1
|
Kaneko R, Kishimoto Y, Ishikawa O, Funahashi N, Koshikawa N. Laminin-γ2-NR6A1 Fusion Protein Promotes Metastatic Potential in Non-Small-Cell Lung Carcinoma Cells without Epidermal Growth Factor Receptor Mutation. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00113-0. [PMID: 40252971 DOI: 10.1016/j.ajpath.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Laminin-γ2 fusion gene (Lm-γ2F), formed by translocation between LAMC2 and NR6A1, functions as an epidermal growth factor receptor (EGFR) ligand. However, its expression and impact on cancers beyond the initially studied contexts remain unclear. This study focused on Lm-γ2F protein secretion and its role in non-small-cell lung carcinoma (NSCLC), where EGFR signaling plays a pivotal role in malignancy progression. Lm-γ2F secretion was confirmed in serum-free conditioned medium from six NSCLC cell lines by Western blot analysis and further validated in NCI-H1650 cells. Hypothesizing that Lm-γ2F functions as an EGFR ligand, its effects in NSCLC cells lacking EGFR mutations were explored. In EKVX and RERF-LC-KJ cell lines, Lm-γ2F overexpression significantly enhanced cell growth, survival, motility, and invasiveness through EGFR signaling activation compared with controls. Conversely, no effects were observed in VMRC-LCD cells lacking EGFR expression. Additionally, increased membrane-type 1 matrix metalloproteinase expression was detected in Lm-γ2F-expressing EKVX cells. In vivo, these cells exhibited elevated metastatic activity in a lung metastasis model. These findings suggested that ectopic Lm-γ2F expression contributes to malignant progression in NSCLC cells without EGFR mutations. Furthermore, EGFR tyrosine kinase inhibitors may suppress metastasis in these contexts. This study provides novel insights into the oncogenic role of Lm-γ2F in NSCLC, highlighting its potential as a therapeutic target to mitigate tumor progression and metastasis.
Collapse
Affiliation(s)
- Ryo Kaneko
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
| | - Yuri Kishimoto
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
| | - Ozora Ishikawa
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
| | - Nobuaki Funahashi
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan.
| | - Naohiko Koshikawa
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan; Clinical Cancer Proteomics Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan.
| |
Collapse
|
2
|
Haq F, Bychkov A, Mete O, Jeon S, Jung CK. Identification of Specific Biomarkers for Anaplastic Thyroid Carcinoma Through Spatial Transcriptomic and Immunohistochemical Profiling. Endocr Pathol 2025; 36:14. [PMID: 40244468 DOI: 10.1007/s12022-025-09858-z] [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] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Anaplastic thyroid carcinoma (ATC) is an aggressive malignancy with a poor prognosis. Despite its rarity, identifying predictive molecular markers that distinguish ATC from follicular cell-derived non-anaplastic thyroid carcinomas is critical for improving diagnosis and treatment strategies. This study aimed to identify and validate key mRNA and protein markers associated with ATC progression and dedifferentiation. We performed spatial transcriptomic analysis on an index case of ATC coexisting with papillary thyroid carcinoma (PTC) and identified eight differentially expressed mRNA markers. These findings were validated in a large cohort using immunohistochemistry on tissue microarrays across various thyroid tumor types, including follicular adenoma, PTC, poorly differentiated thyroid carcinoma, medullary thyroid carcinoma, and ATC. Additionally, the impact of BRAF p.V600E mutation status on these markers was evaluated. COL7A1, LAMC2, SPHK1, and SRPX2 mRNA and protein levels were significantly overexpressed in ATCs. Conversely, CD24, EPHX1, GPX3, and RBM47 mRNA and protein levels were markedly downregulated in ATCs. Functional enrichment analysis, based on mRNA expression data, identified the role of these proteins in tumor invasion, epithelial-mesenchymal transition, extracellular matrix remodeling, and immune evasion. The expression levels of these markers were independent of BRAF p.V600E mutation status, highlighting their potential as diagnostic markers. In summary, this study identified eight molecular markers that can distinguish ATC from other thyroid tumors. The validation of these markers at both the mRNA and protein levels underscores their clinical relevance in ATC diagnosis and tumor characterization. These findings provide a foundation for future biomarker-driven diagnostic and therapeutic strategies for ATC.
Collapse
Affiliation(s)
- Faridul Haq
- Department of Hospital Pathology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-Gu, Seoul, 06591, Republic of Korea
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- College of Medicine, Cancer Research Institute, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Andrey Bychkov
- Department of Pathology, Kameda Medical Center, Kamogawa City, Chiba, 296 - 8602, Japan
| | - Ozgur Mete
- Department of Pathology, University Health Network, University of Toronto, Toronto, ON, M5G 2C4, Canada
| | - Sora Jeon
- College of Medicine, Cancer Research Institute, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-Gu, Seoul, 06591, Republic of Korea.
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- College of Medicine, Cancer Research Institute, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
3
|
Xu L, Wang B, Wang C, Mao N, Huang Y, Fu X, Feng T, He Q, Zhang Y, You G, Ma X, Peng X, Su J. A model of basement membrane-related regulators for prediction of prognoses in esophageal cancer and verification in vitro. BMC Cancer 2025; 25:696. [PMID: 40234833 PMCID: PMC11998150 DOI: 10.1186/s12885-025-14081-4] [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: 12/17/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
Emerging evidence suggests the importance of basement membrane components in cancer metastasis; however, their specific roles in esophageal carcinoma remain underexplored. To investigate this, we analyzed 152 esophageal cancer and 11 normal esophageal tissue samples, identifying basement membrane-related prognostic signatures through differential gene expression profiling and Least Absolute Shrinkage and Selection Operator regression. A six-gene panel (LAMC2, GPC2, AGRN, ITGA3, LAMA3, and LOXL4) demonstrated robust predictive capacity, which we subsequently integrated with clinical features via nomogram modeling to predict overall survival. Our computational analyses revealed distinct tumor microenvironment immune cell profiles and chemotherapeutic drug sensitivities across risk strata. We performed an immunohistochemical assay to confirm increased tumor tissue expression, thereby reinforcing the clinical relevance of these biomarkers. Experimental validation using KYSE-150 esophageal squamous carcinoma cells demonstrated that while LAMC2 knockdown attenuated cellular migration, AGRN, GPC2, ITGA3, LAMA3, and LOXL4 suppression enhanced migratory capacity. Proliferation assays further revealed increased growth rates upon GPC2, ITGA3, and LAMA3 expression inhibition. Our results established a basement membrane-derived risk model for esophageal carcinoma and revealed the roles of the model genes in tumor progression regulation. This model advances prognostic stratification and provides insights into therapeutic targets.
Collapse
Affiliation(s)
- Lang Xu
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, 511400, China
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Bingna Wang
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, 511400, China
- School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China
| | - Chen Wang
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, 511400, China
| | - Nan Mao
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, 511400, China
| | - Yating Huang
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, 511400, China
| | - Xihua Fu
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, 511400, China
| | - Tao Feng
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Qiming He
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Yang Zhang
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, 511400, China
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Guoxing You
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Xiaojun Ma
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Xinsheng Peng
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Jianfen Su
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, 511400, China.
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
- School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
4
|
Kraft A, Kirschner MB, Orlowski V, Ronner M, Bodmer C, Boeva V, Opitz I, Meerang M. Exploring RNA cargo in extracellular vesicles for pleural mesothelioma detection. BMC Cancer 2025; 25:212. [PMID: 39920655 PMCID: PMC11804012 DOI: 10.1186/s12885-025-13617-y] [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: 11/29/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Pleural Mesothelioma (PM) is a highly aggressive cancer, for which effective early detection remains a challenge due to limited screening options and low sensitivity of biomarkers discovered so far. While extracellular vesicles (EVs) have emerged as promising candidates for blood-based biomarkers, their role in PM has not been studied yet. In this study, we characterized the transcriptomic profile of EVs secreted by PM primary cells and explored their potential as a biomarker source for PM detection. METHODS We collected cell culture supernatant from early-passage PM cell cultures derived from the pleural effusion of 4 PM patients. EVs were isolated from the supernatant using Qiagen exoEasy Maxi kit. RNA isolation from EVs was done using the mirVana PARIS kit. Finally, single-end RNA sequencing was done with Illumina Novaseq 6000. RESULTS We identified a range of RNA species expressed in EVs secreted by PM cells, including protein-coding RNA (80%), long non-coding RNA (13%), pseudogenes (4.5%), and short non-coding RNA (1.6%). We detected a subset of genes associated with the previously identified epithelioid (32 genes) and sarcomatoid molecular components (36 genes) in PM-EVs. To investigate whether these markers could serve as biomarkers for PM detection in blood, we compared the RNA content of PM-EVs with the cargo of EVs isolated from the plasma of healthy donors (publicly available data). Majority of upregulated genes in PM-EVs were protein-coding and long non-coding RNAs. Interestingly, 25 of them were the sarcomatoid and epithelioid marker genes. Finally, functional analysis revealed that the PM-EV RNA cargo was associated with Epithelial-Mesenchymal transition, glycolysis, and hypoxia. CONCLUSIONS This is the first study to characterize the transcriptomic profile of EVs secreted by PM primary cell cultures, demonstrating their potential as biomarker source for early detection. Further investigation of the functional role of PM-EVs will provide new insights into disease biology and therapeutic avenues.
Collapse
Affiliation(s)
- Agnieszka Kraft
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Institute for Machine Learning, Department of Computer Science, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Michaela B Kirschner
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Vanessa Orlowski
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Manuel Ronner
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Caroline Bodmer
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Valentina Boeva
- Institute for Machine Learning, Department of Computer Science, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Zurich, Switzerland
- ETH AI Center, ETH Zurich, Zurich, Switzerland
- UMR 8104, UMR-S1016, Cochin InstituteCNRSParis Descartes University, Inserm U1016, 75014, Paris, France
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Mayura Meerang
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Balakrishnan K, Xiao Y, Chen Y, Dong J. Elevated Expression of Cell Adhesion, Metabolic, and Mucus Secretion Gene Clusters Associated with Tumorigenesis, Metastasis, and Poor Survival in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:4049. [PMID: 39682235 DOI: 10.3390/cancers16234049] [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: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
OBJECTIVES Technological advances in identifying gene expression profiles are being applied to study an array of cancers. The goal of this study was to identify differentially expressed genes in pancreatic ductal adenocarcinoma (PDAC) and examine their potential role in tumorigenesis and metastasis. METHODS The transcriptomic profiles of PDAC and non-tumorous tissue samples were derived from the gene expression omnibus (GEO), which is a public repository. The GEO2R tool was used to further derive differentially expressed genes from those profiles. RESULTS In this study, a total of 68 genes were derived from upregulated PDAC genes in three or more transcriptomic profiles and were considered PDAC gene sets. The identified PDAC gene sets were examined in the molecular signatures database (MSigDB) for ontological investigation, which revealed that these genes were involved in the extracellular matrix and associated with the cell adhesion process in PDAC tumorigenesis. The gene set enrichment analysis showed greater enrichment scores for the gene sets. Moreover, the identified gene sets were examined for protein-protein interaction using the STRING database. Based on functional k-means clustering, the following three functional cluster groups were identified in this study: extracellular matrix/cell adhesion, metabolic, and mucus secretion-related protein groups. The receiver operating characteristic (ROC) curve revealed greater specificity and sensitivity for these cluster genes in predicting PDAC tumorigenesis and metastases. In addition, the expression of the cluster genes affects the overall survival rate of PDAC patients. Using the cancer genome atlas (TCGA) database, the associations between expression levels and clinicopathological features were validated. CONCLUSIONS Overall, the genes identified in this study appear to be critical in PDAC development and can serve as potential diagnostic and prognostic targets for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Karthik Balakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Xiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yuanhong Chen
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
6
|
Liang X, Ma X, Luan F, Gong J, Zhao S, Pan Y, Liu Y, Liu L, Huang J, An Y, Hu S, Yang J, Dong D. Identification of new subtypes of breast cancer based on vasculogenic mimicry related genes and a new model for predicting the prognosis of breast cancer. Heliyon 2024; 10:e36565. [PMID: 39263085 PMCID: PMC11387377 DOI: 10.1016/j.heliyon.2024.e36565] [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: 04/18/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Breast cancer is a malignant tumor that poses a serious threat to women's health, and vasculogenic mimicry (VM) is strongly associated with bad prognosis in breast cancer. However, the relationship between VM and immune infiltration in breast cancer and the underlying mechanisms have not been fully studied. On the basis of the Cancer Genome Atlas (TCGA), Fudan University Shanghai Cancer Center (FUSCC) database, GSCALite database, and gene set enrichment analysis (GSEA) datasets, we investigated the potential involvement of VM-related genes in the development and progression of breast cancer. We analyzed the differential expression, mutation status, methylation status, drug sensitivity, tumor mutation burden (TMB), microsatellite instability (MSI), immune checkpoints, tumor microenvironment (TME), and immune cell infiltration levels associated with VM-related genes in breast cancer. We created two VM subclusters out of breast cancer patients using consensus clustering, and discovered that patients in Cluster 1 had better survival outcomes compared to those in Cluster 2. The infiltration levels of T cells CD4 memory resting and T cells CD8 were higher in Cluster 1, indicating an immune-active state in this cluster. Additionally, we selected three prognostic genes (LAMC2, PIK3CA, and TFPI2) using Lasso, univariate, and multivariate Cox regression and constructed a risk model, which was validated in an external dataset. The prognosis of patients is strongly correlated with aberrant expression of VM-related genes, which advances our knowledge of the tumor immune milieu and enables us to identify previously unidentified breast cancer subtypes. This could direct more potent immunotherapy approaches.
Collapse
Affiliation(s)
- Xiao Liang
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xinyue Ma
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Feiyang Luan
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Jin Gong
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Shidi Zhao
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yiwen Pan
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yijia Liu
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Lijuan Liu
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Jing Huang
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yiyang An
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Sirui Hu
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Jin Yang
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Danfeng Dong
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| |
Collapse
|
7
|
Hayderi A, Zegeye MM, Meydan S, Sirsjö A, Kumawat AK, Ljungberg LU. TNF Induces Laminin-332-Encoding Genes in Endothelial Cells and Laminin-332 Promotes an Atherogenic Endothelial Phenotype. Int J Mol Sci 2024; 25:8699. [PMID: 39201392 PMCID: PMC11354388 DOI: 10.3390/ijms25168699] [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: 07/09/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Laminins are essential components of the basement membranes, expressed in a tissue- and cell-specific manner under physiological conditions. During inflammatory circumstances, such as atherosclerosis, alterations in laminin composition within vessels have been observed. Our study aimed to assess the influence of tumor necrosis factor-alpha (TNF), a proinflammatory cytokine abundantly found in atherosclerotic lesions, on endothelial laminin gene expression and the effects of laminin-332 (LN332) on endothelial cells' behavior. We also evaluated the expression of LN332-encoding genes in human carotid atherosclerotic plaques. Our findings demonstrate that TNF induces upregulation of LAMB3 and LAMC2, which, along with LAMA3, encode the LN332 isoform. Endothelial cells cultured on recombinant LN332 exhibit decreased claudin-5 expression and display a loosely connected phenotype, with an elevated expression of chemokines and leukocyte adhesion molecules, enhancing their attractiveness and adhesion to leukocytes in vitro. Furthermore, LAMB3 and LAMC2 are upregulated in human carotid plaques and show a positive correlation with TNF expression. In summary, TNF stimulates the expression of LN332-encoding genes in human endothelial cells and LN332 promotes an endothelial phenotype characterized by compromised junctional integrity and increased leukocyte interaction. These findings highlight the importance of basement membrane proteins for endothelial integrity and the potential role of LN332 in atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Liza U. Ljungberg
- Cardiovascular Research Centre, Department of Medical Sciences, School of Medicine, Örebro University, 70362 Örebro, Sweden; (A.H.); (S.M.); (A.S.); (A.K.K.)
| |
Collapse
|
8
|
Cheng L, Li X, Dong W, Yang J, Li P, Qiang X, Yin J, Guo L. LAMC2 regulates the proliferation, invasion, and metastasis of gastric cancer via PI3K/Akt signaling pathway. J Cancer Res Clin Oncol 2024; 150:230. [PMID: 38703300 PMCID: PMC11069487 DOI: 10.1007/s00432-024-05720-7] [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: 09/27/2023] [Accepted: 03/21/2024] [Indexed: 05/06/2024]
Abstract
OBJECTIVES Gastric cancer (GC) is a prevalent malignant tumor widely distributed globally, exhibiting elevated incidence and fatality rates. The gene LAMC2 encodes the laminin subunit gamma-2 chain and is found specifically in the basement membrane of epithelial cells. Its expression is aberrant in multiple types of malignant tumors. This research elucidated a link between LAMC2 and the clinical characteristics of GC and investigated the potential involvement of LAMC2 in GC proliferation and advancement. MATERIALS AND METHODS LAMC2 expressions were detected in GC cell lines and normal gastric epithelial cell lines via qRT-PCR. Silencing and overexpression of the LAMC2 were conducted by lentiviral transfection. A xenograft mouse model was also developed for in vivo analysis. Cell functional assays were conducted to elucidate the involvement of LAMC2 in cell growth, migration, and penetration. Further, immunoblotting was conducted to investigate the impact of LAMC2 on the activation of signal pathways after lentiviral transfection. RESULTS In the findings, LAMC2 expression was markedly upregulated in GC cell lines as opposed to normal gastric epithelial cells. In vitro analysis showed that sh-LAMC2 substantially inhibited GC cell growth, migration, and invasion, while oe-LAMC2 displayed a contrasting effect. Xenograft tumor models demonstrated that oe-LAMC2 accelerated tumor growth via high expression of Ki-67. Immunoblotting analysis revealed a substantial decrease in various signaling pathway proteins, PI3K, p-Akt, and Vimentin levels upon LAMC2 knockdown, followed by increased E-cadherin expression. Conversely, its overexpression exhibited contrasting effects. Besides, epithelial-mesenchymal transition (EMT) was accelerated by LAMC2. CONCLUSION This study provides evidence indicating that LAMC2, by stimulating signaling pathways, facilitated EMT and stimulated the progression of GC cells in laboratory settings and mouse models. Research also explored that the abnormal LAMC2 expression acts as a biomarker for GC.
Collapse
Affiliation(s)
- Lulu Cheng
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Xiaofei Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Wenhui Dong
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Jing Yang
- Department of Pathology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Pengmei Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Xihui Qiang
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Jiajun Yin
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China.
| | - Lianyi Guo
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China.
| |
Collapse
|
9
|
Park SS, Lee YK, Choi YW, Lim SB, Park SH, Kim HK, Shin JS, Kim YH, Lee DH, Kim JH, Park TJ. Cellular senescence is associated with the spatial evolution toward a higher metastatic phenotype in colorectal cancer. Cell Rep 2024; 43:113912. [PMID: 38446659 DOI: 10.1016/j.celrep.2024.113912] [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: 08/03/2023] [Revised: 12/21/2023] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
In this study, we explore the dynamic process of colorectal cancer progression, emphasizing the evolution toward a more metastatic phenotype. The term "evolution" as used in this study specifically denotes the phenotypic transition toward a higher metastatic potency from well-formed glandular structures to collective invasion, ultimately resulting in the development of cancer cell buddings at the invasive front. Our findings highlight the spatial correlation of this evolution with tumor cell senescence, revealing distinct types of senescent tumor cells (types I and II) that play different roles in the overall cancer progression. Type I senescent tumor cells (p16INK4A+/CXCL12+/LAMC2-/MMP7-) are identified in the collective invasion region, whereas type II senescent tumor cells (p16INK4A+/CXCL12+/LAMC2+/MMP7+), representing the final evolved form, are prominently located in the partial-EMT region. Importantly, type II senescent tumor cells associate with local invasion and lymph node metastasis in colorectal cancer, potentially affecting patient prognosis.
Collapse
Affiliation(s)
- Soon Sang Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea; Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon 16499, Korea
| | - Young-Kyoung Lee
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea; Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon 16499, Korea
| | - Yong Won Choi
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon 16499, Korea; Department of Hematology and Oncology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Su Bin Lim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea; Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon 16499, Korea
| | - So Hyun Park
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon 16499, Korea; Department of Pathology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Han Ki Kim
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea; Department of Brain Science and Neurology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Jun Sang Shin
- Department of Surgery, Ajou University School of Medicine, Suwon 16499, Korea
| | - Young Hwa Kim
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon 16499, Korea; Department of Pathology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Dong Hyun Lee
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea; Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon 16499, Korea
| | - Jang-Hee Kim
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon 16499, Korea; Department of Pathology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Tae Jun Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea; Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon 16499, Korea.
| |
Collapse
|
10
|
Nisar H, Labonté FM, Roggan MD, Schmitz C, Chevalier F, Konda B, Diegeler S, Baumstark-Khan C, Hellweg CE. Hypoxia Modulates Radiosensitivity and Response to Different Radiation Qualities in A549 Non-Small Cell Lung Cancer (NSCLC) Cells. Int J Mol Sci 2024; 25:1010. [PMID: 38256084 PMCID: PMC10816011 DOI: 10.3390/ijms25021010] [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: 11/23/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Hypoxia-induced radioresistance reduces the efficacy of radiotherapy for solid malignancies, including non-small cell lung cancer (NSCLC). Cellular hypoxia can confer radioresistance through cellular and tumor micro-environment adaptations. Until recently, studies evaluating radioresistance secondary to hypoxia were designed to maintain cellular hypoxia only before and during irradiation, while any handling of post-irradiated cells was carried out in standard oxic conditions due to the unavailability of hypoxia workstations. This limited the possibility of simulating in vivo or clinical conditions in vitro. The presence of molecular oxygen is more important for the radiotoxicity of low-linear energy transfer (LET) radiation (e.g., X-rays) than that of high-LET carbon (12C) ions. The mechanisms responsible for 12C ions' potential to overcome hypoxia-induced radioresistance are currently not fully understood. Therefore, the radioresistance of hypoxic A549 NSCLC cells following exposure to X-rays or 12C ions was investigated along with cell cycle progression and gene expression by maintaining hypoxia before, during and after irradiation. A549 cells were incubated under normoxia (20% O2) or hypoxia (1% O2) for 48 h and then irradiated with X-rays (200 kV) or 12C ions (35 MeV/n, LET ~75 keV/µm). Cell survival was evaluated using colony-forming ability (CFA) assays immediately or 24 h after irradiation (late plating). DNA double-strand breaks (DSBs) were analyzed using γH2AX immunofluorescence microscopy. Cell cycle progression was determined by flow cytometry of 4',6-diamidino-2-phenylindole-stained cells. The global transcription profile post-irradiation was evaluated by RNA sequencing. When hypoxia was maintained before, during and after irradiation, hypoxia-induced radioresistance was observed only in late plating CFA experiments. The killing efficiency of 12C ions was much higher than that of X-rays. Cell survival under hypoxia was affected more strongly by the timepoint of plating in the case of X-rays compared to 12C ions. Cell cycle arrest following irradiation under hypoxia was less pronounced but more prolonged. DSB induction and resolution following irradiation were not significantly different under normoxia and hypoxia. Gene expression response to irradiation primarily comprised cell cycle regulation for both radiation qualities and oxygen conditions. Several PI3K target genes involved in cell migration and cell motility were differentially upregulated in hypoxic cells. Hypoxia-induced radioresistance may be linked to altered cell cycle response to irradiation and PI3K-mediated changes in cell motility and migration in A549 cells rather than less DNA damage or faster repair.
Collapse
Affiliation(s)
- Hasan Nisar
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Department of Medical Sciences, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad 44000, Pakistan
| | - Frederik M. Labonté
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Marie Denise Roggan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Claudia Schmitz
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - François Chevalier
- UMR6252 CIMAP, CEA-CNRS-ENSICAEN-University of Caen Normandy, 14000 Caen, France;
| | - Bikash Konda
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - Sebastian Diegeler
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christa Baumstark-Khan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - Christine E. Hellweg
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| |
Collapse
|
11
|
Tong D, Zhou J, Zhou J, Wang X, Gao B, Rui X, Liu L, Chen Q, Huang C. LAMC2 mitigates ER stress by enhancing ER-mitochondria interaction via binding to MYH9 and MYH10. Cancer Gene Ther 2024; 31:43-57. [PMID: 37891404 PMCID: PMC10794146 DOI: 10.1038/s41417-023-00680-5] [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: 07/21/2023] [Revised: 10/01/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023]
Abstract
Highly proliferative and metastatic tumors are constantly exposed to both intrinsic and extrinsic factors that induce adaptation to stressful conditions. Chronic adaptation to endoplasmic reticulum (ER) ER stress is common to many different types of cancers, and poses a major challenge for acquired drug resistance. Here we report that LAMC2, an extracellular matrix protein upregulated in many types of cancers, is localized in the ER of lung, breast, and liver cancer cells. Under tunicamycin-induced ER stress, protein level of LAMC2 is upregulated. Transfection of cancer cells with LAMC2 resulted in the attenuation of ER stress phenotype, accompanied by elevation in mitochondrial membrane potential as well as reduction in reactive oxygen species (ROS) levels and apoptosis. In addition, LAMC2 forms protein complexes with MYH9 and MYH10 to promote mitochondrial aggregation and increased ER-mitochondria interaction at the perinuclear region. Moreover, overexpression of LAMC2 counteracts the effects of ER stress and promotes tumor growth in vivo. Taken together, our results revealed that in complex with MYH9 and MYH10, LAMC2 is essential for promoting ER-mitochondria interaction to alleviate ER stress and allow cancer cells to adapt and proliferate under stressful conditions. This study provides new insights and highlights the promising potential of LAMC2 as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Dongdong Tong
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Jun Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Jing Zhou
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Xiaofei Wang
- Biomedical Experimental Center of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Beibei Gao
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Xiaoyi Rui
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Liying Liu
- Biomedical Experimental Center of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - QiaoYi Chen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China.
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China.
- Biomedical Experimental Center of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, 710061, Xi'an, China.
| |
Collapse
|
12
|
Gogoi RP, Galoforo S, Fox A, Morris C, Ramos H, Gogoi VK, Chehade H, Adzibolosu NK, Shi C, Zhang J, Tedja R, Morris R, Alvero AB, Mor G. A Novel Role of Connective Tissue Growth Factor in the Regulation of the Epithelial Phenotype. Cancers (Basel) 2023; 15:4834. [PMID: 37835529 PMCID: PMC10571845 DOI: 10.3390/cancers15194834] [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: 08/28/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is a biological process where epithelial cells lose their adhesive properties and gain invasive, metastatic, and mesenchymal properties. Maintaining the balance between the epithelial and mesenchymal stage is essential for tissue homeostasis. Many of the genes promoting mesenchymal transformation have been identified; however, our understanding of the genes responsible for maintaining the epithelial phenotype is limited. Our objective was to identify the genes responsible for maintaining the epithelial phenotype and inhibiting EMT. METHODS RNA seq was performed using an vitro model of EMT. CTGF expression was determined via qPCR and Western blot analysis. The knockout of CTGF was completed using the CTGF sgRNA CRISPR/CAS9. The tumorigenic potential was determined using NCG mice. RESULTS The knockout of CTGF in epithelial ovarian cancer cells leads to the acquisition of functional characteristics associated with the mesenchymal phenotype such as anoikis resistance, cytoskeleton remodeling, increased cell stiffness, and the acquisition of invasion and tumorigenic capacity. CONCLUSIONS We identified CTGF is an important regulator of the epithelial phenotype, and its loss is associated with the early cellular modifications required for EMT. We describe a novel role for CTGF, regulating cytoskeleton and the extracellular matrix interactions necessary for the conservation of epithelial structure and function. These findings provide a new window into understanding the early stages of mesenchymal transformation.
Collapse
Affiliation(s)
- Radhika P. Gogoi
- Karmanos Cancer Institute, Wayne State University, 4100 John R St, Detroit, MI 48202, USA;
| | - Sandra Galoforo
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Alexandra Fox
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Colton Morris
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Harry Ramos
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Vir K. Gogoi
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Nicholas K. Adzibolosu
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Chenjun Shi
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA; (C.S.); (J.Z.)
| | - Jitao Zhang
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA; (C.S.); (J.Z.)
| | - Roslyn Tedja
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Robert Morris
- Karmanos Cancer Institute, Wayne State University, 4100 John R St, Detroit, MI 48202, USA;
| | - Ayesha B. Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| |
Collapse
|
13
|
Kohtamäki L, Leivonen SK, Mäkelä S, Juteau S, Leppä S, Hernberg M. Intra-patient evolution of tumor microenvironment in the pathogenesis of treatment-naïve metastatic melanoma patients. Acta Oncol 2023; 62:1008-1013. [PMID: 37624703 DOI: 10.1080/0284186x.2023.2248371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023]
Affiliation(s)
- Laura Kohtamäki
- Department of Oncology, Helsinki University Hospital, Comprehensive Cancer Center, University of Helsinki, Finland
| | | | - Siru Mäkelä
- Department of Oncology, Helsinki University Hospital, Comprehensive Cancer Center, University of Helsinki, Finland
| | | | - Sirpa Leppä
- Department of Oncology, Helsinki University Hospital, Comprehensive Cancer Center, University of Helsinki, Finland
- Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Micaela Hernberg
- Department of Oncology, Helsinki University Hospital, Comprehensive Cancer Center, University of Helsinki, Finland
| |
Collapse
|
14
|
Rekowska AK, Obuchowska K, Bartosik M, Kimber-Trojnar Ż, Słodzińska M, Wierzchowska-Opoka M, Leszczyńska-Gorzelak B. Biomolecules Involved in Both Metastasis and Placenta Accreta Spectrum-Does the Common Pathophysiological Pathway Exist? Cancers (Basel) 2023; 15:cancers15092618. [PMID: 37174083 PMCID: PMC10177254 DOI: 10.3390/cancers15092618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
The process of epithelial-to-mesenchymal transition (EMT) is crucial in the implantation of the blastocyst and subsequent placental development. The trophoblast, consisting of villous and extravillous zones, plays different roles in these processes. Pathological states, such as placenta accreta spectrum (PAS), can arise due to dysfunction of the trophoblast or defective decidualization, leading to maternal and fetal morbidity and mortality. Studies have drawn parallels between placentation and carcinogenesis, with both processes involving EMT and the establishment of a microenvironment that facilitates invasion and infiltration. This article presents a review of molecular biomarkers involved in both the microenvironment of tumors and placental cells, including placental growth factor (PlGF), vascular endothelial growth factor (VEGF), E-cadherin (CDH1), laminin γ2 (LAMC2), the zinc finger E-box-binding homeobox (ZEB) proteins, αVβ3 integrin, transforming growth factor β (TGF-β), β-catenin, cofilin-1 (CFL-1), and interleukin-35 (IL-35). Understanding the similarities and differences in these processes may provide insights into the development of therapeutic options for both PAS and metastatic cancer.
Collapse
Affiliation(s)
- Anna K Rekowska
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Karolina Obuchowska
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Magdalena Bartosik
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Żaneta Kimber-Trojnar
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Magdalena Słodzińska
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | | | | |
Collapse
|