1
|
Feng K, Cai X, Qiao G. Platelet-derived growth factor subunit B overexpression promotes lung cancer tumor growth and metastasis: The role of glucose metabolism. Cytojournal 2025; 22:33. [PMID: 40260069 PMCID: PMC12010884 DOI: 10.25259/cytojournal_190_2024] [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: 09/14/2024] [Accepted: 01/09/2025] [Indexed: 04/23/2025] Open
Abstract
Objective Lung cancer represents a formidable global health challenge due to its substantial prevalence and mortality rates. Metabolic reprogramming, especially the transition to aerobic glycolysis, plays a pivotal role in the progression of lung cancer by sustaining the energy demands for rapid tumor proliferation. The prominent involvement of platelet-derived growth factor subunit B (PDGFB) in promoting the growth and metastasis of lung cancer through specific signaling cascades is well established in. Nonetheless, further research is imperative to elucidate the intricate regulatory mechanisms of PDGFB in glucose metabolism and its implications for the advancement of lung cancer. Our study is dedicated to exploring the effect of PDGFB on lung cancer by modulating glucose metabolism. Material and Methods First, we determined the expression patterns of PDGFB in various lung cancer cell lines (A549, H460, HCC827, and H1975) using quantitative real-time polymerase chain reaction and Western blot analyses. We measured the expression levels of PDGFB and Ki-67 in tumor tissues from lung cancer patients through immunohistochemistry. We then transfected lung cancer cells with a PDGFB overexpression (PDGFB OE) plasmid. The effects of PDGFB OE and galactose + PDGFB OE co-treatment on cell migration and invasion characteristics were assessed using wound healing and Transwell assays. The impact of PDGFB OE and galactose + PDGFB OE co-treatment on the proliferation capacity of lung cancer cells was evaluated through colony formation and 5-ethynyl-2'-deoxyuridine staining assays. We also measured the effects of PDGFB OE on mitochondrial function and glycolytic capacity in lung cancer cells using extracellular acidification rate assay (ECAR) measurement methods. Results Elevated levels of PDGFB expression were markedly detected in various lung cancer cell lines, notably A549 and H460 (P < 0.001). This observation was validated by the analysis of tumor samples from patients with lung cancer who exhibited heightened PDGFB expression in tumor tissues (P < 0.001). Moreover, an association was found between increased levels of Ki67 expression and elevated PDGFB expression (P < 0.001). The upregulation of PDGFB was linked to heightened migratory (P < 0.001), invasive (P < 0.001), and proliferative (P < 0.001) capacities of the cells. Furthermore, an elevation in lactate levels and ECAR (P < 0.001) was noted in the PDGFB OE group, along with increased levels of glycolysis-related regulatory proteins. The inhibition of aerobic glycolysis with galactose effectively mitigated the PDGFB-induced enhancement of lung cancer cell proliferation and migration. Conclusion By affecting glucose metabolism, PDGFB drives the growth and metastasis of lung cancer, underscoring its potential as a promising therapeutic target for the management of this complex disease.
Collapse
Affiliation(s)
- Kai Feng
- Department of Thoracic Surgery II, Binzhou People’s Hospital, Binzhou, China
| | - Xiaoping Cai
- Department of Oncology II, Binzhou People’s Hospital, Binzhou, China
| | - Gaofeng Qiao
- Department of Thoracic Surgery, Shandong Public Health Clinical Center, Shandong University, Jinan City, Shandong, China
| |
Collapse
|
2
|
Bhattacharjee K, Ghosh A. Identification of key regulators in pancreatic ductal adenocarcinoma using network theoretical approach. PLoS One 2025; 20:e0313738. [PMID: 39869563 PMCID: PMC11771905 DOI: 10.1371/journal.pone.0313738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/30/2024] [Indexed: 01/29/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a devastating disease with poor clinical outcomes, which is mainly because of delayed disease detection, resistance to chemotherapy, and lack of specific targeted therapies. The disease's development involves complex interactions among immunological, genetic, and environmental factors, yet its molecular mechanism remains elusive. A major challenge in understanding PDAC etiology lies in unraveling the genetic profiling that governs the PDAC network. To address this, we examined the gene expression profile of PDAC and compared it with that of healthy controls, identifying differentially expressed genes (DEGs). These DEGs formed the basis for constructing the PDAC protein interaction network, and their network topological properties were calculated. It was found that the PDAC network self-organizes into a scale-free fractal state with weakly hierarchical organization. Newman and Girvan's algorithm (leading eigenvector (LEV) method) of community detection enumerated four communities leading to at least one motif defined by G (3,3). Our analysis revealed 33 key regulators were predominantly enriched in neuroactive ligand-receptor interaction, Cell adhesion molecules, Leukocyte transendothelial migration pathways; positive regulation of cell proliferation, positive regulation of protein kinase B signaling biological functions; G-protein beta-subunit binding, receptor binding molecular functions etc. Transcription Factor and mi-RNA of the key regulators were obtained. Recognizing the therapeutic potential and biomarker significance of PDAC Key regulators, we also identified approved drugs for specific genes. However, it is imperative to subject Key regulators to experimental validation to establish their efficacy in the context of PDAC.
Collapse
Affiliation(s)
| | - Aryya Ghosh
- Department of Chemistry, Ashoka University, Sonipat, Haryana, India
| |
Collapse
|
3
|
Huang Y, Cao D, Zhang M, Yang Y, Niu G, Tang L, Shen Z, Zhang Z, Bai Y, Min D, He A. Exploring the impact of PDGFD in osteosarcoma metastasis through single-cell sequencing analysis. Cell Oncol (Dordr) 2024; 47:1715-1733. [PMID: 38652223 PMCID: PMC11467127 DOI: 10.1007/s13402-024-00949-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2024] [Indexed: 04/25/2024] Open
Abstract
PURPOSE The overall survival rate for metastatic osteosarcoma hovers around 20%. Responses to second-line chemotherapy, targeted therapies, and immunotherapies have demonstrated limited efficacy in metastatic osteosarcoma. Our objective is to validate differentially expressed genes and signaling pathways between non-metastatic and metastatic osteosarcoma, employing single-cell RNA sequencing (scRNA-seq) and additional functional investigations. We aim to enhance comprehension of metastatic mechanisms and potentially unveil a therapeutic target. METHODS scRNA-seq was performed on two primary osteosarcoma lesions (1 non-metastatic and 1 metastatic). Seurat package facilitated dimensionality reduction and cluster identification. Copy number variation (CNV) was predicted using InferCNV. CellChat characterized ligand-receptor-based intercellular communication networks. Differentially expressed genes underwent GO function enrichment analysis and GSEA. Validation was achieved through the GSE152048 dataset, which identified PDGFD-PDGFRB as a common ligand-receptor pair with significant contribution. Immunohistochemistry assessed PDGFD and PDGFRB expression, while multicolor immunofluorescence and flow cytometry provided insight into spatial relationships and the tumor immune microenvironment. Kaplan-Meier survival analysis compared metastasis-free survival and overall survival between high and low levels of PDGFD and PDGFRB. Manipulation of PDGFD expression in primary osteosarcoma cells examined invasion abilities and related markers. RESULTS Ten clusters encompassing osteoblasts, osteoclasts, osteocytes, fibroblasts, pericytes, endothelial cells, myeloid cells, T cells, B cells, and proliferating cells were identified. Osteoblasts, osteoclasts, and osteocytes exhibited heightened CNV levels. Ligand-receptor-based communication networks exposed significant fibroblast crosstalk with other cell types, and the PDGF signaling pathway was activated in non-metastatic osteosarcoma primary lesion. These results were corroborated by the GSE152048 dataset, confirming the prominence of PDGFD-PDGFRB as a common ligand-receptor pair. Immunohistochemistry demonstrated considerably greater PDGFD expression in non-metastatic osteosarcoma tissues and organoids, correlating with extended metastasis-free and overall survival. PDGFRB expression showed no significant variation between non-metastatic and metastatic osteosarcoma, nor strong correlations with survival times. Multicolor immunofluorescence suggested co-localization of PDGFD with PDGFRB. Flow cytometry unveiled a highly immunosuppressive microenvironment in metastatic osteosarcoma. Manipulating PDGFD expression demonstrated altered invasive abilities and marker expressions in primary osteosarcoma cells from both non-metastatic and metastatic lesions. CONCLUSIONS scRNA-seq illuminated the activation of the PDGF signaling pathway in primary lesion of non-metastatic osteosarcoma. PDGFD displayed an inhibitory effect on osteosarcoma metastasis, likely through the suppression of the EMT signaling pathway.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Oncology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dongyan Cao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Biliary-Pancreatic Surgery, the Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Manxue Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Biliary-Pancreatic Surgery, the Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yue Yang
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou, Gansu Province, China
| | | | - Lina Tang
- Department of Oncology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zan Shen
- Department of Oncology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhichang Zhang
- Department of Orthopaedic, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yueqing Bai
- Department of Pathology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Daliu Min
- Department of Oncology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Aina He
- Department of Oncology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Zhou W, Zeng T, Chen J, Tang X, Yuan Y, Hu D, Zhang Y, Li Y, Zou J. Aberrant angiogenic signaling pathways: Accomplices in ovarian cancer progression and treatment. Cell Signal 2024; 120:111240. [PMID: 38823664 DOI: 10.1016/j.cellsig.2024.111240] [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: 05/09/2024] [Revised: 05/23/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Ovarian cancer is one of the most common malignant tumors in women, and treatment options are limited. Despite efforts to adjust cancer treatment models and develop new methods, including tumor microenvironment (TME) therapy, more theoretical support is needed. Increasing attention is being given to antiangiogenic measures for TME treatment. Another important concept in ovarian cancer TME is angiogenesis, where tumor cells obtain nutrients and oxygen from surrounding tissues through blood vessels to support further expansion and metastasis. Many neovascularization signaling pathways become imbalanced and hyperactive during this process. Inhibiting these abnormal pathways can yield ideal therapeutic effects in patients, even by reversing drug resistance. However, these deep TME signaling pathways often exhibit crosstalk and correlation. Understanding these interactions may be an important strategy for further treating ovarian cancer. This review summarizes the latest progress and therapeutic strategies for these angiogenic signaling pathways in ovarian cancer.
Collapse
Affiliation(s)
- Wenchao Zhou
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tian Zeng
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Junling Chen
- Department of Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Yuwei Yuan
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Daopu Hu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yue Zhang
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| | - Juan Zou
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
5
|
Brand J, Haro M, Lin X, Rimel B, McGregor SM, Lawrenson K, Dinh HQ. Fallopian tube single cell analysis reveals myeloid cell alterations in high-grade serous ovarian cancer. iScience 2024; 27:108990. [PMID: 38384837 PMCID: PMC10879678 DOI: 10.1016/j.isci.2024.108990] [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: 10/02/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Most high-grade serous ovarian cancers (HGSCs) likely initiate from fallopian tube (FT) epithelia. While epithelial subtypes have been characterized using single-cell RNA-sequencing (scRNA-Seq), heterogeneity of other compartments and their involvement in tumor progression are poorly defined. Integrated analysis of human FT scRNA-Seq and HGSC-related tissues, including tumors, revealed greater immune and stromal transcriptional diversity than previously reported. We identified abundant monocytes in FTs across two independent cohorts. The ratio of macrophages to monocytes is similar between benign FTs, ovaries, and adjacent normal tissues but significantly greater in tumors. FT-defined monocyte and macrophage signatures, cell-cell communication, and gene set enrichment analyses identified monocyte- and macrophage-specific interactions and functional pathways in paired tumors and adjacent normal tissues. Further reanalysis of HGSC scRNA-Seq identified monocyte and macrophage subsets associated with neoadjuvant chemotherapy. Taken together, our work provides data that an altered FT myeloid cell composition could inform the discovery of early detection markers for HGSC.
Collapse
Affiliation(s)
- Joshua Brand
- McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI 53705, USA
| | - Marcela Haro
- Women’s Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xianzhi Lin
- Women’s Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- RNA Biology Group, Division of Natural and Applied Sciences and Global Health Research Center, Duke Kunshan University, Kunshan 215316, Jiangsu Province, China
| | - B.J. Rimel
- Women’s Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stephanie M. McGregor
- Department of Pathology and Laboratory Medicine, University of Wisconsin – Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Kate Lawrenson
- Women’s Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Huy Q. Dinh
- McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI 53705, USA
| |
Collapse
|
6
|
Gendrau‐Sanclemente N, Figueras A, Gracova K, Lahiguera Á, Alsina‐Sanchís E, Marín‐Jiménez JA, Vidal A, Matias‐Guiu X, Fernandez‐Gonzalez S, Barahona M, Martí L, Ponce J, Viñals F. Ovarian cancer relies on the PDGFRβ-fibronectin axis for tumorsphere formation and metastatic spread. Mol Oncol 2024; 18:136-155. [PMID: 38010623 PMCID: PMC10766197 DOI: 10.1002/1878-0261.13556] [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: 08/17/2023] [Revised: 10/10/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is the deadliest gynecological malignancy. The most common form of metastatic spread of HGSOC is transcoelomic dissemination. In this process, detached cells from the primary tumor aggregate as tumorspheres and promote the accumulation of peritoneal ascites. This represents an early event in HGSOC development and is indicative of poor prognosis. In this study, based on tumorspheres isolated from ascitic liquid samples from HGSOC patients, ovarian cancer spheroid 3D cultures, and in vivo models, we describe a key signal for tumorsphere formation in HGSOC. We report that platelet-derived growth factor receptor beta (PDGFRβ) is essential for fibronectin-mediated cell clustering of ovarian cancer cells into tumorspheres. This effect is mediated by the kinase NUAK family SNF1-like kinase 1 (NUAK1) and blocked by PDGFRβ pharmacological or genetic inhibition. In the absence of PDGFRβ, ovarian cancer cells can be provided with fibronectin by cancer-associated fibroblasts to generate chimeric spheroids. This work provides new insights that uncover potential targets to prevent peritoneal dissemination, the main cause of advanced disease in HGSOC patients.
Collapse
Affiliation(s)
- Núria Gendrau‐Sanclemente
- Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of Oncology (ICO), Hospital Duran i ReynalsBarcelonaSpain
- Oncobell ProgramInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL)BarcelonaSpain
| | - Agnès Figueras
- Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of Oncology (ICO), Hospital Duran i ReynalsBarcelonaSpain
- Oncobell ProgramInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL)BarcelonaSpain
| | - Kristina Gracova
- Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of Oncology (ICO), Hospital Duran i ReynalsBarcelonaSpain
- Oncobell ProgramInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL)BarcelonaSpain
| | - Álvaro Lahiguera
- Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of Oncology (ICO), Hospital Duran i ReynalsBarcelonaSpain
- Oncobell ProgramInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL)BarcelonaSpain
| | - Elisenda Alsina‐Sanchís
- Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of Oncology (ICO), Hospital Duran i ReynalsBarcelonaSpain
- Oncobell ProgramInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL)BarcelonaSpain
| | - Juan A. Marín‐Jiménez
- Cancer Immunotherapy (CIT) Group‐ProCUREBellvitge Biomedical Research Institute (IDIBELL) – OncoBellBarcelonaSpain
- Department of Medical OncologyCatalan Institute of Oncology (ICO)BarcelonaSpain
| | - August Vidal
- Oncobell ProgramInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL)BarcelonaSpain
- Department of PathologyUniversity Hospital of Bellvitge (IDIBELL)BarcelonaSpain
- CIBERONCInstituto de Salud Carlos IIIMadridSpain
| | - Xavier Matias‐Guiu
- Oncobell ProgramInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL)BarcelonaSpain
- Department of PathologyUniversity Hospital of Bellvitge (IDIBELL)BarcelonaSpain
- CIBERONCInstituto de Salud Carlos IIIMadridSpain
| | | | - Marc Barahona
- Department of GynaecologyUniversity Hospital of Bellvitge (IDIBELL)BarcelonaSpain
| | - Lola Martí
- Department of GynaecologyUniversity Hospital of Bellvitge (IDIBELL)BarcelonaSpain
| | - Jordi Ponce
- Department of GynaecologyUniversity Hospital of Bellvitge (IDIBELL)BarcelonaSpain
| | - Francesc Viñals
- Program Against Cancer Therapeutic Resistance (ProCURE)Catalan Institute of Oncology (ICO), Hospital Duran i ReynalsBarcelonaSpain
- Oncobell ProgramInstitut d'Investigació Biomèdica de Bellvitge (IDIBELL)BarcelonaSpain
- Departament de Ciències FisiològiquesUniversitat de BarcelonaSpain
| |
Collapse
|
7
|
Kozłowski M, Borzyszkowska D, Mirko J, Turoń-Skrzypińska A, Piotrowska K, Tołoczko-Grabarek A, Kwiatkowski S, Tarnowski M, Rotter I, Cymbaluk-Płoska A. Preoperative Serum Levels of PDGF-AB, PDGF-BB, TGF-α, EGF and ANG-2 in the Diagnosis of Endometrial Cancer. Cancers (Basel) 2023; 15:4815. [PMID: 37835508 PMCID: PMC10571811 DOI: 10.3390/cancers15194815] [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: 08/05/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
(1) Background: It is relevant to find new diagnostic biomarkers for endometrial cancer. This study aimed to investigate whether PDGF-AB, PDGF-BB, TGF-α, EGF and ANG-2 could be considered new useful markers for diagnosis and survival of endometrial cancer. (2) Methods: A total of 93 women diagnosed with endometrial cancer (EC) and 66 patients with non-cancerous endometrial lesions (NCEL) were included in this study. (3) Results: Median serum levels of PDGF-AB, PDGF-BB, TGF-α, EGF and ANG-2 were significantly higher in the EC group compared to the NCEL group (for PDGF-AB, PDGF-BB, TGF-α and ANG-2, p = 0.0000; for EGF, p = 0.0186). The cut-off level of PDGF-AB was set at 127.69 pg/mL with a sensitivity of 87.1% and a specificity of 66.67% (AUC = 0.78, p < 0.000001). The cut-off level of PDGF-BB was set at 207.86 ng/L with a sensitivity of 82.8% and a specificity of 75.76% (AUC = 0.85, p < 0.000001). The cut-off level of TGF-α was set at 33.85 ng/L with a sensitivity of 82.8% and a specificity of 75.76% (AUC = 0.82, p < 0.000001). The cut-off level of EGF was set at 934.76 pg/mL with a sensitivity of 83.87% and a specificity of 28.79% (AUC = 0.61, p = 0.018472). The cut-off level of ANG-2 was set at 3120.68 pg/mL with a sensitivity of 72.04% and a specificity of 93.94% (AUC = 0.87, p < 0.000001). (4) Conlusion: It was concluded that all the proteins studied could be potential diagnostic markers in endometrial cancer.
Collapse
Affiliation(s)
- Mateusz Kozłowski
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland (A.C.-P.)
| | - Dominika Borzyszkowska
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland (A.C.-P.)
| | - Justyna Mirko
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland (A.C.-P.)
| | - Agnieszka Turoń-Skrzypińska
- Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University in Szczecin, Żołnierska 48, 71-210 Szczecin, Poland
| | - Katarzyna Piotrowska
- Department of Physiology, Pomeranian Medical University in Szczecin, al. Powstancow Wlkp. 72, 70-111 Szczecin, Poland
| | | | - Sebastian Kwiatkowski
- Department of Obstetrics and Gynecology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Żołnierska 54, 70-210 Szczecin, Poland
| | - Iwona Rotter
- Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University in Szczecin, Żołnierska 48, 71-210 Szczecin, Poland
| | - Aneta Cymbaluk-Płoska
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland (A.C.-P.)
| |
Collapse
|
8
|
Zhao Z, Li T, Yuan Y, Zhu Y. What is new in cancer-associated fibroblast biomarkers? Cell Commun Signal 2023; 21:96. [PMID: 37143134 PMCID: PMC10158035 DOI: 10.1186/s12964-023-01125-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/05/2023] [Indexed: 05/06/2023] Open
Abstract
The tumor microenvironment is one of the important drivers of tumor development. Cancer-associated fibroblasts (CAFs) are a major component of the tumor stroma and actively participate in tumor development, invasion, metastasis, drug resistance, and other biological behaviors. CAFs are a highly heterogeneous group of cells, a reflection of the diversity of their origin, biomarkers, and functions. The diversity of CAF origin determines the complexity of CAF biomarkers, and CAF subpopulations expressing different biomarkers may play contrasting roles in tumor progression. In this review, we provide an overview of these emerging CAF biomarkers and the biological functions that they suggest, which may give a better understanding of the relationship between CAFs and tumor cells and be of great significance for breakthroughs in precision targeted therapy for tumors. Video Abstract.
Collapse
Affiliation(s)
- Zehua Zhao
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), No. 44 of Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Tianming Li
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), No. 44 of Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, No. 155 of Nanjing Road, Heping District, Shenyang, 110001, China.
| | - Yanmei Zhu
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), No. 44 of Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| |
Collapse
|
9
|
Mei C, Gong W, Wang X, Lv Y, Zhang Y, Wu S, Zhu C. Anti-angiogenic therapy in ovarian cancer: Current understandings and prospects of precision medicine. Front Pharmacol 2023; 14:1147717. [PMID: 36959862 PMCID: PMC10027942 DOI: 10.3389/fphar.2023.1147717] [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: 01/19/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Ovarian cancer (OC) remains the most fatal disease of gynecologic malignant tumors. Angiogenesis refers to the development of new vessels from pre-existing ones, which is responsible for supplying nutrients and removing metabolic waste. Although not yet completely understood, tumor vascularization is orchestrated by multiple secreted factors and signaling pathways. The most central proangiogenic signal, vascular endothelial growth factor (VEGF)/VEGFR signaling, is also the primary target of initial clinical anti-angiogenic effort. However, the efficiency of therapy has so far been modest due to the low response rate and rapidly emerging acquiring resistance. This review focused on the current understanding of the in-depth mechanisms of tumor angiogenesis, together with the newest reports of clinical trial outcomes and resistance mechanism of anti-angiogenic agents in OC. We also emphatically summarized and analyzed previously reported biomarkers and predictive models to describe the prospect of precision therapy of anti-angiogenic drugs in OC.
Collapse
Affiliation(s)
- Chao Mei
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijing Gong
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Xu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongning Lv
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sanlan Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Chunqi Zhu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|