and immunotherapy response in gastric cancer (GC). This study aimed to develop hypoxia- and immune-related gene signatures for prognostic evaluation in GC. Methods: Transcriptomic data from TCGA-STAD were [...] Read more.
Background: Hypoxia and immune components significantly shape the tumor microenvironment and influence prognosis and immunotherapy response in gastric cancer (GC). This study aimed to develop hypoxia- and immune-related gene signatures for prognostic evaluation in GC. Methods: Transcriptomic data from TCGA-STAD were integrated with hypoxia- and immune-related genes from InnateDB and MSigDB. A prognostic gene signature was constructed using Cox regression analyses and validated on an independent GSE84437 cohort and single-cell RNA dataset. We further analyzed immune cell infiltration, molecular characteristics of different risk groups, and their association with immunotherapy response. Single-cell RNA-seq data from the TISCH database were used to explore gene expression patterns across cell types. Results: Five genes (TGFB3, INHA, SERPINE1, GPC3, SRPX) were identified. The risk score effectively stratified patients by prognosis, with the high-risk group showing lower overall survival and lower T-cell expression. The gene signature had an association with immune suppression, ARID1A mutation, EMT features, and poorer response to immunotherapy. Gene signature, especially SRPX was enriched in fibroblasts. Conclusions: We developed a robust hypoxia- and immune-related gene signature that predicts prognosis and may help guide immunotherapy strategies for GC patients. Full article
(This article belongs to the Special Issue Linking Genomic Changes with Cancer in the NGS Era, 2nd Edition)
Show Figures

Figure 1

15 pages, 4034 KiB  
Article
Establishment of Human Lung Cancer Organoids Using Small Biopsy and Surgical Tissues
by Mina Hwang, Junsu Choe, Yong Jae Shin, Bo-Gyeong Seo, Kyung-Mi Park, Sun Hye Shin, Byung Woo Jhun, Hongseok Yoo, Byeong-Ho Jeong, Kyeongman Jeon, Kyungjong Lee, Junghee Lee, Yeong Jeong Jeon, Jong Ho Cho, Seong Yong Park, Hong Kwan Kim and Sang-Won Um
Cancers 2025, 17(14), 2291; https://doi.org/10.3390/cancers17142291 - 10 Jul 2025
Viewed by 632
Abstract
Background/Objectives: Lung cancer is a highly diverse disease, and reliable preclinical models that accurately reflect tumor characteristics are essential for studying lung cancer biology and testing new therapies. This study aimed to establish patient-derived tumor organoids (PDTOs) using small biopsy samples and surgical [...] Read more.
Background/Objectives: Lung cancer is a highly diverse disease, and reliable preclinical models that accurately reflect tumor characteristics are essential for studying lung cancer biology and testing new therapies. This study aimed to establish patient-derived tumor organoids (PDTOs) using small biopsy samples and surgical specimens to create a model system that preserves the genetic and histological features of the original tumors. Methods: PDTOs were generated from 163 lung cancer specimens, including 109 samples obtained using endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) or bronchoscopy, 52 surgical specimens, and 2 pleural fluid samples. The organoid establishment rate beyond passage three was assessed, and histological subtypes and genetic profiles were analyzed using immunohistochemical staining and targeted exome sequencing. Results: The overall PDTO establishment rate was 34.4% (56/163), and 44.6% (25/56) of these organoids retained the histological and genetic features of the parental tumors. Genetic analysis identified key mutations, including KRAS G12C, EGFR L858R, MET exon 14 skipping mutation, and ROS1 fusion. PDTOs successfully formed tumors in mice while maintaining the genetic characteristics of the original tumors. Co-culture of PDTOs with cancer-associated fibroblasts (CAFs) resulted in increased resistance to paclitaxel. In the co-culture model of PDTOs with immune cells, dose-dependent growth inhibition of PDTOs was observed in response to immune checkpoint inhibitors. Conclusions: PDTOs established from small biopsy and surgical specimens serve as a valuable model for studying lung cancer biology, tumor microenvironment interactions, and drug response. This model has the potential to improve personalized treatment strategies. Full article
(This article belongs to the Special Issue New Perspectives in the Treatment of Thoracic Cancers)
Show Figures

Figure 1

18 pages, 1010 KiB  
Review
Molecular Mechanisms of Lymph Node Metastasis in Gallbladder Cancer: Insights into the Tumor Microenvironment
by Qingyu Tang, Yichen Guan, Yubo Ma, Qi Li and Zhimin Geng
Biomedicines 2025, 13(6), 1372; https://doi.org/10.3390/biomedicines13061372 - 4 Jun 2025
Viewed by 949
Abstract
Gallbladder cancer (GBC) is a highly aggressive malignancy with a propensity for lymph node metastasis (LNM), which significantly worsens prognosis. This review explores the molecular mechanisms underlying LNM in GBC, focusing on the roles of vascular endothelial growth factors (VEGFs), chemokines, cancer-associated fibroblasts [...] Read more.
Gallbladder cancer (GBC) is a highly aggressive malignancy with a propensity for lymph node metastasis (LNM), which significantly worsens prognosis. This review explores the molecular mechanisms underlying LNM in GBC, focusing on the roles of vascular endothelial growth factors (VEGFs), chemokines, cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), hypoxia-inducible factors (HIFs), and non-coding RNAs (ncRNAs) in shaping the tumor microenvironment (TME). Unique features of GBC, such as its bile-rich microenvironment and hypoxia-driven lymphangiogenesis, are highlighted. We discuss how these factors promote lymphangiogenesis, immune evasion, and extracellular matrix (ECM) remodeling, collectively facilitating LNM. Potential therapeutic targets, including VEGF-C/D pathways, matrix metalloproteinase (MMP) inhibitors, and immune-modulating therapies, are also reviewed. Future research integrating single-cell omics and patient-derived organoid models is essential for advancing precision medicine in GBC. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 1

22 pages, 11326 KiB  
Article
Isolation and Characterization of the Adamantinomatous Craniopharyngioma Primary Cells with Cancer-Associated Fibroblast Features
by Dongting Chen, Ting Lei, Yulin Wang, Zaitao Yu, Siqi Liu, Ling Ye, Wanfang Li, Qin Yang, Hongtao Jin, Fangjun Liu and Yan Li
Biomedicines 2025, 13(4), 912; https://doi.org/10.3390/biomedicines13040912 - 9 Apr 2025
Cited by 1 | Viewed by 894
Abstract
Backgrounds: Adamantinomatous craniopharyngiomas (ACPs) are benign intracranial tumors that behave aggressively due to their location, infiltration of the surrounding nervous tissue and high capacity for recurrence. In this study, we aimed to construct ACP primary cell models for further investigation of tumorigenic [...] Read more.
Backgrounds: Adamantinomatous craniopharyngiomas (ACPs) are benign intracranial tumors that behave aggressively due to their location, infiltration of the surrounding nervous tissue and high capacity for recurrence. In this study, we aimed to construct ACP primary cell models for further investigation of tumorigenic and recurrent mechanisms. Methods: Primary cells were isolated from primary (one case) and recurrent (one case) ACP. Short tandem repeat (STR) analysis was used to clarify the identity of the ACP primary cells we isolated. Whole exome sequencing (WES), immunofluorescence (IF) and immunohistochemistry (IHC) were performed on primary cells and corresponding ACP tissues, to determine the mutational profile and to clarify the tissue origin and phenotypic of primary cells. Transcriptome RNA-seq was performed to obtain the gene expression characteristics of ACP primary cells. Subsequently, a heterotopic ACP xenograft mouse model was established to confirm the tumorigenesis capacity of ACP primary cells. Results: ACP primary cells were successfully cultured. The genetic variants were similar to the original tumor tissue, and they owned expression of cancer-associated fibroblast (CAF) markers (FSP1/S100A4, Vimentin) and nuclear translocation β-catenin. Meanwhile, they had an high level expression of extracellular matrix components (Fibronectin). The tumor formation ability of ACP primary cells was verified. The transcriptional signatures of ACP primary cells were also explored. Conclusions: We successfully isolated and characterized ACP primary cells that acquired multiple CAF features and demonstrated stable propagation through dozens of passages. These PDC models laid the foundation for further research on ACP. Full article
(This article belongs to the Special Issue Clinical, Radiological, and Molecular Insights into Craniopharyngioma)
Show Figures

Figure 1

17 pages, 8059 KiB  
Article
Cancer-Associated Fibroblasts Genes and Transforming Growth Factor Beta Pathway in Gastric Cancer for Novel Therapeutic Strategy
by Hiroyuki Minoura, Riku Okamoto, Naoki Hiki and Keishi Yamashita
Cancers 2025, 17(5), 795; https://doi.org/10.3390/cancers17050795 - 26 Feb 2025
Cited by 2 | Viewed by 1486
Abstract
Background-Objective: Cancer-associated fibroblasts (CAFs) play a crucial role in the tumor microenvironment of gastric cancer (GC). Understanding the molecular characteristics of CAFs-associated genes (CAFGs) is essential for elucidating their role in tumor progression and prognosis. This review aims to summarize the current knowledge [...] Read more.
Background-Objective: Cancer-associated fibroblasts (CAFs) play a crucial role in the tumor microenvironment of gastric cancer (GC). Understanding the molecular characteristics of CAFs-associated genes (CAFGs) is essential for elucidating their role in tumor progression and prognosis. This review aims to summarize the current knowledge on CAFGs, highlighting their expression patterns, prognostic significance, and potential functional mechanisms. Methods: A comprehensive review of existing literature was conducted, focusing on molecular features of CAFGs in GC. Single-cell RNA sequencing (scRNA-seq) analyses were examined to assess the expression patterns of CAFGs in broad-sense CAFs, which include both CAFs and pericytes. Additionally, clinicopathological studies validating the prognostic significance of CAFGs were reviewed. Results: ScRNA-seq analyses revealed that CAFGs are not necessarily restricted to CAFs alone but may also reflect the activation status of surrounding cells. Several CAFGs, including SPARC, THBS2, COL1A1, COL3A1, INHBA, PDGFC, and SDC2, have been validated for their prognostic relevance in GC. However, compared with other cancers, the functional mechanisms of these genes in GC remain poorly understood. While CAFGs exhibit synchronized expression with TGFB1 in colorectal cancer (CRC), such patterns have yet to be confirmed in GC due to the limitations of available microdissected data. Conclusions: A comprehensive understanding of CAFGs and their interaction with the TGFB pathway, including LTBP family genes, may be critical for developing novel therapeutic strategies for GC. Further research is needed to elucidate their functional mechanisms and therapeutic potential. Full article
(This article belongs to the Special Issue Gastric Cancer Metastases)
Show Figures

Figure 1

36 pages, 43463 KiB  
Article
A Multi-Omics-Based Exploration of the Predictive Role of MSMB in Prostate Cancer Recurrence: A Study Using Bayesian Inverse Convolution and 10 Machine Learning Combinations
by Shan Huang and Hang Yin
Biomedicines 2025, 13(2), 487; https://doi.org/10.3390/biomedicines13020487 - 16 Feb 2025
Cited by 1 | Viewed by 1170
Abstract
Background: Prostate cancer (PCa) is a prevalent malignancy among elderly men. Biochemical recurrence (BCR), which typically occurs after radical treatments such as radical prostatectomy or radiation therapy, serves as a critical indicator of potential disease progression. However, reliable and effective methods for predicting [...] Read more.
Background: Prostate cancer (PCa) is a prevalent malignancy among elderly men. Biochemical recurrence (BCR), which typically occurs after radical treatments such as radical prostatectomy or radiation therapy, serves as a critical indicator of potential disease progression. However, reliable and effective methods for predicting BCR in PCa patients remain limited. Methods: In this study, we used Bayesian deconvolution combined with 10 machine learning algorithms to build a five-gene model for predicting PCa progression. The model and the five selected genes were externally validated. Various analyses such as prognosis, clinical subgroups, tumor microenvironment, immunity, genetic variants, and drug sensitivity were performed on MSMB/Epithelial_cells subgroups. Results: Our model outperformed 102 previously published prognostic features. Notably, PCa patients with a high proportion of MSMB/epithelial cells were characterized by a greater progression-free Interval (PFI), a higher proportion of early-stage tumors, a lower stromal component, and a reduced presence of tumor-associated fibroblasts (CAF). The high proportion of MSMB/epithelial cells was also associated with higher frequencies of SPOP and TP53 mutations. Drug sensitivity analysis revealed that patients with a poorer prognosis and lower MSMB/epithelial cell ratio showed increased sensitivity to cyclophosphamide, cisplatin, and dasatinib. Conclusions: The model developed in this study provides a robust and accurate tool for predicting PCa progression. It offers significant potential for enhancing risk stratification and informing personalized treatment strategies for PCa patients. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 1

30 pages, 12176 KiB  
Article
Targeting Fibroblast-Derived Interleukin 6: A Strategy to Overcome Epithelial-Mesenchymal Transition and Radioresistance in Head and Neck Cancer
by Xinyang Li, Hugues Escoffier, Thomas Sauter and Mahvash Tavassoli
Cancers 2025, 17(2), 267; https://doi.org/10.3390/cancers17020267 - 15 Jan 2025
Cited by 1 | Viewed by 1542
Abstract
Background: Cancer-associated fibroblasts have been reported to play a central role in driving cancer progression, promoting metastasis, and conferring resistance to therapy in HNSCC. Methods: Indirect and direct co-culture models of HPV-positive and HPV-negative HNSCC cells with fibroblasts were developed to study the [...] Read more.
Background: Cancer-associated fibroblasts have been reported to play a central role in driving cancer progression, promoting metastasis, and conferring resistance to therapy in HNSCC. Methods: Indirect and direct co-culture models of HPV-positive and HPV-negative HNSCC cells with fibroblasts were developed to study the effect of fibroblasts on cancer cells. ELISA was used to measure IL-6 secretion in these models. To dissect the underlying signalling mechanisms, the effects of IL-6, an IL-6 receptor (IL-6R) inhibitor, a MAPK/ERK inhibitor, and a JAK/STAT inhibitor were evaluated. Epithelial-to-mesenchymal transition (EMT) was assessed by measuring EMT markers and conducting scratch assays and spheroid assays. Radioresistance was evaluated using clonogenic assays. Additionally, radioresistant (RR) cell lines were established from parental cells to examine the correlation between radioresistance and EMT. Results: Fibroblasts were found to drive EMT-like changes and heightened radioresistance in HNSCC cells through IL-6 secretion. Remarkably, these Fb-driven effects were robustly reversed using IL-6R and MAPK/ERK inhibitors in both HPV-positive and HPV-negative cell lines, whereas JAK/STAT inhibitors proved effective only in HPV-negative cells. RR cell lines exhibit a more aggressive phenotype than their parental counterparts, marked by pronounced EMT features and heightened resistance to radiotherapy. Importantly, these aggressive characteristics were substantially attenuated by targeting IL-6R or MAPK/ERK pathways. Conclusions: This study highlights the critical role of fibroblast-secreted IL-6 in driving and maintaining EMT and radioresistance in HNSCC, resulting in a more aggressive tumour phenotype. Targeting the IL-6/IL-6R/ERK pathway emerges as a promising therapeutic approach for combating CAF-driven tumour progression and improving clinical outcomes in patients with aggressive, therapy-resistant HNSCC. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

20 pages, 4167 KiB  
Article
Transcriptome Analysis Suggests PKD3 Regulates Proliferative Glucose Metabolism, Calcium Homeostasis and Microtubule Dynamics After MEF Spontaneous Immortalization
by Jocshan Loaiza-Moss, Ursula Braun and Michael Leitges
Int. J. Mol. Sci. 2025, 26(2), 596; https://doi.org/10.3390/ijms26020596 - 12 Jan 2025
Cited by 1 | Viewed by 1205
Abstract
Cell immortalization corresponds to a biologically relevant clinical feature that allows cells to acquire a high proliferative potential during carcinogenesis. In multiple cancer types, Protein Kinase D3 (PKD3) has often been reported as a dysregulated oncogenic kinase that promotes cell proliferation. Using mouse [...] Read more.
Cell immortalization corresponds to a biologically relevant clinical feature that allows cells to acquire a high proliferative potential during carcinogenesis. In multiple cancer types, Protein Kinase D3 (PKD3) has often been reported as a dysregulated oncogenic kinase that promotes cell proliferation. Using mouse embryonic fibroblasts (MEFs), in a spontaneous immortalization model, PKD3 has been demonstrated as a critical regulator of cell proliferation after immortalization. However, the mechanisms by which PKD3 regulates proliferation in immortalized MEFs require further elucidation. Using a previously validated Prkd3-deficient MEF model, we performed a poly-A transcriptomic analysis to identify putative Prkd3-regulated biological processes and downstream targets in MEFs after spontaneous immortalization. To this end, differentially expressed genes (DEGs) were identified and further analyzed by gene ontology (GO) enrichment and protein–protein interaction (PPI) network analyses to identify potential hub genes. Our results suggest that Prkd3 modulates proliferation through the regulation of gene expression associated with glucose metabolism (Tnf, Ucp2, Pgam2, Angptl4), calcium homeostasis and transport (Calcr and P2rx7) and microtubule dynamics (Stmn2 and Map10). These candidate processes and associated genes represent potential mechanisms involved in Prkd3-induced proliferation in spontaneously immortalized cells as well as clinical targets in several cancer types. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

20 pages, 6263 KiB  
Article
Aging-Related Gene-Based Prognostic Model for Lung Adenocarcinoma: Insights into Tumor Microenvironment and Therapeutic Implications
by Jin Wang, Hailong Zhang, Yaohui Feng, Xian Gong, Xiangrong Song, Meidan Wei, Yaoyu Hu and Jianxiang Li
Int. J. Mol. Sci. 2024, 25(24), 13572; https://doi.org/10.3390/ijms252413572 - 18 Dec 2024
Cited by 1 | Viewed by 1596
Abstract
Lung cancer remains the leading cause of cancer-related mortality globally, with a poor prognosis primarily due to late diagnosis and limited treatment options. This research highlights the critical demand for advanced prognostic tools by creating a model centered on aging-related genes (ARGs) to [...] Read more.
Lung cancer remains the leading cause of cancer-related mortality globally, with a poor prognosis primarily due to late diagnosis and limited treatment options. This research highlights the critical demand for advanced prognostic tools by creating a model centered on aging-related genes (ARGs) to improve prediction and treatment strategies for lung adenocarcinoma (LUAD). By leveraging datasets from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), we developed a prognostic model that integrates 14 ARGs using the least absolute shrinkage and selection operator (LASSO) alongside Cox regression analyses. The model exhibited strong predictive performance, achieving area under the curve (AUC) values greater than 0.8 for one-year survival in both internal and external validation cohorts. The risk scores generated by our model were significantly correlated with critical features of the tumor microenvironment, including the presence of cancer-associated fibroblasts (CAFs) and markers of immune evasion, such as T-cell dysfunction and exclusion. Higher risk scores correlated with a more tumor-promoting microenvironment and increased immune suppression, highlighting the model’s relevance in understanding LUAD progression. Additionally, XRCC6, a protein involved in DNA repair and cellular senescence, was found to be upregulated in LUAD. Functional assays demonstrated that the knockdown of XRCC6 led to decreased cell proliferation, whereas its overexpression alleviated DNA damage, highlighting its significance in tumor biology and its potential therapeutic applications. This study provides a novel ARG-based prognostic model for LUAD, offering valuable insights into tumor dynamics and the tumor microenvironment, which may guide the development of targeted therapies and improve patient outcomes. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

17 pages, 14804 KiB  
Article
A Role for Periostin Pathological Variants and Their Interaction with HSP70-1a in Promoting Pancreatic Cancer Progression and Chemoresistance
by Yasuo Tsunetoshi, Fumihiro Sanada, Yuko Kanemoto, Kana Shibata, Atsushi Masamune, Yoshiaki Taniyama, Koichi Yamamoto and Ryuichi Morishita
Int. J. Mol. Sci. 2024, 25(23), 13205; https://doi.org/10.3390/ijms252313205 - 8 Dec 2024
Cited by 2 | Viewed by 1697
Abstract
Pancreatic ductal adenocarcinoma (PDAC) characterized by an abundant cancer stroma is an aggressive malignancy with a poor prognosis. Periostin (Pn) is a key extracellular matrix (ECM) protein in various tumor progression. Previously, we described the role of Pn alternative splicing variants (ASVs) with [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) characterized by an abundant cancer stroma is an aggressive malignancy with a poor prognosis. Periostin (Pn) is a key extracellular matrix (ECM) protein in various tumor progression. Previously, we described the role of Pn alternative splicing variants (ASVs) with specific functional features in breast cancer. Pn is known to associate with a chemoresistance of PDAC, but the functions of the Pn-ASVs remain largely unknown. In this study, we focused on physiological and pathological Pn-ASVs, and examined the characteristics of Pn-expressing cells and the difference in function of each ASV. We found that cancer-associated fibroblasts (CAFs) are a main source of Pn synthesis, which selectively secrete pathological Pn-ASVs with exon 21 both in mouse and human samples. RNA sequencing identified a gene signature of Pn-positive CAFs associated with ECM-related genes and chemokines, factors that shape the chemoresistance tumor microenvironment (TME). Additionally, only pathological Pn-ASVs interacted with heat shock protein 70-1a (HSP70-1a), leading to significant rescue of gemcitabine-induced PDAC apoptosis. In silico analysis revealed that the presence or absence of exon 21 changes the tertiary structure of Pn and the binding sites for HSP70-1a. Altogether, Pn-ASVs with exon 21 secreted from CAFs play a key role in supporting tumor growth by interacting with cancer cell-derived HSP70-1a, indicating that Pn-ASVs with exon 21 might be a potential therapeutic and diagnostic target in PDAC patients with rich stroma. Full article
Show Figures

Figure 1

20 pages, 15501 KiB  
Article
Pan-Cancer Analysis Reveals the Potential of PLOD1 as a Prognostic and Immune Biomarker for Human Cancer
by Zhao Zhai, Shuo Wang, Yudong Cao, Jia Liu, Qiang Zhao, Yongpeng Ji, Xiao Yang, Xingxing Tang, Jinchao Ma and Peng Du
Biomedicines 2024, 12(12), 2653; https://doi.org/10.3390/biomedicines12122653 - 21 Nov 2024
Viewed by 1153
Abstract
Background/Objectives: Procollagen-lysine, 2-oxoglutarate 5-dioxygenase 1 (PLOD1) is known as an enhancer of collagen fiber deposition and cross-linking stability. However, there is limited information on its function in tumors. In this study, we aimed to elucidate the function and potential mechanism of action of [...] Read more.
Background/Objectives: Procollagen-lysine, 2-oxoglutarate 5-dioxygenase 1 (PLOD1) is known as an enhancer of collagen fiber deposition and cross-linking stability. However, there is limited information on its function in tumors. In this study, we aimed to elucidate the function and potential mechanism of action of PLOD1 across cancers. Methods: We assessed the pan-cancer expression, mutation, methylation and prognostic value of PLOD1 through multiple online databases. In addition, we performed correlation analyses of its immunological features, as well as functional assessment analyses of PLOD1. Finally, we assessed the effect of PLOD1 knockdown on bladder tumor cells using in vitro experiments. Results: Our findings suggest that PLOD1 is aberrantly expressed in multiple cancer types, accompanied by a poor prognosis. Epigenetic alterations in PLOD1 are highly heterogeneous across a wide range of tumors, and aberrant methylation and copy number variants correlate with a poor prognosis. In the tumor microenvironment, PLOD1 expression correlated positively with the infiltration level of various immunosuppressive cells (e.g., monocytes, macrophages and tumor-associated fibroblasts) and negatively with immune-killing cells (e.g., CD8+ T cells, B cells and CD4+ T cells). In addition, PLOD1 expression was associated with immune checkpoints and immunomodulatory genes. Finally, in vitro experiments demonstrated that knockdown of PLOD1 reduced the proliferation, migration and antiapoptotic abilities of T24 cells. Conclusions: The results of this study demonstrate that PLOD1 is a potential oncogene and prognostic biomarker in pan-cancer; tumor tissues with high PLOD1 expression reveal a relatively immunosuppressive tumor microenvironment. Full article
(This article belongs to the Special Issue Epigenetic Regulation and Its Impact for Medicine)
Show Figures

Figure 1

21 pages, 4671 KiB  
Article
Extracellular Vesicles from Lung Adenocarcinoma Cells Induce Activation of Different Cancer-Associated Fibroblast Subtypes
by Jessica Angelina Trejo Vazquez, Rebecca Towle, Dylan Andrew Farnsworth, Masih Sarafan, William Wallace Lockwood and Cathie Garnis
Biomedicines 2024, 12(11), 2523; https://doi.org/10.3390/biomedicines12112523 - 4 Nov 2024
Cited by 1 | Viewed by 1642
Abstract
Background: Lung cancer, including the major subtype lung adenocarcinoma (LUAD), is the leading cause of cancer deaths worldwide, largely due to metastasis. Improving survival rates requires new treatment strategies and a deeper understanding of the mechanisms that drive tumor progression within the tumor [...] Read more.
Background: Lung cancer, including the major subtype lung adenocarcinoma (LUAD), is the leading cause of cancer deaths worldwide, largely due to metastasis. Improving survival rates requires new treatment strategies and a deeper understanding of the mechanisms that drive tumor progression within the tumor microenvironment (TME). This study investigated the impact of extracellular vesicles (EVs) derived from LUAD cells on lung fibroblasts. Methods: EVs were isolated from LUAD cell lines via ultracentrifugation and characterized using nanoparticle tracking analysis and Western blotting. Lung fibroblasts were treated with PBS, TGFβ, or EVs, and their activation was assessed through protein (Western blotting) and RNA analyses (RNA seq and RT-qPCR). Results: The results confirmed the TGFβ induced activation and showed that LUAD EVs could also activate fibroblasts, increasing cancer-associated fibroblast (CAF) markers. While EV-induced CAF activation displayed unique features, like an increase in proliferation-related genes, the EV and TGFβ treatments also shared some differentially expressed genes. The EV groups induced a higher expression of ECM remodeling and EMT-associated genes, but some of those genes were also upregulated in the TGFβ group. Mesenchymal genes POSTN and SPOCK1 were significantly upregulated in TGFβ- and EV-treated fibroblasts. Their secretion as proteins from the TGFβ- and EV-induced CAFs was not significant, confirmed through ELISA. Conclusions: These findings suggest that LUAD EVs play a role in CAF activation through both shared and distinct pathways compared to canonical TGFβ activation, potentially identifying novel gene expressions involved in CAF activation. Additionally, optimal protein secretion conditions of confirmed CAF-upregulated genes need to be established to determine their contribution to the TME. Full article
(This article belongs to the Special Issue Fibroblasts: Insights from Molecular and Pathophysiology Perspectives)
Show Figures

Figure 1

25 pages, 1116 KiB  
Review
Insights into CSF-1R Expression in the Tumor Microenvironment
by Caterina Tomassetti, Gaia Insinga, Francesca Gimigliano, Andrea Morrione, Antonio Giordano and Emanuele Giurisato
Biomedicines 2024, 12(10), 2381; https://doi.org/10.3390/biomedicines12102381 - 18 Oct 2024
Cited by 9 | Viewed by 3555
Abstract
The colony-stimulating factor 1 receptor (CSF-1R) plays a pivotal role in orchestrating cellular interactions within the tumor microenvironment (TME). Although the CSF-1R has been extensively studied in myeloid cells, the expression of this receptor and its emerging role in other cell types in [...] Read more.
The colony-stimulating factor 1 receptor (CSF-1R) plays a pivotal role in orchestrating cellular interactions within the tumor microenvironment (TME). Although the CSF-1R has been extensively studied in myeloid cells, the expression of this receptor and its emerging role in other cell types in the TME need to be further analyzed. This review explores the multifaceted functions of the CSF-1R across various TME cellular populations, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), cancer-associated fibroblasts (CAFs), endothelial cells (ECs), and cancer stem cells (CSCs). The activation of the CSF-1R by its ligands, colony-stimulating factor 1 (CSF-1) and Interleukin-34 (IL-34), regulates TAM polarization towards an immunosuppressive M2 phenotype, promoting tumor progression and immune evasion. Similarly, CSF-1R signaling influences MDSCs to exert immunosuppressive functions, hindering anti-tumor immunity. In DCs, the CSF-1R alters antigen-presenting capabilities, compromising immune surveillance against cancer cells. CSF-1R expression in CAFs and ECs regulates immune modulation, angiogenesis, and immune cell trafficking within the TME, fostering a pro-tumorigenic milieu. Notably, the CSF-1R in CSCs contributes to tumor aggressiveness and therapeutic resistance through interactions with TAMs and the modulation of stemness features. Understanding the diverse roles of the CSF-1R in the TME underscores its potential as a therapeutic target for cancer treatment, aiming at disrupting pro-tumorigenic cellular crosstalk and enhancing anti-tumor immune responses. Full article
Show Figures

Figure 1

13 pages, 5403 KiB  
Article
Activation of Mammary Epithelial and Stromal Fibroblasts upon Exposure to Escherichia coli Metabolites
by Jamilah H. Alshehri, Huda K. Al-Nasrallah, Mysoon M. Al-Ansari and Abdelilah Aboussekhra
Cells 2024, 13(20), 1723; https://doi.org/10.3390/cells13201723 - 17 Oct 2024
Cited by 4 | Viewed by 1202
Abstract
Breast cancer is the leading cause of cancer death among women worldwide. The mammary gland is composed of various types of cells including luminal cells, fibroblasts, immune cells, adipocytes, and specific microbiota. The reciprocal interaction between these multiple types of cells can dictate [...] Read more.
Breast cancer is the leading cause of cancer death among women worldwide. The mammary gland is composed of various types of cells including luminal cells, fibroblasts, immune cells, adipocytes, and specific microbiota. The reciprocal interaction between these multiple types of cells can dictate the initiation and progression of cancer, as well as metastasis and response to therapy. In the present report, we have shown that Escherichia coli-conditioned media (E-CM) can directly activate human mammary luminal epithelial cells (HMLEs), by inducing epithelial-to-mesenchymal transition (EMT), a process associated with increased proliferation and invasion capacities, as well as stemness features. Additionally, it has been shown that E-CM has an indirect pro-carcinogenic effect, mediated by the activation of normal breast fibroblasts (NBFs). Indeed, E-CM upregulated various markers of active fibroblasts (FAP-α, GPR77, and CD10), and enhanced the proliferation, migration, and invasion capacities of NBFs. Furthermore, E-CM induced an inflammatory response in NBFs by activating the pro-inflammatory NF-kB transcription factor and several of its downstream target cytokines including IL-1β, IL-6, and IL-8. This E-CM-dependent activation of NBFs was confirmed by showing their paracrine pro-carcinogenic effects through inducing EMT and stemness features in normal breast epithelial cells. Interestingly, similar effects were obtained by recombinant human IL-1β. These results provide the first indication that E. coli can initiate breast carcinogenesis through the activation of breast stromal fibroblasts and their paracrine pro-carcinogenic effects. Full article
(This article belongs to the Section Cell Microenvironment)
Show Figures

Figure 1

Back to TopTop