1
|
Contreras L, Rodríguez-Gil A, Muntané J, de la Cruz J. Sorafenib-associated translation reprogramming in hepatocellular carcinoma cells. RNA Biol 2025; 22:1-11. [PMID: 40116042 PMCID: PMC11934173 DOI: 10.1080/15476286.2025.2483484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Abstract
Sorafenib (Sfb) is a multikinase inhibitor regularly used for the management of patients with advanced hepatocellular carcinoma (HCC) that has been shown to increase very modestly life expectancy. We have shown that Sfb inhibits protein synthesis at the level of initiation in cancer cells. However, the global snapshot of mRNA translation following Sorafenib-treatment has not been explored so far. In this study, we performed a genome-wide polysome profiling analysis in Sfb-treated HCC cells and demonstrated that, despite global translation repression, a set of different genes remain efficiently translated or are even translationally induced. We reveal that, in response to Sfb inhibition, translation is tuned, which strongly correlates with the presence of established mRNA cis-acting elements and the corresponding protein factors that recognize them, including DAP5 and ARE-binding proteins. At the level of biological processes, Sfb leads to the translational down-regulation of key cellular activities, such as those related to the mitochondrial metabolism and the collagen synthesis, and the translational up-regulation of pathways associated with the adaptation and survival of cells in response to the Sfb-induced stress. Our findings indicate that Sfb induces an adaptive reprogramming of translation and provides valuable information that can facilitate the analysis of other drugs for the development of novel combined treatment strategies based on Sfb therapy.
Collapse
Affiliation(s)
- Laura Contreras
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | - Alfonso Rodríguez-Gil
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| | - Jordi Muntané
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús de la Cruz
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
2
|
Pawlina-Tyszko K, Semik-Gurgul E, Podstawski P, Herc W, Witkowski M, Ropka-Molik K. Altered expression of collagen gene family members and its epigenetic background in equine Sarcoids. Res Vet Sci 2025; 190:105656. [PMID: 40288239 DOI: 10.1016/j.rvsc.2025.105656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Alterations in the genes involved in the creation of the extracellular matrix (ECM) were observed in our earlier transcriptome studies of sarcoids and their cell culture model. For a complete characterization of the underlying molecular pathways, it is imperative to comprehend the involvement of ECM modifications in the oncogenic transformation of sarcoid fibroblasts. Thus, the aim of this investigation was to describe the expression patterns of a set of genes that are essential for the rearrangements of the extracellular matrix, namely collagen genes, and elucidate possible mechanisms underlying the observed disruptions. To this end, we applied the RT-qPCR method on BPV-negative skin samples and sarcoid samples (n = 6 and 7; respectively) to perform relative quantification of the expression level of eight genes belonging to the collagen family and carried out an integrative analysis of the obtained data with previously characterized epigenetic signatures. The results showed aberrations in the level of chosen collagen genes in the sarcoids compared to the control, manifesting in their elevated levels in the tumor samples (p-value≤0.05). The upregulation of Col1A2, Col11A1, Col6A3, Col5A2, Col4A1, Col6A6, Col5A1, Col6A2 genes was detected in sarcoid samples. The identified changes were statistically significant (p-value≤0.05) and ranged from 1.43 (Col6A2) to 1.88 (Col6A3). Further investigation into the potential involvement of epigenetic mechanisms in the regulation of collagen gene levels in sarcoids revealed compelling evidence of DNA methylation and microRNAs playing significant roles. The findings suggest a complex interplay between gene expression, epigenetic regulation, and the dysregulation of the ECM in sarcoid pathogenesis.
Collapse
Affiliation(s)
- Klaudia Pawlina-Tyszko
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 st, 32-083 Balice, Poland
| | - Ewelina Semik-Gurgul
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 st, 32-083 Balice, Poland
| | - Przemysław Podstawski
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 st, 32-083 Balice, Poland
| | - Weronika Herc
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 st, 32-083 Balice, Poland
| | - Maciej Witkowski
- Institute of Veterinary Medicine, University Centre of Veterinary Medicine JU-AU, Mickiewicza 24/28 st., 30-059 Kraków, Poland; Horse Clinic Służewiec, Puławska 266 st, 02-684 Warsaw, Poland
| | - Katarzyna Ropka-Molik
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 st, 32-083 Balice, Poland.
| |
Collapse
|
3
|
Wang S, Sun Z, Wang C, Zhang A, Zhang C, Hou S, Lin N, Li Q. The JAK1/STAT3 pathway mediates the effects of SERPINH1 on glioma EMT. Int Immunopharmacol 2025; 157:114731. [PMID: 40334628 DOI: 10.1016/j.intimp.2025.114731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025]
Abstract
Glioma is marked by swift cell proliferation, extensive invasion and poor outcomes. Serine protease inhibitor H1 (SERPINH1) encoding heat shock protein 47, a collagen-specific molecular chaperone, plays a role in a number of cancers. However, its definite role in glioma remains unclear. The aim of the present study was to investigate the role of SERPINH1 in glioma progression, especially its impact on cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT). The glioma cell lines LN229, T98, U251 and U87MG were transfected with lentivirus for stable knockdown or overexpression of SERPINH1. Assays assessing cell proliferation, migration and invasion were conducted to investigate the role of SERPINH1 in these processes. Bioinformatic analysis was conducted using The Cancer Genome Atlas and the Chinese Glioma Genome Atlas databases to identify potential molecular pathways associated with SERPINH1. Western blotting (WB) was employed to examine the expression of significant proteins in the JAK1/STAT3 signaling pathway and EMT markers. Nude mice were used for in vivo experiments to evaluate tumor growth and changes related to EMT. Overexpression of SERPINH1 notably increased glioma cell proliferation, migration and invasion, whereas knockdown suppressed these activities. Bioinformatic analyses revealed that SERPINH1 is closely associated with the JAK1/STAT3 signaling pathway and EMT-related genes. WB results confirmed that SERPINH1 regulates the activation of JAK1/STAT3 and influences the levels of EMT markers such as N- and E-cadherin. The JAK1/STAT3 agonist RO8191 partially rescued glioma cell behavior in the SERPINH1 knockdown group, while the inhibitor STATTIC partially weakened the enhanced effects in the SERPINH1 overexpression group. In vivo, SERPINH1 overexpression accelerated tumor growth and EMT progression, while knockdown resulted in a reduction in tumor size and the expression of EMT markers. SERPINH1 is essential for glioma progression, enhancing cell proliferation, migration, invasion and EMT by activating the JAK1/STAT3 signaling pathway. These results indicate that targeting SERPINH1 could provide a promising new approach for glioma therapy.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Zhiming Sun
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Chao Wang
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Antian Zhang
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Chao Zhang
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Shiqiang Hou
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China
| | - Ning Lin
- Department of Neurosurgery, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China.
| | - Qun Li
- Health Examination Center, The First People's Hospital of Chuzhou, Anhui, China, The Affiliated Chuzhou Hospital of Anhui Medical University, 12 Zhongyou Road, Chuzhou 239001, China.
| |
Collapse
|
4
|
Li B, Zhao R, Jiang X, Liu C, Ma Y, Zhang H. Phytochemical investigation of Jie-Geng-Tang and regulatory role in the TNF-α pathway in mitigating pulmonary fibrosis using UPLC-Q-TOF/MS. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:7251-7262. [PMID: 39729206 DOI: 10.1007/s00210-024-03755-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Jie-Geng-Tang (JGT), composed of Platycodon grandiflorus (Jacq.) A. DC and Glycyrrhiza uralensis Fisch, is widely used in traditional Chinese medicine for its potential effects in preventing pulmonary fibrosis (PF). This study systematically explored the effects of JGT's water and 70% EtOH extracts in bleomycin (BLM)-induced PF models. In vitro, the 70% EtOH extract significantly reversed BLM-induced reductions in cell viability and apoptosis, whereas the water extract had limited impact. In vivo, the EtOH extract markedly reduced fibrosis markers, such as α-SMA and collagen-I, alleviating lung tissue damage and collagen deposition. UPLC-Q-TOF/MS analysis revealed that the EtOH extract contained a higher abundance of flavonoids compared to the water extract. Through network pharmacology analysis of the EtOH extract, four key flavonoids-apigenin, kaempferol, kaempferol 3-glucuronoside, and quercetin-were identified as crucial compounds. These flavonoids were found to reverse BLM-induced cell viability loss, with apigenin showing the most pronounced effect by modulating the TNF-α signaling pathway and inhibiting caspase-3 activation. Apigenin, as a primary active component derived from JGT, holds significant potential as a preventive agent against pulmonary fibrosis.
Collapse
Affiliation(s)
- Bingxin Li
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China
| | - Ruining Zhao
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China
| | - Xiaojie Jiang
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China
| | - Chang Liu
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China
| | - Yun Ma
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China
| | - Haijun Zhang
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China.
| |
Collapse
|
5
|
Lan JT, Liu CX, Zhang J, Ma YH, Xiao G, Wang SM, Yang G, Cui GB, Hu YC. Predicting the risk grades of thymic epithelial tumours using T1 mapping and diffusion-weighted MRI. Br J Radiol 2025; 98:947-955. [PMID: 40085784 DOI: 10.1093/bjr/tqaf060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 12/25/2024] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVE To explore the value of native T1 and apparent diffusion coefficients (ADCs) for predicting subtypes and stages of thymic epithelial tumours (TETs). METHODS Sixty-seven patients with TETs confirmed by pathological analysis were retrospectively enrolled. The mean native T1 (T1mean), global native T1 (ADCtotal), relative minimum ADC (ADCmin), and global ADC (ADCtotal) values of the tumour were measured and compared for differences among low-risk thymoma (LRT), high-risk thymoma (HRT), and thymic carcinoma (TC). The differentiating efficacy was determined using receiver operating characteristic curve analysis. RESULTS The native T1 values in thymoma or early stage were significantly higher than those in TC (P < 0.05/3) or advanced stage of TETs (P < 0.01). The ADC values in LRT or early stage were significantly higher than those in TC (P < 0.05/3) or advanced stage of TETs (P < 0.001). For differentiating the thymoma or HRT from TC, a combination of native T1 and ADC achieved the highest efficacy with an area under the curve (AUC) of 0.891 and 0.851, respectively. For determining the TET stage, ADCmin achieved a relatively high diagnostic efficacy with an AUC of 0.933, and a combination of native T1 and ADC obtained an AUC of 0.876. CONCLUSION The combination of native T1 and ADC values could be helpful in clinical practice regarding evaluating TETs before treatment. ADVANCES IN KNOWLEDGE The combination of T1 mapping and diffusion-weighted imaging (DWI) can improve diagnostic accuracy and help guide clinical practice in providing the best individual treatment for TET patients.
Collapse
Affiliation(s)
- Jiang-Tao Lan
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, Xi'an, 710038 Shaanxi, P.R. China
| | - Chen-Xi Liu
- Department of Radiology, Xi'an Traditional Chinese Medicine Hospital, Xi'an, 710038 Shaanxi, P.R. China
| | - Jie Zhang
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, Xi'an, 710038 Shaanxi, P.R. China
| | - Yu-Hui Ma
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, Xi'an, 710038 Shaanxi, P.R. China
| | - Gang Xiao
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, Xi'an, 710038 Shaanxi, P.R. China
| | - Shu-Mei Wang
- Department of Pathology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038 Shaanxi, P.R. China
| | - Guang Yang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038 Shaanxi, P.R. China
| | - Guang-Bin Cui
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, Xi'an, 710038 Shaanxi, P.R. China
| | - Yu-Chuan Hu
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, Xi'an, 710038 Shaanxi, P.R. China
| |
Collapse
|
6
|
Dinkel Z, Baker A, Akins A, King KL, Harrington M, Kunkel D, Gao Z, Ye T, Dunn H. Collagen architecture in triple negative breast cancer. PLoS One 2025; 20:e0324655. [PMID: 40397870 DOI: 10.1371/journal.pone.0324655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/28/2025] [Indexed: 05/23/2025] Open
Abstract
This study evaluated collagen properties in TNBC samples collected from different racial groups to determine the presence of variance in matrix architecture. African American (AA) breast cancer patients have a significantly higher mortality rate and nearly a three-fold increased prevalence of triple negative breast cancer (TNBC) when compared to Caucasian (C) patients. The extracellular matrix region surrounding tumors contains abundant collagen fibers, and these fibers undergo remodeling throughout cancer progression, promote metastasis, and impede treatment response. High mammographic density, commonly known as dense breast tissue, is hypothesized to be more prevalent in AA women and characterized by increased collagen deposition and associated with more aggressive cancers. The aim of this research was to investigate fibrillar collagen architecture in TNBC samples from AA and C patients using two-photon microscopy with second harmonic generation (SHG), an intrinsic optical signal produced by fibrillar collagen. Twenty tissue regions per tumor sample were randomly selected for SHG microscopy, and image processing was conducted using the Fiji macro TWOMBLI to quantify mesoscopic fibrillar morphological properties and nanoscopic fibrillar properties with the Forward-Backward SHG ratio. Compared to the images from C tumor samples, those from AA samples exhibited a significant increase in parameters including fiber area, total length, and number of endpoints and branchpoints, but had decreased lacunarity. Collagen microstructure, including fibril arrangement and packing density, did not significantly differ between the groups. These results illustrate that the TNBC samples analyzed from AA patients may have macrostructural collagen characteristics associated with aggressive phenotype tumor formation.
Collapse
Affiliation(s)
- Zoë Dinkel
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Adam Baker
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Alannah Akins
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Kylie L King
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Matthew Harrington
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Deborah Kunkel
- School of Mathematical and Statistical Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Zhi Gao
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Tong Ye
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Heather Dunn
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| |
Collapse
|
7
|
Qusairy Z, Rada M. Glycosylation in cancer: mechanisms, diagnostic markers, and therapeutic applications. Mol Cell Biochem 2025:10.1007/s11010-025-05303-1. [PMID: 40389792 DOI: 10.1007/s11010-025-05303-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 05/04/2025] [Indexed: 05/21/2025]
Abstract
Glycosylation, a key post-translational modification, plays a pivotal role in cancer progression by influencing critical processes such as protein folding, immune modulation, and intercellular signaling. Altered glycosylation patterns are increasingly recognized as fundamental drivers of tumorigenesis, contributing to key cancer hallmarks like enhanced tumor migration, metastasis, and immune evasion. These aberrant glycosylation signatures not only offer insights into cancer biology but also serve as valuable diagnostic markers and potential therapeutic targets across a range of malignancies. This review explores the mechanisms underlying glycosylation alterations in cancer. We discuss the molecular basis of these changes, including genetic mutations, epigenetic regulation, and oncogene-driven shifts in glycosylation pathways. Additionally, we highlight recent advancements in glycomics research, with a focus on how these alterations influence tumor progression, angiogenesis, and the tumor microenvironment. Furthermore, the review considers the clinical implications of glycosylation changes, including their role in resistance to anti-cancer therapies and their potential as biomarkers for personalized treatment strategies. By bridging fundamental glycosylation research with clinical applications, this review underscores the promise of glycosylation as both a diagnostic tool and a therapeutic target in oncology, offering new avenues for improved patient stratification and precision medicine.
Collapse
Affiliation(s)
- Zahraa Qusairy
- McGill University Health Center Research Institute, Montreal, QC, H4A 3J1, Canada
| | - Miran Rada
- Medical Laboratory Science, Komar University of Science and Technology, Qularaisi, Sulaimani, Sulaymaniyah, Kurdistan Region, Iraq.
- Komar Cancer Research Program, Komar University of Science and Technology, Qularaisi, Sulaimani, Sulaymaniyah, Kurdistan Region, Iraq.
| |
Collapse
|
8
|
Liu Y, Qian S, Wei J, He J, Li M, Gao X, Cai H, Wang Y, Han Y, Tan T, Yang M. The Expression and Molecular Roles of MAMDC2 in MSS Colorectal Cancer with a High Tumor Stromal Ratio. Biomedicines 2025; 13:1217. [PMID: 40427044 PMCID: PMC12109205 DOI: 10.3390/biomedicines13051217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Revised: 05/02/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Colorectal cancer (CRC) heterogeneity is strongly influenced by molecular subtypes and tumor stroma interactions. The meprin/A5/PTPmu (MAM) domain, a conserved structural motif in transmembrane proteins, remains undercharacterized in CRC pathogenesis. Methods: We analyzed RNA-seq data from TCGA-COAD to evaluate MAM domain gene expression. Immunohistochemistry and Western blotting were conducted to validate the results of the database analysis. Results: Bioinformatics analysis revealed that MAM domain-containing protein 2 (MAMDC2) was enriched in mesenchymal subtype 4 (CMS4) colorectal cancer (p < 0.001). IHC confirmed MAMDC2 overexpression in MSS colorectal cancer with a high tumor stroma ratio (TSR) and peritoneal metastatic lesions (p < 0.01). WB and real-time PCR analyses confirmed that MAMDC2 has a role in regulating epithelial-mesenchymal transition (EMT) development in CRC. Importantly, we identified that cancer cell-derived MAMDC2 promotes MYLK expression in cancer-associated fibroblasts (CAFs) through paracrine signaling. Conclusions: Our findings suggest MAMDC2 may function as a stromal-associated regulator in MSS colorectal cancer with a high tumor stromal ratio (TSR).
Collapse
Affiliation(s)
- Yiling Liu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shengnan Qian
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jia Wei
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianting He
- Department of Pathology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Minghui Li
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaobing Gao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hong Cai
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yiqing Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yue Han
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tianyuan Tan
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Minhui Yang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
9
|
Li M, Meyer L, Meier N, Witte J, Maldacker M, Seredynska A, Schueler J, Schilling O, Föll M. Spatial Proteomics by Parallel Accumulation-Serial Fragmentation Supported MALDI MS/MS Imaging: A First Glance Into Multiplexed and Spatial Peptide Identification. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2025; 39:e10006. [PMID: 39910729 PMCID: PMC11799399 DOI: 10.1002/rcm.10006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/07/2025]
Abstract
RATIONALE In spatial proteomics, matrix-assisted laser desorption/ionization (MALDI) imaging enables rapid and cost-effective peptide measurements. Yet, in situ peptide identification remains challenging. Therefore, this study aims to integrate the trapped ion mobility spectrometry (TIMS)-based parallel accumulation-serial fragmentation (PASEF) into MALDI imaging of tryptic peptides to enable multiplexed MS/MS imaging. METHODS An initial MALDI TIMS MS1 survey measurement was performed, followed by a manual generation of a precursor list containing mass over charge values and ion mobility windows. Inside the dual TIMS system, submitted precursors were trapped, separately eluted by their ion mobility and analyzed in a quadrupole time-of-flight device, thereby enabling multiplexed MALDI MS/MS imaging. Finally, precursors were identified by peptide to spectrum matching. RESULTS This study presents the first multiplexed MALDI TIMS MS/MS imaging (iprm-PASEF) of tryptic peptides. Its applicability was showcased on two histomorphologically distinct tissue specimens in a four-plex and five-plex setup. Precursors were successfully identified by the search engine MASCOT in one single MALDI imaging experiment for each respective tissue. Peptide identifications were corroborated by liquid-chromatography tandem mass spectrometry experiments and fragment colocalization analyses. CONCLUSIONS In this study, we present a novel pipeline, based on iprm-PASEF, that allows the multiplexed and spatial identification of tryptic peptides in MALDI imaging. Hence, it marks a first step towards the integration of MALDI imaging into the emerging field of spatial proteomics.
Collapse
Affiliation(s)
- Mujia Jenny Li
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre FreiburgUniversity of FreiburgFreiburgGermany
- Institute for Pharmaceutical SciencesUniversity of FreiburgFreiburgGermany
| | - Larissa Chiara Meyer
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre FreiburgUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Nadine Meier
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre FreiburgUniversity of FreiburgFreiburgGermany
| | - Jannik Witte
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre FreiburgUniversity of FreiburgFreiburgGermany
| | - Maximilian Maldacker
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre FreiburgUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Adrianna Seredynska
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre FreiburgUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
- German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Julia Schueler
- Therapeutic Area Lead OncologyCharles River Laboratories Germany GmbHFreiburgGermany
| | - Oliver Schilling
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre FreiburgUniversity of FreiburgFreiburgGermany
- German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Melanie Christine Föll
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre FreiburgUniversity of FreiburgFreiburgGermany
- German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
10
|
Tang X, Khan H, Niewola-Staszkowska K, Wuest F, Brindley DN. Inhibition of autotaxin activity with IOA-289 decreases fibrosis in mouse E0771 breast tumors. Int J Cancer 2025. [PMID: 40345856 DOI: 10.1002/ijc.35471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025]
Abstract
Tumor-associated fibrosis contributes to an immunosuppressive microenvironment that hinders effective anti-tumor immune responses. This study investigates the potential of IOA-289, a novel autotaxin (ATX) inhibitor, which blocks lysophosphatidate (LPA) production and signaling, in modulating fibrosis in breast tumors. Bioinformatic analysis of human breast tumors revealed a strong correlation between levels of LPA1,-4 receptors and extracellular matrix (ECM) genes. Interaction of ECM molecules and integrin β1/CD44 between myofibroblasts and other cell types had the highest contribution to cell-cell communication. We showed that LPA induced α-smooth muscle actin mRNA in mouse mammary fibroblasts and increased expressions of collagen type-I α1 chain (COL1A1) and lamininγ1. IOA-289 decreased the expressions of COL1A1, fibronectin-1, and transforming growth factor β1 (TGFβ1) in E0771 breast tumors in mice. Masson's trichrome staining revealed a marked decrease in collagen deposition within breast tumors of IOA-289-treated mice. Decreased tumor fibrosis aligns with previous findings that IOA-289 enhanced the infiltration of CD8+ cytotoxic T cells and decreased fibrotic factors including leukemia inhibitory factor and transforming growth factor-beta1 in tumors. We also demonstrated that E0771 cells express negligible ATX and LPA receptors. Therefore, ATX inhibition did not affect cancer cells directly in our model. These results underscore the potential of ATX inhibitors in reprogramming the tumor microenvironment to favor anti-tumor immunity and attenuate fibrosis. ATX inhibitors are in clinical trials for treating idiopathic pulmonary fibrosis and pancreatic cancer. Our results support the development of ATX inhibitors as a strategy for improving the treatment of breast cancer and other diseases involving fibrosis.
Collapse
Affiliation(s)
- Xiaoyun Tang
- Cancer Research Institute of Northern Alberta, Edmonton, Alberta, Canada
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Humayara Khan
- Cancer Research Institute of Northern Alberta, Edmonton, Alberta, Canada
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Frank Wuest
- Cancer Research Institute of Northern Alberta, Edmonton, Alberta, Canada
- Department of Oncology, Division of Oncologic Imaging, University of Alberta, Edmonton, Alberta, Canada
| | - David N Brindley
- Cancer Research Institute of Northern Alberta, Edmonton, Alberta, Canada
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
11
|
Wimmers DG, Huebner K, Dale T, Papargyriou A, Reichert M, Hartmann A, Schneider-Stock R. A floating collagen matrix triggers ring formation and stemness characteristics in human colorectal cancer organoids. Pathol Res Pract 2025; 269:155890. [PMID: 40073643 DOI: 10.1016/j.prp.2025.155890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/06/2025] [Accepted: 03/01/2025] [Indexed: 03/14/2025]
Abstract
Intestinal organoids reflect the 3D structure and function of their original tissues. Organoid are typically cultured in Matrigel, an extracellular matrix (ECM) mimicking the basement membrane, which is suitable for epithelial cells but does not accurately mimic the tumour microenvironment of colorectal cancer (CRC). The ECM and particularly collagen type I is crucial for CRC progression and invasiveness. Given that efforts to examine CRC organoid invasion in a more physiologically relevant ECM have been limited, we used a floating collagen type I matrix (FC) to study organoid invasion in three patient-derived CRC organoid lines. In FC gel, organoids contract, align, and fuse into macroscopic ring structures, initiating minor branch formation and invasion fronts, phenomena unique for the collagen ECM and otherwise not observed in Matrigel-grown CRC organoids. In contrast to Matrigel, FC organoids showed basal extrusion with improper actin localization, but without change in the organoid polarity. Moreover, small clusters of vital invading cells were observed. Gene expression analysis revealed that the organoids cultured in a FC matrix presented more epithelial and stem cell-like characteristics. This novel technique of cultivating CRC organoids in a FC matrix represents an in-vitro model for studying cancer organization and matrix remodelling with increased organoid stemness potential.
Collapse
Affiliation(s)
- Daniel Gerhard Wimmers
- Experimental Tumorpathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany; Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Kerstin Huebner
- Experimental Tumorpathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany; Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Trevor Dale
- Cardiff University, European Cancer Stem Cell Research Institute (ECSCRI), School of Bioscience, Cardiff, United Kingdom
| | - Aristeidis Papargyriou
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Technical University of Munich, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Munich, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Maximilian Reichert
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Technical University of Munich, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Munich, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, 85764, Germany; Center for Organoid Systems, Technical University of Munich, Garching, Germany; Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich, Garching, Germany; German Center for Translational Cancer Research (DKTK), Munich, Germany; Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Arndt Hartmann
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany; Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Regine Schneider-Stock
- Experimental Tumorpathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany; Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany; Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| |
Collapse
|
12
|
Gostomczyk K, Drozd M, Marsool Marsool MD, Pandey A, Tugas K, Chacon J, Tayyab H, Ullah A, Borowczak J, Szylberg Ł. Biomarkers for the detection of circulating tumor cells. Exp Cell Res 2025; 448:114555. [PMID: 40228709 DOI: 10.1016/j.yexcr.2025.114555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Circulating tumor cells (CTCs) have emerged as a key biomarker in cancer detection and prognosis, and their molecular profiling is gaining importance in precision oncology. Liquid biopsies, which allow the extraction of CTCs, circulating tumor DNA (ctDNA) or cell-free DNA (cfDNA), have measurable advantages over traditional tissue biopsies, especially when molecular material is difficult to obtain. However, this method is not without limitations. Difficulties in differentiating between primary and metastatic lesions, uncertain predictive values and the complexity of the biomarkers used can prove challenging. Recently, high cell heterogeneity has been identified as the main obstacle to achieving high diagnostic accuracy. Because not all cells undergo epithelial-mesenchymal transition (EMT) at the same time, there is a large population of hybrid CTCs that express both epithelial and mesenchymal markers. Since traditional diagnostic tools primarily detect epithelial markers, they are often unable to detect cells with a hybrid phenotype; therefore, additional markers may be required to avoid false negatives. In this review, we summarize recent reports on emerging CTCs markers, with particular emphasis on their use in cancer diagnosis. Most of them, including vimentin, TWIST1, SNAI1, ZEB1, cadherins, CD44, TGM2, PD-L1 and GATA, hold promise for the detection of CTCs, but are also implicated in cancer progression, metastasis, and therapeutic resistance. Therefore, understanding the nature and drivers of epithelial-mesenchymal plasticity (EMP) is critical to advancing our knowledge in this field.
Collapse
Affiliation(s)
- Karol Gostomczyk
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Tumor Pathology and Pathomorphology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland.
| | - Magdalena Drozd
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland
| | | | - Anju Pandey
- Memorial Sloan Kettering Cancer Center, New York, USA
| | | | - Jose Chacon
- American University of Integrative Sciences, Saint Martin, Cole Bay, Barbados
| | | | - Ashraf Ullah
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jędrzej Borowczak
- Department of Clinical Oncology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland
| | - Łukasz Szylberg
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Tumor Pathology and Pathomorphology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland
| |
Collapse
|
13
|
Lindgren M, Ljuslinder I, Jonsson P, Nyström H. Type IV collagen, carcinoembryonic antigen, osteopontin, and hepatocyte growth factor as biomarkers for liver metastatic colorectal cancer. Int J Biol Markers 2025:3936155251329590. [PMID: 40289465 DOI: 10.1177/03936155251329590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
IntroductionDiagnosis and monitoring of metastatic colorectal cancer (mCRC) depend on diagnostic imaging. Circulating carcinoembryonic antigen (CEA) can be analyzed but no optimal, non-invasive biomarker exists. Circulating collagen IV (COL IV) is a promising biomarker in patients with colorectal liver metastases (CLM). This study aimed to evaluate COL IV and other cancer-related and stroma-derived proteins as biomarkers for mCRC.Materials & methodsPlasma COL IV and 10 other proteins were analyzed with ELISA and Luminex multiplex assays.ResultsmCRC patients have elevated levels of circulating COL IV, CEA, interleukin-8 (IL-8), hepatocyte growth factor (HGF), cytokeratin-19 fragments (CYFRA 21-1), osteopontin (OPN), and migration inhibitory factor (MIF) compared to primary CRC (pCRC) patients. COL IV is elevated in mCRC patients compared to healthy individuals. Levels of COL IV, CEA, OPN, CYFRA 21-1, IL-8, and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) were dependent on the metastatic site. OPN, CEA, and HGF are very good at discriminating between mCRC patients and pCRC controls. COL IV is very good at distinguishing between mCRC patients and healthy controls. The combination of OPN + CEA is superior at detecting mCRC than CEA alone. High HGF and COL IV levels correlate to poor prognosis.ConclusionOPN, CEA, and HGF are potential biomarkers for mCRC. COL IV is a potential biomarker for CLM. The combination of OPN with CEA is superior to CEA alone in detecting mCRC. Levels of circulating proteins depend on metastatic localization, implying that a combination of markers is better than single markers in detecting mCRC disease. High levels of COL IV and HGF have potential prognostic value.
Collapse
Affiliation(s)
- Moa Lindgren
- Department of Diagnostics and Intervention/Surgery, Umeå University, SE-901 85, Sweden
| | - Ingrid Ljuslinder
- Department of Diagnostics and Intervention/Oncology, Umeå University, SE-901 87, Sweden
| | - Pär Jonsson
- Department of Chemistry, Umeå University, SE-907 36, Sweden
| | - Hanna Nyström
- Department of Diagnostics and Intervention/Surgery, Umeå University, SE-901 85, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, SE-901 87, Sweden
| |
Collapse
|
14
|
Veliz L, Lambin C, Cooper TT, McCarvell WM, Lajoie GA, Postovit LM, Lagugné-Labarthet F. Emerging SERS and TERS MoS 2 platforms for the characterization of plasma-derived extracellular vesicles. NANOSCALE 2025; 17:9926-9936. [PMID: 40163166 DOI: 10.1039/d4nr04926h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Extracellular vesicles (EVs) play a crucial role in intercellular communication processes. In addition, their biomolecular cargoes such as lipids, proteins, and nucleic acids are useful for identifying potential biomarkers related to different stages of cancer disease. However, the small size and heterogenicity of tumor-related EVs represent a major challenge in properly identifying the content of EVs' cargoes with common characterization protocols. To address these issues, surface-enhanced Raman spectroscopy (SERS) and tip-enhanced Raman spectroscopy (TERS) are powerful alternatives to assign the vibrational fingerprints to the biomolecules contained in cancer EVs, providing high specificity and spatial resolution. Transition metal dichalcogenides are particularly interesting as SERS and TERS substrates due to the high sensitivity of their 2D surface through coulombic and van der Waals interactions when in contact with an analyte or small object such as the charged membranes of EVs. These interactions induce subtle changes in the work function of the flakes which can be measured through drastic changes of optical processes. We investigate the use of MoS2 flakes synthesized by atmospheric pressure chemical vapor deposition as a potential label-free SERS and TERS platform for the identification of plasma EVs. To exemplify this technology, we isolated plasma EV samples from donors with early-stage [FIGO (I/II)] with high-grade serous carcinoma (HGSC) by size exclusion chromatography (SEC). Both surface- and tip-enhanced measurements were conducted individually, enabling the identification of a series of markers from ovarian cancer donors, highlighting the complementarity of SERS and TERS measurements.
Collapse
Affiliation(s)
- Lorena Veliz
- Department of Chemistry, Western University (The University of Western Ontario), 1151 Richmond Street, London, ON, N6A 5B7, Canada.
| | - Cédric Lambin
- Department of Chemistry, Western University (The University of Western Ontario), 1151 Richmond Street, London, ON, N6A 5B7, Canada.
| | - Tyler T Cooper
- Department of Biochemistry, Western University, 1151 Richmond Street, London, ON, N6A 5B7, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, 99 University Ave, Kingston, ON K7L 3N6, Canada
| | - W Michael McCarvell
- Department of Chemistry, Western University (The University of Western Ontario), 1151 Richmond Street, London, ON, N6A 5B7, Canada.
| | - Gilles A Lajoie
- Department of Biochemistry, Western University, 1151 Richmond Street, London, ON, N6A 5B7, Canada
| | - Lynne-Marie Postovit
- Department of Biomedical and Molecular Sciences, Queen's University, 99 University Ave, Kingston, ON K7L 3N6, Canada
| | - François Lagugné-Labarthet
- Department of Chemistry, Western University (The University of Western Ontario), 1151 Richmond Street, London, ON, N6A 5B7, Canada.
| |
Collapse
|
15
|
Hsieh HC, Han Q, Brenes D, Bishop KW, Wang R, Wang Y, Poudel C, Glaser AK, Freedman BS, Vaughan JC, Allbritton NL, Liu JTC. Imaging 3D cell cultures with optical microscopy. Nat Methods 2025:10.1038/s41592-025-02647-w. [PMID: 40247123 DOI: 10.1038/s41592-025-02647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/16/2025] [Indexed: 04/19/2025]
Abstract
Three-dimensional (3D) cell cultures have gained popularity in recent years due to their ability to represent complex tissues or organs more faithfully than conventional two-dimensional (2D) cell culture. This article reviews the application of both 2D and 3D microscopy approaches for monitoring and studying 3D cell cultures. We first summarize the most popular optical microscopy methods that have been used with 3D cell cultures. We then discuss the general advantages and disadvantages of various microscopy techniques for several broad categories of investigation involving 3D cell cultures. Finally, we provide perspectives on key areas of technical need in which there are clear opportunities for innovation. Our goal is to guide microscope engineers and biomedical end users toward optimal imaging methods for specific investigational scenarios and to identify use cases in which additional innovations in high-resolution imaging could be helpful.
Collapse
Affiliation(s)
- Huai-Ching Hsieh
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Qinghua Han
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - David Brenes
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Kevin W Bishop
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Rui Wang
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Chetan Poudel
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Adam K Glaser
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Benjamin S Freedman
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Nephrology, Kidney Research Institute and Institute for Stem Cell and Regenerative Medicine, Seattle, WA, USA
- Plurexa LLC, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan T C Liu
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
16
|
Kato A, Nomura S, Takahashi M, Sukowati EW, Harashima H, Mukai H. Tumor-stromal opening via S. typhimurium VNP20009 administration for complete inhibition of refractory tumor growth with liposomal anticancer drugs. J Control Release 2025; 380:1152-1163. [PMID: 39993636 DOI: 10.1016/j.jconrel.2025.02.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025]
Abstract
Many clinical tumors exhibit a vascular endothelium covered by mural cells and stroma with abundant collagen fibers, which greatly inhibit the penetration of nanoparticle drug delivery systems (DDS) formulations deep into the tumors. We previously found that Salmonella typhimurium VNP20009 attracting attention as live bacterial therapeutics, which is a novel pharmaceutical modality for cancer treatment, can grow within deep tumors with abundant stroma and tight vasculature. Because this finding interestingly indicates that VNP20009 administration disrupts vascular and stromal structures even in refractory tumors, we investigated the possibility that VNP20009 administration improves DDS formulations migrations into tumors in this study. VNP20009 co-administration drastically improved the translocation and diffusion of liposomes deep into the tumors, particularly in stroma-rich xenografted tumors, indicating its tumor stromal opening ability. Furthermore, this approach can completely inhibit tumors in various refractory tumor models, including pancreatic cancers, using liposomal doxorubicin (Doxil®) and liposomal irinotecan (Onivyde®). Notably, this remarkable anticancer effect is not simply attributed to the therapeutic effects of liposomal anticancer drugs and VNP20009, but it involves an additional effect, improving the intratumor pharmacokinetics of liposomal anticancer drugs following VNP20009 co-administration. The unique tumor stromal opening ability of VNP20009 demonstrated in this study is a promising strategy for resolving the major challenges faced by tumor DDS.
Collapse
Affiliation(s)
- Akari Kato
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Hokkaido 060-0812, Japan
| | - Shoko Nomura
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Department of Pharmaceutical Informatics, Graduate School of Biomedical Science, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, Nagasaki 852-8588, Japan
| | - Maiko Takahashi
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Erike Widyasari Sukowati
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Hokkaido 060-0812, Japan
| | - Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Department of Pharmaceutical Informatics, Graduate School of Biomedical Science, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, Nagasaki 852-8588, Japan.
| |
Collapse
|
17
|
Vakhrusheva O, Zhao F, Markowitsch SD, Slade KS, Brandt MP, Tsaur I, Cinatl J, Michaelis M, Efferth T, Blaheta RA, Haferkamp A, Juengel E. Artesunate Inhibits Metastatic Potential in Cisplatin-Resistant Bladder Cancer Cells by Altering Integrins. Cells 2025; 14:570. [PMID: 40277897 PMCID: PMC12026051 DOI: 10.3390/cells14080570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/28/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
The survival of patients with locally advanced and metastatic bladder cancer (BCa) is persistently low. Hence, new treatment options are urgently needed. Artesunate (ART) a derivative of artemisinin, used in Traditional Chinese Medicine, shows anti-tumor activity extending over a broad spectrum of human cancers. As we have previously shown, ART inhibits growth in cisplatin-sensitive (parental) and cisplatin-resistant BCa cells. However, how ART acts on the metastatic potential of BCa remained unclear. To clarify, we applied ART to parental and cisplatin-resistant RT4, RT112, T24, and TCCSup BCa cell lines. We examined tumor cell adhesion to vascular endothelium and immobilized collagen and evaluated chemotactic activity, migration, and invasive activity of the BCa cells. Adhesion receptors, integrin α and β subtypes, integrin-linked kinase (ILK), and focal adhesion kinase (FAK) were investigated. The functional relevance of integrin expression altered by ART was determined by blocking studies. ART significantly reduced tumor cell adhesion to vascular endothelium and immobilized collagen in parental as well as in cisplatin-resistant BCa cells. Depending on cell type, ART suppressed tumor cell motility and diminished integrin expression (surface and total). Functional blocking of integrins altered by ART reduced cell adhesion and invasion of the BCa cells. Thus, the metastatic potential of parental and cisplatin-resistant BCa cells was significantly inhibited by ART, making it a promising treatment option for patients with advanced or therapy-resistant BCa.
Collapse
Affiliation(s)
- Olesya Vakhrusheva
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | - Fuguang Zhao
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Sascha Dennis Markowitsch
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Kimberly Sue Slade
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Maximilian Peter Brandt
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Igor Tsaur
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | - Jindrich Cinatl
- Interdisciplinary Laboratory for Paediatric Tumour and Virus Research, Dr. Petra Joh Research Institute, 60529 Frankfurt am Main, Germany; (J.C.J.); (M.M.)
| | - Martin Michaelis
- Interdisciplinary Laboratory for Paediatric Tumour and Virus Research, Dr. Petra Joh Research Institute, 60529 Frankfurt am Main, Germany; (J.C.J.); (M.M.)
- School of Natural Sciences, University of Kent, Canterbury CT2 7NJ, UK
| | - Thomas Efferth
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany;
| | - Roman Alexander Blaheta
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Axel Haferkamp
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Eva Juengel
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| |
Collapse
|
18
|
Zhang M, Zhang B. Extracellular matrix stiffness: mechanisms in tumor progression and therapeutic potential in cancer. Exp Hematol Oncol 2025; 14:54. [PMID: 40211368 PMCID: PMC11984264 DOI: 10.1186/s40164-025-00647-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
Tumor microenvironment (TME) is a complex ecosystem composed of both cellular and non-cellular components that surround tumor tissue. The extracellular matrix (ECM) is a key component of the TME, performing multiple essential functions by providing mechanical support, shaping the TME, regulating metabolism and signaling, and modulating immune responses, all of which profoundly influence cell behavior. The quantity and cross-linking status of stromal components are primary determinants of tissue stiffness. During tumor development, ECM stiffness not only serves as a barrier to hinder drug delivery but also promotes cancer progression by inducing mechanical stimulation that activates cell membrane receptors and mechanical sensors. Thus, a comprehensive understanding of how ECM stiffness regulates tumor progression is crucial for identifying potential therapeutic targets for cancer. This review examines the effects of ECM stiffness on tumor progression, encompassing proliferation, migration, metastasis, drug resistance, angiogenesis, epithelial-mesenchymal transition (EMT), immune evasion, stemness, metabolic reprogramming, and genomic stability. Finally, we explore therapeutic strategies that target ECM stiffness and their implications for tumor progression.
Collapse
Affiliation(s)
- Meiling Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China
| | - Bin Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China.
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.
| |
Collapse
|
19
|
Dutour A, Pasello M, Farrow L, Amer MH, Entz-Werlé N, Nathrath M, Scotlandi K, Mittnacht S, Gomez-Mascard A. Microenvironment matters: insights from the FOSTER consortium on microenvironment-driven approaches to osteosarcoma therapy. Cancer Metastasis Rev 2025; 44:44. [PMID: 40210800 PMCID: PMC11985652 DOI: 10.1007/s10555-025-10257-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
Osteosarcoma (OS), a prevalent malignant bone tumor, has seen limited progress in treatment efficacy and patient outcomes over decades. Recent insights into the tumor microenvironment (TME) have revealed its crucial role in tumor progression and therapeutic resistance, particularly in OS. This review offers a comprehensive exploration of the OS microenvironment, meticulously dissecting its crucial components: the mesenchymal stromal TME, the immune microenvironment, hypoxia-induced adaptations, and the impact of the physical microenvironment. By demonstrating how these elements collectively drive tumor proliferation, immune evasion, and invasion, this review explores the intricate molecular and cellular dynamics at play. Furthermore, innovative approaches targeting the OS microenvironment, such as immunotherapies, are presented. This review highlights the importance of the TME in OS progression and its potential as a source of novel therapeutic strategies, offering new hope for improved patient outcomes.
Collapse
Affiliation(s)
- Aurelie Dutour
- Childhood Cancer & Cell Death Team, Centre de Recherche en Cancérologie de Lyon (CRCL), Centre Léon Bérard, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, 69008, Lyon, France
| | - Michela Pasello
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Luke Farrow
- University College London Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley St, London, WC1E 6DD, UK
| | - Mahetab H Amer
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Natacha Entz-Werlé
- Pediatric Onco-Hematology Unit, University Hospitals of Strasbourg, Strasbourg, France
- Translational, Transversal and Therapeutic Oncology Team, Laboratory of Bioimaging and Pathologies, Faculty of Pharmacy, CNRS UMR 7021, Illkirch, France
| | - Michaela Nathrath
- Department of Pediatric Hemato-Oncology, Psychosomatics and Systemic Diseases, Children's Hospital Kassel, Kassel, Germany
- Department of Pediatrics, Children'S Cancer Research Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sibylle Mittnacht
- University College London Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley St, London, WC1E 6DD, UK
| | - Anne Gomez-Mascard
- Department of Pathology, CHU, IUCT-Oncopole, University of Toulouse, Eq19. ONCOSARC CRCT, UMR 1037 Inserm/UT3, ERL 5294 CNRS, 1 Avenue Irène Joliot-Curie, 31059, Toulouse Cedex 9, France.
| |
Collapse
|
20
|
Kong X, Xie X, Wu J, Wang X, Zhang W, Wang S, Abbasova DV, Fang Y, Jiang H, Gao J, Wang J. Combating cancer immunotherapy resistance: a nano-medicine perspective. Cancer Commun (Lond) 2025. [PMID: 40207650 DOI: 10.1002/cac2.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 03/24/2025] [Accepted: 03/30/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer immunotherapy offers renewed hope for treating this disease. However, cancer cells possess inherent mechanisms that enable them to circumvent each stage of the immune cycle, thereby evading anti-cancer immunity and leading to resistance. Various functionalized nanoparticles (NPs), modified with cationic lipids, pH-sensitive compounds, or photosensitizers, exhibit unique physicochemical properties that facilitate the targeted delivery of therapeutic agents to cancer cells or the tumor microenvironment (TME). These NPs are engineered to modify immune activity. The crucial signal transduction pathways and mechanisms by which functionalized NPs counteract immunotherapy resistance are outlined, including enhancing antigen presentation, boosting the activation and infiltration of tumor-specific immune cells, inducing immunogenic cell death, and counteracting immunosuppressive conditions in the TME. Additionally, this review summarizes current clinical trials involving NP-based immunotherapy. Ultimately, it highlights the potential of nanotechnology to advance cancer immunotherapy.
Collapse
Affiliation(s)
- Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, P. R. China
| | - Xintong Xie
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, P. R. China
| | - Juan Wu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Wenxiang Zhang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Shuowen Wang
- Department of Skin and Breast Tumor, University Clinical Hospital No. 4 affiliated with the First Moscow State Medical University named after I.M. Sechenov, Moscow, Russia Federation
| | - Daria Valerievna Abbasova
- Department of Skin and Breast Tumor, University Clinical Hospital No. 4 affiliated with the First Moscow State Medical University named after I.M. Sechenov, Moscow, Russia Federation
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Hongnan Jiang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, P. R. China
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, P. R. China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
21
|
Hu G, Niu W, Ge J, Xuan J, Liu Y, Li M, Shen H, Ma S, Li Y, Li Q. Identification of thyroid cancer biomarkers using WGCNA and machine learning. Eur J Med Res 2025; 30:244. [PMID: 40186253 PMCID: PMC11971869 DOI: 10.1186/s40001-025-02466-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/17/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE The incidence of thyroid cancer (TC) is increasing in China, largely due to overdiagnosis from widespread screening and improved ultrasound technology. Identifying precise TC biomarkers is crucial for accurate diagnosis and effective treatment. METHODS TC patient data were obtained from TCGA. DEGs were analyzed using DESeq2, and WGCNA identified gene modules associated with TC. Machine learning algorithms (XGBoost, LASSO, RF) identified key biomarkers, with ROC and AUC > 0.95 indicating strong diagnostic performance. Immune cell infiltration and biomarker correlation were analyzed using CIBERSORT. RESULTS Four key genes (P4HA2, TFF3, RPS6KA5, EYA1) were found as potential biomarkers. High P4HA2 expression was associated with suppressed anti-tumor immune responses and promoted disease progression. In vitro studies showed that P4HA2 upregulation increased TC cell growth and migration, while its suppression reduced these activities. CONCLUSION Through bioinformatics and experimental validation, we identified P4HA2 as a key potential thyroid cancer biomarker. This finding provides new molecular targets for diagnosis and treatment. P4HA2 has the potential to be a diagnostic or therapeutic target, which could have significant implications for improving clinical outcomes in thyroid cancer patients.
Collapse
Affiliation(s)
- Gaofeng Hu
- Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wenyuan Niu
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jiaming Ge
- Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Jie Xuan
- Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yanyang Liu
- Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Mengjia Li
- Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Huize Shen
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shang Ma
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Yuanqiang Li
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Qinglin Li
- Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
| |
Collapse
|
22
|
Cao J, Zhou C, Mao H, Zhang X. Leveraging machine learning and bioinformatics to identify diagnostic biomarkers connected to hypoxia-related genes in preeclampsia. Comput Methods Biomech Biomed Engin 2025:1-19. [PMID: 40181664 DOI: 10.1080/10255842.2025.2484572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/26/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
PE is a serious form of pregnancy-related hypertension. Hypoxia can induce cellular dysfunction, adversely affecting both the infant and the mother. This study aims to investigate the relationship between HRGs and the diagnosis of PE, seeking to enhance our understanding of potential molecular mechanisms and offer new perspectives for the detection and treatment of the condition. A WGCNA network was established to identify key genes significantly associated with traits of PE. LASSO, SVM-RFE, and RF were utilized to identify feature genes. Calibration curves and DCA were employed to assess the diagnostic performance of the comprehensive nomogram. Consensus clustering was applied to identify subtypes of PE. GSEA and the construction of a ceRNA network were used to explore the potential biological functions and regulatory mechanisms of the identified feature genes. Furthermore, ssGSEA was conducted to investigate the immune landscape associated with PE. We successfully identified three potential diagnostic biomarkers for PE: P4HA1, NDRG1, and BHLHE40. Furthermore, the nomogram exhibited strong diagnostic performance. In patients with PE, the abundance of pro-inflammatory immune cells was significantly elevated, reflecting characteristics of high infiltration. The levels of immune cells infiltration were significantly correlated with the expression of the identified feature genes. Notably, these feature genes may be closely linked to mitochondrial-related biological functions. In conclusion, our findings enhance the understanding of the pathological mechanisms underlying PE and open innovative avenues for the diagnosis and treatment of PE.
Collapse
Affiliation(s)
- Jianfang Cao
- Department of Prenatal and Postnatal Care, Jinhua Maternal and Child Health Hospital, Jinhua, Zhejiang, China
| | - Chaofen Zhou
- Department of Prenatal and Postnatal Care, Jinhua Maternal and Child Health Hospital, Jinhua, Zhejiang, China
| | - Heshui Mao
- Department of Burns and Plastic Surgery, Jinhua Central Hospital, Jinhua, Zhejiang, China
| | - Xia Zhang
- Department of Oncology, Jinhua Central Hospital, Jinhua, Zhejiang, China
| |
Collapse
|
23
|
Cerce DDP, Cândido GC, de Almeida MB, Silva JL, Dias FGG, Rodrigues MA. Exploring the relationship between histological grading, fibrillar collagen alterations and nuclear phenotypes in canine mammary carcinomas. J Comp Pathol 2025; 218:1-11. [PMID: 40022855 DOI: 10.1016/j.jcpa.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/05/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025]
Abstract
We evaluated collagen deposition and nuclear phenotypes in non-inflammatory, metastasis-free canine mammary carcinomas at the time of tumour resection. A retrospective cohort analysis was conducted on 68 female dogs diagnosed with mammary carcinomas between January 2013 and December 2021, excluding cases of mammary sarcoma, carcinosarcoma, inflammatory mammary cancer and metastases. Tumours were classified into histological subtypes using the Peña grading system and assigned grades accordingly. Software-assisted video image analysis was utilized to quantitatively assess collagen deposition, organization and nuclear phenotypes. Histological grading was performed by three independent examiners to ensure reproducibility and minimize observer bias. Significant differences in collagen deposition and nuclear phenotypes were observed across histological grades. Grade III carcinomas had significantly greater collagen deposition, both within the tumour core and at the tumour periphery, compared with grades I and II. Collagen organization was markedly increased in grade III carcinomas. Nuclear phenotype analysis revealed distinct features that allowed clear differentiation between grade II and grade III tumours. Software-assisted image analysis effectively identified distinct patterns of collagen deposition, organization and nuclear phenotypes associated with canine mammary carcinomas of various grades, providing important information about tumour biology.
Collapse
Affiliation(s)
- Danielle D P Cerce
- Franca University, UNIFRAN, Avenida Dr. Armando de Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | - Gabriela C Cândido
- Franca University, UNIFRAN, Avenida Dr. Armando de Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | - Maysa B de Almeida
- Franca University, UNIFRAN, Avenida Dr. Armando de Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | - Jhuan L Silva
- Franca University, UNIFRAN, Avenida Dr. Armando de Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | - Fernanda G G Dias
- Franca University, UNIFRAN, Avenida Dr. Armando de Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil
| | - Marcela A Rodrigues
- Franca University, UNIFRAN, Avenida Dr. Armando de Salles Oliveira, 201, Parque Universitário, Franca, São Paulo, 14404-600, Brazil.
| |
Collapse
|
24
|
Taylor H, Spruill L, Jensen-Smith H, Rujchanarong D, Hulahan T, Ivey A, Siougiannis A, Bethard JR, Ball LE, Sandusky GE, Hollingsworth MA, Barth JL, Mehta AS, Drake RR, Marks JR, Nakshatri H, Ford M, Angel PM. Spatial localization of collagen hydroxylated proline site variation as an ancestral trait in the breast cancer microenvironment. Matrix Biol 2025; 136:71-86. [PMID: 39863086 DOI: 10.1016/j.matbio.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 12/27/2024] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Collagen stroma interactions within the extracellular microenvironment of breast tissue play a significant role in breast cancer, including risk, progression, and outcomes. Hydroxylation of proline (HYP) is a common post-translational modification directly linked to breast cancer survival and progression. Changes in HYP status lead to alterations in epithelial cell signaling, extracellular matrix remodeling, and immune cell recruitment. In the present study, we test the hypothesis that the breast cancer microenvironment presents unique PTMs of collagen, which form bioactive domains at these sites that are associated with spatial histopathological characteristics and influence breast epithelial cell signaling. Mass spectrometry imaging proteomics targeting collagens were paired with comprehensive proteomic methods to identify novel breast cancer-related collagen domains based on spatial localization and regulation in 260 breast tissue samples. As ancestry plays a significant role in breast cancer outcomes, these methods were performed on ancestry diverse breast cancer tissues. Lumpectomies from the Cancer Genome Atlas (TCGA; n=10) reported increased levels of prolyl 4-hydroxylase subunit alpha-3 (P4HA3) accompanied by spatial regulation of fibrillar collagen protein sequences. A concise set of triple negative breast cancer lumpectomies (n=10) showed spatial regulation of specific domain sites from collagen alpha-1(I) chain. Tissue microarrays identified proteomic alterations around post-translationally modified collagen sites in healthy breast (n=81) and patient matched normal adjacent (NAT; n=76) and invasive ductal carcinoma (n=83). A collagen alpha-1(I) chain domain encompassing amino acids 506-514 with site-specific proline hydroxylation reported significant alteration between patient matched normal adjacent tissue and invasive breast cancer. Functional testing of domain 506-514 on breast cancer epithelial cells showed proliferation, chemotaxis and cell signaling response dependent on site localization of proline hydroxylation within domain 506-514 variants. These findings support site localized collagen HYP forms novel bioactive domains that are spatially distributed within the breast cancer microenvironment and may play a role in ancestral traits of breast cancer.
Collapse
Affiliation(s)
- Harrison Taylor
- Department of Pharmacology & Immunology, Proteomics Center, Medical University of South Carolina, Charleston, SC, United States
| | - Laura Spruill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Heather Jensen-Smith
- Eppley Institute for Cancer Research & Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Denys Rujchanarong
- Department of Pharmacology & Immunology, Proteomics Center, Medical University of South Carolina, Charleston, SC, United States
| | - Taylor Hulahan
- Department of Pharmacology & Immunology, Proteomics Center, Medical University of South Carolina, Charleston, SC, United States
| | - Ashlyn Ivey
- Department of Pharmacology & Immunology, Proteomics Center, Medical University of South Carolina, Charleston, SC, United States
| | - Alex Siougiannis
- College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jennifer R Bethard
- Department of Pharmacology & Immunology, Proteomics Center, Medical University of South Carolina, Charleston, SC, United States
| | - Lauren E Ball
- Department of Pharmacology & Immunology, Proteomics Center, Medical University of South Carolina, Charleston, SC, United States
| | - George E Sandusky
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, United States
| | - M A Hollingsworth
- Eppley Institute for Cancer Research & Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jeremy L Barth
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Anand S Mehta
- Department of Pharmacology & Immunology, Proteomics Center, Medical University of South Carolina, Charleston, SC, United States
| | - Richard R Drake
- Department of Pharmacology & Immunology, Proteomics Center, Medical University of South Carolina, Charleston, SC, United States
| | - Jeffrey R Marks
- Department of Surgery, Duke Cancer Institute, Duke University, Durham, NC, United States
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, United States
| | - Marvella Ford
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Peggi M Angel
- Department of Pharmacology & Immunology, Proteomics Center, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
25
|
Wang H, Li J, Jing S, Lin P, Qiu Y, Yan X, Yuan J, Tang Z, Li Y, Zhang H, Chen Y, Wang Z, Li H. SOAPy: a Python package to dissect spatial architecture, dynamics, and communication. Genome Biol 2025; 26:80. [PMID: 40158115 PMCID: PMC11954224 DOI: 10.1186/s13059-025-03550-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
Advances in spatial omics enable deeper insights into tissue microenvironments while posing computational challenges. Therefore, we developed SOAPy, a comprehensive tool for analyzing spatial omics data, which offers methods for spatial domain identification, spatial expression tendency, spatiotemporal expression pattern, cellular co-localization, multi-cellular niches, cell-cell communication, and so on. SOAPy can be applied to diverse spatial omics technologies and multiple areas in physiological and pathological contexts, such as tumor biology and developmental biology. Its versatility and robust performance make it a universal platform for spatial omics analysis, providing diverse insights into the dynamics and architecture of tissue microenvironments.
Collapse
Affiliation(s)
- Heqi Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiarong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Siyu Jing
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ping Lin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yiling Qiu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xi Yan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiao Yuan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - ZhiXuan Tang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yujie Chen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhen Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
26
|
Mivehchi H, Eskandari-Yaghbastlo A, Ghazanfarpour M, Ziaei S, Mesgari H, Faghihinia F, Zokaei Ashtiani N, Afjadi MN. Microenvironment-based immunotherapy in oral cancer: a comprehensive review. Med Oncol 2025; 42:140. [PMID: 40153139 DOI: 10.1007/s12032-025-02694-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/19/2025] [Indexed: 03/30/2025]
Abstract
Oral cancer, a prevalent form of head and neck malignancy, accounts for 4% of global cancer cases. The most common type, oral squamous cell carcinoma (OSCC), has a survival rate of about 50%. Even though emerging molecular therapies show promise for managing oral cancer, current treatments like surgery, radiotherapy, and chemotherapy have significant side effects. In addition, the complex tumor microenvironment (TME), involving the extracellular matrix (ECM) and cells like fibroblasts and stromal cells like immune cells, promotes tumor growth and inhibits immune responses, complicating treatment. Nonetheless, immunotherapy is crucial in cancer treatment, especially in oral cancers. Indeed, its effectiveness lies in targeting immune checkpoints such as PD-1 and CTLA-4 inhibitors, as well as monoclonal antibodies like pembrolizumab and cetuximab, adoptive cell transfer methods (including CAR-T cell therapy), cytokine therapy such as IL-2, and tumor vaccines. Thus, these interventions collectively regulate tumor proliferation and metastasis by targeting the TME through autocrine-paracrine signaling pathways. Immunotherapy indeed aims to stimulate the immune system, leveraging both innate and adaptive immunity to counteract cancer cell signals and promote tumor destruction. This review will explore how the TME controls tumor proliferation and metastasis via autocrine-paracrine signaling pathways. It will then detail the effectiveness of immunotherapy in oral cancers, focusing on immune checkpoints, targeted monoclonal antibodies, adoptive cell transfer, cytokine therapy, and tumor vaccines.
Collapse
Affiliation(s)
- Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - SeyedMehdi Ziaei
- Faculty of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hassan Mesgari
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Islamic Azad University, Tehran Branch, Tehran, Iran
| | - Farbod Faghihinia
- School of Dentistry, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
27
|
Kataki AD, Gupta PG, Cheema U, Nisbet A, Wang Y, Kocher HM, Pérez-Mancera PA, Velliou EG. Mapping Tumor-Stroma-ECM Interactions in Spatially Advanced 3D Models of Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16708-16724. [PMID: 40052705 PMCID: PMC11931495 DOI: 10.1021/acsami.5c02296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/21/2025]
Abstract
Bioengineering-based in vitro tumor models are increasingly important as tools for studying disease progression and therapy response for many cancers, including the deadly pancreatic ductal adenocarcinoma (PDAC) that exhibits a tumor/tissue microenvironment of high cellular/biochemical complexity. Therefore, it is crucial for in vitro models to capture that complexity and to enable investigation of the interplay between cancer cells and factors such as extracellular matrix (ECM) proteins or stroma cells. Using polyurethane (PU) scaffolds, we performed a systematic study on how different ECM protein scaffold coatings impact the long-term cell evolution in scaffolds containing only cancer or only stroma cells (activated stellate and endothelial cells). To investigate potential further changes in those biomarkers due to cancer-stroma interactions, we mapped their expression in dual/zonal scaffolds consisting of a cancer core and a stroma periphery, spatially mimicking the fibrotic/desmoplastic reaction in PDAC. In our single scaffolds, we observed that the protein coating affected the cancer cell spatial aggregation, matrix deposition, and biomarker upregulation in a cell-line-dependent manner. In single stroma scaffolds, different levels of fibrosis/desmoplasia in terms of ECM composition/quantity were generated depending on the ECM coating. When studying the evolution of cancer and stroma cells in our dual/zonal model, biomarkers linked to cell aggressiveness/invasiveness were further upregulated by both cancer and stroma cells as compared to single scaffold models. Collectively, our study advances the understanding of how different ECM proteins impact the long-term cell evolution in PU scaffolds. Our findings show that within our bioengineered models, we can stimulate the cells of the PDAC microenvironment to develop different levels of aggressiveness/invasiveness, as well as different levels of fibrosis. Furthermore, we highlight the importance of considering spatial complexity to map cell invasion. Our work contributes to the design of in vitro models with variable, yet biomimetic, tissue-like properties for studying the tumor microenvironment's role in cancer progression.
Collapse
Affiliation(s)
- Anna-Dimitra Kataki
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| | - Priyanka G. Gupta
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
- School
of Life and Health Sciences, Whitelands College, University of Roehampton, London SW15 4JD, U.K.
| | - Umber Cheema
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| | - Andrew Nisbet
- Department
of Medical Physics and Biomedical Engineering, University College London, London WC1E 6BT, U.K.
| | - Yaohe Wang
- Centre
for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Hemant M. Kocher
- Centre
for Tumour Biology and Experimental Cancer Medicine, Barts Cancer
Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Pedro A. Pérez-Mancera
- Department
of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
| | - Eirini G. Velliou
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| |
Collapse
|
28
|
Sinha AP, Jurrius P, van Schelt AS, Darwish O, Shifa B, Annio G, Peterson Z, Jeffery H, Welsh K, Metafa A, Spence J, Kothari A, Hamed H, Bitsakou G, Karydakis V, Thorat M, Shaari E, Sever A, Rigg A, Ng T, Pinder S, Sinkus R, Purushotham A. Tumor Biomechanics Quantified Using MR Elastography to Predict Response to Neoadjuvant Chemotherapy in Individuals with Breast Cancer. Radiol Imaging Cancer 2025; 7:e240138. [PMID: 39950962 PMCID: PMC11966563 DOI: 10.1148/rycan.240138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 11/13/2024] [Accepted: 12/13/2024] [Indexed: 04/05/2025]
Abstract
Purpose To evaluate the ability of MR elastography (MRE) to noninvasively quantify tissue biomechanics and determine the added diagnostic value of biomechanics for predicting response throughout neoadjuvant chemotherapy (NAC). Materials and Methods In this prospective study (between September 2020 and August 2023; registration no. NCT03238144), participants with breast cancer scheduled to undergo NAC underwent five MRE scans at different time points alongside clinical dynamic contrast-enhanced MRI (DCE MRI). Regions of interest were drawn over the tumor region for the first two scans, while for the post-NAC scan, the initial pre-NAC tumor footprint was used. Biomechanics, specifically tumor stiffness and phase angle within these regions of interest, were quantified as well as the corresponding ratios relative to before NAC (tumor-stiffness ratio and phase-angle ratio, respectively). Postsurgical pathologic analysis was used to determine complete and partial responders. Furthermore, a repeatability analysis was performed for 18 participants. Results Datasets of 41 female participants (mean age, 47 years ± 12.5 [SD]) were included in this analysis. The tumor-stiffness ratio following NAC decreased significantly for complete responders and increased for partial responders (0.76 ± 0.16 and 1.14 ± 0.24, respectively; P < .001). The phase-angle ratio after the first cycle of the first NAC regimen compared with before NAC predicted pathologic response (1.23 ± 0.31 vs 0.91 ± 0.34; P < .001). Combining the tumor stiffness ratio with DCE MRI improved specificity compared with DCE MRI alone (96% vs 44%) while maintaining the high sensitivity of DCE MRI (94%). Repeatability analysis showed excellent agreement for elasticity (repeatability coefficient, 8.3%) and phase angle (repeatability coefficient, 5%). Conclusion MRE-derived phase-angle ratio and tumor stiffness ratio were associated with pathologic complete response in participants with breast cancer undergoing NAC, and a combined DCE MRI plus MRE approach significantly enhanced specificity for identification of complete responders after NAC, while maintaining high sensitivity. Keywords: Breast Cancer, MR Elastography, Neoadjuvant Chemotherapy, Dynamic Contrast-enhanced MRI Supplemental material is available for this article. Clinical trials registration no. NCT03238144 Published under a CC BY 4.0 license.
Collapse
Affiliation(s)
- Aaditya P. Sinha
- School of Cancer and Pharmaceutical Sciences,
King’s College London, London, United Kingdom
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Patriek Jurrius
- School of Cancer and Pharmaceutical Sciences,
King’s College London, London, United Kingdom
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Anne-Sophie van Schelt
- School of Biomedical Engineering and Imaging Sciences,
King’s College London, London, United Kingdom
| | - Omar Darwish
- School of Biomedical Engineering and Imaging Sciences,
King’s College London, London, United Kingdom
| | - Belul Shifa
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Giacomo Annio
- LVTS, Inserm U1148, University Paris Diderot, Paris,
France
| | - Zhane Peterson
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Hannah Jeffery
- School of Cancer and Pharmaceutical Sciences,
King’s College London, London, United Kingdom
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Karen Welsh
- School of Biomedical Engineering and Imaging Sciences,
King’s College London, London, United Kingdom
- Department of Radiology, Guy’s and St Thomas NHS
Foundation Trust, London, United Kingdom
| | - Anna Metafa
- Breast Unit, King’s College Hospital NHS
Foundation Trust, London, United Kingdom
| | - John Spence
- Department of Radiology, Guy’s and St Thomas NHS
Foundation Trust, London, United Kingdom
| | - Ashutosh Kothari
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Hisham Hamed
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Georgina Bitsakou
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Vasileios Karydakis
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Mangesh Thorat
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Elina Shaari
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Ali Sever
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Anne Rigg
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Tony Ng
- School of Cancer and Pharmaceutical Sciences,
King’s College London, London, United Kingdom
| | - Sarah Pinder
- School of Cancer and Pharmaceutical Sciences,
King’s College London, London, United Kingdom
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| | - Ralph Sinkus
- School of Biomedical Engineering and Imaging Sciences,
King’s College London, London, United Kingdom
- LVTS, Inserm U1148, University Paris Diderot, Paris,
France
| | - Arnie Purushotham
- School of Cancer and Pharmaceutical Sciences,
King’s College London, London, United Kingdom
- Breast Unit, Guy’s and St Thomas NHS Foundation
Trust, Guy’s Hospital, Great Maze Pond, London SE1 9RT, United
Kingdom
| |
Collapse
|
29
|
Lai GY, Lee YC, Weng HJ, Lai KH, Hsiang MC, Hsu KY, Liao CP. Discoidin domain receptor inhibitor DDR1-IN-1 induces autophagy and necroptotic cell death in malignant peripheral nerve sheath tumor. Cell Death Discov 2025; 11:83. [PMID: 40025071 PMCID: PMC11873111 DOI: 10.1038/s41420-025-02367-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/14/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Malignant peripheral nerve sheath tumor (MPNST) is a soft tissue sarcoma commonly associated with the tumor-predisposition disorder neurofibromatosis 1. The extracellular matrix collagens contribute to many fibrotic tumors; however, the role of collagen signaling in MPNST was unclear. This study investigated the effects of blocking the interaction between collagens and their receptors in MPNST. We first analyzed the expressions of collagen family proteins in MPNSTs and found an overall increase compared to neurofibroma. Treatment of DDR1-IN-1, a small molecule inhibitor for the collagen receptor discoidin domain receptor, induced a robust MPNST cell death, highlighting the dependence of MPNST survival on collagen signaling. DDR1-IN-1 induced MPNST cell death by activating autophagy and necroptosis signaling. Treatment of necroptosis inhibitors necrostatin-1 or necrosulfonamide reduced the numbers of DDR1-IN-1-induced necrotic cells and autolysosomes, suggesting that the autophagic process depends on necroptosis activation. Combinations of DDR1-IN-1 with other anti-MPNST agents revealed synergistic activities against MPNST. In summary, this study discovered a critical MPNST death signaling induced by the small molecule DDR1-IN-1, which might shed light on future MPNST therapeutic strategies.
Collapse
Affiliation(s)
- Guan-Yi Lai
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yu-Cheng Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Hao-Jui Weng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Dermatology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, 23561, Taiwan
- Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kuei-Hung Lai
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Min-Chen Hsiang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kai-Yu Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chung-Ping Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Cancer Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, 11696, Taiwan.
| |
Collapse
|
30
|
Li Y, Wu R, Wang H, Zhong M, Qing Y, Lu S, Zhang Z, Ma T, Luo J, Xiao H, Qiu J, Li K. Non-epithelial Circulating Tumor Cells Enhance Disease Progression in High-risk Prostate Cancer through EMT and COL1A1 Expression. Int J Med Sci 2025; 22:1562-1573. [PMID: 40093812 PMCID: PMC11905264 DOI: 10.7150/ijms.107703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction: Circulating tumor cells (CTCs) are important prognostic indicators for malignancies. However, a reliable positive/negative cutoff value of non-epithelial (NE+: hybrid and mesenchymal) CTCs phenotype in prostate cancer (PCa) patients has not been established. Here, we aimed to determine the cutoff value and the prognostic value of NE+ CTCs in high-risk prostate cancer (HRPC) patients after radical prostatectomy (RP). Methods: The cutoff value of NE+ CTCs was established in spiking experiments, and CTCs were detected in 208 HRPC patients using the CanPatrolTM platform. The expression and function of COL1A1 in PCa were examined via qRT-PCR, Western blot, wound healing assay, Transwell assay, and immunohistochemistry (IHC). Results: The cutoff value of NE+ CTCs was determined to be 45% by spiking experiments. In 208 HRPC patients, the NE+ CTCs positive group had higher prostate-specific antigen (PSA) levels, more advanced pathological tumor stage, and lymph node stage (P < 0.001, P = 0.002 and 0.002, respectively). Besides, patients with NE+ CTCs ≥ 45% had a shorter median progression-free survival (PFS) than those with NE+ CTCs < 45% (44.5 vs. 51.0 months, hazard ratio = 3.31, P < 0.05). Moreover, we identified that COL1A1 was associated with a high proportion of NE+ CTCs in HRPC patients via an EMT mechanism. Conclusion: Our findings suggest that NE+ CTCs represent a reliable prognostic indicator for HRPC patients and that targeting COL1A1 may prevent the formation of NE+ CTCs.
Collapse
Affiliation(s)
- Yiyuan Li
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruji Wu
- Department of Urology, SSL Central Hospital of Dongguan City, Dongguan, China
| | - Hua Wang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meinong Zhong
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunhao Qing
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuo Lu
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zixiao Zhang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tan Ma
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jieheng Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hengjun Xiao
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianguang Qiu
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke Li
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
31
|
Kakani P, Dhamdhere SG, Pant D, Joshi R, Mishra S, Pandey A, Notani D, Shukla S. Hypoxia-induced CTCF mediates alternative splicing via coupling chromatin looping and RNA Pol II pause to promote EMT in breast cancer. Cell Rep 2025; 44:115267. [PMID: 39913285 DOI: 10.1016/j.celrep.2025.115267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 12/16/2024] [Accepted: 01/15/2025] [Indexed: 02/28/2025] Open
Abstract
Hypoxia influences the epithelial-mesenchymal transition (EMT) through the remodeling of the chromatin structure, epigenetics, and alternative splicing. Hypoxia drives CCCTC-binding factor (CTCF) induction through hypoxia-inducible factor 1-alpha (HIF1α), which promotes EMT, although the underlying mechanisms remain unclear. We find that hypoxia significantly increases CTCF occupancy at various EMT-related genes. We present a CTCF-mediated intricate mechanism promoting EMT wherein CTCF binding at the collagen type V alpha 1 chain (COL5A1) promoter is crucial for COL5A1 upregulation under hypoxia. Additionally, hypoxia drives exon64A inclusion in a mutually exclusive alternative splicing event of COL5A1exon64 (exon64A/64B). Notably, CTCF mediates COL5A1 promoter-alternatively spliced exon upstream looping that regulates DNA demethylation at distal exon64A. This further regulates the CTCF-mediated RNA polymerase II pause at COL5A1exon64A, leading to its inclusion in promoting the EMT under hypoxia. Genome-wide study indicates the association of gained CTCF occupancy with the alternative splicing of many cancer-related genes, similar to the proposed model. Specifically, disrupting the HIF1α-CTCF-COL5A1exon64A axis through the dCas9-DNMT3A system alleviates the EMT in hypoxic cancer cells and may represent a novel therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Parik Kakani
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India
| | - Shruti Ganesh Dhamdhere
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India
| | - Deepak Pant
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India
| | - Rushikesh Joshi
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India
| | - Sachin Mishra
- National Center for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, Karnataka 560065, India
| | - Anchala Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India
| | - Dimple Notani
- National Center for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, Karnataka 560065, India
| | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India.
| |
Collapse
|
32
|
Chen Y, Ali M, Tayyab MB, Nazir MM, Umar M, Khan S, Ismail DM, Abdel-Maksoud MA, Ebaid H, Alamri A, Almutairi S, Almanaa TN, Kiani BH. The role of Prolyl 3-Hydroxylase 1 (P3H1) in tumor development and prognosis: a pan-cancer analysis with validation in colonic adenocarcinoma. Am J Transl Res 2025; 17:770-790. [PMID: 40092085 PMCID: PMC11909568 DOI: 10.62347/suvc1606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 12/16/2024] [Indexed: 03/19/2025]
Abstract
BACKGROUND Cancer is a multifaceted disease characterized by unregulated cell proliferation, evasion of apoptosis, and metastasis. Recent studies have highlighted the importance of extracellular matrix remodeling and post-translational modifications in tumorigenesis. Prolyl 3-hydroxylase 1 (P3H1), an enzyme involved in collagen hydroxylation, has gained attention for its role in cancer progression. METHODS This study investigates P3H1 expression, prognostic value, and functional relevance across multiple human cancers using a combination of bioinformatic and experimental approaches. RESULTS Using The Cancer Genome Atlas (TCGA) data from TIMER2.0 and UALCAN databases, we observed a significant upregulation of P3H1 mRNA and protein in various cancers. Prognostic analysis using GEPIA2 and KM plotter revealed that high P3H1 expression correlates with poorer overall survival in colon adenocarcinoma (COAD), kidney renal clear cell carcinoma (KIRC), and liver hepatocellular carcinoma (LIHC). Further, genetic and promoter methylation analyses showed low mutation frequencies and reduced methylation of P3H1 in specific cancer types. Functional and pathway enrichment analyses indicated that P3H1 is involved in collagen formation, endoplasmic reticulum activity, and pathways such as ECM-receptor interaction and PI3K-Akt signaling. Validation by enzyme linked immunosorbent assay in COAD patient serum samples demonstrated significantly elevated P3H1 levels compared to healthy controls, with an AUC approaching 1.0 by receiver operating characteristic (ROC) curve analysis. This suggests its potential as a diagnostic biomarker. Additionally, functional experiments were conducted in COAD cells to assess P3H1's role in tumorigenesis. Knockdown of P3H1 in HCT116 cells resulted in a significant reduction in cell proliferation, colony formation, and migratory abilities of these cells. CONCLUSION These findings emphasize P3H1's relevance in COAD, KIRC, and LIHC pathogenesis and possible utility in clinical diagnosis.
Collapse
Affiliation(s)
- Yanqin Chen
- Nanjing Drum Tower HospitalNanjing 210000, Jiangsu, China
| | - Moazzam Ali
- Department of Gastroenterology, Hayatabad Medical Complex PeshawarPeshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Bilal Tayyab
- Institute of Drug Discovery and Development, Zhengzhou UniversityZhengzhou 450001, Henan, China
| | | | - Muhammad Umar
- Department of Neurosurgery, Allied Hospital FaisalabadFaisalabad 37521, Punjab, Pakistan
| | - Salman Khan
- DHQ Teaching Hospital, GMCDikah, Abbottabad 22010, Khyber Pakhtunkhwa, Pakistan
| | | | - Mostafa A Abdel-Maksoud
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh 11451, Saudi Arabia
| | - Hossam Ebaid
- Department of Zoology, College of Science, King Saud UniversityP.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdulaziz Alamri
- Department of Biochemistry, College of Science, King Saud UniversityP.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Saeedah Almutairi
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh 11451, Saudi Arabia
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh 11451, Saudi Arabia
| | - Bushra Hafeez Kiani
- Department of Biology and Biotechnology, Worcester Polytechnic InstituteWorcester, Massachuesetts 01609, USA
| |
Collapse
|
33
|
Zhang J, Wang Y, Liu Y, Chen H, Chai N, Zhao Y, Li Y, Li X, He J, Zhang H. Proteomic profiling reveals biological processes and biomarkers involved in the pathogenesis of occult breast cancer. BMC Cancer 2025; 25:231. [PMID: 39930421 PMCID: PMC11812265 DOI: 10.1186/s12885-025-13657-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Occult breast cancer (OBC) is defined as axillary lymph node metastasis without any evidence of a primary tumor in the breast. Because of the limited number of clinical cases, the clinicopathological features and treatment recommendations of OBC are still controversial. In addition, its natural history is poorly understood and its proteomic signature remains unknown. MATERIALS AND METHODS We compared the clinicopathological features and prognosis of OBC patients and Non-OBC patients from the Surveillance, Epidemiology, and End Results (SEER) database and analyzed the effects of local treatment on the survival outcomes of OBC patients. Additionally, we performed a quantitative proteomic analysis for tissue samples of metastatic lymph nodes from OBC patients (OBC-LN), and paired tissue samples of metastatic lymph nodes (Non-OBC-LN) and primary tumors (Non-OBC-PT) from Non-OBC patients. We identified differentially expressed proteins in different comparable groups using Student's t test. Functional enrichment and protein-protein interaction network analyses were used to interpret the functions and interactions of the differentially expressed proteins in the comparison of OBC-LN vs Non-OBC-LN. Immunohistochemistry was used for the validation of the hub proteins. RESULTS Analysis of data from the SEER database demonstrated that OBC patients had a better prognosis than Non-OBC patients did and that either mastectomy or radiation therapy improved the outcomes of OBC patients. A total of 7208 comparable proteins were successfully quantified. Compared with those of the Non-OBC-LN samples, the OBC-LN protein profiles exhibited an active extracellular matrix and a thoroughly upregulated epithelial-mesenchymal transition phenotype. COL1A1, COL1A2, COL3A1, MMP2 and LUM were overexpressed in the OBC-LN samples and were identified as hub proteins. Immunohistochemical staining demonstrated that the five hub proteins were overexpressed in OBC-LN samples. CONCLUSION Our results provide insights for the clinical management of OBC and the proteomic signature of OBC offers molecular basis for further biological research.
Collapse
Affiliation(s)
- Jingyi Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - Yidi Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - Yu Liu
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - He Chen
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Na Chai
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - Yuting Zhao
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yijun Li
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - Xiaoqian Li
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China
| | - Jianjun He
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China.
| | - Huimin Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, People's Republic of China.
| |
Collapse
|
34
|
Chen X, Miao J, Huang X, Han X, Zheng L, Chen J, Chen L, Li L. Multiphoton imaging-based quantifiable collagen signatures for predicting outcomes in patients with pancreatic ductal adenocarcinoma. Sci Rep 2025; 15:4414. [PMID: 39910233 PMCID: PMC11799447 DOI: 10.1038/s41598-025-88984-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/03/2025] [Indexed: 02/07/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents a clinical challenge due to its poor prognosis and high mortality rate. Here, we aimed to enhance the prognostic prediction of patients with PDAC by studying collagen features in tumor microenvironment using multiphoton microscopy (MPM) combining with image processing technique. We identified eight distinct tumor-associated collagen signatures (TACS1-8) from multiphoton images of PDAC tissues and developed an optical biomarker, TACS-score, based on the TACS1-8 using ridge regression analysis. Additionally, we also extracted 142 microscopic TACS (M-TACS) from second-harmonic generation (SHG) images and constructed a new robust biomarker, M-TACS-score, using the least absolute shrinkage and selection operator (LASSO) regression analysis. Our statistical results demonstrate that as two new optical biomarkers, TACS- and M-TACS-score, are independent prognostic factors and have good discriminatory ability (high AUC) as well as risk stratification (high HR) comparing with traditional clinical model (combining seven clinical risk factors, age, sex, TNM stage, tumor location and differentiation, perineural and lymph-vascular invasion) in predicting overall survival (OS) of patients with PDAC, highlighting their potential prognostic and predictive value. A combination of label-free multiphoton imaging technique and computer-aided image processing method may offer a novel and promising approach for finding new biomarkers to improve prognosis prediction and thereby tailor treatment strategies more effectively.
Collapse
Affiliation(s)
- Xiwen Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Jikui Miao
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Xingxin Huang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Xiahui Han
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Liqin Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Linying Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, China.
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China.
| |
Collapse
|
35
|
Diniz-Filho JF, Silva ACM, Teixeira AL, Sousa BLN, Santos-Oliveira R, Silva GEB, dos Santos CC, Alencar LMR. Penile Cancer: Innovations in Ultrastructural and Vibrational Markers. ACS OMEGA 2025; 10:3449-3461. [PMID: 39926554 PMCID: PMC11800048 DOI: 10.1021/acsomega.4c07293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/11/2025]
Abstract
Penile cancer (PCa) is a disease that manifests predominantly as squamous cell carcinomas (SCCs), which, although rare, represents a significant public health problem, especially in regions with less socioeconomic development. One of the biggest challenges in managing this disease is the difficulty in differentiating tumor subtypes, making accurate diagnosis and treatment challenging. In this context, new characterization techniques are needed to investigate these tumors more completely. Atomic force microscopy (AFM) and Raman spectroscopy (RS) are valuable in this context, providing quantitative and qualitative ultrastructural data and vibrational signatures of the analyzed samples. In this study, AFM and RS techniques were employed to investigate subtypes of penile cancer, including the highly aggressive basaloid subtype, which is closely associated with human papillomavirus (HPV), and the sarcomatoid subtype, comparing them with nontumorous tissues. The AFM results revealed nanoscale changes in the ultrastructural properties of tumor samples, such as increased roughness in tumor tissues, with emphasis on the basaloid type associated with the HPV virus, and reduction in the surface area and volume of tumor tissues at the nanoscale, suggesting deeper tissue infiltration and greater deformability of tumor samples at the nanoscale. RS results detected significant spectral differences between normal and cancerous tissues and between tumor subtypes, particularly in vibrational modes related to proteins and lipids. Principal component analysis (PCA) confirmed a strong discriminative power between control and PCa groups. The data presented here offers new insights into the characteristics of penile tumors that, when integrated with clinical analyses, could improve the understanding of penile cancer behavior, contributing to more accurate diagnostic methods and targeted treatments.
Collapse
Affiliation(s)
- Joel Félix
Silva Diniz-Filho
- Biophysics
and Nanosystems Laboratory, Department of Physics, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Ana Caroline Muniz Silva
- Biophysics
and Nanosystems Laboratory, Department of Physics, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Antônio
Augusto Lima Teixeira
- Immunofluorescence
and Electron Microscopy Laboratory (LIME/HUUFMA), Department of Medicine, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Bruna Larissa Nolêto Sousa
- Immunofluorescence
and Electron Microscopy Laboratory (LIME/HUUFMA), Department of Medicine, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Ralph Santos-Oliveira
- Brazilian
Nuclear Energy Commission, Institute of
Nuclear Engineering, Laboratory of Nanoradiopharmacy and Synthesis
of New Radiopharmaceuticals, Rio de Janeiro 21941906, Brazil
- State
University of Rio de Janeiro, Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Rio de Janeiro 21941906, Brazil
| | - Gyl Eanes Barros Silva
- Immunofluorescence
and Electron Microscopy Laboratory (LIME/HUUFMA), Department of Medicine, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Clenilton Costa dos Santos
- Biophysics
and Nanosystems Laboratory, Department of Physics, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Luciana Magalhães Rebelo Alencar
- Biophysics
and Nanosystems Laboratory, Department of Physics, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| |
Collapse
|
36
|
Ghannam SF, Rutland CS, Allegrucci C, Mather ML, Alsaleem M, Bateman‐Price TD, Patke R, Ball G, Mongan NP, Rakha E. Geometric characteristics of stromal collagen fibres in breast cancer using differential interference contrast microscopy. J Microsc 2025; 297:135-152. [PMID: 39359124 PMCID: PMC11733853 DOI: 10.1111/jmi.13361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
Breast cancer (BC) is characterised by a high level of heterogeneity, which is influenced by the interaction of neoplastic cells with the tumour microenvironment. The diagnostic and prognostic role of the tumour stroma in BC remains to be defined. Differential interference contrast (DIC) microscopy is a label-free imaging technique well suited to visualise weak optical phase objects such as cells and tissue. This study aims to compare stromal collagen fibre characteristics between in situ and invasive breast tumours using DIC microscopy and investigate the prognostic value of collagen parameters in BC. A tissue microarray was generated from 200 cases, comprising ductal carcinoma in situ (DCIS; n = 100) and invasive tumours (n = 100) with an extra 50 (25 invasive BC and 25 DCIS) cases for validation was utilised. Two sections per case were used: one stained with haematoxylin and eosin (H&E) stain for histological review and one unstained for examination using DIC microscopy. Collagen fibre parameters including orientation angle, fibre alignment, fibre density, fibre width, fibre length and fibre straightness were measured. Collagen fibre density was higher in the stroma of invasive BC (161.68 ± 11.2 fibre/µm2) compared to DCIS (p < 0.0001). The collagen fibres were thinner (13.78 ± 1.08 µm), straighter (0.96 ± 0.006, on a scale of 0-1), more disorganised (95.07° ± 11.39°) and less aligned (0.20 ± 0.09, on a 0-1 scale) in the invasive BC compared to DCIS (all p < 0.0001). A model considering these features was developed that could distinguish between DCIS and invasive tumours with 94% accuracy. There were strong correlations between fibre characteristics and clinicopathological parameters in both groups. A statistically significant association between fibre characteristics and patients' outcomes (breast cancer specific survival, and recurrence free survival) was observed in the invasive group but not in DCIS. Although invasive BC and DCIS were both associated with stromal reaction, the structural features of collagen fibres were significantly different in the two disease stages. Analysis of the stroma fibre characteristics in the preoperative core biopsy specimen may help to differentiate pure DCIS from those associated with invasion.
Collapse
Affiliation(s)
- Suzan F. Ghannam
- Academic Unit for Translational Medical SciencesSchool of MedicineUniversity of NottinghamNottinghamUK
- Faculty of MedicineDepartment of Histology and Cell BiologySuez Canal UniversityIsmailiaEgypt
- Nottingham Breast Cancer Research CentreBiodiscovery InstituteUniversity of NottinghamNottinghamUK
| | - Catrin Sian Rutland
- Nottingham Breast Cancer Research CentreBiodiscovery InstituteUniversity of NottinghamNottinghamUK
- School of Veterinary Medicine and ScienceUniversity of NottinghamNottinghamUK
| | - Cinzia Allegrucci
- Nottingham Breast Cancer Research CentreBiodiscovery InstituteUniversity of NottinghamNottinghamUK
- School of Veterinary Medicine and ScienceUniversity of NottinghamNottinghamUK
| | - Melissa L. Mather
- Optics and Photonics Research GroupFaculty of EngineeringUniversity of NottinghamNottinghamUK
| | - Mansour Alsaleem
- Department of Applied Medical ScienceApplied CollegeQassim UniversityQassimSaudi Arabia
| | - Thomas D. Bateman‐Price
- Optics and Photonics Research GroupFaculty of EngineeringUniversity of NottinghamNottinghamUK
| | - Rodhan Patke
- School of Veterinary Medicine and ScienceUniversity of NottinghamNottinghamUK
- Biodiscovery InstituteUniversity of NottinghamUniversity ParkNottinghamUK
| | - Graham Ball
- Medical Technology Research CentreAnglia Ruskin UniversityChelmsfordUK
| | - Nigel P. Mongan
- School of Veterinary Medicine and ScienceUniversity of NottinghamNottinghamUK
- Department of PharmacologyWeill Cornell MedicineNew YorkNew YorkUSA
| | - Emad Rakha
- Academic Unit for Translational Medical SciencesSchool of MedicineUniversity of NottinghamNottinghamUK
- Nottingham Breast Cancer Research CentreBiodiscovery InstituteUniversity of NottinghamNottinghamUK
- Cellular Pathology DepartmentNottingham University Hospitals NHS TrustNottinghamUK
- Pathology DepartmentHamad Medical CorporationDohaQatar
| |
Collapse
|
37
|
Wang P, Wang G, Li H, Yuan Y, Chen H, Wang S, Sun Z, Meng F, Li Y, Yang F, Wang J, Chen K, Qiu M. Nicotinamide N-methyltransferase negatively regulates metastasis-promoting property of cancer-associated fibroblasts in lung adenocarcinoma. Cancer Commun (Lond) 2025; 45:110-137. [PMID: 39623600 PMCID: PMC11833673 DOI: 10.1002/cac2.12633] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 02/19/2025] Open
Abstract
BACKGROUND Recurrence and metastasis remain significant challenges in lung adenocarcinoma (LUAD) after radical resection. The mechanisms behind the recurrence and metastasis of LUAD remain elusive, and deregulated cellular metabolism is suspected to play a significant role. This study explores the metabolic and epigenetic regulation mediated by nicotinamide N-methyl transferase (NNMT) in LUAD. METHODS Untargeted metabolomic analyses were performed to detect metabolism irregularities. Single-cell RNA sequencing (RNA-seq) databases and multiplex immunofluorescence analysis were used to identify the location of NNMT within the tumor microenvironment. The biological functions of NNMT were investigated both in vitro and in vivo, with RNA-seq and chromatin immunoprecipitation-PCR providing insights into underlying mechanisms. Finally, single-cell RNA-seq data and immunohistochemistry of primary tumors were analyzed to validate the main findings. RESULTS Untargeted metabolomic analyses revealed metabolic aberrations in amino acids, organic acids, lipids, and nicotinamide pathways, which are linked to metastasis of non-small cell lung cancer. NNMT is a key enzyme in nicotinamide metabolism, and we found the bulk tissue mRNA level of NNMT gene was inversely associated with LUAD metastasis. NNMT was proved to be predominantly expressed in cancer-associated fibroblasts (CAFs) within the stromal regions of LUAD, and a low stromal NNMT expression was identified as a predictor of poor disease-free survival following radical resection of LUAD. The isolation and primary culture of CAFs from LUAD enabled in vitro and in vivo experiments, which confirmed that NNMT negatively regulated the metastasis-promoting properties of CAFs in LUAD. Mechanistically, the downregulation of NNMT led to an increase in intracellular methyl groups by reducing the activity of the methionine cycle, resulting in heightened methylation at H3K4me3. This alteration triggered the upregulation of genes involved in extracellular matrix remodeling in CAFs, including those encoding collagens, integrins, laminins, and matrix metalloproteinases, thereby facilitating cancer cell invasion and metastasis. Reanalysis of single-cell RNA-seq data and immunohistochemistry assays of primary LUAD tissues substantiated NNMT's negative regulation of these genes in CAFs. CONCLUSIONS This study provides novel insights into the metabolic and epigenetic regulatory functions of NNMT in CAFs, expanding the current understanding of LUAD metastasis regulation and suggesting potential avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Peiyu Wang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
- Department of Thoracic SurgeryThe First Affiliated Hospital of Zhengzhou University, ZhengzhouHenanP. R. China
| | - Guangxi Wang
- Department of Pathology, School of Basic Medical SciencesInstitute of Systems Biomedicine, Peking‐Tsinghua Center for Life Sciences, Peking University Health Science CenterBeijingP. R. China
| | - Haoran Li
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Yuyao Yuan
- Department of Pathology, School of Basic Medical SciencesInstitute of Systems Biomedicine, Peking‐Tsinghua Center for Life Sciences, Peking University Health Science CenterBeijingP. R. China
| | - Haiming Chen
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Shaodong Wang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Zewen Sun
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Fanjie Meng
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chao Yang HospitalCapital Medical UniversityBeijingP. R. China
| | - Yun Li
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Fan Yang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Jun Wang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Kezhong Chen
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Mantang Qiu
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| |
Collapse
|
38
|
Fisher M, Duhon BH, Nguyen HTN, Tonniges JR, Wu KC, Ren Y. Quantitative Assessment of Collagen Architecture to Determine Role of Tumor Stroma During Vestibular Schwannoma Progression. Otolaryngol Head Neck Surg 2025; 172:614-622. [PMID: 39506612 PMCID: PMC11773437 DOI: 10.1002/ohn.1018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 09/15/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE The primary objective was to characterize the abundance and architecture of collagen in the extracellular matrix in vestibular schwannoma (VS). The secondary objective was to investigate the association between collagen architecture and tumor size. STUDY DESIGN Retrospective cohort study. SETTING Academic referral center. METHODS Tumor samples were obtained from patients with sporadic VS undergoing microsurgical resection. Histological analyses were performed including picrosirius red (PSR) staining under polarized light. Collagen architecture was quantified using an automated fiber detection software. Second Harmonic Generation (SHG) microscopy and immunofluorescence (IF) were utilized to characterize collagen architecture. RESULTS Eleven tumor specimens were included (mean tumor diameter = 2.80 cm, range 1.5-4.0 cm), and were divided into large (mean diameter = 3.5 ± 0.4 cm) and small (mean tumor diameter = 2.0 ± 0.4 cm) cohorts based on size. The large VS cohort showed significantly higher collagen density (27.65% vs 12.73%, P = .0043), with more thick fibers (mature Type I, 24.54% vs 12.97%, P = .0022) and thin fibers (immature Type I or mature Type III, 23.55% vs 12.27%, P = .026). Tumor volume correlated with greater degree of collagen fiber disorganization (P = .0413, r2 = 0.298). Specifically, collagen type I intensity was significantly higher in large VS compared to small tumors (P < .001) and peripheral nerve (P = .028). CONCLUSION Larger VS exhibit increased collagen abundance in the tumor stroma, and a more disorganized collagen architecture compared to smaller VS and normal peripheral nerve tissue. This finding indicates that collagen organization may play a significant role in extracellular matrix remodeling and the progression of VS.
Collapse
Affiliation(s)
- Melanie Fisher
- Department of Otolaryngology–Head and Neck Surgery, Division of Otology, Neurotology and Cranial Base SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Bailey H. Duhon
- Department of Otolaryngology–Head and Neck Surgery, Division of Otology, Neurotology and Cranial Base SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Han T. N. Nguyen
- Department of Otolaryngology–Head and Neck Surgery, Division of Otology, Neurotology and Cranial Base SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Jeffrey R. Tonniges
- Campus Microscopy and Imaging Facility, Comprehensive Cancer CenterThe Ohio State UniversityColumbusOhioUSA
| | - Kyle C. Wu
- Department of NeurosurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Yin Ren
- Department of Otolaryngology–Head and Neck Surgery, Division of Otology, Neurotology and Cranial Base SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|
39
|
Namakshenas P, Crezee J, Tuynman JB, Tanis PJ, Oei AL, Kok HP. Computational Evaluation of Improved HIPEC Drug Delivery Kinetics via Bevacizumab-Induced Vascular Normalization. Pharmaceutics 2025; 17:155. [PMID: 40006522 PMCID: PMC11859678 DOI: 10.3390/pharmaceutics17020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Oxaliplatin-based hyperthermic intraperitoneal chemotherapy (HIPEC) using the original 30 min protocol has shown limited benefits in patients with peritoneal metastasis of colorectal cancer (PMCRC), likely due to the short duration, which limits drug penetration into tumor nodules. Bevacizumab, an antiangiogenic antibody that modifies the tumor microenvironment, may improve drug delivery during HIPEC. This in silico study evaluates the availability of oxaliplatin within tumor nodules when HIPEC is performed after bevacizumab treatment. Methods: Using a computational fluid dynamics (CFD) model of HIPEC, the temperature and oxaliplatin distribution within the rat abdomen were calculated, followed by a model of drug transport within tumor nodules located at various sites in the peritoneum. The vascular normalization effect of the bevacizumab treatment was incorporated by adjusting the biophysical parameters of the tumor nodules. The effective penetration depth values, including the thermal enhancement ratio of cytotoxicity, were then compared between HIPEC alone and HIPEC combined with the bevacizumab treatment. Results: After bevacizumab treatments at doses of 0.5 mg/kg and 5 mg/kg, the oxaliplatin availability increased by up to 20% and 45% when HIPEC was performed during the vascular normalization phase, with the penetration depth increasing by 1.5-fold and 2.3-fold, respectively. Tumors with lower collagen densities and larger vascular pore sizes showed higher oxaliplatin enhancement after the combined treatment. Bevacizumab also enabled a reduction in the oxaliplatin dose (up to half at 5 mg/kg bevacizumab) while maintaining effective drug levels in the tumor nodules, potentially reducing systemic toxicity. Conclusions: These findings suggest that administering oxaliplatin-based HIPEC during bevacizumab-induced vascular normalization could significantly improve drug penetration and enhance treatment efficacy.
Collapse
Affiliation(s)
- Pouya Namakshenas
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.C.); (A.L.O.); (H.P.K.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Treatment and Quality of Life, 1105 AZ Amsterdam, The Netherlands; (J.B.T.); (P.J.T.)
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.C.); (A.L.O.); (H.P.K.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Treatment and Quality of Life, 1105 AZ Amsterdam, The Netherlands; (J.B.T.); (P.J.T.)
| | - Jurriaan B. Tuynman
- Cancer Center Amsterdam, Treatment and Quality of Life, 1105 AZ Amsterdam, The Netherlands; (J.B.T.); (P.J.T.)
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Pieter J. Tanis
- Cancer Center Amsterdam, Treatment and Quality of Life, 1105 AZ Amsterdam, The Netherlands; (J.B.T.); (P.J.T.)
- Department of Surgery, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Arlene L. Oei
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.C.); (A.L.O.); (H.P.K.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), 1105 AZ Amsterdam, The Netherlands
| | - H. Petra Kok
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.C.); (A.L.O.); (H.P.K.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Treatment and Quality of Life, 1105 AZ Amsterdam, The Netherlands; (J.B.T.); (P.J.T.)
| |
Collapse
|
40
|
Wang Y, Wang J. The Dynamic Changes of COL11A1 Expression During the Carcinogenesis and Development of Breast Cancer and as a Candidate Diagnostic and Prognostic Marker. Breast J 2025; 2025:7861864. [PMID: 39845732 PMCID: PMC11752105 DOI: 10.1155/tbj/7861864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 08/12/2024] [Accepted: 12/27/2024] [Indexed: 01/24/2025]
Abstract
Purpose: Collagen type XI alpha 1 (COL11A1), a critical member of the collagen superfamily, is essential for tissue structure and integrity. This study aimed to validate previously identified variations in COL11A1 expression during breast cancer carcinogenesis and progression, as well as elucidate their clinical implications. Methods: COL11A1 mRNA expression levels were assessed using real-time reverse transcription-PCR (RT-PCR) in 30 pairs of normal breast tissue and primary breast cancer, 30 pairs of primary breast cancer and lymph node metastases, 30 benign tumors, and 107 primary breast cancers. COL11A1 protein expression was evaluated by Western blot in six matched trios of normal tissue, primary cancer, and lymph node metastasis. Results: COL11A1 mRNA levels were significantly higher in primary breast cancer tissues (n = 30) than in adjacent normal breast tissues (p < 0.001). Conversely, lymph node metastases (n = 30) showed significantly lower COL11A1 mRNA levels compared to their primary breast cancer counterparts (p=0.005). In a larger cohort, primary breast cancers (n = 107) had significantly elevated COL11A1 mRNA levels relative to adjacent normal tissues (n = 30) and benign tumors (n = 30) (p < 0.001). Benign tumors also demonstrated higher levels compared to normal tissues (p=0.012). The protein expression patterns were consistent with the mRNA findings. Receiver operating characteristic (ROC) curve analysis confirmed the diagnostic relevance of COL11A1 expression levels. Significant associations were found between COL11A1 mRNA levels and clinical parameters including lymph node involvement (p=0.046), clinical stage (p=0.004), and progesterone receptor status (p=0.048). Overexpression of COL11A1 was correlated with poor prognosis. Conclusions: COL11A1 expression varies during breast tumor initiation and progression, with elevated levels linked to worse prognoses. These findings underscore COL11A1's potential as a biomarker in breast cancer, suggesting its assessment could enhance diagnostic and prognostic strategies for more personalized patient management.
Collapse
Affiliation(s)
- Yuli Wang
- Medical Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jing Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
41
|
Wang Z, Wang W, Luo Q, Song G. High matrix stiffness accelerates migration of hepatocellular carcinoma cells through the integrin β1-Plectin-F-actin axis. BMC Biol 2025; 23:8. [PMID: 39789506 PMCID: PMC11721467 DOI: 10.1186/s12915-025-02113-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Abundant research indicates that increased extracellular matrix (ECM) stiffness significantly enhances the malignant characteristics of hepatocellular carcinoma (HCC) cells. Plectin, an essential cytoskeletal linker protein, has recently emerged as a promoter of cancer progression, particularly in the context of cancer cell invasion and metastasis. However, the responsiveness of plectin to changes in ECM stiffness and its impact on HCC progression remain unclear. In this study, we aimed to investigate whether plectin responds to variations in ECM stiffness and to explore its involved molecular mechanisms in regulating HCC cell migration. RESULTS Our results showed that, when compared with control group (7 kPa), high ECM stiffness (53 kPa) boosts HCC cell migration by upregulating plectin and integrin β1 expression and increasing F-actin polymerization. Knockdown of integrin β1 negated the high stiffness-upregulated plectin expression. Furthermore, reducing either plectin or integrin β1 levels, or using latrunculin A, effectively prevented the high ECM stiffness-induced F-actin polymerization and HCC cell migration. CONCLUSIONS These findings demonstrate that integrin β1-plectin-F-actin axis is necessary for high matrix stiffness-driven migration of HCC cells, and provide evidence for the critical role of plectin in mechanotransduction in HCC cells.
Collapse
Affiliation(s)
- Zhihui Wang
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China
| | - Wenbin Wang
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China
| | - Qing Luo
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China
| | - Guanbin Song
- College of Bioengineering, Chongqing University, Chongqing, 400030, China.
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
42
|
Hansen R, Ervik Ø, Amini N, Solberg S, Berg S, Ayyalasomayajula V, Skallerud B, Leira HO, Sorger H, Avdal J. Pulse-Echo Ultrasound for Quantitative Measurements of Two Uncorrelated Elastic Parameters. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:15-25. [PMID: 39370356 DOI: 10.1016/j.ultrasmedbio.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 10/08/2024]
Abstract
OBJECTIVE This paper describes the relationship between elastic tissue properties and strain and presents an initial investigation of pulse-echo ultrasound to measure two uncorrelated elastic parameters in tissue-mimicking phantoms. The two elastic parameters are the shear modulus, related to deformation of shape, and what we in the paper define as the nonlinear compressibility, related to deformation of volume. METHODS We prepared tissue-mimicking phantoms containing lesions of variable shear modulus and variable nonlinear compressibility. An in-house framework for shear wave imaging was developed using ultrasound radiation force at 4.5 MHz to induce shear waves and plane wave imaging with pulses in a frequency band centered around 12.5 MHz to track the shear waves. For measurements of nonlinear compressibility, co-propagating dual-frequency pulse complexes at 0.7 MHz and 14 MHz were applied. Algorithms were implemented on a Verasonics Vantage ultrasound scanner and a custom-made multi-frequency ultrasound transducer was used. Mechanical indentation measurements were performed to validate ultrasound measurements of the shear modulus. For the nonlinear compressibility, ultrasound measurements were compared to results derived from the literature. RESULTS We found good agreement in elasticity results from ultrasound measurements and mechanical indentation as well as when comparing with results derived from the literature. CONCLUSION Results of the current investigation were promising. We plan patient studies involving thyroid lesions and liver steatosis to explore whether measurements of elastic parameters related both to shape deformation and volume deformation are useful in clinical practice.
Collapse
Affiliation(s)
- Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Øyvind Ervik
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Clinic of Medicine, Nord-Trøndelag Hospital Trust, Levanger Hospital, Levanger, Norway
| | | | | | - Sigrid Berg
- Department of Health Research, SINTEF Digital, Trondheim, Norway
| | - Venkat Ayyalasomayajula
- Department of Structural Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjørn Skallerud
- Department of Structural Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Håkon Olav Leira
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Thoracic Medicine, St Olavs Hospital, Trondheim, Norway
| | - Hanne Sorger
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Clinic of Medicine, Nord-Trøndelag Hospital Trust, Levanger Hospital, Levanger, Norway
| | - Jørgen Avdal
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
43
|
Li J, Bai L, Chen Y, Cao J, Zhu J, Zhi W, Cheng Q. Detecting Collagen by Machine Learning Improved Photoacoustic Spectral Analysis for Breast Cancer Diagnostics: Feasibility Studies With Murine Models. JOURNAL OF BIOPHOTONICS 2025; 18:e202400371. [PMID: 39600191 DOI: 10.1002/jbio.202400371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/05/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024]
Abstract
Collagen, a key structural component of the extracellular matrix, undergoes significant remodeling during carcinogenesis. However, the important role of collagen levels in breast cancer diagnostics still lacks effective in vivo detection techniques to provide a deeper understanding. This study presents photoacoustic spectral analysis improved by machine learning as a promising non-invasive diagnostic method, focusing on exploring collagen as a salient biomarker. Murine model experiments revealed more profound associations of collagen with other cancer components than in normal tissues. Moreover, an optimal set of feature wavelengths was identified by a genetic algorithm for enhanced diagnostic performance, among which 75% were from collagen-dominated absorption wavebands. Using optimal spectra, the diagnostic algorithm achieved 72% accuracy, 66% sensitivity, and 78% specificity, surpassing full-range spectra by 6%, 4%, and 8%, respectively. The proposed photoacoustic methods examine the feasibility of offering valuable biochemical insights into existing techniques, showing great potential for early-stage cancer detection.
Collapse
Affiliation(s)
- Jiayan Li
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, People's Republic of China
| | - Lu Bai
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yingna Chen
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, People's Republic of China
| | - Junmei Cao
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, People's Republic of China
| | - Jingtao Zhu
- School of Physics Science and Engineering, Tongji University, Shanghai, People's Republic of China
| | - Wenxiang Zhi
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Qian Cheng
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, People's Republic of China
- National Key Laboratory of Autonomous Intelligent Unmanned Systems, Shanghai, People's Republic of China
- Frontiers Science Center for Intelligent Autonomous Systems, Ministry of Education, Shanghai, People's Republic of China
| |
Collapse
|
44
|
Zhu B, Li F, Yu J, Liang Z, Ke X, Wang Y, Song Z, Li Z, Li G, Guo Y. PIEZO1 mediates matrix stiffness-induced tumor progression in kidney renal clear cell carcinoma by activating the Ca 2+/Calpain/YAP pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119871. [PMID: 39490703 DOI: 10.1016/j.bbamcr.2024.119871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVE The significance of physical factors in the onset and progression of tumors has been increasingly substantiated by a multitude of studies. The extracellular matrix, a pivotal component of the tumor microenvironment, has been the subject of extensive investigation in connection with the advancement of KIRC (Kidney Renal Clear Cell Carcinoma) in recent years. PIEZO1, a mechanosensitive ion channel, has been recognized as a modulator of diverse physiological processes. Nonetheless, the precise function of PIEZO1 as a transducer of mechanical stimuli in KIRC remains poorly elucidated. METHODS A bioinformatics analysis was conducted using data from The Cancer Genome Atlas (TCGA) and the Clinical Proteomic Tumor Analysis Consortium (CPTAC) to explore the correlation between matrix stiffness indicators, such as COL1A1 and LOX mRNA levels, and KIRC prognosis. Expression patterns of mechanosensitive ion channels, particularly PIEZO1, were examined. Collagen-coated polyacrylamide hydrogel models were utilized to simulate varying stiffness environments and study their effects on KIRC cell behavior in vitro. Functional experiments, including PIEZO1 knockdown and overexpression, were performed to investigate the molecular mechanisms underlying matrix stiffness-induced cellular changes. Interventions in the Ca2+/Calpain/YAP Pathway were conducted to evaluate their effects on cell growth, EMT, and stemness characteristics. RESULTS Our findings indicate a significant correlation between matrix stiffness and the prognosis of KIRC patients. It is observed that higher mechanical stiffness can facilitate the growth and metastasis of KIRC cells. Notably, we have also observed that the deficiency of PIEZO1 hinders the proliferation, EMT, and stemness characteristics of KIRC cells induced by a stiff matrix. Our study suggests that PIEZO1 plays a crucial role in mediating KIRC growth and metastasis through the activation of the Ca2+/Calpain/YAP Pathway. CONCLUSION This study elucidates a novel mechanism through which the activation of PIEZO1 leads to calcium influx, subsequent calpain activation, and YAP nuclear translocation, thereby contributing to the progression of KIRC driven by matrix stiffness.
Collapse
Affiliation(s)
- Biqiang Zhu
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China.
| | - Fan Li
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China.
| | - Jiajun Yu
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Zhulin Liang
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Xinwen Ke
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Yong Wang
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Zhengshuai Song
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Zhongyuan Li
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Guohao Li
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Yonglian Guo
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China.
| |
Collapse
|
45
|
Sato A, Takagi K, Yoshida M, Yamaguchi-Tanaka M, Sagehashi M, Miki Y, Miyashita M, Suzuki T. Discoidin Domain Receptor 2 Contributes to Breast Cancer Progression and Chemoresistance by Interacting with Collagen Type I. Cancers (Basel) 2024; 16:4285. [PMID: 39766183 PMCID: PMC11674238 DOI: 10.3390/cancers16244285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Chemoresistance is an important issue to be solved in breast cancer. It is well known that the content and morphology of collagens in tumor tissues are drastically altered following chemotherapy, and discoidin domain receptor 2 (DDR2) is a unique type of receptor tyrosine kinase (RTK). This RTK is activated by collagens, playing important roles in human malignancies. However, the contribution to the chemoresistance of DDR2 in terms of the association with collagens remains largely unclear in breast cancer. Methods: We immunolocalized DDR2 and collagen type I in 224 breast cancer tissues and subsequently conducted in vitro studies to confirm the role of DDR2 in breast cancer chemoresistance using chemosensitive and chemoresistant cell lines. Results: DDR2 immunoreactivity was positively correlated with aggressive behaviors of breast cancer and was significantly associated with an increased risk of recurrence, especially in those who received chemotherapy. Moreover, in vitro experiments demonstrated that DDR2 promoted the proliferative activity of breast cancer cells, and cell viability after epirubicin treatment was significantly maintained by DDR2 in a collagen I-dependent manner. Conclusions: These data suggested that DDR2 could be a poor prognostic factor associated with cell proliferation and chemotherapy resistance in human breast cancer.
Collapse
Affiliation(s)
- Ai Sato
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.S.); (T.S.)
| | - Kiyoshi Takagi
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.S.); (T.S.)
| | - Momoka Yoshida
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.S.); (T.S.)
| | - Mio Yamaguchi-Tanaka
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.S.); (T.S.)
- Personalized Medicine Center, Tohoku University Hospital, Sendai 980-8574, Japan
| | - Mikoto Sagehashi
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.S.); (T.S.)
| | - Yasuhiro Miki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Minoru Miyashita
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan;
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.S.); (T.S.)
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Department of Pathology, Tohoku University Hospital, Sendai 980-8574, Japan
| |
Collapse
|
46
|
Vitacolonna M, Bruch R, Schneider R, Jabs J, Hafner M, Reischl M, Rudolf R. A spheroid whole mount drug testing pipeline with machine-learning based image analysis identifies cell-type specific differences in drug efficacy on a single-cell level. BMC Cancer 2024; 24:1542. [PMID: 39696122 DOI: 10.1186/s12885-024-13329-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The growth and drug response of tumors are influenced by their stromal composition, both in vivo and 3D-cell culture models. Cell-type inherent features as well as mutual relationships between the different cell types in a tumor might affect drug susceptibility of the tumor as a whole and/or of its cell populations. However, a lack of single-cell procedures with sufficient detail has hampered the automated observation of cell-type-specific effects in three-dimensional stroma-tumor cell co-cultures. METHODS Here, we developed a high-content pipeline ranging from the setup of novel tumor-fibroblast spheroid co-cultures over optical tissue clearing, whole mount staining, and 3D confocal microscopy to optimized 3D-image segmentation and a 3D-deep-learning model to automate the analysis of a range of cell-type-specific processes, such as cell proliferation, apoptosis, necrosis, drug susceptibility, nuclear morphology, and cell density. RESULTS This demonstrated that co-cultures of KP-4 tumor cells with CCD-1137Sk fibroblasts exhibited a growth advantage compared to tumor cell mono-cultures, resulting in higher cell counts following cytostatic treatments with paclitaxel and doxorubicin. However, cell-type-specific single-cell analysis revealed that this apparent benefit of co-cultures was due to a higher resilience of fibroblasts against the drugs and did not indicate a higher drug resistance of the KP-4 cancer cells during co-culture. Conversely, cancer cells were partially even more susceptible in the presence of fibroblasts than in mono-cultures. CONCLUSION In summary, this underlines that a novel cell-type-specific single-cell analysis method can reveal critical insights regarding the mechanism of action of drug substances in three-dimensional cell culture models.
Collapse
Affiliation(s)
- Mario Vitacolonna
- CeMOS, Mannheim University of Applied Sciences, 68163, Mannheim, Germany.
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany.
| | - Roman Bruch
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, 76344, Eggen-stein-Leopoldshafen, Germany
| | | | - Julia Jabs
- Merck Healthcare KGaA, 64293, Darmstadt, Germany
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
- Institute of Medical Technology, Medical Faculty Mannheim of Heidelberg University, Mannheim University of Applied Sciences, 68167, Mannheim, Germany
| | - Markus Reischl
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, 76344, Eggen-stein-Leopoldshafen, Germany
| | - Rüdiger Rudolf
- CeMOS, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
| |
Collapse
|
47
|
Lo Buglio G, Lo Cicero A, Campora S, Ghersi G. The Multifaced Role of Collagen in Cancer Development and Progression. Int J Mol Sci 2024; 25:13523. [PMID: 39769286 PMCID: PMC11678882 DOI: 10.3390/ijms252413523] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/05/2025] Open
Abstract
Collagen is a crucial protein in the extracellular matrix (ECM) essential for preserving tissue architecture and supporting crucial cellular functions like proliferation and differentiation. There are twenty-eight identified types of collagen, which are further divided into different subgroups. This protein plays a critical role in regulating tissue homeostasis. However, in solid tumors, the balance can be disrupted, due to an abundance of collagen in the tumor microenvironment, which significantly affects tumor growth, cell invasion, and metastasis. It is important to investigate the specific types of collagens in cancer ECM and their distinct roles in tumor progression to comprehend their unique contribution to tumor behavior. The diverse pathophysiological functions of different collagen types in cancers illustrate collagen's dual roles, offering potential therapeutic options and serving as prognostic markers.
Collapse
Affiliation(s)
- Gabriele Lo Buglio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Alessandra Lo Cicero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
| | - Simona Campora
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
- Abiel srl, 90128 Palermo, Italy
| |
Collapse
|
48
|
Polemidiotou K, Kulkarni SG, Szydlak R, Lekka M, Radmacher M, Gkretsi V, Stylianopoulos T, Stylianou A. Assessing sarcoma cell cytoskeleton remodeling in response to varying collagen concentration. Int J Biol Macromol 2024; 282:136770. [PMID: 39437949 DOI: 10.1016/j.ijbiomac.2024.136770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Sarcomas, rare malignant tumors of mesenchymal origin, are often underdiagnosed and have face diagnostic ambiguities and limited treatment options. The main objective of this study was to define the nanomechanical and biophysical properties of sarcoma cells, particularly examining how the cytoskeleton's remodeling and related cellular processes such as cell migration and invasion in response to environmental stimuli due to collagen content. Utilizing one murine fibrosarcoma and one osteosarcoma cell line we employed atomic force microscopy, immunostaining, advanced image processing, in vitro cellular assays, and molecular techniques to investigate cells' cytoskeleton remodeling in response to varying collagen concentration. Our study focused on how alterations in collagen content affects the cytoskeletal dynamics and correlate with changes in gene expression profiles relevant to metastasis and an aggressive cancer phenotypes. Our findings indicate that despite their shared classification, fibrosarcoma and osteosarcoma cells display distinct biophysical properties and respond differently to mechanical forces. Notably, this difference in cellular behavior renders mechanical properties a potent novel biomarkers. Furthermore, the metastasis-related identified genes related to metastatic capability, could be potential therapeutic targets. This study highlights the significance of understanding the unique traits of sarcoma cells to improve diagnostic precision and expand therapeutic strategies, for this rare type of cancer.
Collapse
Affiliation(s)
- Katerina Polemidiotou
- Cancer Mechanobiology & Applied Biophysics Group, Basic and Translational Cancer Research Center, School of Sciences, European University Cyprus/EUC Research Centre, 2404 Nicosia, Cyprus.
| | - Shruti G Kulkarni
- Institute of Biophysics, University of Bremen, 28359 Bremen, Germany.
| | - Renata Szydlak
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Krakow, Poland; Department of Bioinformatics and Telemedicine, Jagiellonian University Medical College, PL-30688 Krakow, Poland.
| | - Małgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Krakow, Poland.
| | - Manfred Radmacher
- Institute of Biophysics, University of Bremen, 28359 Bremen, Germany.
| | - Vasiliki Gkretsi
- Cancer Metastasis and Adhesion Group, Basic and Translational Cancer Research Center (BTCRC), European University Cyprus, Nicosia, Cyprus.
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, 1678 Nicosia, Cyprus.
| | - Andreas Stylianou
- Cancer Mechanobiology & Applied Biophysics Group, Basic and Translational Cancer Research Center, School of Sciences, European University Cyprus/EUC Research Centre, 2404 Nicosia, Cyprus; Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, 1678 Nicosia, Cyprus.
| |
Collapse
|
49
|
Vargas Garcia AP, Reis LA, Ribeiro BRM, Nunes CB, de Paula AM, Cassali GD. Comparative evaluation of collagen modifications in breast cancer in human and canine carcinomas. Sci Rep 2024; 14:28846. [PMID: 39572729 PMCID: PMC11582713 DOI: 10.1038/s41598-024-79854-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
New diagnostic and therapeutic approaches have been increasingly demanded due to the high morbidity and mortality associated with breast cancer. Recently, changes in the collagen fibres in mammary neoplasms have been shown to provide information that can be helpful for more accurate diagnosis. We aimed to conduct a comparative analysis of the tumour stroma in human and canine mammary neoplasms to assess the relationship between collagen modifications and the behaviour of carcinomas in both species, by multiphoton microscopy. We present a retrospective study of 70 cases of human mammary tumour and 74 cases of canine mammary tumour. We analysed sections stained with haematoxylin and eosin from 1,200 representative areas of normal mammary tissue, fibroadenoma, grade I invasive carcinoma, grade III invasive carcinoma and invasive micropapillary carcinoma in human species and 1,304 representative areas of normal mammary tissue, benign mixed tumour, mixed carcinoma, carcinosarcoma, invasive micropapillary carcinoma and solid carcinoma in canine species. We obtained that both human and canine mammary carcinomas present lower density of collagen fibres, higher density of cells and the collagen fibres are more aligned than in normal tissue. For human mammary carcinomas, the collagen fibres are more linear as compared to normal tissue. In addition, we demonstrated that the carcinomas with unfavourable prognosis present shorter collagen fibres, and these collagen changes correlate with the clinical and pathological data in human and canine species. For dogs, there is a correlation between the mean fibre length with the specific survival times. Thus, we demonstrate that dogs provide an excellent comparative perspective for studying how changes in the tumour stroma affect patient survival.
Collapse
Affiliation(s)
- Ana Paula Vargas Garcia
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901, MG, Brazil
| | - Luana Aparecida Reis
- Department of Physics, Institute of Exact Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901, MG, Brazil
| | - Bárbara Regina Melo Ribeiro
- Department of Physics, Institute of Exact Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901, MG, Brazil
| | - Cristiana Buzelin Nunes
- Department of Anatomic Pathology, Faculty of Medicine, Federal University of Minas Gerais, Av. Prof. Alfredo Balena, 190, Santa Efigênia, Belo Horizonte, 30130-100, MG, Brazil
| | - Ana Maria de Paula
- Department of Physics, Institute of Exact Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901, MG, Brazil.
- Institute of Physics "Gleb Wataghin", University of Campinas, Campinas, SP, Brazil.
| | - Geovanni Dantas Cassali
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901, MG, Brazil
| |
Collapse
|
50
|
Avsharian LC, Loganathan S, Ebelt ND, Shalamzari AF, Rodarte Muñoz I, Manuel ER. Tumor-Colonizing E. coli Expressing Both Collagenase and Hyaluronidase Enhances Therapeutic Efficacy of Gemcitabine in Pancreatic Cancer Models. Biomolecules 2024; 14:1458. [PMID: 39595636 PMCID: PMC11591662 DOI: 10.3390/biom14111458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Desmoplasia is a hallmark feature of pancreatic ductal adenocarcinoma (PDAC) that contributes significantly to treatment resistance. Approaches to enhance drug delivery into fibrotic PDAC tumors continue to be an important unmet need. In this study, we have engineered a tumor-colonizing E. coli-based agent that expresses both collagenase and hyaluronidase as a strategy to reduce desmoplasia and enhance the intratumoral perfusion of anticancer agents. Overall, we observed that the tandem expression of both these enzymes by tumor-colonizing E. coli resulted in the reduced presence of intratumoral collagen and hyaluronan, which likely contributed to the enhanced chemotherapeutic efficacy observed when used in combination. These results highlight the importance of combination treatments involving the depletion of desmoplastic components in PDAC before or during treatment.
Collapse
Affiliation(s)
- Lara C. Avsharian
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (L.C.A.); (S.L.); (N.D.E.)
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010, USA;
| | - Suvithanandhini Loganathan
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (L.C.A.); (S.L.); (N.D.E.)
| | - Nancy D. Ebelt
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (L.C.A.); (S.L.); (N.D.E.)
| | - Azadeh F. Shalamzari
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010, USA;
| | - Itzel Rodarte Muñoz
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (L.C.A.); (S.L.); (N.D.E.)
| | - Edwin R. Manuel
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (L.C.A.); (S.L.); (N.D.E.)
| |
Collapse
|