1
|
Xu ZY, Han J, Yang K, Zhang GM, Jiao MN, Liang SX, Yan YB, Chen W. HSP27 promotes vasculogenic mimicry formation in human salivary adenoid cystic carcinoma via the AKT-MMP-2/9 pathway. Oral Surg Oral Med Oral Pathol Oral Radiol 2024; 137:515-528. [PMID: 38553306 DOI: 10.1016/j.oooo.2024.02.016] [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: 07/16/2023] [Revised: 01/06/2024] [Accepted: 02/20/2024] [Indexed: 04/30/2024]
Abstract
PURPOSE To explore the role and mechanism of heat shock protein 27 (HSP27) in SACC VM formation. STUDY DESIGN Immunohistochemistry and double staining with cluster of differentiation 31 (CD31) and periodic acid-Schiff (PAS) were used to detect HSP27 expression and VM in 70 SACC tissue samples separately. Quantitative real-time polymerase chain reaction (qRT-PCR), western blot analysis, and immunofluorescence were used to detect gene and protein expression. HSP27 in SACC cells were overexpression or downregulated by transfecting HSP27 or short hairpin RNA target HSP27 (sh-HSP27). The migration and invasion abilities of SACC cells were detected using wound healing and Transwell invasion assays. The VM formation ability of the cells in vitro was detected using a Matrigel 3-dimensional culture. RESULTS HSP27 expression was positively correlated with VM formation and affected the prognosis of patients. In vitro, HSP27 upregulation engendered VM formation and the invasion and migration of SACC cells. Mechanistically, HSP27 upregulation increased Akt phosphorylation and subsequently increased downstream matrix metalloproteinase 2 and 9 expressions. CONCLUSION HSP27 may plays an important role in VM formation in SACC via the AKT-MMP-2/9 signalling pathway.
Collapse
Affiliation(s)
- Zhao-Yuan Xu
- Department of Oral Medical Center, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241000, China; Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241000, China; Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Jing Han
- Department of Oral Implantology, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Kun Yang
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Guan-Meng Zhang
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Mai-Ning Jiao
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China
| | - Su-Xia Liang
- Department of Operative Dentistry and Endodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China.
| | - Ying-Bin Yan
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China.
| | - Wei Chen
- Department of Oromaxillofacial-Head and Neck Surgery, Tianjin Stomatological Hospital, School of Medicine, Nankai University, 75 Dagu Road, Heping District, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Road, Heping District, Tianjin 300041, China.
| |
Collapse
|
2
|
Shevchenko JA, Nazarov KV, Alshevskaya AA, Sennikov SV. Erythroid Cells as Full Participants in the Tumor Microenvironment. Int J Mol Sci 2023; 24:15141. [PMID: 37894821 PMCID: PMC10606658 DOI: 10.3390/ijms242015141] [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: 09/12/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The tumor microenvironment is an important factor that can determine the success or failure of antitumor therapy. Cells of hematopoietic origin are one of the most important mediators of the tumor-host interaction and, depending on the cell type and functional state, exert pro- or antitumor effects in the tumor microenvironment or in adjacent tissues. Erythroid cells can be full members of the tumor microenvironment and exhibit immunoregulatory properties. Tumor growth is accompanied by the need to obtain growth factors and oxygen, which stimulates the appearance of the foci of extramedullary erythropoiesis. Tumor cells create conditions to maintain the long-term proliferation and viability of erythroid cells. In turn, tumor erythroid cells have a number of mechanisms to suppress the antitumor immune response. This review considers current data on the existence of erythroid cells in the tumor microenvironment, formation of angiogenic clusters, and creation of optimal conditions for tumor growth. Despite being the most important life-support function of the body, erythroid cells support tumor growth and do not work against it. The study of various signaling mechanisms linking tumor growth with the mobilization of erythroid cells and the phenotypic and functional differences between erythroid cells of different origin allows us to identify potential targets for immunotherapy.
Collapse
Affiliation(s)
- Julia A. Shevchenko
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (J.A.S.); (K.V.N.)
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution, Ministry of Health of the Russian Federation, Higher Education I.M. Sechenov First Moscow State Medical University, Sechenov University, 119048 Moscow, Russia;
| | - Kirill V. Nazarov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (J.A.S.); (K.V.N.)
| | - Alina A. Alshevskaya
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution, Ministry of Health of the Russian Federation, Higher Education I.M. Sechenov First Moscow State Medical University, Sechenov University, 119048 Moscow, Russia;
| | - Sergey V. Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution, Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (J.A.S.); (K.V.N.)
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution, Ministry of Health of the Russian Federation, Higher Education I.M. Sechenov First Moscow State Medical University, Sechenov University, 119048 Moscow, Russia;
| |
Collapse
|
3
|
Grizzi F, Hegazi MAAA, Zanoni M, Vota P, Toia G, Clementi MC, Mazzieri C, Chiriva-Internati M, Taverna G. Prostate Cancer Microvascular Routes: Exploration and Measurement Strategies. Life (Basel) 2023; 13:2034. [PMID: 37895416 PMCID: PMC10608780 DOI: 10.3390/life13102034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Angiogenesis is acknowledged as a pivotal feature in the pathology of human cancer. Despite the absence of universally accepted markers for gauging the comprehensive angiogenic activity in prostate cancer (PCa) that could steer the formulation of focused anti-angiogenic treatments, the scrutiny of diverse facets of tumoral blood vessel development may furnish significant understanding of angiogenic processes. Malignant neoplasms, encompassing PCa, deploy a myriad of strategies to secure an adequate blood supply. These modalities range from sprouting angiogenesis and vasculogenesis to intussusceptive angiogenesis, vascular co-option, the formation of mosaic vessels, vasculogenic mimicry, the conversion of cancer stem-like cells into tumor endothelial cells, and vascular pruning. Here we provide a thorough review of these angiogenic mechanisms as they relate to PCa, discuss their prospective relevance for predictive and prognostic evaluations, and outline the prevailing obstacles in quantitatively evaluating neovascularization via histopathological examinations.
Collapse
Affiliation(s)
- Fabio Grizzi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
| | - Mohamed A. A. A. Hegazi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Matteo Zanoni
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Paolo Vota
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Giovanni Toia
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Maria Chiara Clementi
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Cinzia Mazzieri
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Maurizio Chiriva-Internati
- Departments of Gastroenterology, Hepatology & Nutrition, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Gianluigi Taverna
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| |
Collapse
|
4
|
Wei Y, Jiao Z, Sun T, Lai Z, Wang X. Molecular Mechanisms Behind Vascular Mimicry as the Target for Improved Breast Cancer Management. Int J Womens Health 2023; 15:1027-1038. [PMID: 37465721 PMCID: PMC10350405 DOI: 10.2147/ijwh.s406327] [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: 01/29/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023] Open
Abstract
Introduction Breast cancer has a high incidence and mortality rate in women due to metastasis and drug resistance which is associated with vasculogenic mimicry (VM). We purposed to explore VM formulation in breast cancer and mechanism of which is involved in EphA2/PIK3R1/CTNNB1 in the present study. Methods The expression of EphA2/PIK3R1/CTNNB1 and breast cancer patient prognosis was analyzed from TCGA data, both gene and protein expression as well as VM were measured in human breast cancer tissue samples collected in our study. The relationship between EphA2/PIK3R1/CTNNB1 and the formation of VM in breast cancer and its possible regulatory mechanism was explored. Results The results of the bioinformatics analysis based on TCGA showed that the expression of PIK3R1/ CTNNB1/ PECAM1 (CD31) in tumor tissues was significantly lower than that in normal tissues. EphA2 was positively correlated with PIK3R1, PIK3R1 with CTNNB1, and CTNNB1 with PECAM1 expression in breast cancer tissues. The results of detection in breast cancer and adjacent tissues indicated that the expression of EphA2/PIK3R1/CTNNB1 in cancer tissues was significantly lower than that in adjacent tissues. The expression of PIK3R1 was positively correlated with EphA2 and CTNNB1 expression, respectively, as well as EphA2 expression correlated with CTNNB1 expression positively. VM formation was significantly increased in breast cancer tissues compared with adjacent tissues. Conclusion Our results suggested that the formation of VM in breast cancer may be related to the EphA2/PIK3R1/CTNNB1 molecular signaling pathway.
Collapse
Affiliation(s)
- Yali Wei
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei Province, People’s Republic of China
| | - Zheng Jiao
- Department of Neurosurgery, Youanmen Hospital, Beijing, People’s Republic of China
| | - Tianpei Sun
- Clinical School of Medicine, Hebei University, Baoding, Hebei Province, People’s Republic of China
| | - Zhiwei Lai
- Department of Thoracic Surgery, Shanghai Sixth People’s Hospital Fujian Campus, Jinjiang, Fujian Province, People’s Republic of China
| | - Xiaochun Wang
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei Province, People’s Republic of China
| |
Collapse
|
5
|
Delgado-Bellido D, Oliver FJ, Vargas Padilla MV, Lobo-Selma L, Chacón-Barrado A, Díaz-Martin J, de Álava E. VE-Cadherin in Cancer-Associated Angiogenesis: A Deceptive Strategy of Blood Vessel Formation. Int J Mol Sci 2023; 24:ijms24119343. [PMID: 37298296 DOI: 10.3390/ijms24119343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Tumor growth depends on the vascular system, either through the expansion of blood vessels or novel adaptation by tumor cells. One of these novel pathways is vasculogenic mimicry (VM), which is defined as a tumor-provided vascular system apart from endothelial cell-lined vessels, and its origin is partly unknown. It involves highly aggressive tumor cells expressing endothelial cell markers that line the tumor irrigation. VM has been correlated with high tumor grade, cancer cell invasion, cancer cell metastasis, and reduced survival of cancer patients. In this review, we summarize the most relevant studies in the field of angiogenesis and cover the various aspects and functionality of aberrant angiogenesis by tumor cells. We also discuss the intracellular signaling mechanisms involved in the abnormal presence of VE-cadherin (CDH5) and its role in VM formation. Finally, we present the implications for the paradigm of tumor angiogenesis and how targeted therapy and individualized studies can be applied in scientific analysis and clinical settings.
Collapse
Affiliation(s)
- Daniel Delgado-Bellido
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
- Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, 41013 Seville, Spain
| | - F J Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain
| | | | - Laura Lobo-Selma
- Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, 41013 Seville, Spain
| | | | - Juan Díaz-Martin
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
- Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, 41013 Seville, Spain
| | - Enrique de Álava
- Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
- Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, 41013 Seville, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
| |
Collapse
|
6
|
Li F, Sun H, Yu Y, Che N, Han J, Cheng R, Zhao N, Guo Y, Huang C, Zhang D. RIPK1-dependent necroptosis promotes vasculogenic mimicry formation via eIF4E in triple-negative breast cancer. Cell Death Dis 2023; 14:335. [PMID: 37217473 PMCID: PMC10203343 DOI: 10.1038/s41419-023-05841-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/24/2023]
Abstract
Necroptosis is a caspase-independent form of programmed cell death. Receptor interacting protein kinase 1 (RIPK1) is a key molecule in the initiation of necroptosis and the formation of the necrotic complex. Vasculogenic mimicry (VM) provides a blood supply to tumor cells that is not dependent on endothelial cells. However, the relationship between necroptosis and VM in triple-negative breast cancer (TNBC) is not fully understood. In this study, we found that RIPK1-dependent necroptosis promoted VM formation in TNBC. Knockdown of RIPK1 significantly suppressed the number of necroptotic cells and VM formation. Moreover, RIPK1 activated the p-AKT/eIF4E signaling pathway during necroptosis in TNBC. eIF4E was blocked by knockdown of RIPK1 or AKT inhibitors. Furthermore, we found that eIF4E promoted VM formation by promoting epithelial-mesenchymal transition (EMT) and the expression and activity of MMP2. In addition to its critical role in necroptosis-mediated VM, eIF4E was essential for VM formation. Knockdown of eIF4E significantly suppressed VM formation during necroptosis. Finally, through clinical significance, the results found that eIF4E expression in TNBC was positively correlated with the mesenchymal marker vimentin, the VM marker MMP2, and the necroptosis markers MLKL and AKT. In conclusion, RIPK1-dependent necroptosis promotes VM formation in TNBC. Necroptosis promotes VM formation by activating RIPK1/p-AKT/eIF4E signaling in TNBC. eIF4E promotes EMT and MMP2 expression and activity, leading to VM formation. Our study provides a rationale for necroptosis-mediated VM and also providing a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Fan Li
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Huizhi Sun
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China
| | - Yihui Yu
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Na Che
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Jiyuan Han
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Runfen Cheng
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Yuhong Guo
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China
| | - Chongbiao Huang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China.
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China.
| |
Collapse
|
7
|
Zhang X, Wang Q, Zhang R, Kong Z. DAB2IP-knocking down resulted in radio-resistance of breast cancer cells is associated with increased hypoxia and vasculogenic mimicry formation. Int J Radiat Biol 2023; 99:1595-1606. [PMID: 36947637 DOI: 10.1080/09553002.2023.2194390] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 03/16/2023] [Indexed: 03/24/2023]
Abstract
PURPOSE As a part of breast-conserving therapy (BCT), postoperative radiotherapy is one of the main means to improve the clinical efficacy of breast cancer (BCa). However, ionizing radiation (IR) may induce BCa cells to develop radioresistance, which causes tumor recurrence and metastasis after treatment. Recently, DOC-2/DAB2 interactive protein (DAB2IP) has been reported often down-regulated in a variety of cancers and is related to tumor tolerance to radiotherapy. In this study, BCa cell lines were introduced to study how DAB2IP deficient influenced BCa cell radiosensitivity in vitro and in vivo and discuss the possible mechanism. METHODS AND MATERIALS Small RNA interference system (siRNA) was employed to decrease DAB2IP expression in two BCa cell lines, MDA-MB-231 and 4T1. Cells in response to IR or antineoplastics were detected by clone formation assay or MTT method, respectively. For in vivo studies, siDAB2IP or siControl cells were subcutaneously injected into the right flank of each female mouse. Sphere formation assay, soft agar colony anchoring assay and in vivo tumorigenesis assay were implemented to examine the stem cell-like features of BCa cells. Tube formation assay as well as immunofluorescence assay (IFA) were respectively applied to determine the angiogenesis of tumor cells in vitro and in vivo. The expression of a series of angiogenesis-related molecules was analyzed by qRT-PCR, western blot and IFA. RESULTS It was observed that the downregulation of DAB2IP could significantly improve the clone formation ability of BCa cells, reduce their sensitivity to radiation and chemotherapy drugs, enhance their migration and invasion abilities and increase their stemness characteristics. It was also noted that either DAB2IP-knocking down or treated with the conditioned medium from DAB2IP-deficient BCa cells could promote the tube-forming ability of the endothelial cell. Similarly, in vivo studies showed that tumors developed from siDAB2IP BCa cells had higher tumor microvascular density (MVD) and more severe oxygen deficiency than that in DAB2IP- sufficient tumors. Meanwhile, Knock-down of DAB2IP inhibited vascular maturation and promoted the formation of vasculogenic mimicry (VM) in BCa tissues. Down-regulation of STAT3 could enhance siDAB2IP cells sensitivity to IR, accompanied by the decrease of VEGF expression. CONCLUSIONS Our data support that loss of DAB2IP confers radio-resistance of BCa could be due to increased hypoxia, inhibited vascular maturation and promoted VM formation. STAT3 inhibition could be a potential way to overcome such DAB2IP-deficient induced tolerance in BCT.
Collapse
Affiliation(s)
- Xiangyan Zhang
- Department of Radiobiology, Institute of Radiation Medicine, Fudan University, Shanghai, P.R. China
| | - Quanxin Wang
- Department of Radiobiology, Institute of Radiation Medicine, Fudan University, Shanghai, P.R. China
| | - Ruiqi Zhang
- Department of Radiobiology, Institute of Radiation Medicine, Fudan University, Shanghai, P.R. China
| | - Zhaolu Kong
- Department of Radiobiology, Institute of Radiation Medicine, Fudan University, Shanghai, P.R. China
| |
Collapse
|
8
|
Rhizoma Paridis saponins suppresses vasculogenic mimicry formation and metastasis in osteosarcoma through regulating miR-520d-3p/MIG-7 axis. J Pharmacol Sci 2022; 150:180-190. [DOI: 10.1016/j.jphs.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/25/2022] [Accepted: 08/25/2022] [Indexed: 11/19/2022] Open
|
9
|
Zhou X, Zhou M, Zheng M, Tian S, Yang X, Ning Y, Li Y, Zhang S. Polyploid giant cancer cells and cancer progression. Front Cell Dev Biol 2022; 10:1017588. [PMID: 36274852 PMCID: PMC9581214 DOI: 10.3389/fcell.2022.1017588] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/21/2022] [Indexed: 12/02/2022] Open
Abstract
Polyploid giant cancer cells (PGCCs) are an important feature of cellular atypia, the detailed mechanisms of their formation and function remain unclear. PGCCs were previously thought to be derived from repeated mitosis/cytokinesis failure, with no intrinsic ability to proliferate and divide. However, recently, PGCCs have been confirmed to have cancer stem cell (CSC)-like characteristics, and generate progeny cells through asymmetric division, which express epithelial-mesenchymal transition-related markers to promote invasion and migration. The formation of PGCCs can be attributed to multiple stimulating factors, including hypoxia, chemotherapeutic reagents, and radiation, can induce the formation of PGCCs, by regulating the cell cycle and cell fusion-related protein expression. The properties of CSCs suggest that PGCCs can be induced to differentiate into non-tumor cells, and produce erythrocytes composed of embryonic hemoglobin, which have a high affinity for oxygen, and thereby allow PGCCs survival from the severe hypoxia. The number of PGCCs is associated with metastasis, chemoradiotherapy resistance, and recurrence of malignant tumors. Targeting relevant proteins or signaling pathways related with the formation and transdifferentiation of adipose tissue and cartilage in PGCCs may provide new strategies for solid tumor therapy.
Collapse
Affiliation(s)
- Xinyue Zhou
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Mingming Zhou
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Shifeng Tian
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
- *Correspondence: Shiwu Zhang,
| |
Collapse
|
10
|
Palkina N, Aksenenko M, Zemtsov D, Lavrentev S, Zinchenko I, Belenyuk V, Kirichenko A, Savchenko A, Ruksha T. miR-204-5p in vivo inhibition cause diminished CD45RO cells rate in lungs of melanoma B16-bearing mice. Noncoding RNA Res 2022; 7:133-141. [PMID: 35756165 PMCID: PMC9188961 DOI: 10.1016/j.ncrna.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 10/25/2022] Open
|
11
|
Xie H, Li W, Liu H, Chen Y, Ma M, Wang Y, Luo Y, Song D, Hou Q, Lu W, Bai Y, Li B, Ma J, Huang C, Yang T, Liu Z, Zhao X, Ding P. Erythrocyte Membrane-Coated Invisible Acoustic-Sensitive Nanoparticle for Inducing Tumor Thrombotic Infarction by Precisely Damaging Tumor Vascular Endothelium. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201933. [PMID: 35789094 DOI: 10.1002/smll.202201933] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/16/2022] [Indexed: 06/15/2023]
Abstract
Selective induction of tumor thrombus infarction is a promising antitumor strategy. Non-persistent embolism due to non-compacted thrombus and activated fibrinolytic system within the tumor large blood vessels and tumor margin recurrence are the main therapeutic bottlenecks. Herein, an erythrocyte membrane-coated invisible acoustic-sensitive nanoparticle (TXA+DOX/PFH/RBCM@cRGD) is described, which can induce tumor thrombus infarction by precisely damaging tumor vascular endothelium. It is revealed that TXA+DOX/PFH/RBCM@cRGD can effectively accumulate on the endothelial surface of tumor vessels with the help of the red blood cell membrane (RBCM) stealth coating and RGD cyclic peptide (cRGD), which can be delivered in a targeted manner as nanoparticle missiles. As a kind of phase-change material, perfluorohexane (PFH) nanodroplets possess excellent acoustic responsiveness. Acoustic-sensitive missiles can undergo an acoustic phase transition and intense cavitation with response to low-intensity focused ultrasound (LIFU), damaging the tumor vascular endothelium, rapidly initiating the coagulation cascade, and forming thromboembolism in the tumor vessels. The drugs loaded in the inner water phase are released explosively. Tranexamic acid (TXA) inhibits the fibrinolytic system, and doxorubicin (DOX) eliminates the margin survival. In summary, a stealthy and acoustically responsive multifunctional nanoparticle delivery platform is successfully developed for inducing thrombus infarction by precisely damaging tumor vascular endothelium.
Collapse
Affiliation(s)
- Huichao Xie
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wan Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hui Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yongfeng Chen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Mengrui Ma
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yichen Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yucen Luo
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Di Song
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qianqian Hou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wenwen Lu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yu Bai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Bao Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jizhuang Ma
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chi Huang
- Ultrasound Department of Shengjing Hospital, China Medical University, Shenyang, 110016, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, 04401, USA
| | - Zhining Liu
- Ultrasound Department, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Xiaoyun Zhao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| |
Collapse
|
12
|
Liu Q, Zhao E, Geng B, Gao S, Yu H, He X, Li X, Dong G, You B. Tumor-associated macrophage-derived exosomes transmitting miR-193a-5p promote the progression of renal cell carcinoma via TIMP2-dependent vasculogenic mimicry. Cell Death Dis 2022; 13:382. [PMID: 35443741 PMCID: PMC9021253 DOI: 10.1038/s41419-022-04814-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/23/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022]
Abstract
Previous studies have investigated whether tumor-associated macrophages (TAMs) play tumorigenic and immunosuppressive roles to encourage cancer development, but the role of TAMs in regulating vasculogenic mimicry (VM) in clear-cell renal cell carcinoma (ccRCC) cells has not been completely clarified. We conducted immunostaining of the tumor-associated macrophage biomarkers CD68/CD163 and double staining for PAS/CD31 in ccRCC human specimens to find that higher TAM infiltration was positively correlated with VM formation. Then we demonstrated that TAM-derived exosomes downregulate TIMP2 expression in RCC cells to promote VM and invasion by shuttling miR-193a-5p. Mechanistic analysis indicated that HIF-1α upregulation in macrophages could transcriptionally increase miR-193a-5p expression. Exosome-shuttled miR-193a-5p then targeted the 3′ untranslated region (UTR) of TIMP2 mRNA to suppress its translation. A preclinical study using an in vivo orthotopic xenograft model of ccRCC in mice substantiated that TAM-derived exosomes enhance VM and enable tumor progression, which confirmed our in vitro data. Suppressing TAM-derived exosomal miR-193a-5p successfully inhibited tumor progression and metastasis. Overall, miR-193a-5p from TAM-derived exosomes downregulates the TIMP2 gene to facilitate the development of RCC, which provides a novel perspective for developing therapeutic strategies for RCC.
Collapse
Affiliation(s)
- Qing Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China.,Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Enyang Zhao
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Bo Geng
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Shan Gao
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Hongyang Yu
- Department of Radiation Oncology, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Xinyang He
- Department of Radiation Oncology, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Xuedong Li
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China.
| | - Guanglu Dong
- Department of Radiation Oncology, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China.
| | - Bosen You
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China. .,Department of Urology, The Second Affiliated Hospital of Harbin Medical University, 150001, Harbin, China.
| |
Collapse
|
13
|
Li F, Shi Y, Zhang Y, Yang X, Wang Y, Jiang K, Hua C, Wu C, Sun C, Qin Y, Liu S. Investigating the mechanism of Xian-ling-lian-xia-fang for inhibiting vasculogenic mimicry in triple negative breast cancer via blocking VEGF/MMPs pathway. Chin Med 2022; 17:44. [PMID: 35379271 PMCID: PMC8981688 DOI: 10.1186/s13020-022-00597-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/20/2022] [Indexed: 11/12/2022] Open
Abstract
Background Xian-ling-lian-xia-fang (XLLXF), a Chinese medicine decoction, is widely used in the treatment of triple negative breast cancer (TNBC). However, the underlying mechanism of XLLXF in TNBC treatment has not been totally elucidated. Methods Here, network pharmacology and molecular docking were used to explore the mechanism of Traditional Chinese medicine in the treatment of TNBC. Then, biological experiments were integrated to verify the results of network pharmacology. Results Network pharmacology showed that the candidate active ingredients mainly included quercetin, kaempferol, stigmasterol, and β-sitosterol through the “XLLXF–active ingredients–targets” network. Vascular endothelial growth factor A (VEGFA) and matrix metalloproteinase (MMP) 2 were the potential therapeutic targets obtained through the protein–protein interaction (PPI) network. Molecular docking confirmed that quercetin, kaempferol, stigmasterol, and β-sitosterol could stably combine with VEGFA and MMP2. Experimental verification showed that XLLXF could inhibit proliferation, colony ability, and vasculogenic mimicry (VM) formation and promote cell apoptosis in TNBC. Laser confocal microscopy found that XLLXF impaired F-actin cytoskeleton organization and inhibited epithelial mesenchymal transition. Animal experiments also found that XLLXF could inhibit tumor growth and VM formation in TNBC xenograft model. Western blot analysis and immunohistochemical staining showed that XLLXF inhibited the protein expression of VEGFA, MMP2, MMP9, Vimentin, VE-cadherin, and Twist1 and increased that of E-cadherin, tissue inhibitors of metalloproteinase (TIMP)-1, and TIMP-3 in vitro and in vivo. Conclusions Integrating the analysis of network pharmacology and experimental validation revealed that XLLXF could inhibit VM formation via downregulating the VEGF/MMPs signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s13020-022-00597-5.
Collapse
Affiliation(s)
- Feifei Li
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Youyang Shi
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Yang Zhang
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Xiaojuan Yang
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Yi Wang
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Kexin Jiang
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Ciyi Hua
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Chunyu Wu
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Chenping Sun
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Yuenong Qin
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China.
| | - Sheng Liu
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China.
| |
Collapse
|
14
|
He M, Yang H, Shi H, Hu Y, Chang C, Liu S, Yeh S. Sunitinib increases the cancer stem cells and vasculogenic mimicry formation via modulating the lncRNA-ECVSR/ERβ/Hif2-α signaling. Cancer Lett 2022; 524:15-28. [PMID: 34461182 DOI: 10.1016/j.canlet.2021.08.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022]
Abstract
Sunitinib is the first-line drug for treating renal cell carcinoma (RCC), and it functions mainly through inhibition of tumor angiogenesis. However, the patients may become insensitive or develop resistance toward sunitinib treatment, but the underlying mechanisms have not yet been fully elucidated. Herein, it was found that sunitinib could have adverse effects of promoting RCC progression by increasing vascular mimicry (VM) formation of RCC cells. Mechanism dissection revealed that sunitinib can increase the expression of a long non-coding RNA (lncRNA), lncRNA-ECVSR, thereby enhancing the stability of estrogen receptor β (ERβ) mRNA. Subsequently, the increased ERβ expression can then function via transcriptional up-regulation of Hif2-α. Notably, sunitinib-increased lncRNA-ECVSR/ERβ/Hif2-α signaling resulted in an increased cancer stem cell (CSC) phenotype, thereby promoting VM formation. Furthermore, the sunitinib/lncRNA-ECVSR-increased ERβ expression can transcriptionally regulate lncRNA-ECVSR expression via a positive-feedback loop. Supportively, preclinical studies using RCC mouse xenografts demonstrated that combining sunitinib with the small molecule anti-estrogen PHTPP can increase sunitinib efficacy with reduced VM formation. Collectively, the findings of this study may aid in the development of potential biomarker(s) and novel therapies to better monitor and suppress RCC progression.
Collapse
Affiliation(s)
- Miao He
- Departments of Urology, Pathology and the Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA; Department of Urology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huan Yang
- Department of Urology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hangchuan Shi
- Departments of Urology, Pathology and the Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Yixi Hu
- Departments of Urology, Pathology and the Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Chawnshang Chang
- Departments of Urology, Pathology and the Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA; Sex Hormone Research Center, China Medical University and Hospital, Taichung, 404, Taiwan
| | - Shunfang Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuyuan Yeh
- Departments of Urology, Pathology and the Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
15
|
Aspriţoiu VM, Stoica I, Bleotu C, Diaconu CC. Epigenetic Regulation of Angiogenesis in Development and Tumors Progression: Potential Implications for Cancer Treatment. Front Cell Dev Biol 2021; 9:689962. [PMID: 34552922 PMCID: PMC8451900 DOI: 10.3389/fcell.2021.689962] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is a multi-stage process of new blood vessel development from pre-existing vessels toward an angiogenic stimulus. The process is essential for tissue maintenance and homeostasis during embryonic development and adult life as well as tumor growth. Under normal conditions, angiogenesis is involved in physiological processes, such as wound healing, cyclic regeneration of the endometrium, placental development and repairing certain cardiac damage, in pathological conditions, it is frequently associated with cancer development and metastasis. The control mechanisms of angiogenesis in carcinogenesis are tightly regulated at the genetic and epigenetic level. While genetic alterations are the critical part of gene silencing in cancer cells, epigenetic dysregulation can lead to repression of tumor suppressor genes or oncogene activation, becoming an important event in early development and the late stages of tumor development, as well. The global alteration of the epigenetic spectrum, which includes DNA methylation, histone modification, chromatin remodeling, microRNAs, and other chromatin components, is considered one of the hallmarks of cancer, and the efforts are concentrated on the discovery of molecular epigenetic markers that identify cancerous precursor lesions or early stage cancer. This review aims to highlight recent findings on the genetic and epigenetic changes that can occur in physiological and pathological angiogenesis and analyze current knowledge on how deregulation of epigenetic modifiers contributes to tumorigenesis and tumor maintenance. Also, we will evaluate the clinical relevance of epigenetic markers of angiogenesis and the potential use of "epi-drugs" in modulating the responsiveness of cancer cells to anticancer therapy through chemotherapy, radiotherapy, immunotherapy and hormone therapy as anti-angiogenic strategies in cancer.
Collapse
Affiliation(s)
| | - Ileana Stoica
- Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Coralia Bleotu
- Faculty of Biology, University of Bucharest, Bucharest, Romania.,Romanian Academy, Stefan S. Nicolau Institute of Virology, Bucharest, Romania
| | | |
Collapse
|
16
|
Geng B, Zhu Y, Yuan Y, Bai J, Dou Z, Sui A, Luo W. Artesunate Suppresses Choroidal Melanoma Vasculogenic Mimicry Formation and Angiogenesis via the Wnt/CaMKII Signaling Axis. Front Oncol 2021; 11:714646. [PMID: 34476217 PMCID: PMC8406848 DOI: 10.3389/fonc.2021.714646] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/26/2021] [Indexed: 01/14/2023] Open
Abstract
Angiogenesis and vasculogenic mimicry (VM) are considered to be the main processes to ensure tumor blood supply during the proliferation and metastasis of choroidal melanoma (CM). The traditional antimalarial drug artesunate (ART) has some potential anti-CM effects; however, the underlying mechanisms remain unclarified. Recent studies have shown that the Wnt5a/calmodulin-dependent kinase II (CaMKII) signaling pathway has a close correlation with angiogenesis and VM formation. This study demonstrated that ART eliminated VM formation by inhibiting the aforementioned signaling pathway in CM cells. The microvessel sprouting of the mouse aortic rings and the microvessel density of chicken chorioallantoic membrane (CAM) decreased significantly after ART treatment. VM formation assay and periodic acid schiff (PAS) staining revealed that ART inhibited VM formation in CM. Moreover, ART downregulated the expression levels of the angiogenesis-related proteins vascular endothelial growth factor receptor (VEGFR) 2, platelet-derived growth factor receptor (PDGFR) and vascular endothelial growth factor (VEGF) A, and VM-related proteins ephrin type-A receptor (EphA) 2 and vascular endothelial (VE)-cadherin. The expression of hypoxia-inducible factor (HIF)-1α, Wnt5a, and phosphorylated CaMKII was also downregulated after ART treatment. In addition, we further demonstrated that ART inhibited the proliferation, migration, and invasion of OCM-1 and C918 cells. Collectively, our results suggested that ART inhibited angiogenesis and VM formation of choroidal melanoma likely by regulating the Wnt5a/CaMKII signaling pathway. These findings further supported the feasibility of ART for cancer therapy.
Collapse
Affiliation(s)
- Bochao Geng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuanzhang Zhu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingying Yuan
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jingyi Bai
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhizhi Dou
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aihua Sui
- Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenjuan Luo
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
17
|
Hasegawa T, Sugihara T, Hoshino Y, Tarumoto R, Matsuki Y, Kanda T, Takata T, Nagahara T, Matono T, Isomoto H. Photosensitizer verteporfin inhibits the growth of YAP- and TAZ-dominant gastric cancer cells by suppressing the anti-apoptotic protein Survivin in a light-independent manner. Oncol Lett 2021; 22:703. [PMID: 34457058 PMCID: PMC8358589 DOI: 10.3892/ol.2021.12964] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/07/2021] [Indexed: 12/11/2022] Open
Abstract
Yes-associated protein (YAP) positivity indicates a poor prognosis in gastric cancer. Transcriptional co-activator with a PDZ-binding domain (TAZ), a YAP paralog, is highly expressed in gastric signet ring cell carcinoma. Verteporfin (VP), a clinical photosensitizer, was recently shown to inhibit YAP/TAZ. In the present study, the therapeutic potential of VP treatment was explored using two gastric cancer cell lines: MKN-45 (TAZ-dominant) and MKN-74 (YAP-dominant). Cell proliferation was evaluated by MTS assay. Vascular mimicry was evaluated by the tube formation assay. Gene and protein expression levels of YAP/TAZ downstream effectors [such as Survivin, Cysteine-rich angiogenic inducer 61 (CYR61), and connective tissue growth factor (CTGF)] were measured. YAP or TAZ localization was evaluated by immunofluorescence. Cell death was assessed by immunofluorescent staining of Annexin V. YAP and TAZ expression were knocked down by small interfering RNA. The current results demonstrate that MKN-45, a poorly differentiated TAZ-dominant gastric cancer cell line, was more sensitive to VP than MKN-74, a moderately differentiated YAP-dominant gastric cancer cell line. VP changed the localization of YAP/TAZ, promoted its degradation and significantly decreased the protein level of Survivin in both cell lines. Cell death was induced by VP treatment in a dose-dependent manner. Vascular mimicry was inhibited in both cell lines. Proliferation in both cell lines decreased in response to YAP/TAZ knockdown. The present study indicated that VP has potential as a therapeutic agent in YAP- and TAZ-dominant gastric cancers due to its ability to suppress the anti-apoptotic protein Survivin via inhibition of YAP and TAZ.
Collapse
Affiliation(s)
- Takashi Hasegawa
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Takaaki Sugihara
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Yoshiki Hoshino
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Ryohei Tarumoto
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Yukako Matsuki
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Tsutomu Kanda
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Tomoaki Takata
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Takakazu Nagahara
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Tomomitsu Matono
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Hajime Isomoto
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| |
Collapse
|
18
|
Zhu N, Xu X, Wang Y, Zeng MS, Yuan Y. EBV latent membrane proteins promote hybrid epithelial-mesenchymal and extreme mesenchymal states of nasopharyngeal carcinoma cells for tumorigenicity. PLoS Pathog 2021; 17:e1009873. [PMID: 34407150 PMCID: PMC8405006 DOI: 10.1371/journal.ppat.1009873] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 08/30/2021] [Accepted: 08/07/2021] [Indexed: 12/30/2022] Open
Abstract
EBV-encoded LMPs are consistently detected in nasopharyngeal carcinoma (NPC). Recent evidence suggests potential roles of LMP1 and LMP2A in Epithelial-to-mesenchymal transition (EMT) process in NPC. EMT engages in the generation and maintenance of cancer stem cells (CSCs) and confers on cancer cells increased tumor-initiating and metastatic potential, and higher resistance to anticancer therapies. However, how LMP1 and LMP2A regulate the EMT process to generate cells with different EMT states and its implications for tumor progression remain unclear. Here we report that LMP1 and LMP2A promote EMT that drives NPC cells from the epithelial-like state (E) (CD104+, CD44low) to epithelial-mesenchymal hybrid (E/M) state (CD104+, CD44high). Furthermore, LMP2A possesses an additional function in stabilizing LMP1 and increasing the level of LMP1 in NPC cells. The elevated LMP1 further forces the EMT to generate extreme-mesenchymal (xM) state cells (CD104-, CD44high). To define the tumorigenic features of cancer stem cells at different states in the EMT spectrum, E, E/M and xM subpopulations were isolated and tested for tumorigenic capability in a tumor xenograft animal model. We found that the cells with E/M phenotypes possess the highest tumor initiating capacity. However, the xM subpopulation exhibits increased vasculogenic mimicry, a hallmark of metastatic cancers. Taken together, coordinated action of LMP1 and LMP2A generates an array of intermediate subpopulations in the EMT spectrum that are responsible for distinct tumorigenic features of NPC such as tumor-initiation, vasculogenesis, and metastasis. Intratumoral heterogeneity, characterized by the existence of distinct cellular populations within tumor lesions, poses a significant challenge for the treatment of high-grade cancers. Using an EBV-associated nasopharyngeal carcinoma (NPC) model, we sought to elucidate how a virus or its oncoproteins promote the establishment of cancer stem cells that comprises heterogeneous subpopulations. We found that the coordinated action of EBV membrane proteins LMP1 and LMP2A generates heterogeneous subpopulations of cancer stem cells in nasopharyngeal carcinoma by activating the epithelial-to-mesenchymal transition (EMT). Furthermore, the contributions of the different subpopulations to NPC oncogenesis were investigated. Results showed that cells with an epithelial/mesenchymal hybrid state (E/M) possess the highest tumor initiating capacity; and a highly mesenchymal state (xM) subpopulation exhibits increased vasculogenic mimicry. These finding suggest that cancer stem cells residing at various EMT states are responsible for distinct tumorigenic features such as tumor-initiation, vasculogenesis, and metastasis.
Collapse
Affiliation(s)
- Nannan Zhu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoting Xu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yan Wang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Yuan
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Basic and Translational Sciences, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
19
|
Yu P, Zhu X, Zhu JL, Han YB, Zhang H, Zhou X, Yang L, Xia YZ, Zhang C, Kong LY. The Chk2-PKM2 axis promotes metabolic control of vasculogenic mimicry formation in p53-mutated triple-negative breast cancer. Oncogene 2021; 40:5262-5274. [PMID: 34244606 DOI: 10.1038/s41388-021-01933-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/15/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023]
Abstract
Vasculogenic mimicry (VM) formation, which participates in the process of neovascularization, is highly activated in p53-mutated triple-negative breast cancer (TNBC). Here, we show that Chk2 is negatively correlated with VM formation in p53-mutated TNBC. Its activation by DNA-damaging agents such as cisplatin, etoposide, and DPT reduces VM formation. Mechanistically, the Chk2-PKM2 axis plays an important role in the inhibition of VM formation at the level of metabolic regulation. Chk2 promotes the Chk2-PKM2 interaction through the Chk2 SCD (SQ/TQ cluster domain) and the PKM2 C domain. Furthermore, Chk2 promotes the nuclear export of PKM2 by phosphorylating PKM2 at Ser100. P-PKM2 S100 reduces VM formation by decreasing glucose flux, and the PKM2 S100A mutation abolishes the inhibition of glucose flux and VM formation induced by Chk2 activation. Overall, this study proposes a novel strategy of VM suppression through Chk2 induction, which prevents PKM2-mediated glucose flux in p53-mutated TNBC.
Collapse
Affiliation(s)
- Pei Yu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiong Zhu
- Medical and Chemical Institute, China Pharmaceutical University, Nanjing, China
| | - Jia-Le Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yu-Bao Han
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiang Zhou
- Department of Science, China Pharmaceutical University, Nanjing, China
| | - Lei Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuan-Zheng Xia
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
20
|
Wu M, Sun X, Wang T, Zhang M, Li P. TRPS1 knockdown inhibits angiogenic vascular mimicry in human triple negative breast cancer cells. Clin Transl Oncol 2021; 24:145-153. [PMID: 34216368 DOI: 10.1007/s12094-021-02676-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/22/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE Vascular mimicry (VM) tubules are lumen structures comprised of malignant tumor cells without the participation of endothelial cells. VM simulates blood vessel function in tumors to deliver a sufficient blood supply for proliferation, invasion, and metastasis of malignant tumors, thereby reducing the clinical effects of anti-angiogenic treatments. The elimination or prevention of malignant tumor VM development therefore represents an urgent research goal as a therapeutic strategy to and cut off nutrients required for tumor growth. The GATA transcription factor TRPS1 is abnormally up-regulated in breast cancer, osteosarcoma, prostate cancer, and other tumor tissues, and is instrumental in regulating cell proliferation, differentiation, and tissue growth and development. METHODS Here, we explored the effects of TRPS1 knockdown on VM and the proteins underlying its development in triple-negative breast cancer cell line MDA-MB-231. RESULTS We found that TRPS1 knockdown resulted in obvious inhibition of VM development. Fluorescence microscopy of F-actin and tubulin revealed that loss of TRPS1 function resulted in disruption of cytoskeleton and microtubule formation, respectively. In addition, TRPS1-suppressed cells exhibited reduced accumulation of VM-associated proteins EphA2, MMP-2, MMP-9, VEGF, and VE-cadherin. Moreover, it is interesting to know that the capacity for migration and invasion were limited in MDA-MB-231cells after TRPS1 knockdown and that the average number of VM tubules, their length, and number of intersections were also significantly decreased. CONCLUSIONS Based on our results, and in light of previous studies, we thus proposed that TRPS1 suppression negatively affects vascular mimicry possibly through reduced TRPS1-mediated transcriptional regulation of VM-related protein VEGF-A.
Collapse
Affiliation(s)
- M Wu
- Chinese Integrative Medicine Oncology Department, The First Affiliated Hospital of Anhui Medical University, No. 120 Wanshui Road, Shushan District, Hefei, 230022, Anhui Province, China
| | - X Sun
- Chinese Integrative Medicine Oncology Department, The First Affiliated Hospital of Anhui Medical University, No. 120 Wanshui Road, Shushan District, Hefei, 230022, Anhui Province, China
| | - T Wang
- Chinese Integrative Medicine Oncology Department, The First Affiliated Hospital of Anhui Medical University, No. 120 Wanshui Road, Shushan District, Hefei, 230022, Anhui Province, China
| | - M Zhang
- Chinese Integrative Medicine Oncology Department, The First Affiliated Hospital of Anhui Medical University, No. 120 Wanshui Road, Shushan District, Hefei, 230022, Anhui Province, China
| | - P Li
- Chinese Integrative Medicine Oncology Department, The First Affiliated Hospital of Anhui Medical University, No. 120 Wanshui Road, Shushan District, Hefei, 230022, Anhui Province, China.
| |
Collapse
|
21
|
Yue Y, Lou Y, Liu X, Peng X. Vasculogenic mimicry in head and neck tumors: a narrative review. Transl Cancer Res 2021; 10:3044-3052. [PMID: 35116612 PMCID: PMC8798303 DOI: 10.21037/tcr-21-34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/10/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To discuss the role and mechanism of vasculogenic mimicry (VM) and to provide reference for the further research of VM in head and neck tumors. BACKGROUND Head and neck tumors are common in the clinic, and tumor metastasis is clinically difficult to treat. VM is another tumor blood supply mode that is different from angiogenesis and plays an important role in tumor growth, metastasis, and invasion. At present, studies on VM have mainly focused on breast cancer, melanoma, glioblastoma, and other cancers. With time, VM has become a hotspot in head and neck tumor research. METHODS We searched published English literatures from 2015 to 2020 on PubMed. In this paper, we review the progress of VM in head and neck tumors from 7 different perspectives. VM has two distinct types, namely tubular type and patterned matrix type. VM is associated with high tumor grade, tumor progression, invasion, metastasis, and poor prognosis in patients with head and neck tumors. We discuss the recent studies on the effects of immune cells and Epstein-Barr virus on VM in head and neck tumors. Furthermore, we also summarize the molecular mechanism of VM formation in head and neck tumors. Finally, we discussed the possibility of VM-targeted therapy in the clinical treatment of head and neck tumors. CONCLUSIONS VM plays a critical role in tumor invasion, metastasis, and poor prognosis in patients with head and neck tumors. There is potential for VM as a potential new antitumor target. VM has become a hotspot in head and neck tumor research.
Collapse
Affiliation(s)
- Yuan Yue
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yunfan Lou
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiong Liu
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaohong Peng
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Hong D, Yang J, Guo J, Zhang Y, Chen Z. Ultrasound-Targeted Microbubble Destruction Enhances Inhibitory Effect of Apatinib on Angiogenesis in Triple Negative Breast Carcinoma Xenografts. Anal Cell Pathol (Amst) 2021; 2021:8837950. [PMID: 33959473 PMCID: PMC8075700 DOI: 10.1155/2021/8837950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/19/2021] [Accepted: 04/07/2021] [Indexed: 12/31/2022] Open
Abstract
Ultrasound-targeted microbubble destruction (UTMD) has been proven as an effective technique to assist drugs to cross the vascular wall and cell membrane. This study was aimed at evaluating the synergistic antiangiogenic and growth-inhibiting effects of apatinib (APA) and UTMD on the triple negative breast cancer (TNBC). The TNBC xenograft model was established in nude mice (n = 40) which were then randomly divided into the APA plus UTMD (APA-U) group, UTMD group, APA group, and model control (M) group (n = 10 per group). Corresponding treatment was done once daily for 14 consecutive days. The general condition and body weight of tumor-bearing nude mice were monitored. Routine blood test and detection of liver and kidney function were done after treatments. The tumor size and microcirculation were examined by two-dimensional ultrasonography (2DUS) and contrast-enhanced ultrasonography (CEUS), respectively. Then, the tumor tissues were harvested for the detection of vascular endothelial growth factor (VEGF) by immunohistochemistry and for CD31-PAS double staining to assess microvessel density (MVD) and heterogeneous vascular positivity rate. After treatments, the tumor growth and angiogenesis were significantly inhibited in the APA group and the APA-U group, and these effects were more obvious in the APA-U group. The tumor volume, CEUS parameters, VEGF expression, and MVD in the APA-U group were significantly lower than those in the APA group (P < 0.05), while there were no marked differences in the heterogeneous vascular positivity rate, body weight, and blood parameters between the two groups (P > 0.05). In the UTMD group, the tumor growth and angiogenesis were not significantly inhibited, and all the parameters were similar to those in the M group (P > 0.05). During the experiment, all mice survived and generally had good condition. In conclusion, APA combined with UTMD may exert synergistic antiangiogenic and growth-inhibiting effects on the TNBC and not increase the heterogeneous vasculature and the severity of APA-related systemic side effects.
Collapse
Affiliation(s)
- Dengke Hong
- Department of Vascular Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jiajia Yang
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jingjing Guo
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yu Zhang
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Zhikui Chen
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
23
|
Hong KO, Oh KY, Yoon HJ, Swarup N, Jung M, Shin JA, Kim JH, Chawla K, Lee JI, Cho SD, Hong SD. SOX7 blocks vasculogenic mimicry in oral squamous cell carcinoma. J Oral Pathol Med 2021; 50:766-775. [PMID: 33733517 DOI: 10.1111/jop.13176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/16/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Vasculogenic mimicry (VM) is the formation of an alternative circulatory system by aggressive tumor cells. The characteristics of VM and its underlying mechanism in oral squamous cell carcinoma (OSCC) remain unclear. This study aims to determine the relationship between VM in OSCC tissues and clinical outcomes and to investigate the biological role of SOX7 in VM in OSCC cells. METHODS CD31/PAS staining was performed to evaluate VM in OSCC tissue. The relationships between VM and clinicopathological variables, and VM and SOX7 levels were analyzed. The correlation between SOX7 levels and cancer cohorts was investigated using in silico analysis. VM formation assay was performed to observe VM in vitro. To investigate the role of SOX7 in VM formation, SOX7 was transiently over-expressed in SCC-9 cells. VM-modulating genes were identified by Western blotting. RESULTS We found a positive correlation between VM and lymph node metastasis and patient survival in OSCC (p = 0.003). In silico analysis from The Cancer Genome Atlas and Gene Expression Omnibus database showed that down-regulation of SOX7 expression was significantly correlated with OSCC patients (p = 0.0187) and lymph node metastasis (p = 0.0017). We also found that the presence of VM in OSCC tissue was inversely associated with SOX7 expression (p = 0.020). We observed that overexpression of SOX7 impaired VM formation by reducing the expression of VE-cadherin, thereby inhibiting cell migration and invasion. CONCLUSION These results suggest that SOX7 plays an important role in the regulation of VM formation and may inhibit OSCC metastasis.
Collapse
Affiliation(s)
- Kyoung-Ok Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Kyu-Young Oh
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Hye-Jung Yoon
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Neeti Swarup
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Minjung Jung
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Jung-Hwan Kim
- Department of Pharmacology, College of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju, Korea
| | - Kunal Chawla
- Department of Computer Science, School of Interactive Computing, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jae-Il Lee
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Seong-Doo Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
24
|
Zheng N, Zhang S, Wu W, Zhang N, Wang J. Regulatory mechanisms and therapeutic targeting of vasculogenic mimicry in hepatocellular carcinoma. Pharmacol Res 2021; 166:105507. [PMID: 33610718 DOI: 10.1016/j.phrs.2021.105507] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is a typical hyper-vascular solid tumor; aberrantly rich in tumor vascular network contributes to its malignancy. Conventional anti-angiogenic therapies seem promising but transitory and incomplete efficacy on HCC. Vasculogenic mimicry (VM) is one of functional microcirculation patterns independent of endothelial vessels which describes the plasticity of highly aggressive tumor cells to form vasculogenic-like networks providing sufficient blood supply for tumor growth and metastasis. As a pivotal alternative mechanism for tumor vascularization when tumor cells undergo lack of oxygen and nutrients, VM has an association with the malignant phenotype and poor clinical outcome for HCC, and may challenge the classic anti-angiogenic treatment of HCC. Current studies have contributed numerous findings illustrating the underlying molecular mechanisms and signaling pathways supporting VM in HCC. In this review, we summarize the correlation between epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs) and VM, the role of hypoxia and extracellular matrix remodeling in VM, the involvement of adjacent non-cancerous cells, cytokines and growth factors in VM, as well as the regulatory influence of non-coding RNAs on VM in HCC. Moreover, we discuss the clinical significance of VM in practice and the potential therapeutic strategies targeting VM for HCC. A better understanding of the mechanism underlying VM formation in HCC may optimize anti-angiogenic treatment modalities for HCC.
Collapse
Affiliation(s)
- Ning Zheng
- Department of Pharmacology, The School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Shaoqin Zhang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Wenda Wu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Nan Zhang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Jichuang Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China.
| |
Collapse
|
25
|
Zhao J, Wu J, Qin Y, Zhang W, Huang G, Qin L. LncRNA PVT1 induces aggressive vasculogenic mimicry formation through activating the STAT3/Slug axis and epithelial-to-mesenchymal transition in gastric cancer. Cell Oncol (Dordr) 2020; 43:863-876. [PMID: 32729021 DOI: 10.1007/s13402-020-00532-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2020] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Vasculogenic mimicry (VM), a vessel-like network formed by highly aggressive tumor cells, plays an important role in accelerating cancer progression. This special vascularization pattern is closely associated with a poor prognosis in various cancers. As yet, however, the regulatory mechanism of VM formation is largely unknown. In this study, we assess whether the long noncoding RNA PVT1 is involved in VM generation in gastric cancer. METHODS VM formation was determined by immunohistochemistry using PAS/CD31 double staining in gastric cancers and Matrigel tube formation in vitro. qRT-PCR and Western blotting were used to assess mRNA and protein expression. Interaction between PVT1 and STAT3 was determined using a RNA pull-down assay. Luciferase reporter and chromatin immunoprecipitation assays were performed to evaluate transcriptional activity of STAT3 on the Slug gene promoter. RESULTS We found that PVT1 can induce VM generation both in vitro and in vivo. Mechanistically, we found that PVT1 interacted with and activated STAT3 through a 850-1770 nt fragment. PVT1 facilitated STAT3 recruitment to the Slug promoter and transcriptionally enhanced Slug expression, thereby triggering epithelial-to-mesenchymal transition (EMT) and VM capillary formation. STAT3 inhibition effectively blocked PVT1-mediated VM. In primary gastric cancer samples, a positive correlation was found between PVT1 and Slug upregulation, and patients with a high PVT1 and Slug expression exhibited markedly shorter survival times. CONCLUSION Our results shed light on the role of PVT1 in gastric cancer cell-dependent VM formation. Our findings provide valuable clues for the design of new anti-angiogenic therapeutic strategies. The PVT1/STAT3 axis may serve as a potential target in gastric cancer treatment.
Collapse
Affiliation(s)
- Jing Zhao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China.,Cancer Metastasis Institute, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jing Wu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Yunyun Qin
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Wenhong Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China.
| | - Guangjian Huang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China.
| | - Lunxiu Qin
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China. .,Cancer Metastasis Institute, Fudan University, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
26
|
Lim D, Do Y, Kwon BS, Chang W, Lee MS, Kim J, Cho JG. Angiogenesis and vasculogenic mimicry as therapeutic targets in ovarian cancer. BMB Rep 2020. [PMID: 32438972 PMCID: PMC7330806 DOI: 10.5483/bmbrep.2020.53.6.060] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tumor angiogenesis is an essential process for growth and metastasis of cancer cells as it supplies tumors with oxygen and nutrients. During tumor angiogenesis, many pro-angiogenic factors are secreted by tumor cells to induce their own vascularization via activation of pre-existing host endothelium. However, accumulating evidence suggests that vasculogenic mimicry (VM) is a key alternative mechanism for tumor vascularization when tumors are faced with insufficient supply of oxygen and nutrients. VM is a tumor vascularization mechanism in which tumors create a blood supply system, in contrast to tumor angiogenesis mechanisms that depend on pre-existing host endothelium. VM is closely associated with tumor progression and poor prognosis in many cancers. Therefore, inhibition of VM may be a promising therapeutic strategy and may overcome the limitations of anti-angiogenesis therapy for cancer patients. In this review, we provide an overview of the current anti-angiogenic therapies for ovarian cancer and the current state of knowledge regarding the links between microRNAs and the VM process, with a focus on the mechanism that regulates associated signaling pathways in ovarian cancer. Moreover, we discuss the potential for VM as a therapeutic strategy against ovarian cancer.
Collapse
Affiliation(s)
- Dansaem Lim
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Yeojin Do
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Byung Su Kwon
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Korea
| | - Myeong-Sok Lee
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Jongmin Kim
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Jin Gu Cho
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| |
Collapse
|
27
|
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: 32] [Impact Index Per Article: 8.0] [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
|
28
|
Regulation of cancer stem cell properties, angiogenesis, and vasculogenic mimicry by miR-450a-5p/SOX2 axis in colorectal cancer. Cell Death Dis 2020; 11:173. [PMID: 32144236 PMCID: PMC7060320 DOI: 10.1038/s41419-020-2361-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 11/30/2022]
Abstract
Growing evidence indicates that a small number of cancer cells express stem cell markers and possess stem cell-like properties that promote malignant progression. Sex-determining region Y-box2 (SOX2) is a stem cell transcription factor essential for maintaining the properties of cancer stem cell (CSC). As CSC properties have been associated with angiogenesis and vasculogenic mimicry (VM), we aimed to comprehensively investigate whether SOX2 regulates CSC properties, angiogenesis, and VM in colorectal carcinoma (CRC) and its potential mechanism in this study. For this study, sphere formation assay, flow cytometry, cell survival analysis, tube formation, 3D culture, immunoblot, mouse model, and luciferase reporter assay were performed in vivo and in vitro. Expressions of SOX2 and miR-450a-5p in CRC tissue samples were examined through immunohistochemistry. First, the expression of SOX2 was not only associated with poor differentiation and prognosis but also promoted angiogenesis and VM. Knockdown of SOX2 ceased stemness properties, angiogenesis, and VM, along with decreased expression of CD133, CD31, and VE-cadherin as observed in functional experiments. Downregulation of SOX2 was found to inhibit tumorigenesis in vivo. Second, miR-450a-5p suppressed the expression of SOX2 by targeting its 3’UTR region directly and hence restrained SOX2-induced CSC properties, angiogenesis, and VM. Moreover, SOX2 overexpression preserved the miR-450a-5p-induced inhibition of CRC properties, angiogenesis, and VM. Finally, clinical samples exhibited a negative correlation between miR-450a-5p and SOX2. Patients with higher SOX2 and lower miR-450a-5p expressions had a poorer prognosis than patients with inverse expressions. Conclusively, we elucidated a unique mechanism of miR-450a-5p-SOX2 axis in the regulation of stemness, angiogenesis, and VM, which may act as a potential therapeutic practice in CRC.
Collapse
|
29
|
Vartanian AA, Khochenkov DA, Khochenkova YA, Machkova YS, Khachatryan DS, Kolotaev AV, Balaev AN, Ohmanovich KA, Osipov VN. Effect of Derivatives of Hydroxamic Acids on Vasculogenic Mimicry. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2020. [DOI: 10.1134/s106816202002017x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Li X, Sun B, Zhao X, An J, Zhang Y, Gu Q, Zhao N, Wang Y, Liu F. Function of BMP4 in the Formation of Vasculogenic Mimicry in Hepatocellular Carcinoma. J Cancer 2020; 11:2560-2571. [PMID: 32201526 PMCID: PMC7066000 DOI: 10.7150/jca.40558] [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: 09/23/2019] [Accepted: 01/21/2020] [Indexed: 01/18/2023] Open
Abstract
Vasculogenic mimicry (VM) is linked to vascular invasion of human hepatocellular carcinoma (HCC). BMP4, one BMP family member, is upregulated in several cancers. The purpose of this report is to identify the function of BMP4 in the formation of VM in HCC and the mechanism underling this regulation. In our report, BMP4 up-regulation resulted in an increase in migration, invasion and channel-like structure formation as well as induced epithelial-mesenchymal transition (EMT) process and stem cell-associated proteins OCT4 and SOX2 expression in HCC cells. In addition, The VM-associated proteins, including EphA2, VE-cadherin and MMP2, also could be effectively enhanced by the overexpression of BMP4. Furthermore, according to the TCGA database, higher expression of BMP4 is seen in HCC in contrast to normal liver samples. Immunohistochemistry revealed that BMP4 was positively associated with VM formation, age, histological differentiation, HCC stage, and shorter survival duration. These data demonstrated that BMP4 could promote VM network formation in HCC through induction of stemness in EMT and modulating the EphA2/VE-cadherin/MMP2 signaling pathway.
Collapse
Affiliation(s)
- Xiao Li
- Department Of Pathology, General Hospital Of Tianjin Medical University, Tianjin, 300052, China.,Department Of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Baocun Sun
- Department Of Pathology, General Hospital Of Tianjin Medical University, Tianjin, 300052, China.,Department Of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Xiulan Zhao
- Department Of Pathology, General Hospital Of Tianjin Medical University, Tianjin, 300052, China.,Department Of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Jindan An
- Department Of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, 300060, China
| | - Yanhui Zhang
- Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, 300060, China
| | - Qiang Gu
- Department Of Pathology, General Hospital Of Tianjin Medical University, Tianjin, 300052, China.,Department Of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Nan Zhao
- Department Of Pathology, General Hospital Of Tianjin Medical University, Tianjin, 300052, China.,Department Of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Yong Wang
- Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, 300060, China
| | - Fang Liu
- Department Of Pathology, General Hospital Of Tianjin Medical University, Tianjin, 300052, China.,Department Of Pathology, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
31
|
Cheng L, Wang Q, Tao X, Qin Y, Wu Q, Zheng D, Chai D, Zhang Y, Lu D, Ci H, Wang Z, Ma J, Wang D, Cheng Z, Wu S, Tao Y. FOXM 1 induces Vasculogenic mimicry in esophageal cancer through β-catenin /Tcf4 signaling. Diagn Pathol 2020; 15:14. [PMID: 32035486 PMCID: PMC7007660 DOI: 10.1186/s13000-020-00929-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 02/04/2020] [Indexed: 12/31/2022] Open
Abstract
Objective To investigate the role of FOXM1, β-catenin and TCF4 in esophageal cancer (EC) and their relationship to VM (Vasculogenic Mimicry). Methods CCK-8 were performed to examine EC cell proliferation in FOXM1 silenced cells. EC cell migration and invasion were investigated through wound healing and Transwell assays, respectively. The formation of pipe like structures were assessed in 3D cultures. The expression of Foxm1, β-catenin, Tcf4 and E-cadherin were investigated through western blot, RT-qPCR and immunohistochemistry (IHC) staining. The relationship between FOXM1 expression, clinic-pathological features, and overall survival (OS) were further analyzed. Results A loss of FOXM1 expression correlated with the OS of ESCC patients. FOXM1 silencing led to a loss of cell growth and suppressed cell migration and invasion in ESCC cells. VM structures were identified in ESCC tissues and human EC cell lines. Mechanistically, FOXM1 was found to promote tumorigenesis through the regulation of β-catenin, Tcf4, and E-cadherin in EC cells, leading to the formation of VM structures. Conclusions These findings highlight FoxM1 as a novel therapeutic target in ESCC.
Collapse
Affiliation(s)
- Lili Cheng
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Qi Wang
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Xiaoying Tao
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Yanzi Qin
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Qiong Wu
- Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Dafang Zheng
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Damin Chai
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Yong Zhang
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Dongbing Lu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Hongfei Ci
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Zhiwei Wang
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China
| | - Jia Ma
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China
| | - Danna Wang
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Zenong Cheng
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Shiwu Wu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China.,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China
| | - Yisheng Tao
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Changhuai road 287, Bengbu, Anhui, 233000, People's Republic of China. .,Department of Pathology, Bengbu Medical College, 2600 Donghai Avenue, Bengbu, Anhui Province, China.
| |
Collapse
|
32
|
Haiaty S, Rashidi MR, Akbarzadeh M, Maroufi NF, Yousefi B, Nouri M. Targeting vasculogenic mimicry by phytochemicals: A potential opportunity for cancer therapy. IUBMB Life 2020; 72:825-841. [PMID: 32026601 DOI: 10.1002/iub.2233] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 01/06/2020] [Indexed: 12/18/2022]
Abstract
Vasculogenic mimicry (VM) is regarded as a process where very aggressive cancer cells generate vascular-like patterns without the presence of endothelial cells. It is considered as the main mark of malignant cancer and has pivotal role in cancer metastasis and progression in various types of cancers. On the other hand, resistance to the antiangiogenesis therapies leads to the cancer recurrence. Therefore, development of novel chemotherapies and their combinations is urgently needed for abolition of VM structures and also for better tumor therapy. Hence, identifying compounds that target VM structures might be superior therapeutic factors for cancers treatment and controlling the recurrence and metastasis. In recent times, naturally occurring compounds, especially phytochemicals have obtained great attention due to their safe properties. Phytochemicals are also capable of targeting VM structure and also their main signaling pathways. Consequently, in this review article, we illustrated key signaling pathways in VM, and the phytochemicals that affect these structures including curcumin, genistein, lycorine, luteolin, columbamine, triptolide, Paris polyphylla, dehydroeffusol, jatrorrhizine hydrochloride, grape seed proanthocyanidins, resveratrol, isoxanthohumol, dehydrocurvularine, galiellalactone, oxacyclododecindione, brucine, honokiol, ginsenoside Rg3, and norcantharidin. The recognition of these phytochemicals and their safety profile may lead to new therapeutic agents' development for VM elimination in different types of tumors.
Collapse
Affiliation(s)
- Sanya Haiaty
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Akbarzadeh
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Nazila F Maroufi
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
33
|
Markkanen E. Know Thy Model: Charting Molecular Homology in Stromal Reprogramming Between Canine and Human Mammary Tumors. Front Cell Dev Biol 2019; 7:348. [PMID: 31921858 PMCID: PMC6927989 DOI: 10.3389/fcell.2019.00348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/03/2019] [Indexed: 12/24/2022] Open
Abstract
Spontaneous canine simple mammary tumors (CMTs) are often viewed as models of human breast cancer. Cancer-associated stroma (CAS) is central for initiation and progression of human cancer, and is likely to play a key role in canine tumors as well. Until recently, however, canine CAS in general, and in CMT in particular, lacked detailed characterization and it remained unclear how canine and human CAS compare. This void in knowledge regarding canine CAS and the resulting lack of unbiased cross-species analysis of molecular homologies and differences undermined the validity of the canine model for human disease. To assess stromal reprogramming in canine breast tumors, we have recently established a protocol to specifically isolate and analyze CAS and matched normal stroma from archival, formalin-fixed paraffin embedded (FFPE) clinical tumor samples using laser-capture microdissection followed by next-generation RNA-sequencing. Using this approach, we have analyzed stromal reprogramming in both malignant canine mammary carcinomas (mCAs) as well as benign canine mammary adenomas in a series of studies. Our results demonstrate strong stromal reprogramming in CMTs and identify high-grade molecular homology between human and canine CAS. Here, I aim to give a short background on the value of comparative oncology in general, and spontaneous CMT in particular. This will be followed by a concise review of the current knowledge of stromal reprogramming in both malignant canine mCA and benign adenoma. Finally, I will conclude with insights on highly conserved aspects of stromal reprogramming between CMT and human breast cancer that accentuate the relevance of CAS in CMT as a model for the human disease.
Collapse
Affiliation(s)
- Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Yang Z, Chen J, Xie H, Liu T, Chen Y, Ma Z, Pei X, Yang W, Li L. Androgen receptor suppresses prostate cancer metastasis but promotes bladder cancer metastasis via differentially altering miRNA525-5p/SLPI-mediated vasculogenic mimicry formation. Cancer Lett 2019; 473:118-129. [PMID: 31843555 DOI: 10.1016/j.canlet.2019.12.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022]
Abstract
Early studies suggest that the androgen receptor (AR) may play differential roles in influencing prostate cancer (PCa) and bladder cancer (BCa) metastasis, but the underlying mechanisms remain unclear. Here, we found that the AR might function via differentially altering vasculogenic mimicry (VM) formation to either decrease PCa metastasis or increase BCa metastasis. Mechanism dissection showed that the AR could differentially alter the expression of the VM marker SLPI through miR-525-5p to regulate SLPI; moreover, it could either increase miR-525-5p transcription in PCa or decrease it in BCa via binding to different androgen-response-elements (AREs) located at different positions in the miR-525 precursor promoter. Further, results from liquid chromatography-mass spectrometry (LC-MS) showed that the co-factors of AR in PCa and BCa are NFIX and HDAC2, respectively. Together, these results provide the first detailed mechanism of how the AR can differentially alter PCa and BCa metastasis; thus, targeting the newly identified AR-miR-525-5p-SLPI axis may help suppress metastasis.
Collapse
Affiliation(s)
- Zhao Yang
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jiaqi Chen
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hongjun Xie
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tianjie Liu
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yule Chen
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhenkun Ma
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xinqi Pei
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wenjie Yang
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lei Li
- Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
35
|
Zhu H, Dai C, He L, Xu A, Chen T. Iron (II) Polypyridyl Complexes as Antiglioblastoma Agents to Overcome the Blood-Brain Barrier and Inhibit Cell Proliferation by Regulating p53 and 4E-BP1 Pathways. Front Pharmacol 2019; 10:946. [PMID: 31551768 PMCID: PMC6733960 DOI: 10.3389/fphar.2019.00946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/24/2019] [Indexed: 12/28/2022] Open
Abstract
Background and Purpose: It is urgently required to develop promising candidates to permeate across blood-brain barrier (BBB) efficiently with simultaneous disrupting vasculogenic mimicry capability of gliomas. Previously, a series of iron (II) complexes were synthesized through a modified method. Hence, the aim of this study was to evaluate anticancer activity of Fe(PIP)3SO4 against glioma cancer cells. Methods: Cytotoxic effects were determined via MTT assay, and IC50 values were utilized to evaluate the cytotoxicity. Cellular uptake of Fe(PIP)3SO4 between U87 and HEB cells was conducted by subtracting content of the complex remaining in the cell culture supernatants. Propidium Iodide (PI)-flow cytometric analysis was used to analyze cell cycle proportion of U87 cells treated with Fe(PIP)3SO4. The reactive oxygen species levels induced by Fe(PIP)3SO4 were measured by 2'-deoxycoformycin (DCF) probe; ABTS assay was utilized to examine the radical scavenge capacity of Fe(PIP)3SO4. To study the bind efficiency to thioredoxin reductase (TrxR), Fe(PIP)3SO4 was introduced into solution containing TrxR. To verify if Fe(PIP)3SO4 could penetrate BBB, HBMEC/U87 coculture as BBB model was established, and penetrating capability of Fe(PIP)3SO4 was tested. In vitro U87 tumor spheroids were formed to test the permeating ability of Fe(PIP)3SO4. Acute toxicity and biodistribution of Fe(PIP)3SO4 were tested on mice for 72 h. Protein profiles associated with U87 cells treated with Fe(PIP)3SO4 were determined by Western blotting analysis. Results: Results showed that Fe(PIP)3SO4 could suppress cell proliferation by inducing G2/M phase cycle retardation and apoptotic pathways, which was related with expression of p53 and initiation factor 4E binding protein 1. In addition, Fe complex could suppress cell proliferation by downregulating reactive oxygen species levels via scavenging free radicals and interaction with TrxR. Furthermore, Fe(PIP)3SO4 could permeate across BBB and simultaneously inhibited the vasculogenic mimicry-channel of U87 cells, suggesting favorable antiglioblastoma efficacy. Acute toxicity manifested lower degree of the complex compared with cisplatin and temozolomide. Conclusion: Fe(PIP)3SO4 exhibited favorable anticancer activity against glioma cells associated with p53 and 4E binding protein 1, accompanied with negligible toxic effects on normal tissues. Herein, Fe(PIP)3SO4 could be developed as a promising metal-based chemotherapeutic agent to overcome BBB and antagonize glioblastomas.
Collapse
Affiliation(s)
- Huili Zhu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Chengli Dai
- The First Affiliated Hospital and the Department of Chemistry, Jinan University, Guangzhou, China
| | - Lizhen He
- The First Affiliated Hospital and the Department of Chemistry, Jinan University, Guangzhou, China
| | - Anding Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Tianfeng Chen
- The First Affiliated Hospital and the Department of Chemistry, Jinan University, Guangzhou, China
| |
Collapse
|
36
|
Massimini M, De Maria R, Malatesta D, Romanucci M, D'Anselmo A, Della Salda L. Establishment of three-dimensional canine osteosarcoma cell lines showing vasculogenic mimicry and evaluation of biological properties after treatment with 17-AAG. Vet Comp Oncol 2019; 17:376-384. [PMID: 31006970 DOI: 10.1111/vco.12482] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/11/2019] [Accepted: 04/14/2019] [Indexed: 12/17/2022]
Abstract
Vasculogenic mimicry (VM) is an alternative type of blood perfusion characterized by formation of non-endothelial cell-lined microcirculatory channels and is responsible for aggressive tumour biology and increased tumour-related mortality. VM-correlated genes are associated with vascular endothelial grown factor receptor 1 (VEGFR1), and hypoxia-related (hypoxia inducible factor 1 α-HIF1α) signalling pathways, whose molecules are client proteins of Hsp90 (heat shock protein 90) and are potential therapeutic targets. This pilot study was aimed to investigate vasculogenic mimicry in a three-dimensional (3D) cell culture system of two aggressive canine osteosarcoma (OSA) cell lines (D22 and D17), and to evaluate the response of these cells to 17-AAG (17-N-allylamino-17-demethoxygeldanamycin) treatment in relation to tubular-like structure formation in vitro. Only D17 cell line formed hollow matrix channels in long-term 3D cultures and assumed endothelial morphology, with cells expressing both Hsp90 and VEGFR1, but lacking expression of endothelial marker CD31. 17-AAG treatment inhibited migration of D17 OSA cells, also decreasing VM markers in vitro and inducing a reduction of HIF1α transcript and protein in this cell line. Taken together, these preliminary data indicate that the biological effects of 17-AAG on D17 3D culture and on HIF1α regulation can provide interesting information to translate these findings from the basic research to clinical approach for the treatment of canine OSA as a model in comparative oncology.
Collapse
Affiliation(s)
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | | | | | - Angela D'Anselmo
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | | |
Collapse
|
37
|
A Novel Peptide from Abalone ( Haliotis discus hannai) to Suppress Metastasis and Vasculogenic Mimicry of Tumor Cells and Enhance Anti-Tumor Effect In Vitro. Mar Drugs 2019; 17:md17040244. [PMID: 31022939 PMCID: PMC6520751 DOI: 10.3390/md17040244] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/06/2019] [Accepted: 04/19/2019] [Indexed: 12/17/2022] Open
Abstract
Vasculogenic mimicry (VM) formed by tumor cells plays a vital role in the progress of tumor, because it provides nutrition for tumor cells and takes away the metabolites. Therefore, the inhibition of VM is crucial to the clinical treatment of tumors. In this study, we investigated the anti-tumor effect of a novel peptide, KVEPQDPSEW (AATP), isolated from abalone (Haliotis discus hannai) on HT1080 cells by migration, invasion analysis and the mode of action. The results showed that AATP effectively inhibited MMPs by blocking MAPKs and NF-κB pathways, leading to the downregulation of metastasis of tumor cells. Moreover, AATP significantly inhibited VM and pro-angiogenic factors, including VEGF and MMPs by suppression of AKT/mTOR signaling. In addition, molecular docking was used to study the interaction of AATP and HIF-1α, and the results showed that AATP was combined with an active site of HIF-1α by a hydrogen bond. The effect of AATP on anti-metastatic and anti-vascular in HT1080 cells revealed that AATP may be a potential lead compound for treatment of tumors in the future.
Collapse
|
38
|
Yang F, Wen M, Pan D, Lin X, Mo J, Dong X, Liao S, Ma Y. IL-33/ST2 Axis Regulates Vasculogenic Mimicry via ERK1/2-MMP-2/9 Pathway in Melanoma. Dermatology 2019; 235:225-233. [PMID: 30928981 DOI: 10.1159/000498857] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 02/08/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Melanoma, an extremely malignant form of cancer, poses a significant health risk. Vasculogenic mimicry (VM), blood vessels formed by tumor cells instead of endothelial cells, is an important factor for the rapid progression of melanoma. Interleukin (IL)-33 is an inflammatory factor commonly found in the tumor microenvironment and plays an important role in the progression of many tumors. IL-33 acts on immune cells and tumor cells through its receptor ST2. This study hypothesized that IL-33 directly affects the progression of melanoma. OBJECTIVES This study was designed to investigate the effect of IL-33 on VM of melanoma and its potential mechanism of action. METHODS The expression of ST2 was evaluated in 66 cases of melanoma collected from human patients, and the differences were analyzed. In vitro experiments were conducted to study the effects of the IL-33/ST2 axis on cell migration and invasion and to elucidate possible mechanisms. RESULTS ST2 expression is associated with that of matrix metalloproteinase (MMP)-2 and VM in melanoma of patients. IL-33 increases the abilities of proliferation, migration and invasion of melanoma cells and VM tube formation through ST2. IL-33 induces the production of MMP-2/9 via ERK1/2 phosphorylation. CONCLUSION IL-33 can directly act on melanoma cells and promote its development.
Collapse
Affiliation(s)
- Fuhan Yang
- Department of Operative Surgery, Tianjin Medical University, Tianjin, China
| | - Mingming Wen
- School of Nursing of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dayu Pan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xian Lin
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Jing Mo
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Xueyi Dong
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Shihan Liao
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Yuemei Ma
- Department of Operative Surgery, Tianjin Medical University, Tianjin, China,
| |
Collapse
|
39
|
Yeo C, Lee HJ, Lee EO. Serum promotes vasculogenic mimicry through the EphA2/VE-cadherin/AKT pathway in PC-3 human prostate cancer cells. Life Sci 2019; 221:267-273. [PMID: 30797819 DOI: 10.1016/j.lfs.2019.02.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/14/2019] [Accepted: 02/20/2019] [Indexed: 12/13/2022]
Abstract
AIMS Serum is widely used for in vitro cell culture of eukaryotic cells. Although serum is well known to affect various biological activities in cancer cells, its effect in vasculogenic mimicry (VM) is not yet fully defined. Thus, this study investigated the role of serum in VM in human prostate cancer (PCa) PC-3 cells. MAIN METHODS Invasion assay and 3D culture VM tube formation assay are performed. VM-related molecules are checked by western blot and reverse transcriptase-polymerase chain reaction. Nuclear twist is detected by confocal after twist-FITC/DAPI double staining. KEY FINDINGS Serum dramatically induced not only invasion but also VM. Serum increased the phosphorylation of erythropoietin-producing hepatocellular A2 (EphA2) without affecting EphA2 expression. Both the protein and mRNA expression levels of vascular endothelial cadherin (VE-cadherin) are up-regulated by serum. Twist expression was increased in the nucleus by serum. Serum activated AKT through phosphorylation, despite the unchanged AKT expression. Serum caused an increase in matrix metalloproteinase-2 (MMP-2) and laminin subunit 5 gamma-2 (LAMC2) protein expressions. Wortmannin, a phosphoinositide-3-kinase inhibitor, significantly decreased serum-induced invasion and VM. SIGNIFICANCE These results demonstrated that serum activates EphA2 and up-regulates twist/VE-cadherin, which in turn activate AKT that up-regulates MMP-2 and LAMC2, thereby inducing the invasion and VM of human PCa PC-3 cells.
Collapse
Affiliation(s)
- Changhwan Yeo
- Department of Cancer Preventive Material Development, Graduate school, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Hyo-Jeong Lee
- Department of Cancer Preventive Material Development, Graduate school, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Department of Science in Korean Medicine, Graduate school, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Eun-Ok Lee
- Department of Cancer Preventive Material Development, Graduate school, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; Department of Science in Korean Medicine, Graduate school, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
40
|
Brucine Suppresses Vasculogenic Mimicry in Human Triple-Negative Breast Cancer Cell Line MDA-MB-231. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6543230. [PMID: 30723742 PMCID: PMC6339755 DOI: 10.1155/2019/6543230] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 11/26/2018] [Accepted: 12/23/2018] [Indexed: 11/17/2022]
Abstract
Vasculogenic mimicry (VM) with the pattern of endothelial independent tubular structure formation lined by aggressive tumor cells mimics regular tumor blood vessels to ensure robust blood supply and correlates with the proliferation, invasion, metastasis, and poor prognosis of malignant tumors, which was demonstrated to be a major obstacle for resistance to antiangiogenesis therapy. Therefore, it is urgent to discover methods to abrogate the VM formation of tumors, which possesses important practical significance for improving tumor therapy. Brucine is a traditional medicinal herb extracted from seeds of Strychnos nux-vomica L. (Loganiaceae) exhibiting antitumor activity in a variety of cancer models. In the present study, the effect of brucine on vasculogenic mimicry and the related mechanism are to be investigated. We demonstrated that, in a triple-negative breast cancer cell line MDA-MB-231, brucine induced a dose-dependent inhibitory effect on cell proliferation along with apoptosis induction at higher concentrations. The further study showed that brucine inhibited cell migration and invasion with a dose-dependent manner. Our results for the first time indicated that brucine could disrupt F-actin cytoskeleton and microtubule structure, thereby impairing hallmarks of aggressive tumors, like migration, invasion, and holding a possibility of suppressing vasculogenic mimicry. Hence, the inhibitory effect of brucine on vasculogenic mimicry was further verified. The results illustrated that brucine significantly suppressed vasculogenic mimicry tube formation with a dose-dependent effect indicated by the change of the number of tubules, intersections, and mean length of tubules. The in-depth molecular mechanism of vasculogenic mimicry suppression induced by brucine was finally suggested. It was demonstrated that brucine inhibited vasculogenic mimicry which might be through the downregulation of erythropoietin-producing hepatocellular carcinoma-A2 and matrix metalloproteinase-2 and metalloproteinase-9.
Collapse
|
41
|
Clinical characteristics and preliminary morphological observation of 47 cases of primary anorectal malignant melanomas. Melanoma Res 2018; 28:592-599. [DOI: 10.1097/cmr.0000000000000491] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
42
|
Shao B, Zhao X, Liu T, Zhang Y, Sun R, Dong X, Liu F, Zhao N, Zhang D, Wu L, Wang Y, Wang M, Meng J, Lin X, Sun B. LOXL2 promotes vasculogenic mimicry and tumour aggressiveness in hepatocellular carcinoma. J Cell Mol Med 2018; 23:1363-1374. [PMID: 30506621 PMCID: PMC6349148 DOI: 10.1111/jcmm.14039] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/11/2018] [Accepted: 10/28/2018] [Indexed: 12/11/2022] Open
Abstract
Lysyl oxidase‐like 2 (LOXL2) has shown to promote metastasis and poor prognosis in hepatocellular carcinoma (HCC). Also, we have previously reported that vasculogenic mimicry (VM) is associated with invasion, metastasis and poor survival in HCC patients. In the present study, we investigated molecular function of LOXL2 in HCC and VM. We used the immunohistochemical and CD31/periodic acid‐Schiff double staining to detect the relationship between LOXL2 and VM formation. We performed the gain and loss of function studies and analysed the migratory, invasion and tube formation in HCC cell lines. We analysed the function of LOXL2 in VM formation and HCC metastasis both in vitro and in vivo. We have showed that LOXL2 was overexpression in HCC and was positively correlated with tumour grade, metastasis, VM formation and poor survival in 201 HCC patients. Secondly, our studies have showed that LOXL2 overexpression in HCC cells significantly promoted migration, invasion and tube formation. Finally, we found that LOXL2 may increase SNAIL expression, thereby enabling VM. Our study indicated that LOXL2 may promote VM formation and tumour metastasis by collaborating with SNAIL in HCC. What's more, the overexpression of LOXL2 indicated a poor prognosis in HCC patients.
Collapse
Affiliation(s)
- Bing Shao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Tieju Liu
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Yanhui Zhang
- Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, China
| | - Ran Sun
- Tianjin Nankai Hospital, Tianjin, China
| | - Xueyi Dong
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Fang Liu
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Lili Wu
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Yong Wang
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Meili Wang
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Jie Meng
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Xian Lin
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
43
|
De Carvalho N, Welzel J, Schuh S, Themstrup L, Ulrich M, Jemec GBE, Holmes J, Kaleci S, Chester J, Bigi L, Ciardo S, Pellacani G. The vascular morphology of melanoma is related to Breslow index: An in vivo study with dynamic optical coherence tomography. Exp Dermatol 2018; 27:1280-1286. [DOI: 10.1111/exd.13783] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 09/04/2018] [Accepted: 09/10/2018] [Indexed: 01/04/2023]
Affiliation(s)
| | - Julia Welzel
- Department of Dermatology; General Hospital Augsburg; Augsburg Germany
| | - Sandra Schuh
- Department of Dermatology; General Hospital Augsburg; Augsburg Germany
| | - Lotte Themstrup
- Department of Dermatology; Zealand University Hospital; Roskilde Denmark
- Health Sciences Faculty; University of Copenhagen; Copenhagen Denmark
| | | | - Gregor B. E. Jemec
- Department of Dermatology; Zealand University Hospital; Roskilde Denmark
- Health Sciences Faculty; University of Copenhagen; Copenhagen Denmark
| | | | - Shaniko Kaleci
- Department of Dermatology; University of Modena and Reggio Emilia; Modena Italy
| | - Johanna Chester
- Department of Dermatology; University of Modena and Reggio Emilia; Modena Italy
| | - Laura Bigi
- Department of Dermatology; University of Modena and Reggio Emilia; Modena Italy
| | - Silvana Ciardo
- Department of Dermatology; University of Modena and Reggio Emilia; Modena Italy
| | - Giovanni Pellacani
- Department of Dermatology; University of Modena and Reggio Emilia; Modena Italy
| |
Collapse
|
44
|
Yang L, Song X, Gong T, Jiang K, Hou Y, Chen T, Sun X, Zhang Z, Gong T. Development a hyaluronic acid ion-pairing liposomal nanoparticle for enhancing anti-glioma efficacy by modulating glioma microenvironment. Drug Deliv 2018; 25:388-397. [PMID: 29378465 PMCID: PMC6058578 DOI: 10.1080/10717544.2018.1431979] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glioma, one of the most common brain tumors, remains a challenge worldwide. Due to the specific biological barriers such as blood–brain barrier (BBB), cancer stem cells (CSCs), tumor associated macrophages (TAMs), and vasculogenic mimicry channels (VMs), a novel versatile targeting delivery for anti-glioma is in urgent need. Here, we designed a hyaluronic acid (HA) ion-pairing nanoparticle. Then, these nanoparticles were encapsulated in liposomes, termed as DOX-HA-LPs, which showed near-spherical morphology with an average size of 155.8 nm and uniform distribution (PDI = 0.155). HA was proven to specifically bind to CD44 receptor, which is over-expressed on the surface of tumor cells, other associated cells (such as CSCs and TAMs) and VMs. We systematically investigated anti-glioma efficacy and mechanisms in vivo and in vitro. The strong anti-glioma efficacy could attribute to the accumulation in glioma site and the regulation of tumor microenvironment with depletion of TAMs, inhibition of VMs, and elimination of CSCs.
Collapse
Affiliation(s)
- Liuqing Yang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , PR China
| | - Xu Song
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , PR China
| | - Ting Gong
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , PR China
| | - Kejun Jiang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , PR China
| | - Yingying Hou
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , PR China
| | - Tijia Chen
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , PR China
| | - Xun Sun
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , PR China
| | - Zhirong Zhang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , PR China
| | - Tao Gong
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu , PR China
| |
Collapse
|
45
|
Hsp90β promotes aggressive vasculogenic mimicry via epithelial-mesenchymal transition in hepatocellular carcinoma. Oncogene 2018; 38:228-243. [PMID: 30087438 DOI: 10.1038/s41388-018-0428-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 01/20/2023]
Abstract
Hepatocellular carcinoma (HCC) is a typical hypervascular solid tumor. Vasculogenic mimicry (VM) formed by aggressive tumor cells to mimic vasculogenic networks plays an important role in the tumor malignancy of HCC. Hsp90β promotes endothelial cell-dependent angiogenesis in HCC. However, the relationship between Hsp90β and VM formation is unclear. In this study, we found that Hsp90β is positively correlated with VM and EMT marker proteins in HCC tissues and promotes tube formation, cell migration, and invasion in vitro. Hsp90β interacts with Twist1 and promotes its deubiquitination and stabilization to nuclear translocation and enhances the VE-cadherin promoter activity. Results of in vitro analysis indicate that Hsp90β enhances the tumor VM in tumor-burdened mice, and the Hsp90 inhibitor NVP-BEP800 suppresses VM formation by releasing Hsp90β and Twist1 interaction. This study provides a potential antitumor therapy for inhibiting VM by targeting Hsp90β in HCC.
Collapse
|
46
|
Xiao T, Zhang Q, Zong S, Zhong WL, Qin Y, Bi Z, Chen S, Liu HJ, Wei JJ, Zhou BJ, Wang LM, Zhou HG, Liu YR, Sun T, Yang C. Protease-activated receptor-1 (PAR1) promotes epithelial-endothelial transition through Twist1 in hepatocellular carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:185. [PMID: 30081924 PMCID: PMC6091192 DOI: 10.1186/s13046-018-0858-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 07/12/2018] [Indexed: 01/13/2023]
Abstract
Background Tumor cells transfer into endothelial cells by epithelial–endothelial transition (EET), which is characterized by vasculagenic mimicry (VM) in morphology. VM can change tumor microcirculation, progression, and metastasis. However, the molecular mechanisms of endothelial-like transition remain unclear. EET is a subtype of epithelial–mesenchymal transition (EMT). Twist1, a transcriptional regulatory factor of EMT, is an important factor that induces EET in hepatocellular carcinoma(HCC), but the upstream signal of Twist1 is unclear. Methods Expression plasmids, Ca mobilization, and three-dimensional cultures were evaluated. Western blot assay, reporter gene assay, and immunofluorescence staining were conducted. A murine xenograft model was established. Analyses of immunohistochemistry, patient samples, and complementary DNA (cDNA) microarrays were also performed. Results This study demonstrated that protease-activated receptor-1 (PAR1) can increase the expression of endothelial markers and enhance VM formation by upregulating Twist1 both in vitro and in vivo through thrombin binding. Thrombin not only activates PAR1 but also promotes PAR1 internalization in a time-dependent manner. Clinical pathological analysis further confirms that PAR1 expression is directly correlated with the endothelial marker expression, VM formation, and metastasis and indicates poor survival rate of patients with tumors. Conclusion PAR1 promotes EET through Twist1 in HCC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0858-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ting Xiao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Qiang Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Shumin Zong
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Wei-Long Zhong
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Yuan Qin
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Zhun Bi
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Shuang Chen
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Hui-Juan Liu
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Jun-Jie Wei
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Bi-Jiao Zhou
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Lu-Meng Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Hong-Gang Zhou
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Yan-Rong Liu
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China.
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China. .,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China. .,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China.
| |
Collapse
|
47
|
Ren K, Zhang J, Gu X, Wu S, Shi X, Ni Y, Chen Y, Lu J, Gao Z, Wang C, Yao N. Migration-inducing gene-7 independently predicts poor prognosis of human osteosarcoma and is associated with vasculogenic mimicry. Exp Cell Res 2018; 369:80-89. [PMID: 29750896 DOI: 10.1016/j.yexcr.2018.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 05/06/2018] [Accepted: 05/07/2018] [Indexed: 12/19/2022]
Abstract
Vasculogenic mimicry (VM) is a special type of vascular channel formed by tumor cells without endothelial cell participation. Migration-inducing gene 7 (MIG-7) plays an important role in regulating VM. In this study, immunohistochemical staining was used to detect MIG-7 in tissue specimens from 141 primary osteosarcoma patients, and the relationship between MIG-7 and VM was examined. Survival analysis were performed to evaluate the prognoses. MIG-7 knockdown osteosarcoma cells were used for cell proliferation, apoptosis, migration, invasiveness and VM formation assays. A spontaneously metastasizing cell line-derived orthotopic xenograft mouse model was established to evaluate the effect of MIG-7 knockdown on tumorigenesis, VM formation and lung metastasis. MIG-7 expression was associated with VM formation. There were significant differences in overall and metastasis-free survival between the MIG-7-positive and MIG-7-negative groups. The MIG-7 expression was shown to be an independent indicator of both overall and metastasis-free survival. In vitro knockdown of MIG-7 dramatically reduced migration, invasion and VM formation in osteosarcoma cells without any significant effect on cell proliferation and apoptosis. MIG-7 knockdown also exhibited potent antitumor, antimetastasis and anti-VM effects in the orthotopic mouse model of 143B osteosarcoma. Therefore, MIG-7 serves as an independent unfavorable prognostic indicator in osteosarcoma patients and MIG-7 is an important mediator of osteosarcoma VM formation.
Collapse
Affiliation(s)
- Ke Ren
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu Province, PR China
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No.100, Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu Province, PR China; Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, PR China
| | - Xiaojie Gu
- Institute of Biotechnology, School of Environmental and Chemical Engineering, Dalian Jiaotong University, Dalian 116028, Liaoning Province, PR China
| | - Sujia Wu
- Jinling Hospital, Department of Orthopedics, Nanjing University, School of Medicine, Nanjing 210002, Jiangsu Province, PR China
| | - Xin Shi
- Jinling Hospital, Department of Orthopedics, Nanjing University, School of Medicine, Nanjing 210002, Jiangsu Province, PR China
| | - Yicheng Ni
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No.100, Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu Province, PR China; Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, PR China; Department of Radiology, Faculty of Medicine, K.U. Leuven, Leuven 3000, Belgium
| | - Yong Chen
- Jinling Hospital, Department of Orthopedics, Nanjing University, School of Medicine, Nanjing 210002, Jiangsu Province, PR China
| | - Jun Lu
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu Province, PR China
| | - Zengxin Gao
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu Province, PR China
| | - Chen Wang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu Province, PR China.
| | - Nan Yao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No.100, Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu Province, PR China; Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, PR China.
| |
Collapse
|
48
|
Saponaro C, Vagheggini A, Scarpi E, Centonze M, Catacchio I, Popescu O, Pastena MI, Giotta F, Silvestris N, Mangia A. NHERF1 and tumor microenvironment: a new scene in invasive breast carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:96. [PMID: 29716631 PMCID: PMC5930748 DOI: 10.1186/s13046-018-0766-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/19/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND Tumor microenvironment (TME) includes many factors such as tumor associated inflammatory cells, vessels, and lymphocytes, as well as different signaling molecules and extracellular matrix components. These aspects can be de-regulated and consequently lead to a worsening of cancer progression. In recent years an association between the scaffolding protein Na+/H+ exchanger regulatory factor 1 (NHERF1) and tumor microenvironment changes in breast cancer (BC) has been reported. METHODS Subcellular NHERF1 localization, vascular endothelial growth factor (VEGF), its receptor VEGFR1, hypoxia inducible factor 1 alpha (HIF-1α), TWIST1 expression and microvessel density (MVD) in 183 invasive BCs were evaluated, using immunohistochemistry on tissue microarrays (TMA). Immunofluorescence was employed to explore protein interactions. RESULTS Cytoplasmic NHERF1(cNHERF1) expression was directly related to cytoplasmic VEGF and VEGFR1 expression (p = 0.001 and p = 0.027 respectively), and inversely to nuclear HIF-1α (p = 0.021) and TWIST1 (p = 0.001). Further, immunofluorescence revealed an involvement of tumor cells with NHERF1 positive staining in neo-vascular formation, suggesting a "mosaic" structure development of these neo-vessels. Survival analyses showed that loss of nuclear TWIST1 (nTWIST1) expression was related to a decrease of disease free survival (DFS) (p < 0.001), while nTWIST1-/mNHERF1+ presented an increased DFS with respect to nTWIST1+/mNHERF1- phenotype (p < 0.001). Subsequently, the analyses of nTWIST1+/cNHERF1+ phenotype selected a subgroup of patients with a worse DFS compared to nTWIST1-/cNHERF1- patients (p = 0.004). CONCLUSION Resulting data suggested a dynamic relation between NHERF1 and TME markers, and confirmed both the oncosuppressor role of membranous NHERF1 expression and the oncogene activity of cytoplasmic NHERF1.
Collapse
Affiliation(s)
- Concetta Saponaro
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - Alessandro Vagheggini
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST)-IRCCS, 47014, Meldola, FC, Italy
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST)-IRCCS, 47014, Meldola, FC, Italy
| | - Matteo Centonze
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - Ivana Catacchio
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - Ondina Popescu
- Pathology Department, IRCCS-Istituto Tumori "Giovanni PaoloII", 70124, Bari, Italy
| | - Maria Irene Pastena
- Pathology Department, IRCCS-Istituto Tumori "Giovanni PaoloII", 70124, Bari, Italy
| | - Francesco Giotta
- Medical Oncology Unit, IRCCS-Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - Nicola Silvestris
- Scientific Direction, IRCCS-Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | - Anita Mangia
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy.
| |
Collapse
|
49
|
Smeda M, Kieronska A, Proniewski B, Jasztal A, Selmi A, Wandzel K, Zakrzewska A, Wojcik T, Przyborowski K, Derszniak K, Stojak M, Kaczor D, Buczek E, Watala C, Wietrzyk J, Chlopicki S. Dual antiplatelet therapy with clopidogrel and aspirin increases mortality in 4T1 metastatic breast cancer-bearing mice by inducing vascular mimicry in primary tumour. Oncotarget 2018; 9:17810-17824. [PMID: 29707148 PMCID: PMC5915156 DOI: 10.18632/oncotarget.24891] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/25/2018] [Indexed: 12/21/2022] Open
Abstract
Platelet inhibition has been considered an effective strategy for combating cancer metastasis and compromising disease malignancy although recent clinical data provided evidence that long-term platelet inhibition might increase incidence of cancer deaths in initially cancer-free patients. In the present study we demonstrated that dual anti-platelet therapy based on aspirin and clopidogrel (ASA+Cl), a routine regiment in cardiovascular patients, when given to cancer-bearing mice injected orthotopically with 4T1 breast cancer cells, promoted progression of the disease and reduced mice survival in association with induction of vascular mimicry (VM) in primary tumour. In contrast, treatment with ASA+Cl or platelet depletion did reduce pulmonary metastasis in mice, if 4T1 cells were injected intravenously. In conclusion, distinct platelet-dependent mechanisms inhibited by ASA+Cl treatment promoted cancer malignancy and VM in the presence of primary tumour and afforded protection against pulmonary metastasis in the absence of primary tumour. In view of our data, long-term inhibition of platelet function by dual anti-platelet therapy (ASA+Cl) might pose a hazard when applied to a patient with undiagnosed and untreated malignant cancer prone to undergo VM.
Collapse
Affiliation(s)
- Marta Smeda
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Anna Kieronska
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Bartosz Proniewski
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Anna Selmi
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Krystyna Wandzel
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Agnieszka Zakrzewska
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Tomasz Wojcik
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Kamil Przyborowski
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Katarzyna Derszniak
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Dawid Kaczor
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Elzbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Cezary Watala
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Kosciuszki 4, Lodz 90-419, Poland
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Department of Experimental Oncology, Rudolfa Weigla 4, Wroclaw 53-114, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
- Chair of Pharmacology, Jagiellonian University, Medical College, Grzegorzecka 16, Krakow 31-531, Poland
| |
Collapse
|
50
|
Sun B, Zhang D, Zhao N, Zhao X. Epithelial-to-endothelial transition and cancer stem cells: two cornerstones of vasculogenic mimicry in malignant tumors. Oncotarget 2018; 8:30502-30510. [PMID: 27034014 PMCID: PMC5444760 DOI: 10.18632/oncotarget.8461] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/14/2016] [Indexed: 01/11/2023] Open
Abstract
Vasculogenic mimicry (VM) is a functional microcirculation pattern in malignant tumors accompanied by endothelium-dependent vessels and mosaic vessels. VM has been identified in more than 15 solid tumor types and is associated with poor differentiation, late clinical stage and poor prognosis. Classic anti-angiogenic agents do not target endothelium-dependent vessels and are not efficacious against tumors exhibiting VM. Further insight into the molecular signaling that triggers and promotes VM formation could improve anti-angiogenic therapeutics. Recent studies have shown that cancer stem cells (CSCs) and epithelium-to-endothelium transition (EET), a subtype of epithelial-to-mesenchymal transition (EMT), accelerate VM formation by stimulating tumor cell plasticity, remodeling the extracellular matrix (ECM) and connecting VM channels with host blood vessels. VM channel-lining cells originate from CSCs due to expression of EMT inducers such as Twist1, which promote EET and ECM remodeling. Hypoxia and high interstitial fluid pressure in the tumor microenvironment induce a specific type of cell death, linearly patterned programmed cell necrosis (LPPCN), which spatially guides VM and endothelium-dependent vessel networks. This review focuses on the roles of CSCs and EET in VM, and on possible novel anti-angiogenic strategies against alternative tumor vascularization.
Collapse
Affiliation(s)
- Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|