1
|
Ji K, Wen B, Wang X, Chen L, Chen Y, Wang L, Bao J, Pan X, Zhang G, Jiang Y, Liu H. HIF1A facilitates hypoxia-induced changes in H3K27ac modification to promote myometrial contractility. Commun Biol 2025; 8:475. [PMID: 40119120 PMCID: PMC11928739 DOI: 10.1038/s42003-025-07880-9] [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: 08/07/2024] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
Prior studies have established that myometrial hypoxia during labor is pivotal in intensifying contractions, the alterations in gene expression and histone modifications in myometrial cells under hypoxia have yet to be documented. Here, hypoxia's enhancement of cellular contractility was confirmed, and RNA-seq identified 2,262 differentially expressed genes in human myometrial smooth muscle cells (hMSMCs) under hypoxia. Chromatin immunoprecipitation (ChIP), high-throughput chromosome conformation capture followed by ChIP (Hi-ChIP) were employed to investigate the epigenetic changes, specifically histone modifications (H3K27ac, H3K4me1, H3K27me3, and H3K4me3), in hMSMCs under hypoxia. We identified the enhancer and super-enhancer regions in hMSMCs and found HIF1A as the key mediator of these H3K27ac changes under hypoxia. Labor-associated genes regulated by HIF1A have been identified. Validation experiments on these genes such as CXCL8, RUNX1, IL-6, and PTGES3 demonstrated that HIF1A knockdown reduces their expression and associated H3K27ac modifications in peak regions of their promoters or enhancers. These findings indicate that HIF1A probably mediate changes in histone H3K27ac modifications to regulate myometrial cell contractions under hypoxia, providing potential therapeutic and intervention targets for disorders related to parturition.
Collapse
Affiliation(s)
- Kaiyuan Ji
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bolun Wen
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaodi Wang
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lina Chen
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yunshan Chen
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lele Wang
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junjie Bao
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiuyu Pan
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guozheng Zhang
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanmin Jiang
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Huishu Liu
- Guangzhou Key Laboratory of Maternal-Fetal Medicine, Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Yong W, Jian Y, Wang Q, Fei K, Li P. AC092100.1 promotes angiogenesis in pre-eclampsia through YTHDC2/VEGFA signaling. Funct Integr Genomics 2024; 24:157. [PMID: 39237822 DOI: 10.1007/s10142-024-01428-6] [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: 04/17/2024] [Revised: 07/31/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
Aberrant long non-coding RNA (lncRNA) expression has been shown to be involved in the pathological process of pre-eclampsia (PE), yet only a small portion of lncRNAs has been characterized concerning the function and molecular mechanisms involved in PE. This study aimed to investigate the regulatory mechanism of the lncRNA AC092100.1 (AC092100.1) in angiogenesis in PE. In our study, bioinformatics analysis was performed to screen for differentially expressed lncRNAs between normal subjects and PE patients. The levels of AC092100.1 in placental tissues of patients with or without PE were validated using qRT-PCR. The effect of AC092100.1 overexpression on the proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) was investigated. The binding of AC092100.1 and YT521-B homology domain-containing 2 (YTHDC2) was predicted and verified. The effect of AC092100.1/YTHDC2 on the expression of vascular endothelial growth factor-A (VEGFA) in HUVECs was determined. Finally, a PE mice model was conducted. Fetal mouse growth, the abundance of mesenchymal morphology markers, including hypoxia-inducible factor 1-alpha (HIF-1α), soluble fms-like tyrosine kinase-1 (sFlt-1), soluble endoglin (sEng), Slug, and Vimentin, and endothelial markers, including placental growth factor (PLGF), CD31, and vascular endothelial (VE)-cadherin, in placental tissues were assessed. Here, we found that AC092100.1 was abnormally downregulated in placental tissues from PE patients. We established that AC092100.1 overexpression promoted HUVEC proliferation, migration, and tube formation in vitro. Mechanistically, AC092100.1 induced the accumulation of YTHDC2 and VEGFA through binding to YTHDC2 in HUVECs. Inhibition of YTHDC2 or VEGFA reversed AC092100.1-promoted tube formation. AC092100.1 overexpression contributed to alleviating fetal growth disorder, decreased levels of sEng, HIF-1α, sFlt-1, Slug, and Vimentin, and increased levels of VEGFA, PLGF, CD31, and VE-cadherin in PE mice. Our findings provided evidence supporting the role of the AC092100.1/YTHDC2/VEGFA axis in regulating angiogenesis, which demonstrated a therapeutic pathway for PE targeting angiogenesis.
Collapse
Affiliation(s)
- Wenjing Yong
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, Hunan, China
| | - Yu Jian
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, Hunan, China
| | - Qi Wang
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, Hunan, China
| | - Kuilin Fei
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, Hunan, China
| | - Ping Li
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, Hunan, China.
| |
Collapse
|
3
|
Qin Y, Zhao W, Li Q, Cai Z, Wang G, Wang N, Ma L. Inhibition of Chitinase-3-like Protein 1 Reduced Epithelial-Mesenchymal Transition and Vascular Epithelial Cadherin Expression in Oesophageal Squamous Cell Carcinoma. IRANIAN JOURNAL OF BIOTECHNOLOGY 2024; 22:e3693. [PMID: 39737205 PMCID: PMC11682521 DOI: 10.30498/ijb.2024.394737.3693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/16/2024] [Indexed: 01/01/2025]
Abstract
Background Oesophageal cancer (EC) is one of the common malignant tumors, and the prognosis of patients is poor. Further exploration of EC pathogenesis remains warranted. Objective The relationship between vascular epithelial cadherin (VE-cadherin) and chitinase-3-like protein 1 (CHI3L1) in EC is currently unknown. To further explore the relationship, immunohistochemical staining was performed to detect the expression level of CHI3L1 and VE-cadherin in oesophageal squamous cell carcinoma ( ESCC). Materials and Methods Small interfering RNAs (siRNAs) inhibited CHI3L1 expression in KYSE-150 and TE1 cells. Western blot and quantitative fluorescence polymerase chain reaction were used to detect the levels of CHI3L1, VE-cadherin and epithelial-mesenchymal transition (EMT)-related proteins in vitro and in vivo, and KYSE-150 cells were used to establish an in-vivo model and observe tumour growth. Results High levels of CHI3L1 and VE-cadherin expression were closely associated with the progression of ESCC; the pathologic tumour-node-metastasis stage was also closely related with the progression of ESCC (p < 0.05). High levels of CHI3L1 and VE-cadherin expression led to poor prognosis in patients with EC. In KYSE-150 and TE1 EC cell lines, the invasion, migration and proliferation of cells decreased, and the apoptotic rate increased after CHI3L1 expression was decreased using siRNA. The CHI3L1, VE-cadherin, Snail, Twist1 protein and mRNA expression levels decreased, whereas the E-cadherin levels increased. Conclusions Chitinase-3-like protein 1 could promote the EMT of ESCC, and the inhibition of CHI3L1 decreases the expression of VE-cadherin, which inhibits tumour angiogenesis and tumour progression in ESCC.
Collapse
Affiliation(s)
- Yanzi Qin
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, China
| | - Wenjun Zhao
- Department of Emergency Internal Medicine, The Third the People′s Hospital of Bengbu, Bengbu 233000, China
| | - Qicai Li
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Bengbu Medical University,Bengbu 233000, China
| | - Zhaogeng Cai
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, China
| | - Guowen Wang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Bengbu Medical University,Bengbu 233000, China
| | - Nan Wang
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Li Ma
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, China
| |
Collapse
|
4
|
Shi Y, Li W, Jia Q, Wu J, Wu S, Wu S. Inhibition of PD-L1 expression in non-small cell lung cancer may reduce vasculogenic mimicry formation by inhibiting the epithelial mesenchymal transformation process. Exp Cell Res 2024; 437:113996. [PMID: 38508327 DOI: 10.1016/j.yexcr.2024.113996] [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: 11/01/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024]
Abstract
Non-small cell lung cancer (NSCLC) is a kind of highly malignant tumor. Studies have shown that Vasculogenic mimicry (VM) may be responsible for dismal prognosis in NSCLC. Immunotherapy with programmed death-1 (PD-1) or programmed death ligand-1 (PD-L1) has significantly altered the treatment of assorted cancers, including NSCLC, but its role and mechanism in the formation of Vasculogenic mimicry (VM) in NSCLC remains unclear. This study aimed to investigate the role of the anti-PD-L1 antibody in the formation of VM in NSCLC and its possible mechanisms. The results showed that anti-PD-L1 antibody therapy could inhibit the growth of NSCLC-transplanted tumors and reduce the formation of VMs. In addition, this study found that anti-PD-L1 antibodies could increase the expression of the epithelial-mesenchymal transition (EMT) related factor E-cadherin. zinc finger E-box binding homeobox 1 (ZEB1) is an important transcription factor regulating EMT. Knocking down ZEB1 could significantly inhibit tumor growth, as well as the expression of VE-cadherin and mmp2, while remarkably increase the expression of E-cadherin. During this process, the formation of VM was inhibited by knowing down ZEB1 in both in vitro and in vivo experiments of the constructed ZEB1 knockdown stable transfected cell strains. Therefore, in this study, we found that anti-PD-L1 antibodies may reduce the formation of VMs by inhibiting the EMT process.
Collapse
Affiliation(s)
- Yuqi Shi
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, Anhui, 233000, China; Department of Pathology, School of Basic Medicine, Bengbu Medical University, Anhui, 233000, China; Key Laboratory of Cancer Translational Medicine, Bengbu Medical University, Anhui, 233000, China
| | - Wenjuan Li
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, Anhui, 233000, China
| | - Qianhao Jia
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, Anhui, 233000, China
| | - Jiatao Wu
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital, Bengbu Medical University, 287 Changhuai Road, Bengbu 233004, Anhui, China
| | - Shoufan Wu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, Anhui, 233000, China
| | - Shiwu Wu
- Department of Pathology, Anhui No. 2 Provincial People's Hospital, Anhui, 230000, China; Anhui Province Key Laboratory of Occupational Health, Anhui No. 2 Provincial People's Hospital, Hefei, 230041, China.
| |
Collapse
|
5
|
Dreute J, Pfisterer M, Schmitz ML. A reductionist perspective on HIF-1α's role in cell proliferation under non-hypoxic conditions. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119683. [PMID: 38301905 DOI: 10.1016/j.bbamcr.2024.119683] [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: 10/18/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 02/03/2024]
Abstract
The role of hypoxia-inducible factor (HIF)-1α in the control of proliferation under non-hypoxic conditions has been investigated in numerous studies, but does not yield a coherent picture. Therefore, we conducted this meta-analysis of existing literature to systematically evaluate the role of HIF-1α, based on a number of inclusion and exclusion criteria. Studies analyzing non-transformed, primary cells showed a largely heterogeneous distribution of pro-proliferative, anti-proliferative or absent functions for HIF-1α, which are co-determined by several parameters, including the type and age of the cell and its localization in tissues and organs. In contrast, the analyses of tumor cells showed a predominantly pro-proliferative role of HIF-1α by cell-intrinsic and cell-extrinsic molecular mechanism not yet understood.
Collapse
Affiliation(s)
- Jan Dreute
- Institute of Biochemistry, Justus-Liebig-University Giessen, Germany
| | | | | |
Collapse
|
6
|
Wang J, Zhu L, Zhang Q, Xia T, Yao W, Wei L. LincRNA-P21 knockdown facilitates esophageal squamous cell carcinoma cell progression by upregulating cadherin 5 via YTH domain containing 1. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1797-1805. [PMID: 37766459 PMCID: PMC10686791 DOI: 10.3724/abbs.2023154] [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: 12/27/2022] [Accepted: 05/19/2023] [Indexed: 09/29/2023] Open
Abstract
LincRNA-P21 is a tumor suppressor in esophageal squamous cell carcinoma (ESCC). Cell adhesion modules play vital roles in cell-cell and cell-extracellular matrix (ECM) interactions and malignant cancer progression. In this study, we investigate whether lincRNA-P21 exerts its functions by regulating the cell adhesion molecule cadherin 5 (CDH5) in ESCC. Moreover, the RNA binding protein (RBP) mediators of lincRNA-P21 and CDH5 are further examined. Cell viability, growth and migratory ability are assessed by calcein-AM/PI double staining, CCK-8, EdU, Transwell, and wound healing assays. The expression of collagen I and fibronectin is examined by immunofluorescence (IF). LincRNA-P21 and CDH5 are quantified by RT-qPCR and western blot analysis. Potential lincRNA-P21 targets are identified by RNA sequencing. RBPs that can interact with lincRNA-P21 and CDH5 are identified by RNA immunoprecipitation (RIP) assay. LincRNA-P21 knockdown increases cell viability, growth, cell migration, and collagen I and fibronectin expression in ESCC cells. LincRNA-P21 depletion induces the dysregulation of 316 genes, including CDH5, in TE-1 cells. CDH5 is identified as a downstream molecule of lincRNA-P21 given its close correlation with cell adhesion, ECM reconstruction, and cancer progression. LincRNA-P21 exerts its functions by negatively regulating CDH5 expression. YTH domain containing 1 (YTHDC1) mediates the regulatory effect of lincRNA-P21 on CDH5. LincRNA-P21 knockdown elevates cell viability and growth, promotes cell migration, and induces ECM reorganization by upregulating CDH5 via RBP YTHDC1 in ESCC.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of Thoracic SurgeryHenan Provincial People’s HospitalPeople’s Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhou450003China
| | - Li Zhu
- Department of Thoracic SurgeryZhengzhou University People’s HospitalHenan Provincial People’s HospitalZhengzhou450003China
| | - Quan Zhang
- Department of Thoracic SurgeryHenan Provincial People’s HospitalPeople’s Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhou450003China
| | - Tian Xia
- Department of Thoracic SurgeryHenan Provincial People’s HospitalPeople’s Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhou450003China
| | - Wenjian Yao
- Department of Thoracic SurgeryHenan Provincial People’s HospitalPeople’s Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhou450003China
| | - Li Wei
- Department of Thoracic SurgeryHenan Provincial People’s HospitalPeople’s Hospital of Zhengzhou UniversitySchool of Clinical MedicineHenan UniversityZhengzhou450003China
| |
Collapse
|
7
|
Du Q, Lin Y, Ding C, Wu L, Xu Y, Feng Q. Pharmacological Activity of Matrine in Inhibiting Colon Cancer Cells VM Formation, Proliferation, and Invasion by Downregulating Claudin-9 Mediated EMT Process and MAPK Signaling Pathway. Drug Des Devel Ther 2023; 17:2787-2804. [PMID: 37719361 PMCID: PMC10504061 DOI: 10.2147/dddt.s417077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/25/2023] [Indexed: 09/19/2023] Open
Abstract
Purpose Matrine (Mat), the main active ingredient of traditional Chinese herbal plant Sophora flavescens Ait, has significant antitumor effects, but its pharmacological mechanism on colon cancer (CC) remains unclear. This study aimed to investigate the therapeutic effect of Mat on CC as well as the potential mechanism. Methods The vasculogenic mimicry (VM) of CC cells was observed by three-dimensional (3D) Matrigel cell culture. Cell proliferation, apoptosis, migration, invasion, and actin filament integrity were detected by CCK8, flow cytometry, wound healing, Transwell and Phalloidin staining assays. qRT-PCR and Western blotting were applied to detect the expression of EMT factors. RNA-sequencing was conducted to screen differentially expressed genes (DEGs), and the GO and KEGG pathway enrichment analyses were performed. Then, the expression of the key MAPK pathway genes and the target gene Claudin-9 (Cldn9) were analyzed. RNA interference was used to silence Cldn9 expression, and the effects of Cldn9 silencing and simultaneous treatment with Mat on VM formation, proliferation, apoptosis, invasion, and migration were investigated. Finally, the expression of EMT factors and MAPK pathway key genes was detected. Results CT26 cells formed the most typical VM structure. Mat disrupted the VM of CT26 cells, significantly suppressed their proliferation, migration, invasion, actin filament integrity, induced apoptosis, and inhibited EMT process. RNA-sequencing revealed 163 upregulated genes and 333 downregulated genes in Mat-treated CT26 cells, and the DEGs were significantly enriched in cell adhesion molecules and MAPK signaling pathways. Further confirmed that Mat significantly inhibited the phosphorylation levels of JNK and ERK, and the target gene Cldn9 was significantly upregulated in human CC tissues. Silencing Cldn9 markedly inhibited the VM, proliferative activity, invasiveness, and actin filament integrity of CT26 cells, blocked the EMT process, and downregulated the phosphorylation of JNK and ERK, whereas Mat intervention further strengthened the above trends. Conclusion This study indicated that Mat may synergistically inhibit the EMT process and MAPK signaling pathway through downregulation Cldn9, thereby exerting pharmacological effects on inhibiting VM formation, proliferation, and invasion of CC cells.
Collapse
Affiliation(s)
- Qiu Du
- Department of Neurosurgery, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
- Department of Central Laboratory, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
| | - Yingda Lin
- Department of Pharmacy, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
- Department of Pharmacy, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, People’s Republic of China
| | - Changping Ding
- Department of Medical Laboratory, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
| | - Ling Wu
- Department of Pharmacy, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
| | - Yuan Xu
- Department of Pharmacy, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
| | - Qingling Feng
- Department of Emergency Intensive Care Unit, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
| |
Collapse
|
8
|
Andreucci E, Peppicelli S, Ruzzolini J, Bianchini F, Calorini L. Physicochemical aspects of the tumour microenvironment as drivers of vasculogenic mimicry. Cancer Metastasis Rev 2022; 41:935-951. [PMID: 36224457 PMCID: PMC9758104 DOI: 10.1007/s10555-022-10067-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/04/2022] [Indexed: 01/25/2023]
Abstract
Tumour vascularisation is vital for cancer sustainment representing not only the main source of nutrients and oxygen supply but also an escape route for single or clustered cancer cells that, once detached from the primary mass, enter the blood circulation and disseminate to distant organs. Among the mechanisms identified to contribute to tumour vascularisation, vasculogenic mimicry (VM) is gaining increasing interest in the scientific community representing an intriguing target for cancer treatment. VM indeed associates with highly aggressive tumour phenotypes and strongly impairs patient outcomes. Differently from vessels of healthy tissues, tumour vasculature is extremely heterogeneous and tortuous, impeding efficient chemotherapy delivery, and at the meantime hyperpermeable and thus extremely accessible to metastasising cancer cells. Moreover, tumour vessel disorganisation creates a self-reinforcing vicious circle fuelling cancer malignancy and progression. Because of the inefficient oxygen delivery and metabolic waste removal from tumour vessels, many cells within the tumour mass indeed experience hypoxia and acidosis, now considered hallmarks of cancer. Being strong inducers of vascularisation, therapy resistance, inflammation and metastasis, hypoxia and acidosis create a permissive microenvironment for cancer progression and dissemination. Along with these considerations, we decided to focus our attention on the relationship between hypoxia/acidosis and VM. Indeed, besides tumour angiogenesis, VM is strongly influenced by both hypoxia and acidosis, which could potentiate each other and fuel this vicious circle. Thus, targeting hypoxia and acidosis may represent a potential target to treat VM to impair tumour perfusion and cancer cell sustainment.
Collapse
Affiliation(s)
- Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
9
|
SPHK/HIF-1α Signaling Pathway Has a Critical Role in Chrysin-Induced Anticancer Activity in Hypoxia-Induced PC-3 Cells. Cells 2022; 11:cells11182787. [PMID: 36139362 PMCID: PMC9496844 DOI: 10.3390/cells11182787] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/12/2022] [Accepted: 09/02/2022] [Indexed: 01/30/2023] Open
Abstract
Hypoxia, a typical feature of locally advanced solid tumors including prostate cancer, is a critical contributor to tumor progression and causes resistance to therapy. In this study, we investigated the effects of chrysin on tumor progression in hypoxic PC-3 cells. Chrysin exerted a significant inhibitory effect on 3D cell growth under normoxic and hypoxic conditions. It also decreased the hypoxia-induced vasculogenic mimicry and attenuated the expression of HIF-1α and VE-cadherin. Chrysin inhibited HIF-1α accumulation in a concentration- and time-dependent manner in hypoxic PC-3 cells, while also suppressing the expression of HIF-1α by inhibiting SPHK-1 in both CoCl2 and hypoxic PC-3 cells. At high concentrations of chrysin, there was a greater increase in apoptosis in the hypoxic cells compared to that in normoxic cells, which was accompanied by sub-G1 phase arrest. Chrysin-induced apoptosis inhibited VEGF and Bcl-2 and induced the cleavage of PARP and caspase-3. SPHK-1 knockdown induced apoptosis and inhibited epithelial–mesenchymal transition. Consistent with the in vitro data, 50 mg/kg of chrysin suppressed the tumor growth of PC-3 xenografts by 80.4% compared to that in the untreated control group. The immunohistochemistry of tumor tissues revealed decreased Ki-67, HIF-1α, and VEGF expression in the chrysin-treated group compared to an untreated control. Western blotting data for tumor tissues showed that chrysin treatment decreased SPHK-1, HIF-1α, and PARP expression while inducing caspase-3 cleavage. Overall, our findings suggest that chrysin exerts anti-tumor activity by inhibiting SPHK-1/HIF-1α signaling and thus represents a potent chemotherapeutic agent for hypoxia, which promotes cancer progression and is related to poor prognoses in prostate cancer patients.
Collapse
|
10
|
The Biomarker Like the Correlation between Vasculogenic Mimicry, Vascular Endothelial Cadherin, Sex-DeterminingRegion on Y-Box Transcription Factor 17, and Cyclin D1 in Oesophageal Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:8915503. [PMID: 36072972 PMCID: PMC9444392 DOI: 10.1155/2022/8915503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/22/2022] [Indexed: 12/24/2022]
Abstract
Background This study aimed to explore the relationships between the sex-determining region on Y (SRY) box transcription factor 17 (SOX17), Cyclin D1, vascular endothelial cadherin (VE-cadherin), and vasculogenic mimicry (VM) in the occurrence and development of esophageal squamous cell carcinoma (ESCC). Methods The expressions of SOX17, Cyclin D1, and VE-cadherin, as well as VM, in tissues, were determined using immunohistochemistry. SOX17, Cyclin D1, and VE-cadherin mRNA in ESCC and their corresponding adjacent normal tissues were quantified using quantitative reverse transcription polymerase chain reaction analysis. Cell invasion, migration, and proliferation were determined after the silencing of VE-cadherin. SOX17, Cyclin D1, and VE-cadherin protein were quantified using Western blotting. Results The expression levels of SOX17, Cyclin D1, and VE-cadherin significantly correlated with the clinical characteristics of ESCC. After the VE-cadherin silencing, cell invasion, migration, and proliferation decreased, along with the Cyclin D1 levels, while the SOX17 levels increased. Conclusion SOX17, Cyclin D1, and VE-cadherin are involved in the development of ESCC.
Collapse
|
11
|
Hu H, Ma T, Liu N, Hong H, Yu L, Lyu D, Meng X, Wang B, Jiang X. Immunotherapy checkpoints in ovarian cancer vasculogenic mimicry: Tumor immune microenvironments, and drugs. Int Immunopharmacol 2022; 111:109116. [PMID: 35969899 DOI: 10.1016/j.intimp.2022.109116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 02/09/2023]
Abstract
Vasculogenic mimicry (VM), a vessel-like structure independent of endothelial cells, commonly exists in solid tumors which requires blood vessels to grow. As a special source of blood supply for tumor progression to a more aggressive state, VM has been observed in a variety of human malignant tumors and is tightly associated with tumor proliferation, invasion, metastasis, and poor patient prognosis. So far, various factors, including immune cells and cytokines, were reported to regulate ovarian cancer progression by influencing VM formation. Herein, we review the mechanisms that regulate VM formation in ovarian cancer and the effect of cells, cytokines, and signaling molecules in the tumor microenvironment on VM formation, Furthermore, we summarize the current clinical application of drugs targeting VM formation.
Collapse
Affiliation(s)
- Haitao Hu
- Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, PR China.
| | - Ting Ma
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Nanqi Liu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Hong Hong
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, PR China.
| | - Lujiao Yu
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, PR China.
| | - Dantong Lyu
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, Shenyang 110122, Liaoning Province, PR China.
| | - Biao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning Province, PR China.
| | - Xuefeng Jiang
- Department of Immunology, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
12
|
Wu BC, Hsu ATW, Abadchi SN, Johnson CR, Bengali S, Lay F, Melinosky K, Shao C, Chang KH, Born LJ, Abraham J, Evans D, Ha JS, Harmon JW. Potential Role of Silencing Ribonucleic Acid for Esophageal Cancer Treatment. J Surg Res 2022; 278:433-444. [PMID: 35667884 DOI: 10.1016/j.jss.2022.04.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Esophageal cancer is an aggressive malignancy with high mortality. Optimal treatment of esophageal cancer remains an elusive goal. Ribonucleic acid (RNA) interference is a novel potential targeted approach to treat esophageal cancer. Targeting oncogenes that can alter critical cellular functions with silencing RNA molecules is a promising approach. The silencing of specific oncogenes in esophageal cancer cells in the experimental setting has been shown to decrease the expression of oncogenic proteins. This has resulted in cell apoptosis, reduction in cell proliferation, reduced invasion, migration, epithelial-mesenchymal transition, decrease in tumor angiogenesis and metastasis, and overcoming drug resistance. The Hedgehog (Hh) signaling pathway has been shown to be involved in esophageal adenocarcinoma formation in a reflux animal model. In addition to Hh, we will focus on other targets with clinical potential in the treatment of esophageal cancer. MATERIALS AND METHODS We searched for articles published from 2005 to August 2020 that studied the siRNA effects on inhibiting esophageal cancer formation in experimental settings. We used combinations of the following terms for searching: "esophageal cancer," "RNA interference," "small interfering RNA," "siRNA," "silencing RNA," "Smoothened (Smo)," "Gli," "Bcl-2," "Bcl-XL," "Bcl-W,″ "Mcl-1," "Bfl-1," "STAT3,"and "Hypoxia inducible factor (HIF)". A total of 21 relevant articles were found. RESULTS AND CONCLUSIONS Several proto-oncogenes/oncogenes including Hh pathway mediators, glioma-associated oncogene homolog 1 (Gli-1), Smoothened (Smo), and antiapoptotic Bcl-2 have potential as targets for silencing RNA in the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Bo-Chang Wu
- Bayview Surgical Research Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Angela Ting-Wei Hsu
- Bayview Surgical Research Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sanaz Nourmohammadi Abadchi
- Bayview Surgical Research Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher R Johnson
- Bayview Surgical Research Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of Thoracic Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sameer Bengali
- Bayview Surgical Research Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Frank Lay
- Bayview Surgical Research Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kelsey Melinosky
- Bayview Surgical Research Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Kai-Hua Chang
- Bayview Surgical Research Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Louis J Born
- Department of Bioengineering, University of Maryland, College Park, College Park, Maryland
| | - John Abraham
- Bayview Surgical Research Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Jinny S Ha
- Division of Thoracic Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John W Harmon
- Bayview Surgical Research Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
13
|
Xu Y, Fu L, Pan D, Wei J, Xia H, Wang S, Sun G. Folic Acid Inhibited Vasculogenic Mimicry in Esophageal Cancer Cell Line Eca-109, the One Target Was EphA2. Nutr Cancer 2021; 74:2235-2242. [PMID: 34678082 DOI: 10.1080/01635581.2021.1988992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
The degree of vasculogenic mimicry(VM) is correlated with the prognosis of esophageal cancer, and folic acid supplementation could decrease esophagus cancer deaths among populations. This study aimed to explore the effect of folic acid on VM formation of esophageal cancer cell, and the target. Human esophageal squamous cancer cell lines(Eca-109) were cultured with different concentrations of folic acid (0,1,10,100,200,400, 600,800 μg/ml). A cell counting kit-8 (CCK-8) assay was used to measure the cell proliferation. Then, the amount of VM under the effect of different concentrations of folic acid was observed. Target genes were screened out from several possible targets genes including MMP2, MMP9, EphA2, VE-cad or Ln-5γ2 by employing reverse transcription-quantitative polymerase chain reaction(RT-qPCR). Finally, western blot analysis was used to verify the target proteins. In conclusion, this study found that folic acid inhibited the formation of VM in Eca-109 cells, and the one target protein was EphA2.
Collapse
Affiliation(s)
- YuLing Xu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - LingMeng Fu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Da Pan
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Jie Wei
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Hui Xia
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - ShaoKang Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - GuiJu Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
14
|
Yun BD, Son SW, Choi SY, Kuh HJ, Oh TJ, Park JK. Anti-Cancer Activity of Phytochemicals Targeting Hypoxia-Inducible Factor-1 Alpha. Int J Mol Sci 2021; 22:ijms22189819. [PMID: 34575983 PMCID: PMC8467787 DOI: 10.3390/ijms22189819] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Hypoxia-inducible factor-1 alpha (HIF-1α) is overexpressed in cancer, leading to a poor prognosis in patients. Diverse cellular factors are able to regulate HIF-1α expression in hypoxia and even in non-hypoxic conditions, affecting its progression and malignant characteristics by regulating the expression of the HIF-1α target genes that are involved in cell survival, angiogenesis, metabolism, therapeutic resistance, et cetera. Numerous studies have exhibited the anti-cancer effect of HIF-1α inhibition itself and the augmentation of anti-cancer treatment efficacy by interfering with HIF-1α-mediated signaling. The anti-cancer effect of plant-derived phytochemicals has been evaluated, and they have been found to possess significant therapeutic potentials against numerous cancer types. A better understanding of phytochemicals is indispensable for establishing advanced strategies for cancer therapy. This article reviews the anti-cancer effect of phytochemicals in connection with HIF-1α regulation.
Collapse
Affiliation(s)
- Ba Da Yun
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (B.D.Y.); (S.W.S.); (S.Y.C.)
| | - Seung Wan Son
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (B.D.Y.); (S.W.S.); (S.Y.C.)
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (B.D.Y.); (S.W.S.); (S.Y.C.)
| | - Hyo Jeong Kuh
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Tae-Jin Oh
- Department of Pharmaceutical Engineering and Biotechnology, SunMoon University, 70 Sunmoon-ro 221, Tangjeong-myeon, Asan-si 31460, Korea;
| | - Jong Kook Park
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (B.D.Y.); (S.W.S.); (S.Y.C.)
- Correspondence: ; Tel.: +82-33-248-2114
| |
Collapse
|
15
|
Miao Y, Wang X, Lai Y, Lin W, Huang Y, Yin H, Hou R, Zhang F. Mitochondrial calcium uniporter promotes cell proliferation and migration in esophageal cancer. Oncol Lett 2021; 22:686. [PMID: 34434285 PMCID: PMC8335723 DOI: 10.3892/ol.2021.12947] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 06/14/2021] [Indexed: 12/20/2022] Open
Abstract
Increasing evidence has suggested that mitochondrial calcium uniporter (MCU) is involved in various types of cancer. However, its functions remain unclear in esophageal cancer. The aim of the present study was to explore its abnormal expression and clinical implications in esophageal cancer. A total of 110 patients with esophageal cancer were enrolled in the study. Western blotting was performed to examine the protein expression levels of MCU in 8 pairs of esophageal cancer and adjacent normal tissues. Using immunochemistry, a total of 110 esophageal cancer specimens were analyzed to identify the association between MCU expression and clinicopathological features of patients with esophageal cancer. Furthermore, immunofluorescence of MCU was performed. Pearson's correlation analysis was performed between MCU and hypoxia inducible factor (HIF)-1α/VEGF/E-cadherin/Vimentin expression based on western blotting. After KYSE-150 and TE-1 cells were treated with the MCU agonist Spermine and a small interfering RNA against MCU (si-MCU), a series of functional assays were performed, including Cell Counting Kit-8, colony formation and Transwell assays. The results revealed that, compared with in adjacent normal tissues, MCU was highly expressed in esophageal cancer tissues. MCU expression was significantly associated with depth of invasion, lymph node metastasis, TNM stage and distant metastasis. Moreover, MCU was significantly correlated with HIF-1α/VEGF/E-cadherin/Vimentin in esophageal cancer tissues. MCU overexpression promoted VEGF, MMP2, Vimentin and N-cadherin expression, while it inhibited E-cadherin expression in KYSE-150 and TE-1 cells, and opposite results were observed after transfection with si-MCU. Furthermore, MCU overexpression accelerated the proliferation and migration of KYSE-150 and TE-1 cells. Thus, the current findings suggested that high MCU expression may participate in cell proliferation, migration and epithelial-mesenchymal transition in esophageal cancer.
Collapse
Affiliation(s)
- Yu Miao
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750000, P.R. China
| | - Xiaofei Wang
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yafang Lai
- Department of Gastroenterology, Ordos Center Hospital, Ordos, Inner Mongolia 017000, P.R. China
| | - Wan Lin
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750000, P.R. China
| | - Ying Huang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750000, P.R. China
| | - Hua Yin
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750000, P.R. China
| | - Ruirui Hou
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750000, P.R. China
| | - Feixiong Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750000, P.R. China
| |
Collapse
|
16
|
Davern M, Donlon NE, Power R, Hayes C, King R, Dunne MR, Reynolds JV. The tumour immune microenvironment in oesophageal cancer. Br J Cancer 2021; 125:479-494. [PMID: 33903730 PMCID: PMC8368180 DOI: 10.1038/s41416-021-01331-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 01/16/2021] [Accepted: 02/17/2021] [Indexed: 02/02/2023] Open
Abstract
Oesophageal cancer (OC) is an inflammation-associated malignancy linked to gastro-oesophageal reflux disease, obesity and tobacco use. Knowledge of the microenvironment of oesophageal tumours is relevant to our understanding of the development of OC and its biology, and has major implications for understanding the response to standard therapies and immunotherapies, as well as for uncovering novel targets. In this context, we discuss what is known about the TME in OC from tumour initiation to development and progression, and how this is relevant to therapy sensitivity and resistance in the two major types of OC. We provide an immunological characterisation of the OC TME and discuss its prognostic implications with specific comparison with the Immunoscore and immune-hot, -cold, altered-immunosuppressed and -altered-excluded models. Targeted therapeutics for the TME under pre-clinical and clinical investigation in OCs are also summarised. A deeper understanding of the TME will enable the development of combination approaches to concurrently target the tumour cells and TME delivering precision medicine to OC patients.
Collapse
Affiliation(s)
- Maria Davern
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St James's Cancer Institute, St James's Hospital, Dublin, Ireland
| | - Noel E Donlon
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St James's Cancer Institute, St James's Hospital, Dublin, Ireland
| | - Robert Power
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St James's Cancer Institute, St James's Hospital, Dublin, Ireland
| | - Conall Hayes
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St James's Cancer Institute, St James's Hospital, Dublin, Ireland
| | - Ross King
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St James's Cancer Institute, St James's Hospital, Dublin, Ireland
| | - Margaret R Dunne
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St James's Cancer Institute, St James's Hospital, Dublin, Ireland
| | - John V Reynolds
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland.
- Trinity St James's Cancer Institute, St James's Hospital, Dublin, Ireland.
| |
Collapse
|
17
|
Morales-Guadarrama G, García-Becerra R, Méndez-Pérez EA, García-Quiroz J, Avila E, Díaz L. Vasculogenic Mimicry in Breast Cancer: Clinical Relevance and Drivers. Cells 2021; 10:cells10071758. [PMID: 34359928 PMCID: PMC8304745 DOI: 10.3390/cells10071758] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/24/2022] Open
Abstract
In solid tumors, vasculogenic mimicry (VM) is the formation of vascular structures by cancer cells, allowing to generate a channel-network able to transport blood and tumor cells. While angiogenesis is undertaken by endothelial cells, VM is assumed by cancer cells. Besides the participation of VM in tumor neovascularization, the clinical relevance of this process resides in its ability to favor metastasis and to drive resistance to antiangiogenic therapy. VM occurs in many tumor types, including breast cancer, where it has been associated with a more malignant phenotype, such as triple-negative and HER2-positive tumors. The latter may be explained by known drivers of VM, like hypoxia, TGFB, TWIST1, EPHA2, VEGF, matrix metalloproteinases, and other tumor microenvironment-derived factors, which altogether induce the transformation of tumor cells to a mesenchymal phenotype with a high expression rate of stemness markers. This review analyzes the current literature in the field, including the participation of some microRNAs and long noncoding RNAs in VM-regulation and tumorigenesis of breast cancer. Considering the clinical relevance of VM and its association with the tumor phenotype and clinicopathological parameters, further studies are granted to target VM in the clinic.
Collapse
Affiliation(s)
- Gabriela Morales-Guadarrama
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Edgar Armando Méndez-Pérez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Janice García-Quiroz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Euclides Avila
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico; (G.M.-G.); (E.A.M.-P.); (J.G.-Q.); (E.A.)
- Correspondence: ; Tel.: +52-(55)-5487-0900
| |
Collapse
|
18
|
King R, Hayes C, Donohoe CL, Dunne MR, Davern M, Donlon NE. Hypoxia and its impact on the tumour microenvironment of gastroesophageal cancers. World J Gastrointest Oncol 2021; 13:312-331. [PMID: 34040696 PMCID: PMC8131902 DOI: 10.4251/wjgo.v13.i5.312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/24/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
The malfeasant role of the hypoxic tumour microenvironment (TME) in cancer progression was recognized decades ago but the exact mechanisms that augment the hallmarks of cancer and promote treatment resistance continue to be elucidated. Gastroesophageal cancers (GOCs) represent a major burden of worldwide disease, responsible for the deaths of over 1 million people annually. Disentangling the impact of hypoxia in GOCs enables a better overall understanding of the disease pathogenesis while shining a light on novel therapeutic strategies and facilitating precision treatment approaches with the ultimate goal of improving outcomes for patients with these diseases. This review discusses the underlying principles and processes of the hypoxic response and the effect of hypoxia in promoting the hallmarks of cancer in the context of GOCs. We focus on its bidirectional influence on inflammation and how it drives angiogenesis, innate and adaptive immune evasion, metastasis, and the reprogramming of cellular bioenergetics. The contribution of the hypoxic GOC TME to treatment resistance is examined and a brief overview of the pharmacodynamics of hypoxia-targeted therapeutics is given. The principal methods that are used in measuring hypoxia and how they may enhance prognostication or provide rationale for individually tailored management in the case of tumours with significant hypoxic regions are also discussed.
Collapse
Affiliation(s)
- Ross King
- Department of Surgery, St. James’s Hospital Campus, Trinity Translational Medicine Institute, Dublin D8, Ireland
| | - Conall Hayes
- Department of Surgery, St. James’s Hospital Campus, Trinity Translational Medicine Institute, Dublin D8, Ireland
| | - Claire L Donohoe
- Department of Surgery, St. James’s Hospital Campus, Trinity Translational Medicine Institute, Dublin D8, Ireland
| | - Margaret R Dunne
- Department of Surgery, St. James’s Hospital Campus, Trinity Translational Medicine Institute, Dublin D8, Ireland
| | - Maria Davern
- Department of Surgery, St. James’s Hospital Campus, Trinity Translational Medicine Institute, Dublin D8, Ireland
| | - Noel E Donlon
- Department of Surgery, St. James’s Hospital Campus, Trinity Translational Medicine Institute, Dublin D8, Ireland
| |
Collapse
|
19
|
Nisar MA, Zheng Q, Saleem MZ, Ahmmed B, Ramzan MN, Ud Din SR, Tahir N, Liu S, Yan Q. IL-1β Promotes Vasculogenic Mimicry of Breast Cancer Cells Through p38/MAPK and PI3K/Akt Signaling Pathways. Front Oncol 2021; 11:618839. [PMID: 34055597 PMCID: PMC8160375 DOI: 10.3389/fonc.2021.618839] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Vasculogenic mimicry (VM), a micro vessel-like structure formed by the cancer cells, plays a pivotal role in cancer malignancy and progression. Interleukin-1 beta (IL-1β) is an active pro-inflammatory cytokine and elevated in many tumor types, including breast cancer. However, the effect of IL-1β on the VM of breast cancer has not been clearly elucidated. In this study, breast cancer cells (MCF-7 and MDA-MB-231) were used to study the effect of IL-1β on the changes that can promote VM. The evidence for VM stimulated by IL-1β was acquired by analyzing the expression of VM-associated biomarkers (VE-cadherin, VEGFR-1, MMP-9, MMP-2, c-Fos, and c-Jun) via western blot, immunofluorescent staining, and Immunohistochemistry (IHC). Additionally, morphological evidence was collected via Matrigel-based cord formation assay under normoxic/hypoxic conditions and microvessel examination through Hematoxylin and Eosin staining (H&E). Furthermore, the STRING and Gene Ontology database was also used to analyze the VM-associated interacting molecules stimulated by IL-β. The results showed that the expression of VM biomarkers was increased in both MCF-7 and MDA-MB-231 cells after IL-1β treatment. The increase in VM response was observed in IL-1β treated cells under both normoxia and hypoxia. IL-1β also increased the activation of transcription factor AP-1 complex (c-Fos/c-Jun). The bioinformatics data indicated that p38/MAPK and PI3K/Akt signaling pathways were involved in the IL-1β stimulation. It was further confirmed by the downregulated expression of VM biomarkers and reduced formation of the intersections upon the addition of the signaling pathway inhibitors. The study suggests that IL-1β stimulates the VM and its associated events in breast cancer cells via p38/MAPK and PI3K/Akt signaling pathways. Aiming the VM-associated molecular targets promoted by IL-1β may offer a novel anti-angiogenic therapeutic strategy to control the aggressiveness of breast cancer cells.
Collapse
Affiliation(s)
- Muhammad Azhar Nisar
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qin Zheng
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Muhammad Zubair Saleem
- Department of Pathology and Pathophysiology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Bulbul Ahmmed
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Muhammad Noman Ramzan
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Syed Riaz Ud Din
- Department of Microbiology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Naeem Tahir
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Shuai Liu
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qiu Yan
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, College of Basic Medical Science, Dalian Medical University, Dalian, China
| |
Collapse
|
20
|
Fu R, Du W, Ding Z, Wang Y, Li Y, Zhu J, Zeng Y, Zheng Y, Liu Z, Huang JA. HIF-1α promoted vasculogenic mimicry formation in lung adenocarcinoma through NRP1 upregulation in the hypoxic tumor microenvironment. Cell Death Dis 2021; 12:394. [PMID: 33850110 PMCID: PMC8044151 DOI: 10.1038/s41419-021-03682-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Neovascularization is a key factor that contributes to tumor metastasis, and vasculogenic mimicry (VM) is an important form of neovascularization found in highly invasive tumors, including lung cancer. Despite the increasing number of studies focusing on VM, the mechanisms underlying VM formation remain unclear. Herein, our study explored the role of the HIF-1α/NRP1 axis in mediating lung adenocarcinoma metastasis and VM formation. HIF-1α, NRP1 expression, and VM in lung adenocarcinoma (LUAD) patient samples were examined by immunohistochemical staining. Quantitative real-time (qRT-PCR), western blot, transwell assay, wound healing assay, and tube formation assay were performed to verify the role of HIF-1α/NRP1 axis in LUAD metastasis and VM formation. ChIP and luciferase reporter assay were used to confirm whether NRP1 is a direct target of HIF-1α. In LUAD tissues, we confirmed a positive relationship between HIF-1α and NRP1 expression. Importantly, high HIF-1α and NRP1 expression and the presence of VM were correlated with poor prognosis. We also found that HIF-1α could induce LUAD cell migration, invasion, and VM formation by regulating NRP1. Moreover, we demonstrated that HIF-1α can directly bind to the NRP1 promoter located between −2009 and −2017 of the promoter. Mechanistically, MMP2, VE-cadherin, and Vimentin expression were affected. HIF-1α plays an important role in inducing lung adenocarcinoma cell metastasis and VM formation via upregulation of NRP1. This study highlights the potential therapeutic value of targeting NRP1 for suppressing lung adenocarcinoma metastasis and progression.
Collapse
Affiliation(s)
- Ran Fu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China.,Department of Respiratory Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, China
| | - Wenwen Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Zongli Ding
- Department of Respiratory Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, China
| | - Yi Wang
- Department of Respiratory Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, China
| | - Yue Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China
| | - Jianjie Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Yuanyuan Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Yulong Zheng
- Department of Respiratory Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, China
| | - Zeyi Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China. .,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China. .,Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China.
| | - Jian-An Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China. .,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China. .,Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China.
| |
Collapse
|
21
|
Shakova FM, Kirova YI, Silachev DN, Romanova GA, Morozov SG. Protective Effects of PGC-1α Activators on Ischemic Stroke in a Rat Model of Photochemically Induced Thrombosis. Brain Sci 2021; 11:325. [PMID: 33806692 PMCID: PMC8002020 DOI: 10.3390/brainsci11030325] [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: 12/23/2020] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 11/16/2022] Open
Abstract
The pharmacological induction and activation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α), a key regulator of ischemic brain tolerance, is a promising direction in neuroprotective therapy. Pharmacological agents with known abilities to modulate cerebral PGC-1α are scarce. This study focused on the potential PGC-1α-modulating activity of Mexidol (2-ethyl-6-methyl-3-hydroxypyridine succinate) and Semax (ACTH(4-7) analog) in a rat model of photochemical-induced thrombosis (PT) in the prefrontal cortex. Mexidol (100 mg/kg) was administered intraperitoneally, and Semax (25 μg/kg) was administered intranasally, for 7 days each. The expression of PGC-1α and PGC-1α-dependent protein markers of mitochondriogenesis, angiogenesis, and synaptogenesis was measured in the penumbra via immunoblotting at Days 1, 3, 7, and 21 after PT. The nuclear content of PGC-1α was measured immunohistochemically. The suppression of PGC-1α expression was observed in the penumbra from 24 h to 21 days following PT and reflected decreases in both the number of neurons and PGC-1α expression in individual neurons. Administration of Mexidol or Semax was associated with preservation of the neuron number and neuronal expression of PGC-1α, stimulation of the nuclear translocation of PGC-1α, and increased contents of protein markers for PGC-1α activation. This study opens new prospects for the pharmacological modulation of PGC-1α in the ischemic brain.
Collapse
Affiliation(s)
- Fatima M. Shakova
- Institute of General Pathology and Pathophysiology, Baltiyskaya Str. 8, 125315 Moscow, Russia; (Y.I.K.); (G.A.R.); (S.G.M.)
| | - Yuliya I. Kirova
- Institute of General Pathology and Pathophysiology, Baltiyskaya Str. 8, 125315 Moscow, Russia; (Y.I.K.); (G.A.R.); (S.G.M.)
| | - Denis N. Silachev
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, Leninskye Gory 1, Bldg. 40, 119992 Moscow, Russia;
- Histology, Embryology and Cytology Department, Peoples’ Friendship University of Russia, Miklukho-Maklaya Str. 6, 117198 Moscow, Russia
| | - Galina A. Romanova
- Institute of General Pathology and Pathophysiology, Baltiyskaya Str. 8, 125315 Moscow, Russia; (Y.I.K.); (G.A.R.); (S.G.M.)
| | - Sergey G. Morozov
- Institute of General Pathology and Pathophysiology, Baltiyskaya Str. 8, 125315 Moscow, Russia; (Y.I.K.); (G.A.R.); (S.G.M.)
| |
Collapse
|
22
|
She Q, Hu S, Pu X, Guo Q, Mou C, Yang C. The effect of hepatocellular carcinoma-associated fibroblasts on hepatoma vasculogenic mimicry. Am J Cancer Res 2020; 10:4198-4210. [PMID: 33414995 PMCID: PMC7783763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/19/2020] [Indexed: 06/12/2023] Open
Abstract
Vasculogenic Mimicry (VM) is the main source of blood supply in the early stage of tumor growth. Carcinoma-associated fibroblasts (CAFs) are one of the most important host cells in the tumor microenvironment. Some studies have found that CAFs can promote tumor angiogenesis, but there are few reports on the relationship between CAFs and VM. Tissue samples were collected from 60 cases of hepatocellular carcinoma (HCC) and 10 persons with normal liver function. The relationship between VM expression and clinicopathologic features was analyzed. Furthermore, the relationship between VM expression and vimentin or α-SMA expression was analyzed. Primary culture of hepatocellular CAFs and the collection of conditioned media were carried out. The effects of hepatocellular CAF conditioned medium on the formation of VM and the levels of VM-related proteins and genes in MHCC-97H cells were studied. The positive rate of VM was 35.0% in HCC tissues. There was no VM expression in normal liver tissues. VM expression was related to tumor diameter, Edmondson grade, clinical stage, and liver cirrhosis. The expression of vimentin and α-SMA in VM-positive patients was higher than in VM-negative patients. Different concentrations of hepatocellular CAF conditioned medium could promote the formation of VM and increase the expression of VM-related genes and proteins (MMP2 and EphA2) in MHCC-97H cells. The results show that there was a significant correlation between VM formation and the expression of vimentin or α-SMA in HCC tissues. The conditioned medium of hepatocellular CAFs may promote VM formation and the expression of VM-related genes and proteins (MMP2 and EphA2) in hepatoma cell line MHCC-97H.
Collapse
Affiliation(s)
- Qin She
- Department of Pathology, The Affliated Hospital of Southwest Medical UniversityLuzhou, Sichuan Province, China
| | - Shuya Hu
- Department of Pathology, Sichuan Mianyang 404 HospitalMianyang, Sichuan Province, China
| | - Xia Pu
- Department of Pathology, The Affliated Hospital of Southwest Medical UniversityLuzhou, Sichuan Province, China
| | - Qingxi Guo
- Department of Pathology, The Affliated Hospital of Southwest Medical UniversityLuzhou, Sichuan Province, China
| | - Chunru Mou
- Department of Pathology, The Affliated Hospital of Southwest Medical UniversityLuzhou, Sichuan Province, China
| | - Chengwan Yang
- Department of Pathology, The Affliated Hospital of Southwest Medical UniversityLuzhou, Sichuan Province, China
| |
Collapse
|
23
|
Jiang X, Wang J, Deng X, Xiong F, Zhang S, Gong Z, Li X, Cao K, Deng H, He Y, Liao Q, Xiang B, Zhou M, Guo C, Zeng Z, Li G, Li X, Xiong W. The role of microenvironment in tumor angiogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:204. [PMID: 32993787 PMCID: PMC7526376 DOI: 10.1186/s13046-020-01709-5] [Citation(s) in RCA: 420] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Tumor angiogenesis is necessary for the continued survival and development of tumor cells, and plays an important role in their growth, invasion, and metastasis. The tumor microenvironment—composed of tumor cells, surrounding cells, and secreted cytokines—provides a conducive environment for the growth and survival of tumors. Different components of the tumor microenvironment can regulate tumor development. In this review, we have discussed the regulatory role of the microenvironment in tumor angiogenesis. High expression of angiogenic factors and inflammatory cytokines in the tumor microenvironment, as well as hypoxia, are presumed to be the reasons for poor therapeutic efficacy of current anti-angiogenic drugs. A combination of anti-angiogenic drugs and antitumor inflammatory drugs or hypoxia inhibitors might improve the therapeutic outcome.
Collapse
Affiliation(s)
- Xianjie Jiang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Jie Wang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Xiangying Deng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Fang Xiong
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Shanshan Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ke Cao
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Deng
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yi He
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Can Guo
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China.
| |
Collapse
|
24
|
Ding Y, Ding K, Gong W, Wei H, Mo W, Ding X. WITHDRAWN: Long non-coding RNA LUCAT1 up-regulates the expression of HIF-1α and promotes the proliferation and metastasis of breast cancer cells via sponging miR-199a-5p. Biomed J 2020. [DOI: 10.1016/j.bj.2020.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
25
|
Wechman SL, Emdad L, Sarkar D, Das SK, Fisher PB. Vascular mimicry: Triggers, molecular interactions and in vivo models. Adv Cancer Res 2020; 148:27-67. [PMID: 32723566 DOI: 10.1016/bs.acr.2020.06.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vascular mimicry is induced by a wide array of genes with functions related to cancer stemness, hypoxia, angiogenesis and autophagy. Vascular mimicry competent (VM-competent) cells that form de novo blood vessels are common in solid tumors facilitating tumor cell survival and metastasis. VM-competent cells display increased levels of vascular mimicry selecting for stem-like cells in an O2-gradient-dependent manner in deeply hypoxic tumor regions, while also aiding in maintaining tumor cell metabolism and stemness. Three of the principal drivers of vascular mimicry are EphA2, Nodal and HIF-1α, however, directly or indirectly many of these molecules affect VE-Cadherin (VE-Cad), which forms gap-junctions to bind angiogenic blood vessels together. During vascular mimicry, the endothelial-like functions of VM-competent cancer stem cells co-opt VE-Cad to bind cancer cells together to create cancer cell-derived blood conducting vessels. This process potentially compensates for the lack of access to blood and nutrient in avascular tumors, simultaneously providing nutrients and enhancing cancer invasion and metastasis. Current evidence also supports that vascular mimicry promotes cancer malignancy and metastasis due to the cooperation of oncogenic signaling molecules driving cancer stemness and autophagy. While a number of currently used cancer therapeutics are effective inhibitors of vascular mimicry, developing a new class of vascular mimicry specific inhibitors could allow for the treatment of angiogenesis-resistant tumors, inhibit cancer metastasis and improve patient survival. In this review, we describe the principal vascular mimicry pathways in addition to emphasizing the roles of hypoxia, autophagy and select proangiogenic oncogenes in this process.
Collapse
Affiliation(s)
- Stephen L Wechman
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
26
|
Fathi Maroufi N, Taefehshokr S, Rashidi MR, Taefehshokr N, Khoshakhlagh M, Isazadeh A, Mokarizadeh N, Baradaran B, Nouri M. Vascular mimicry: changing the therapeutic paradigms in cancer. Mol Biol Rep 2020; 47:4749-4765. [PMID: 32424524 DOI: 10.1007/s11033-020-05515-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022]
Abstract
Cancer is a major problem in the health system, and despite many efforts to effectively treat it, none has yet been fully successful. Angiogenesis and metastasis are considered as major challenges in the treatment of various cancers. Researchers have struggled to succeed with anti-angiogenesis drugs for the effective treatment of cancer, although new challenges have emerged in the treatment with the emergence of resistance to anti-angiogenesis and anti-metastatic drugs. Numerous studies have shown that different cancers can resist anti-angiogenesis drugs in a new process called vascular mimicry (VM). The studies have revealed that cells resistant to anti-angiogenesis cancer therapies are more capable of forming VMs in the in vivo and in vitro environment, although there is a link between the presence of VM and poor clinical outcomes. Given the importance of the VM in the challenges facing cancer treatment, researchers are trying to identify factors that prevent the formation of these structures. In this review article, it is attempted to provide a comprehensive overview of the molecules and main signaling pathways involved in VM phenomena, as well as the agents currently being identified as anti-VM and the role of VM in response to treatment and prognosis of cancer patients.
Collapse
Affiliation(s)
- Nazila Fathi Maroufi
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Taefehshokr
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Taefehshokr
- Department of Microbiology and Immunology, Center for Human Immunology, The University of Western Ontario, London, ON, Canada
| | - Mahdieh Khoshakhlagh
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narmin Mokarizadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
27
|
Mammadzada P, Corredoira PM, André H. The role of hypoxia-inducible factors in neovascular age-related macular degeneration: a gene therapy perspective. Cell Mol Life Sci 2020; 77:819-833. [PMID: 31893312 PMCID: PMC7058677 DOI: 10.1007/s00018-019-03422-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022]
Abstract
Understanding the mechanisms that underlie age-related macular degeneration (AMD) has led to the identification of key molecules. Hypoxia-inducible transcription factors (HIFs) have been associated with choroidal neovascularization and the progression of AMD into the neovascular clinical phenotype (nAMD). HIFs regulate the expression of multiple growth factors and cytokines involved in angiogenesis and inflammation, hallmarks of nAMD. This knowledge has propelled the development of a new group of therapeutic strategies focused on gene therapy. The present review provides an update on current gene therapies in ocular angiogenesis, particularly nAMD, from both basic and clinical perspectives.
Collapse
Affiliation(s)
- Parviz Mammadzada
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden
| | - Pablo M Corredoira
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden
| | - Helder André
- Division of Eye and Vision, Department of Clinical Neuroscience, Karolinska Institutet, St. Erik Eye Hospital, Stockholm, Sweden.
| |
Collapse
|
28
|
Andonegui-Elguera MA, Alfaro-Mora Y, Cáceres-Gutiérrez R, Caro-Sánchez CHS, Herrera LA, Díaz-Chávez J. An Overview of Vasculogenic Mimicry in Breast Cancer. Front Oncol 2020; 10:220. [PMID: 32175277 PMCID: PMC7056883 DOI: 10.3389/fonc.2020.00220] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 02/07/2020] [Indexed: 12/24/2022] Open
Abstract
Vasculogenic mimicry (VM) is the formation of vascular channels lacking endothelial cells. These channels are lined by tumor cells with cancer stem cell features, positive for periodic acid-Schiff, and negative for CD31 staining. The term VM was introduced by Maniotis et al. (1), who reported this phenomenon in highly aggressive uveal melanomas; since then, VM has been associated with poor prognosis, tumor aggressiveness, metastasis, and drug resistance in several tumors, including breast cancer. It is proposed that VM and angiogenesis (the de novo formation of blood vessels from the established vasculature by endothelial cells, which is observed in several tumors) rely on some common mechanisms. Furthermore, it is also suggested that VM could constitute a means to circumvent anti-angiogenic treatment in cancer. Therefore, it is important to determinant the factors that dictate the onset of VM. In this review, we describe the current understanding of VM formation in breast cancer, including specific signaling pathways, and cancer stem cells. In addition, we discuss the clinical significance of VM in prognosis and new opportunities of VM as a target for breast cancer therapy.
Collapse
Affiliation(s)
- Marco A Andonegui-Elguera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | - Yair Alfaro-Mora
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Rodrigo Cáceres-Gutiérrez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | | | - Luis A Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico.,Dirección General, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - José Díaz-Chávez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| |
Collapse
|
29
|
Zhu Y, Liu X, Zhao P, Zhao H, Gao W, Wang L. Celastrol Suppresses Glioma Vasculogenic Mimicry Formation and Angiogenesis by Blocking the PI3K/Akt/mTOR Signaling Pathway. Front Pharmacol 2020; 11:25. [PMID: 32116702 PMCID: PMC7025498 DOI: 10.3389/fphar.2020.00025] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
Angiogenesis and vasculogenic mimicry (VM) are thought to be the predominant processes ensuring tumor blood supply during the growth and metastasis of glioblastoma (GBM). Celastrol has potential anti-glioma effects, however the mechanisms underlying these effects remain unclarified. Recent studies have shown that the PI3K/Akt/mTOR signaling pathway is closely related to angiogenesis and VM formation. In the present study, we have demonstrated, for the first time, that celastrol eliminated VM formation by blocking this signaling pathway in glioma cells. By the treatment of celastrol, tumor growth was suppressed, tight junction and basal lamina structures in tumor microvasculature were disarranged in U87 glioma orthotopic xenografts in nude mice. Periodic acid Schiff (PAS)-CD31 staining revealed that celastrol inhibited both VM and angiogenesis in tumor tissues. Additionally, celastrol reduced the expression levels of the angiogenesis-related proteins CD31, vascular endothelial growth factor receptor (VEGFR) 2, angiopoietin (Ang) 2 and VEGFA, VM-related proteins ephrin type-A receptor (EphA) 2, and vascular endothelial (VE)-cadherin. Hypoxia inducible factor (HIF)-1α, phosphorylated PI3K, Akt, and mTOR were also downregulated by treatment with celastrol. In vitro, we further demonstrated that celastrol inhibited the growth, migration, and invasion of U87 and U251 cells, disrupted VM formation, and blocked the activity of PI3K, Akt, and mTOR. Collectively, our data suggest that celastrol inhibits VM formation and angiogenesis likely by regulating the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yingjun Zhu
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Xihong Liu
- Basic Discipline of Integrated Chinese and Western Medicine, Henan University of Chinese Medicine, Henan, China
| | - Peiyuan Zhao
- Basic Discipline of Integrated Chinese and Western Medicine, Henan University of Chinese Medicine, Henan, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Wei Gao
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China.,School of Pharmaceutical Sciences, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Hernández de la Cruz ON, López-González JS, García-Vázquez R, Salinas-Vera YM, Muñiz-Lino MA, Aguilar-Cazares D, López-Camarillo C, Carlos-Reyes Á. Regulation Networks Driving Vasculogenic Mimicry in Solid Tumors. Front Oncol 2020; 9:1419. [PMID: 31993365 PMCID: PMC6970938 DOI: 10.3389/fonc.2019.01419] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/28/2019] [Indexed: 12/21/2022] Open
Abstract
Vasculogenic mimicry (VM) is a mechanism whereby cancer cells form microvascular structures similar to three-dimensional channels to provide nutrients and oxygen to tumors. Unlike angiogenesis, VM is characterized by the development of new patterned three-dimensional vascular-like structures independent of endothelial cells. This phenomenon has been observed in many types of highly aggressive solid tumors. The presence of VM has also been associated with increased resistance to chemotherapy, low survival, and poor prognosis. MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are non-coding RNAs that regulate gene expression at the post-transcriptional level through different pathways. In recent years, these tiny RNAs have been shown to be expressed aberrantly in different human malignancies, thus contributing to the hallmarks of cancer. In this context, miRNAs and lncRNAs can be excellent biomarkers for diagnosis, prognosis, and the prediction of response to therapy. In this review, we discuss the role that the tumor microenvironment and the epithelial-mesenchymal transition have in VM. We include an overview of the mechanisms of VM with examples of diverse types of tumors. Finally, we describe the regulation networks of lncRNAs-miRNAs and their clinical impact with the VM. Knowing the key genes that regulate and promote the development of VM in tumors with invasive, aggressive, and therapy-resistant phenotypes will facilitate the discovery of novel biomarker therapeutics against cancer as well as tools in the diagnosis and prognosis of patients.
Collapse
Affiliation(s)
| | - José Sullivan López-González
- Laboratorio de Cáncer de Pulmón, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico, Mexico
| | - Raúl García-Vázquez
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico, Mexico
| | - Yarely M. Salinas-Vera
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico, Mexico
| | - Marcos A. Muñiz-Lino
- Laboratorio de Patología y Medicina Bucal, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico, Mexico
| | - Dolores Aguilar-Cazares
- Laboratorio de Cáncer de Pulmón, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico, Mexico
| | - Ángeles Carlos-Reyes
- Laboratorio de Cáncer de Pulmón, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico, Mexico
| |
Collapse
|
31
|
Fenretinide reduces angiogenesis by downregulating CDH5, FOXM1 and eNOS genes and suppressing microRNA-10b. Mol Biol Rep 2020; 47:1649-1658. [DOI: 10.1007/s11033-020-05252-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 01/07/2020] [Indexed: 12/13/2022]
|
32
|
Fernández-Cortés M, Delgado-Bellido D, Oliver FJ. Vasculogenic Mimicry: Become an Endothelial Cell "But Not So Much". Front Oncol 2019; 9:803. [PMID: 31508365 PMCID: PMC6714586 DOI: 10.3389/fonc.2019.00803] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/07/2019] [Indexed: 01/04/2023] Open
Abstract
Blood vessels supply all body tissues with nutrients and oxygen, take away waste products and allow the arrival of immune cells and other cells (pericytes, smooth muscle cells) that form part of these vessels around the principal endothelial cells. Vasculogenic mimicry (VM) is a tumor blood supply system that takes place independently of angiogenesis or endothelial cells, and is associated with poor survival in cancer patients. Aberrant expression of VE-cadherin has been strongly associated with VM. Even more, VE-cadherin has constitutively high phosphorylation levels on the residue of Y658 in human malignant melanoma cells. In this review we focus on non-endothelial VE-cadherin and its post-translational modifications as a crucial component in the development of tumor VM, highlighting the signaling pathways that lead to their pseudo-endothelial and stem-like phenotype and the role of tumor microenvironment. We discuss the importance of the tumor microenvironment in VM acquisition, and describe the most recent therapeutic targets that have been proposed for the repression of VM.
Collapse
Affiliation(s)
| | | | - F Javier Oliver
- CSIC, CIBERONC, Instituto de Parasitología y Biomedicina López Neyra, Granada, Spain
| |
Collapse
|
33
|
Qin Y, Zhao W, Cheng L, Wu S, Wu Q, Gao J, Bian Z, Ma L. Clinical significance of vasculogenic mimicry, vascular endothelial cadherin and SOX4 in patients with esophageal squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2462-2473. [PMID: 31934073 PMCID: PMC6949576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 05/22/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Vasculogenic mimicry (VM) plays an important role in invasion and metastasis of malignant tumor. High expression of vascular endothelial cadherin (VE-cahderin) in malignant tumor cells can promote the formation of VM. High expression of SOX4 (sex-determining region Y-related high-mobility group box 4) was found in esophageal squamous cell carcinoma (ESCC). It can promote the development of epithelial stromal transformation. Then, SOX4 can promote the formation of VM in ESCC. METHODS Paraffin-embedded specimens of ESCC (with complete clinicopathological data) and normal esophageal mucosa adjacent to carcinoma (> 5 cm) were collected from January to December 2013. CD34/PAS was used to detect VM. The expression of VE-cadherin and SOX4 was used by immunohistochemistry. The patients were followed up in detail (survival time and survival status). RESULTS SOX4, VM, and VE-cadherin were highly expressed in ESCC. Moreover, they were positively correlated. Survival analysis shows that the expressions of SOX4, VM, and VE-cadherin are associated with the patient's prognosis and can be independent prognostic factors for ESCC. CONCLUSIONS Studies suggests that SOX4, which is highly expressed in ESCC, is involved in the formation of VM. The combined detection of SOX4, VE-cadherin and VM expression can be used as biomarkers for invasion and metastasis of ESCC. These three markers can be used as powerful prognostic factors in patients with ESCC.
Collapse
Affiliation(s)
- Yanzi Qin
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| | - Wenjun Zhao
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu, Anhui Province, China
| | - Lili Cheng
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| | - Shiwu Wu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| | - Qiong Wu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| | - Jin Gao
- Department of Clinical Medicine, Bengbu Medical CollegeAnhui Province, China
| | - Zhaonan Bian
- Department of Clinical Medicine, Bengbu Medical CollegeAnhui Province, China
| | - Li Ma
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| |
Collapse
|
34
|
Wang HF, Wang SS, Zheng M, Dai LL, Wang K, Gao XL, Cao MX, Yu XH, Pang X, Zhang M, Wu JB, Wu JS, Yang X, Tang YJ, Chen Y, Tang YL, Liang XH. Hypoxia promotes vasculogenic mimicry formation by vascular endothelial growth factor A mediating epithelial-mesenchymal transition in salivary adenoid cystic carcinoma. Cell Prolif 2019; 52:e12600. [PMID: 30945361 PMCID: PMC6536414 DOI: 10.1111/cpr.12600] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/27/2019] [Accepted: 02/12/2019] [Indexed: 02/05/2023] Open
Abstract
Objectives To investigate the role of hypoxia in vasculogenic mimicry (VM) of salivary adenoid cystic carcinoma (SACC) and the underlying mechanism involved. Materials and methods Firstly, wound healing, transwell invasion, immunofluorescence and tube formation assays were performed to measure the effect of hypoxia on migration, invasion, EMT and VM of SACC cells, respectively. Then, immunofluorescence and RT‐PCR were used to detect the effect of hypoxia on VE‐cadherin and VEGFA expression. And pro‐vasculogenic mimicry effect of VEGFA was investigated by confocal laser scanning microscopy and Western blot. Moreover, the levels of E‐cadherin, N‐cadherin, Vimentin, CD44 and ALDH1 were determined by Western blot and immunofluorescence in SACC cells treated by exogenous VEGFA or bevacizumab. Finally, CD31/ PAS staining was performed to observe VM and immunohistochemistry was used to determine the levels of VEGFA and HIF‐1α in 95 SACC patients. The relationships between VM and clinicopathological variables, VEGFA or HIF‐1α level were analysed. Results Hypoxia promoted cell migration, invasion, EMT and VM formation, and enhanced VE‐cadherin and VEGFA expression in SACC cells. Further, exogenous VEGFA markedly increased the levels of N‐cadherin, Vimentin, CD44 and ALDH1, and inhibited the expression of E‐cadherin, while the VEGFA inhibitor reversed these changes. In addition, VM channels existed in 25 of 95 SACC samples, and there was a strong positive correlation between VM and clinic stage, distant metastases, VEGFA and HIF‐1α expression. Conclusions VEGFA played an important role in hypoxia‐induced VM through regulating EMT and stemness, which may eventually fuel the migration and invasion of SACC.
Collapse
Affiliation(s)
- Hao-Fan Wang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Sha-Sha Wang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Min Zheng
- Department of Stomatolog, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, China
| | - Lu-Ling Dai
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Ke Wang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Xiao-Lei Gao
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Ming-Xin Cao
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Xiang-Hua Yu
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Xin Pang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Mei Zhang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Jing-Biao Wu
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Jia-Shun Wu
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Xiao Yang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Ya-Jie Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Yu Chen
- State Key Laboratory of Oral Diseases, Department of Oral Pathology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, Department of Oral Pathology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan, China
| |
Collapse
|
35
|
Vasculogenic Mimicry Formation Is Associated with Erythropoietin Expression but Not with Erythropoietin Receptor Expression in Cervical Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1934195. [PMID: 30915348 PMCID: PMC6409067 DOI: 10.1155/2019/1934195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/30/2018] [Accepted: 01/10/2019] [Indexed: 12/24/2022]
Abstract
Background Vasculogenic mimicry (VM), as an endothelium-independent cancer microcirculation, has been observed in many malignancies including cervical cancer. Erythropoietin (EPO) and erythropoietin receptor (EPO-R) could produce an angiogenic effect to promote cervical squamous cell carcinoma (CSCC) progression. However, the association between VM formation and EPO/EPO-R expression in CSCC is poorly explored. Methods Seventy-six paraffin-embedded CSCC samples, 25 high-grade squamous intraepithelial lesion (HSIL) samples, 20 low-grade squamous intraepithelial lesion (LSIL) samples, and 20 normal cervix samples were collected. Immunohistochemistry SP method was performed to detect EPO/EPO-R expression and CD31/periodic acid-Schiff (PAS) double staining was performed to detect VM formation. The associations of EPO/EPO-R and VM with clinicopathological parameters of CSCC were analyzed. The associations between VM formation and EPO/EPO-R expression were also analyzed. Results The positive expression rates of EPO and EPO-R were gradually increasing along the progression of normal cervix-LSIL-HSIL-CSCC sequence (P<0.05). EPO and EPO-R expression were not significantly associated with clinicopathological parameters of CSCC patients (P>0.05). VM was significantly associated with FIGO stage, lymphovascular space involvement, and lymph node metastasis (P<0.05). VM was positively associated with EPO expression (r=0.284, P<0.05) but was not associated with EPO-R expression (P>0.05). Conclusion These data suggest that increased EPO/EPO-R expression may play an important role in cervical carcinogenesis. EPO overexpression may promote VM formation in CSCC.
Collapse
|
36
|
Ci H, Xu Z, Xu J, Wang Y, Wu S. Expressions of KAI1 and E-cadherin in nonsmall cell lung cancer and their correlation with vasculogenic mimicry. Medicine (Baltimore) 2018; 97:e12293. [PMID: 30290593 PMCID: PMC6200519 DOI: 10.1097/md.0000000000012293] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Metastasis and recurrence are the most common reasons for treatment failure of nonsmall cell lung cancer (NSCLC). Vasculogenic mimicry (VM, new blood supply formation in malignant tumors), E-Cadherin (a calcium-dependent transmembrane glycoprotein that mediates intercellular adhesion), KAI1 (a suppressor gene of tumor metastasis) are all valuable factors for metastasis and prognosis in diverse common human cancers. However, the correlation of VM, E-Cadherin, and KAI1 in NSCLC is still unclear. In this study, we analyzed the correlations among these factors as well as their respective correlations with clinicopathological parameters and survival in NSCLC. METHODS The level of VM, E-Cadherin, and KAI1 in 163 tissue samples of NSCLC was examined by immunhistochemistry. Clinical data were also collected. RESULTS Levels of VM was significantly higher, and levels of KAI1 and E-Cadherin significantly lower in NSCLC tissues than in normal lung tissues. Levels of VM were positively associated with lymph node metastasis (LNM), size, grade, and tumor node metastasis (TNM) stages, and negatively associated with patients' overall survival (OS). Levels of KAI1 and E-Cadherin were negatively correlated with LNM, size, grade, and TNM stage, and positively associated with patients' OS. In multivariate analysis, high levels of VM, E-Cadherin, and KAI1, as well as TNM stages were independently correlated with lower OS in patients with NSCLC. CONCLUSION VM and the expression of E-Cadherin and KAI1 may represent promising metastatic and prognostic biomarkers, as well as potential therapeutic targets for NSCLC.
Collapse
Affiliation(s)
- Hongfei Ci
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College
- Department of Pathology, Bengbu Medical College, Bengbu, China
| | - Zhouyi Xu
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College
- Department of Pathology, Bengbu Medical College, Bengbu, China
| | - Jing Xu
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College
- Department of Pathology, Bengbu Medical College, Bengbu, China
| | - Yichao Wang
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College
- Department of Pathology, Bengbu Medical College, Bengbu, China
| | - Shiwu Wu
- Department of Pathology, First Affiliated Hospital of Bengbu Medical College
- Department of Pathology, Bengbu Medical College, Bengbu, China
| |
Collapse
|
37
|
安 海, 林 俊, 孙 海, 许 梨, 苏 嘉, 何 春, 曾 嘉, 梁 佩, 贺 松. [ Biejiajian Pills inhibits hepatoma carcinoma cell vasculogenic mimicry by suppressing RhoA/ROCK signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:997-1001. [PMID: 30187871 PMCID: PMC6744031 DOI: 10.3969/j.issn.1673-4254.2018.08.16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To observe effects of Biejiajian Pills on hepatocarcinoma (HCC) cell vasculogenic mimicry (VM) and explore the molecular mechanism by which Biejiajian Pills inhibits HCC metastasis and invasion. METHODS Forty male SD rats were randomly divided into 4 groups for gastric lavage of normal saline or high, moderate or low doses of Biejiajian Pills (twice daily) for 4 consecutive days. The sera were collected from the rats for treatment of cultured human HCC HepG2 cells. VM formation in the cells was detected using an image acquisition and analysis system 24 h after incubation of the cells with the sera and with the RhoA/ROCK inhibitor Y-27632(P). The expression levels of RhoA and ROCK1 in the cells were detected using Western blotting, and the contents of VE-cadherin and PI3K in the culture supernatant were determined using ELISA. RESULTS Treatment with the sera from Biejiajian Pills-treated rats significantly inhibited formation of VM in HepG2 cells, and the diameters of VM formed were significantly greater than those in the positive control group (P < 0.01). Y-27632 completely inhibited the formation of VM in HepG2 cells (P < 0.01). Treatments with Biejiajian Pills and Y-27632 both inhibited the expression of RhoA and ROCK1 (P < 0.05) and significantly lowered the contents of VE-cadherin and PI3K in the culture supernatant (P < 0.05). CONCLUSIONS Biejiajian Pills can inhibit the formation of VM in HCC cells in vitro possibly by inhibiting the RhoA/ROCK pathways and the expressions of VE-cadherin and PI3K.
Collapse
Affiliation(s)
- 海燕 安
- />南方医科大学中医药学院,广东 广州 510515College of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 俊豪 林
- />南方医科大学中医药学院,广东 广州 510515College of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 海涛 孙
- />南方医科大学中医药学院,广东 广州 510515College of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 梨梨 许
- />南方医科大学中医药学院,广东 广州 510515College of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 嘉琪 苏
- />南方医科大学中医药学院,广东 广州 510515College of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 春雨 何
- />南方医科大学中医药学院,广东 广州 510515College of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 嘉敏 曾
- />南方医科大学中医药学院,广东 广州 510515College of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 佩湘 梁
- />南方医科大学中医药学院,广东 广州 510515College of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - 松其 贺
- />南方医科大学中医药学院,广东 广州 510515College of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
38
|
Wang S, Zhang Z, Qian W, Ji D, Wang Q, Ji B, Zhang Y, Zhang C, Sun Y, Zhu C, Sun Y. Angiogenesis and vasculogenic mimicry are inhibited by 8-Br-cAMP through activation of the cAMP/PKA pathway in colorectal cancer. Onco Targets Ther 2018; 11:3765-3774. [PMID: 29997437 PMCID: PMC6033084 DOI: 10.2147/ott.s164982] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Introduction Vasculogenic mimicry (VM) describes the formation of an epithelial-independent tumor microcirculation system that differs from traditional angiogenesis. Angiogenesis and the formation of VM are closely related through the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway and the epithelial–mesenchymal transition (EMT) process. Materials and methods In this study, 8-Br-cAMP, a cAMP analog and PKA activator, was used to activate the cAMP/PKA pathway to evaluate the effects of cAMP/PKA on angiogenesis and VM in colorectal cancer (CRC) cells. We used a syngeneic model of CRC in BALB/c mice. Results We discovered that treatment with 8-Br-cAMP significantly reduced tumor number compared to control mice after the 7th, 14th, and 28th days of treatment. VM was evaluated by periodic acid–schiff (PAS)–CD31 staining, and we found that VM was inhibited by 8-Br-cAMP treatment in vivo. Immunohistochemistry confirmed the inhibition of vascular endothelial growth factor (VEGF) and cAMP and the activation of PKA by 8-Br-cAMP; quantitative real-time-PCR (qRT-PCR) demonstrated that 8-Br-cAMP regulated the expression of vascular endothelial (VE)-cadherin, matrix metalloproteinase 2 (MMP2), ephrin type-A receptor 2 (EphA2), and VEGF in vivo. Experiments in vitro revealed that treatment with 8-Br-cAMP and U0126 decreased VEGF expression through PKA–ERK in CT26 cells by qRT-PCR. We further confirmed that tube formation of human umbilical vein endothelial cells was inhibited by 8-Br-cAMP in vitro. Discussion This study demonstrates that angiogenesis and VM are inhibited by 8-Br-cAMP treatment. Our data indicate that 8-Br-cAMP acts through the cAMP/PKA–ERK pathway and through EMT processes in CRC. These findings provide an insight into mechanisms of CRC and suggest that the cAMP/PKA–ERK pathway is a novel potential therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Sen Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Zhiyuan Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Wenwei Qian
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Dongjian Ji
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Qingyuan Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Bing Ji
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Yue Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Chuan Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Ye Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Chunyan Zhu
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Yueming Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| |
Collapse
|
39
|
You X, Wang Y, Wu J, Liu Q, Chen D, Tang D, Wang D. Prognostic significance of galectin-1 and vasculogenic mimicry in patients with gastric cancer. Onco Targets Ther 2018; 11:3237-3244. [PMID: 29881296 PMCID: PMC5985771 DOI: 10.2147/ott.s165899] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION We evaluated the expression of galectin-1 (Gal-1) and vasculogenic mimicry (VM) in gastric cancer (GC) and investigated their relationships with the clinicopathological factors and prognostic significance in GC. MATERIALS AND METHODS Immunohistochemical (IHC) staining and CD34-periodic acid-Schiff double stain were used to investigate Gal-1 expression and VM in paraffin-embedded sections from 127 patients with GC of all tumor stages. The relationships between Gal-1 expression and VM, clinicopathological variables, and survival were analyzed. P < 0.05 was considered statistically significant. RESULTS Among the 127 cases, 86 (67.7%) were positive for Gal-1; VM was detected in 29 cases (22.8%). There was a significant association between VM and the Gal-1 IHC staining; all cases with VM were positive for Gal-1 staining. Gal-1 expression and VM in primary GC tissue were associated with tumor size, differentiation, depth of tumor invasion, stage, lymph node metastases, and tumor emboli in microvessels (all, P < 0.05). Kaplan-Meier analysis revealed that the overall survival time was 52.56 ± 2.44 months (95% confidence interval [CI]: 47.77-57.35) for patients with Gal-1-negative and VM-negative primary GC tissue, 43.83 ± 2.17 months (95% CI: 39.58-48.08) for patients with Gal-1-positive but VM-negative primary GC tissue, and 23.97 ± 2.44 months (95% CI: 19.18-28.76) for patients with Gal-1-positive and VM-positive primary GC tissue (χ2 = 60.21, P < 0.01). Gal-1 expression was positively associated with VM in primary GC tissue. CONCLUSION Both Gal-1 expression and VM in primary GC tissue are indicators of poor prognosis for GC after gastrectomy, and Gal-1 may be a novel target for treating VM in GC.
Collapse
Affiliation(s)
- Xiaolan You
- Department of Integrated Traditional Chinese and Western Medicine, Medical College of Yangzhou University, Yangzhou, Jiangsu Province, People’s Republic of China
- Department of Gastrointestinal Surgery, Taizhou People’s Hospital, Taizhou, Jiangsu Province, People’s Republic of China
| | - Yuanjie Wang
- Department of Gastrointestinal Surgery, Taizhou People’s Hospital, Taizhou, Jiangsu Province, People’s Republic of China
| | - Jian Wu
- Department of Gastrointestinal Surgery, Taizhou People’s Hospital, Taizhou, Jiangsu Province, People’s Republic of China
| | - Qinghong Liu
- Department of Gastrointestinal Surgery, Taizhou People’s Hospital, Taizhou, Jiangsu Province, People’s Republic of China
| | - Dehu Chen
- Department of Gastrointestinal Surgery, Taizhou People’s Hospital, Taizhou, Jiangsu Province, People’s Republic of China
| | - Dong Tang
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People’s Hospital of Jiangsu Province), Yangzhou, Jiangsu Province, People’s Republic of China
| | - Daorong Wang
- Department of Gastrointestinal Surgery, Clinical Medical College of Yangzhou University (Subei People’s Hospital of Jiangsu Province), Yangzhou, Jiangsu Province, People’s Republic of China
| |
Collapse
|
40
|
Tang J, Wang J, Fan L, Li X, Liu N, Luo W, Wang J, Wang Y, Wang Y. cRGD inhibits vasculogenic mimicry formation by down-regulating uPA expression and reducing EMT in ovarian cancer. Oncotarget 2018; 7:24050-62. [PMID: 26992227 PMCID: PMC5029683 DOI: 10.18632/oncotarget.8079] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/29/2016] [Indexed: 11/25/2022] Open
Abstract
Vasculogenic minicry (VM), an alternative blood supply modality except to endothelial cells-mediated vascular network, is a potential therapeutic target for ovarian cancer due to VM correlated with poor prognosis in ovarian cancer patients. Accelerated extracellular matrix (ECM) degradation is prerequisite for VM formation induced by epithelial-mesenchymal transition (EMT). Previous reports demonstrate uPA has ability to degrade ECM thereby promoting tumor angiogenesis. Also, exogenous cRGD sequence enables to modulate uPA expression, attenuate EMT and suppress endothelial-lined channels. Till now, the correlation of uPA and VM formation and the effect of exogenous cRGD on VM formation remain unknown. Herein, we validate uPA expression is positively correlated with VM formation in ovarian cancer tissues (90 cases) and ovarian cancer cells (SKOV-3, OVCAR-3 and A2780 cells). In particular, silencing uPA experiments show that down-regulated uPA causes notable decrease for the complete channels formed by SKOV-3 and OVCAR-3 cells. Mechanism study discloses uPA promotes VM formation by regulating AKT/mTOR/MMP-2/Laminin5γ2 signal pathway. The result demonstrates uPA may serve as therapeutic target of VM for ovarian cancer. Also, it is found exogenous cRGD enables to inhibit VM formation in ovarian cancer via not only down-regulating uPA expression but also reducing EMT. Exogenous cRGD may be a promising angiogenic inhibitor for ovarian cancer therapy due to its inhibiting effect on VM formation as well as endothelial cells-mediated vascular network.
Collapse
Affiliation(s)
- Jiao Tang
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.,Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianguo Wang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lin Fan
- Department of Reproductive Medicine Center, The Women and Children Hospital of Guangdong, Guangzhou Medical University, Guangzhou 511442, China
| | - Xiaoxuan Li
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Na Liu
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Wanxian Luo
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jihui Wang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yifeng Wang
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ying Wang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
41
|
Zhang J, Deng G, Qiao L, Luo H, Liu Q, Liang N, Xie J, Zhang J. Effect of galectin-3 on vasculogenic mimicry in esophageal cancer cells. Oncol Lett 2018; 15:4907-4911. [PMID: 29552129 PMCID: PMC5840659 DOI: 10.3892/ol.2018.7959] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 07/14/2017] [Indexed: 12/13/2022] Open
Abstract
Galectin-3 is a multifunctional β-galactoside binding lectin associated with tumor progression. Previous studies confirmed the roles of galecin-3 overexpression and silencing in the biological behavior of Eca109 human esophageal cancer (EC) cells; galectin-3 may serve a critical role in the vasculogenic mimicry (VM) of tumors. Therefore, the present study examined the effects of galectin-3 knockdown using lentivirus vectors on VM in EC. Eca109 and EC9706 EC cells were transfected with a lentiviral vector to inhibit galectin-3 expression, or a control vector. VM formation in vitro was evaluated via 3D culture. Western blotting was used to detect the expression level of galectin-3 following galectin-3 silencing and the expression levels of VE-cadherin, ephrin type-A receptor 2 precursor (EphA2) and matrix metalloproteinase 2 (MMP-2). According to the results of western blot analysis, the Eca109/galectin-3 and EC9706/galectin-3 cells exhibited effective galectin-3 silencing (P<0.05). Eca109 and EC9706 cells formed typical tubular networks; the number of tubular networks markedly decreased subsequent to galectin-3 knockdown. The expression levels of MMP-2 and EphA2 proteins in Eca109/galectin-3 and EC9706/galectin-3 cells were lower compared with those in Eca109, EC9706, and control vector-transfected Eca109 and EC9706 cells (P<0.05); however, there was no significant difference in the expression of VE-cadherin proteins. These results indicated that galectin-3 may modulate VM in EC by regulating the EphA2 expression level, which affects VM formation via MMP-2.
Collapse
Affiliation(s)
- Jingxin Zhang
- Graduate School, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Guodong Deng
- Department of Graduate, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Lili Qiao
- Department of Graduate, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Hui Luo
- Graduate School, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Qiqi Liu
- Department of Graduate, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Ning Liang
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Jian Xie
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Jiandong Zhang
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
42
|
Irani S, Dehghan A. Expression of Vascular Endothelial-Cadherin in Mucoepidermoid Carcinoma: Role in Cancer Development. J Int Soc Prev Community Dent 2017; 7:301-307. [PMID: 29387612 PMCID: PMC5774049 DOI: 10.4103/jispcd.jispcd_323_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/30/2017] [Indexed: 12/29/2022] Open
Abstract
Objectives: Mucoepidermoid carcinoma (MEC) accounts for 35% of all malignant salivary gland tumors. Previous investigations have shown that vasculogenic mimicry (VM) exists in many cancers which can be used as a prognostic factor of poor prognosis. Elevated expression level of vascular endothelial (VE)-cadherin has been implicated in cancer neovascularization, growth, and progression. The current study aimed to study the presence of VE-cadherin in VM channels and tumor cells in different grades of MEC. Materials and Methods: A total of 63 MEC samples (21 samples in each grade) were collected from the archive of pathology department of Besat Educational Hospital, Hamadan, Iran, from 2002 to 2016. Hematoxylin and eosin staining was performed to confirm the previous diagnosis. The specimens were then processed for immunohistochemistry analysis. Then, periodic acid–Schiff staining was performed. Analyses were conducted through SPSS software version 22.0 (SPSS, Inc., Chicago, IL, USA). Chi-square test was used to examine the differences between categorical variables. Significance level was set at 0.05. Pearson's correlation was used to assess the co-localization of the marker. Results: A total of 63 samples (35 men; 55.6%, and 28 women; 44.4%) were used for immunohistochemical study. There were statistically significant differences between tumor grade and the expression levels of VE-cadherin (P = 0.000), between tumor grade and VM formation (P = 0.000), and also between tumor grade and microvessel density (MVD) (P = 0.000). Additionally, there was a strong positive correlation between tumor grade and VE-cadherin expression level (Pearson's r = 0.875, P < 0.000). Conclusions: Our results may disclose a definite relationship between VE-cadherin expression level, VM, epithelial–mesenchymal transition, cancer stem cells, and MVD in MEC samples. Thus, it is reasonable to suggest that VE-cadherin is related to angiogenesis and VM formation in MECs.
Collapse
Affiliation(s)
- Soussan Irani
- Department of Oral Pathology, Dental Research Centre, Research Centre for Molecular Medicine, Dental Faculty, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Arash Dehghan
- Department of Pathology, Besat Hospital, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
43
|
Collateral Damage Intended-Cancer-Associated Fibroblasts and Vasculature Are Potential Targets in Cancer Therapy. Int J Mol Sci 2017; 18:ijms18112355. [PMID: 29112161 PMCID: PMC5713324 DOI: 10.3390/ijms18112355] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/25/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023] Open
Abstract
After oncogenic transformation, tumor cells rewire their metabolism to obtain sufficient energy and biochemical building blocks for cell proliferation, even under hypoxic conditions. Glucose and glutamine become their major limiting nutritional demands. Instead of being autonomous, tumor cells change their immediate environment not only by their metabolites but also by mediators, such as juxtacrine cell contacts, chemokines and other cytokines. Thus, the tumor cells shape their microenvironment as well as induce resident cells, such as fibroblasts and endothelial cells (ECs), to support them. Fibroblasts differentiate into cancer-associated fibroblasts (CAFs), which produce a qualitatively and quantitatively different extracellular matrix (ECM). By their contractile power, they exert tensile forces onto this ECM, leading to increased intratumoral pressure. Moreover, along with enhanced cross-linkage of the ECM components, CAFs thus stiffen the ECM. Attracted by tumor cell- and CAF-secreted vascular endothelial growth factor (VEGF), ECs sprout from pre-existing blood vessels during tumor-induced angiogenesis. Tumor vessels are distinct from EC-lined vessels, because tumor cells integrate into the endothelium or even mimic and replace it in vasculogenic mimicry (VM) vessels. Not only the VM vessels but also the characteristically malformed EC-lined tumor vessels are typical for tumor tissue and may represent promising targets in cancer therapy.
Collapse
|
44
|
Zhu B, Zhou L, Yu L, Wu S, Song W, Gong X, Wang D. Evaluation of the correlation of vasculogenic mimicry, ALDH1, KAI1 and microvessel density in the prediction of metastasis and prognosis in colorectal carcinoma. BMC Surg 2017; 17:47. [PMID: 28431527 PMCID: PMC5399824 DOI: 10.1186/s12893-017-0246-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/13/2017] [Indexed: 01/13/2023] Open
Abstract
Background Metastasis and recurrence are the most common reasons for treatment failure of colorectal carcinoma (CRC). Vasculogenic mimicry (VM, blood supply formation often seen in highly aggressive tumors), Aldehyde dehydrogenase 1 (ALDH1, a biomarker of cancer stem cells), KAI1 (a suppressor gene of tumor metastasis) are all valuable factors for metastasis and prognosis in diverse human cancers. However, the correlation of VM, ALDH1, KAI1 and microvessel density (MVD) in CRC is unclear. In this study, we analyzed the correlations among VM, ALDH1, KAI1 and MVD, as well as their respective correlations with clinicopathological parameters and survival in CRC. Methods The level of VM, ALDH1, KAI1 and MVD in 204 whole tissue samples of CRC were examined by immunhistochemistry. Clinical data was also collected. Results Levels of VM, ALDH1 and MVD were significantly higher, and levels of KAI1 significantly lower, in CRC tissues than in normal colorectal tissues. Levels of VM, ALDH1 and MVD were positively associated with invasion of depth, lymph node metastasis (LNM), distant metastasis and tumor-node-metastasis (TNM) stages, and negatively with patients’ overall survival (OS). Levels of KAI1 was negatively correlated with invasion of depth, LNM, distant metastasis and TNM stages, and the KAI1 positive expression subgroup had significantly longer OS than did the KAI1- subgroup. In multivariate analysis, high levels of VM, ALDH1 and KAI1, as well as TNM stages were independently correlated with lower OS in patients with CRC. Conclusions VM, MVD and the expression of ALDH1 and KAI1 may represent promising metastatic and prognostic biomarkers, as well as potential therapeutic targets for CRC.
Collapse
Affiliation(s)
- Bo Zhu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, Anhui Province, China
| | - Lei Zhou
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, Anhui Province, China
| | - Lan Yu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, Anhui Province, China
| | - Shiwu Wu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, Anhui Province, China.
| | - Wenqing Song
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, Anhui Province, China
| | - Xiaomeng Gong
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, Anhui Province, China
| | - Danna Wang
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, Anhui Province, China
| |
Collapse
|
45
|
Delgado-Bellido D, Serrano-Saenz S, Fernández-Cortés M, Oliver FJ. Vasculogenic mimicry signaling revisited: focus on non-vascular VE-cadherin. Mol Cancer 2017; 16:65. [PMID: 28320399 PMCID: PMC5359927 DOI: 10.1186/s12943-017-0631-x] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 03/06/2017] [Indexed: 12/11/2022] Open
Abstract
Vasculogenic mimicry (VM) is a blood supply system independent of endothelial vessels in tumor cells from different origins. It reflects the plasticity of aggressive tumor cells that express vascular cell markers and line tumor vasculature. The presence of VM is associated with a high tumor grade, short survival, invasion and metastasis. Endothelial cells (ECs) express various members of the cadherin superfamily, in particular vascular endothelial (VE-) cadherin, which is the main adhesion receptor of endothelial adherent junctions. Aberrant extra-vascular expression of VE-cadherin has been observed in certain cancer types associated with VM. In this review we focus on non-endothelial VE-cadherin as a prominent factor involved in the acquisition of tubules-like structures by aggressive tumor cells and we summarize the specific signaling pathways, the association with trans-differentiation and stem-like phenotype and the therapeutic opportunities derived from the in-depth knowledge of the peculiarities of the biology of VE-cadherin and other key components of VM.
Collapse
Affiliation(s)
| | | | | | - F Javier Oliver
- IPBLN, CSIC, CIBERONC, Granada, Spain. .,IPBLN, CSIC, Av. Conocimiento s/n, 18016, Granada, Spain.
| |
Collapse
|
46
|
Yu L, Zhu B, Wu S, Zhou L, Song W, Gong X, Wang D. Evaluation of the correlation of vasculogenic mimicry, ALDH1, KiSS-1, and MACC1 in the prediction of metastasis and prognosis in ovarian carcinoma. Diagn Pathol 2017; 12:23. [PMID: 28253891 PMCID: PMC5335811 DOI: 10.1186/s13000-017-0612-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 02/20/2017] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Recurrence and metastasis are the usual manifestations of treatment failure of epithelial ovarian carcinoma (EOC). Vasculogenic mimicry (VM; blood supply development often seen in highly aggressive cancers), aldehyde dehydrogenase 1 (ALDH1, cancer stem cell biomarker), KiSS-1 (suppressor of tumor metastasis), and metastasis associated in colon cancer-1 (MACC1) are all useful predictive factors for metastasis and prognosis in various cancers. In this study, we analyzed associations among VM, ALDH1, KiSS-1, and MACC1 in EOC, and their respective correlations with clinicopathological characteristics and survival in EOC. METHODS Positive rates of VM, ALDH1, KiSS-1, and MACC1 in 207 whole EOC tissue samples were detected by immunohistochemistry. Patients' clinical data were also collected. RESULTS Levels of VM, ALDH1, and MACC1 were significantly higher, and levels of KiSS-1 significantly lower, in EOC tissues than in benign ovary tumors. Levels of VM, ALDH1, KiSS-1, and MACC1 were associated significantly with tumor/lymph node/metastasis (LNM) grade, implantation, and International Federation of Gynecology and Obstetrics (FIGO) stage, and with patients' overall survival (OS); whereas the KiSS-1+ subgroup had significantly longer OS than did the KiSS-1- subgroup. In multivariate analysis, high VM, ALDH1 or MACC1 levels, FIGO stage, implantation and low KiSS-1 levels were independently associated with shorter OS in patients with EOC. CONCLUSIONS VM and expressions of ALDH1, KiSS-1, and MACC1 represent promising markers for metastasis and prognosis, and potential therapeutic targets for EOC.
Collapse
Affiliation(s)
- Lan Yu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Bo Zhu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Shiwu Wu
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China.
| | - Lei Zhou
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Wenqing Song
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Xiaomeng Gong
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| | - Danna Wang
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, No.287, Changhuai Road, Bengbu, 233003, Anhui Province, China
| |
Collapse
|
47
|
Williamson SC, Metcalf RL, Trapani F, Mohan S, Antonello J, Abbott B, Leong HS, Chester CPE, Simms N, Polanski R, Nonaka D, Priest L, Fusi A, Carlsson F, Carlsson A, Hendrix MJC, Seftor REB, Seftor EA, Rothwell DG, Hughes A, Hicks J, Miller C, Kuhn P, Brady G, Simpson KL, Blackhall FH, Dive C. Vasculogenic mimicry in small cell lung cancer. Nat Commun 2016; 7:13322. [PMID: 27827359 PMCID: PMC5105195 DOI: 10.1038/ncomms13322] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/22/2016] [Indexed: 02/08/2023] Open
Abstract
Small cell lung cancer (SCLC) is characterized by prevalent circulating tumour cells (CTCs), early metastasis and poor prognosis. We show that SCLC patients (37/38) have rare CTC subpopulations co-expressing vascular endothelial-cadherin (VE-cadherin) and cytokeratins consistent with vasculogenic mimicry (VM), a process whereby tumour cells form 'endothelial-like' vessels. Single-cell genomic analysis reveals characteristic SCLC genomic changes in both VE-cadherin-positive and -negative CTCs. Higher levels of VM are associated with worse overall survival in 41 limited-stage patients' biopsies (P<0.025). VM vessels are also observed in 9/10 CTC patient-derived explants (CDX), where molecular analysis of fractionated VE-cadherin-positive cells uncovered copy-number alterations and mutated TP53, confirming human tumour origin. VE-cadherin is required for VM in NCI-H446 SCLC xenografts, where VM decreases tumour latency and, despite increased cisplatin intra-tumour delivery, decreases cisplatin efficacy. The functional significance of VM in SCLC suggests VM regulation may provide new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Stuart C. Williamson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Robert L. Metcalf
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Francesca Trapani
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Sumitra Mohan
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Jenny Antonello
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Benjamin Abbott
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Hui Sun Leong
- Computational Biology Support Team, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Christopher P. E. Chester
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Nicole Simms
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Radoslaw Polanski
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Daisuke Nonaka
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Lynsey Priest
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Alberto Fusi
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Fredrika Carlsson
- University of Southern California Dornsife, Los Angeles, California 90089-3301, USA
| | - Anders Carlsson
- University of Southern California Dornsife, Los Angeles, California 90089-3301, USA
| | - Mary J. C. Hendrix
- Stanley Manne Children's Research Institute, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Richard E. B. Seftor
- Stanley Manne Children's Research Institute, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Elisabeth A. Seftor
- Stanley Manne Children's Research Institute, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Dominic G. Rothwell
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Andrew Hughes
- The Institute of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK
| | - James Hicks
- University of Southern California Dornsife, Los Angeles, California 90089-3301, USA
| | - Crispin Miller
- RNA Biology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Peter Kuhn
- University of Southern California Dornsife, Los Angeles, California 90089-3301, USA
| | - Ged Brady
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Kathryn L. Simpson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Fiona H. Blackhall
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
- The Institute of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK
- Cancer Research UK, Lung Cancer Centre of Excellence, Manchester M20 4BX, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
- Cancer Research UK, Lung Cancer Centre of Excellence, Manchester M20 4BX, UK
| |
Collapse
|
48
|
The hypoxia-related signaling pathways of vasculogenic mimicry in tumor treatment. Biomed Pharmacother 2016; 80:127-135. [DOI: 10.1016/j.biopha.2016.03.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 03/13/2016] [Accepted: 03/13/2016] [Indexed: 12/20/2022] Open
|
49
|
Ju RJ, Zeng F, Liu L, Mu LM, Xie HJ, Zhao Y, Yan Y, Wu JS, Hu YJ, Lu WL. Destruction of vasculogenic mimicry channels by targeting epirubicin plus celecoxib liposomes in treatment of brain glioma. Int J Nanomedicine 2016; 11:1131-46. [PMID: 27042063 PMCID: PMC4809325 DOI: 10.2147/ijn.s94467] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The efficacy of chemotherapy for brain glioma is restricted by the blood–brain barrier (BBB), and surgery or radiotherapy cannot eliminate the glioma cells because of their unique location. Residual brain glioma cells can form vasculogenic mimicry (VM) channels that can cause a recurrence of brain glioma. In the present study, targeting liposomes incorporating epirubicin and celecoxib were prepared and used for the treatment of brain glioma, along with the destruction of their VM channels. Evaluations were performed on the human brain glioma U87MG cells in vitro and on intracranial brain glioma-bearing nude mice. Targeting epirubicin plus celecoxib liposomes in the circulatory blood system were able to be transported across the BBB, and accumulated in the brain glioma region. Then, the liposomes were internalized by brain glioma cells and killed glioma cells by direct cytotoxic injury and the induction of apoptosis. The induction of apoptosis was related to the activation of caspase-8- and -3-signaling pathways, the activation of the proapoptotic protein Bax, and the suppression of the antiapoptotic protein Mcl-1. The destruction of brain glioma VM channels was related to the downregulation of VM channel-forming indictors, which consisted of MMP-2, MMP-9, FAK, VE-Cad, and VEGF. The results demonstrated that the targeting epirubicin plus celecoxib liposomes were able to effectively destroy the glioma VM channels and exhibited significant efficacy in the treatment of intracranial glioma-bearing nude mice. Therefore, targeting epirubicin plus celecoxib liposomes could be a potential nanostructured formulation to treat gliomas and destroy their VM channels.
Collapse
Affiliation(s)
- Rui-Jun Ju
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China; Department of Pharmaceutical Engineering, Beijing Institute of Petrochemical Technology, Beijing, People's Republic of China
| | - Fan Zeng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Lei Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Li-Min Mu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Hong-Jun Xie
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Yao Zhao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Yan Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Jia-Shuan Wu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Ying-Jie Hu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Wan-Liang Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| |
Collapse
|
50
|
miR-27a-3p suppresses tumor metastasis and VM by down-regulating VE-cadherin expression and inhibiting EMT: an essential role for Twist-1 in HCC. Sci Rep 2016; 6:23091. [PMID: 26980408 PMCID: PMC4793289 DOI: 10.1038/srep23091] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/26/2016] [Indexed: 12/30/2022] Open
Abstract
Twist-1 and miRNAs have been reported to be associated with tumor metastasis and angiogenesis. However, the relationship between Twist-1 and miRNAs and the function of miRNAs remain largely undefined. We aimed to reveal the Twist-1-related miRNA expression profile and to determine whether Twist-1 functions in tumor metastasis and vasculogenic mimicry (VM) by regulating miRNA expression in hepatocellular carcinoma (HCC). Results showed that the expression of miR-27a-3p was consistently down-regulated in HCC cell lines and tissue samples displaying high expression of Twist-1. Both loss- and gain-of-function assays revealed suppressive effects of miR-27a-3p. Low miR-27a-3p expression was significantly associated with early metastasis in HCC. Subsequent investigations revealed that miR-27a-3p mediated the inhibition of epithelial–mesenchymal transition (EMT). Additional experiments showed that VE-cadherin is a direct target of miR-27a-3p and further demonstrated the critical role of miR-27a-3p in suppressing tumor metastasis and VM. Conclusions: Twist-1 up-regulation in HepG2 cells resulted in the differential expression of 18 miRNAs. Among them, miR-27a-3p deregulation contributed to VM and metastasis. The miR-27a-3p-mediated down-regulation of VE-cadherin and inhibition of EMT may be essential for Twist-1 to induce tumor metastasis and VM. Our findings highlight the importance of miR-27a-3p and suggest a promising new strategy for anti-HCC therapy.
Collapse
|