1
|
Chen X, Li Q, Fu X, Li J, Deng J, Zhang Q, Qiu M, Lyu X, Cai L, Li H, Li X, Yao K, Wang J, Huang Z, Chen L, Zhang J, Li D. Tumor-Derived EBV-miR-BART2-5p Promotes Nasopharyngeal Carcinoma Metastasis by Inducing Premetastatic Endothelial Cell Pyroptosis. Mol Cancer Res 2025; 23:250-262. [PMID: 39545766 DOI: 10.1158/1541-7786.mcr-24-0165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 09/26/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Extravasation is a key step in tumor metastasis. Epstein‒Barr virus plays a crucial role in nasopharyngeal carcinoma (NPC) metastasis. However, the functions and molecular mechanisms of Epstein‒Barr virus during tumor cell extravasation remain unclear. Here, we showed that the expression of pyroptosis-associated proteins is greater in the endothelial cells of metastatic NPC tissues than in those of nontumor tissues exosomes derived from NPC cells promoted endothelial cell pyroptosis, vascular permeability, and tumor cell extravasation. Moreover, we found that BART2-5p is abundant in serum exosomes from patients with NPC metastasis and in NPC cells and that it regulates endothelial cell pyroptosis in premetastatic organs via MRE11A. Exosomes containing a BART2-5p inhibitor and AAV-MRE11A attenuated endothelial cell pyroptosis and tumor metastasis. Moreover, in the endothelial cells of metastatic tissues from patients with NPC, the BART2-5p level was positively associated with pyroptosis-related protein expression. Collectively, our findings suggest that exosomal BART2-5p is involved in premetastatic niche formation, identifying secreted BART2-5p as a potential therapeutic target for NPC metastasis. Implications: The finding that secreted BART2-5p is involved in premetastatic niche formation may aid the development of a potential therapeutic target for NPC metastasis.
Collapse
Affiliation(s)
- Xingrui Chen
- GuangDong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Qiqi Li
- Department of Pathology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyan Fu
- Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jike Li
- GuangDong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Deng
- Department of Biologic Products of GuangDong Institute for Drug Control, NMPA Key Laboratory for Quality Control of Blood Product, Guangzhou, China
| | - Qianbing Zhang
- GuangDong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Mengying Qiu
- GuangDong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoming Lyu
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Linbo Cai
- GuangDong Sanjiu Brain Hospital, Guangzhou, China
| | - Hainan Li
- GuangDong Sanjiu Brain Hospital, Guangzhou, China
| | - Xin Li
- 9ShenZhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center (CIRC), ShenZhen Hospital, Southern Medical University, Shenzhen, China
| | - Kaitai Yao
- GuangDong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiahong Wang
- GuangDong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhongxi Huang
- GuangDong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Liang Chen
- Department of Otolaryngology, Head and Neck Surgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, China
| | - Jiangyu Zhang
- Department of Pathology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Dengke Li
- GuangDong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Ye H, Hu X, Wen Y, Tu C, Hornicek F, Duan Z, Min L. Exosomes in the tumor microenvironment of sarcoma: from biological functions to clinical applications. J Nanobiotechnology 2022; 20:403. [PMID: 36064358 PMCID: PMC9446729 DOI: 10.1186/s12951-022-01609-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/23/2022] [Indexed: 11/10/2022] Open
Abstract
The current diagnosis and treatment of sarcoma continue to show limited timeliness and efficacy. In order to enable the early detection and management of sarcoma, increasing attentions have been given to the tumor microenvironment (TME). TME is a dynamic network composed of multiple cells, extracellular matrix, vasculature, and exosomes. Exosomes are nano-sized extracellular vesicles derived from various cells in the TME. The major function of exosomes is to promote cancer progress and metastasis through mediating bidirectional cellular communications between sarcoma cells and TME cells. Due to the content specificity, cell tropism, and bioavailability, exosomes have been regarded as promising diagnostic and prognostic biomarkers, and therapeutic vehicles for sarcoma. This review summarizes recent studies on the roles of exosomes in TME of sarcoma, and explores the emerging clinical applications.
Collapse
Affiliation(s)
- Huali Ye
- West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Xin Hu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Wen
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Chongqi Tu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Francis Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedics, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedics, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Li Min
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Mahadevan S, Kwong K, Lu M, Kelly E, Chami B, Romin Y, Fujisawa S, Manova K, Moore MAS, Zoellner H. A Novel Cartesian Plot Analysis for Fixed Monolayers That Relates Cell Phenotype to Transfer of Contents between Fibroblasts and Cancer Cells by Cell-Projection Pumping. Int J Mol Sci 2022; 23:ijms23147949. [PMID: 35887295 PMCID: PMC9316567 DOI: 10.3390/ijms23147949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 01/27/2023] Open
Abstract
We recently described cell-projection pumping as a mechanism transferring cytoplasm between cells. The uptake of fibroblast cytoplasm by co-cultured SAOS-2 osteosarcoma cells changes SAOS-2 morphology and increases cell migration and proliferation, as seen by single-cell tracking and in FACS separated SAOS-2 from co-cultures. Morphological changes in SAOS-2 seen by single cell tracking are consistent with previous observations in fixed monolayers of SAOS-2 co-cultures. Notably, earlier studies with fixed co-cultures were limited by the absence of a quantitative method for identifying sub-populations of co-cultured cells, or for quantitating transfer relative to control populations of SAOS-2 or fibroblasts cultured alone. We now overcome that limitation by a novel Cartesian plot analysis that identifies individual co-cultured cells as belonging to one of five distinct cell populations, and also gives numerical measure of similarity to control cell populations. We verified the utility of the method by first confirming the previously established relationship between SAOS-2 morphology and uptake of fibroblast contents, and also demonstrated similar effects in other cancer cell lines including from melanomas, and cancers of the ovary and colon. The method was extended to examine global DNA methylation, and while there was no clear effect on SAOS-2 DNA methylation, co-cultured fibroblasts had greatly reduced DNA methylation, similar to cancer associated fibroblasts.
Collapse
Affiliation(s)
- Swarna Mahadevan
- The Cellular and Molecular Pathology Research Unit, Oral Pathology and Oral Medicine, School of Dentistry, Faculty of Medicine and Health, The University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia; (S.M.); (K.K.); (M.L.); (E.K.); (B.C.)
| | - Kenelm Kwong
- The Cellular and Molecular Pathology Research Unit, Oral Pathology and Oral Medicine, School of Dentistry, Faculty of Medicine and Health, The University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia; (S.M.); (K.K.); (M.L.); (E.K.); (B.C.)
| | - Mingjie Lu
- The Cellular and Molecular Pathology Research Unit, Oral Pathology and Oral Medicine, School of Dentistry, Faculty of Medicine and Health, The University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia; (S.M.); (K.K.); (M.L.); (E.K.); (B.C.)
| | - Elizabeth Kelly
- The Cellular and Molecular Pathology Research Unit, Oral Pathology and Oral Medicine, School of Dentistry, Faculty of Medicine and Health, The University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia; (S.M.); (K.K.); (M.L.); (E.K.); (B.C.)
| | - Belal Chami
- The Cellular and Molecular Pathology Research Unit, Oral Pathology and Oral Medicine, School of Dentistry, Faculty of Medicine and Health, The University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia; (S.M.); (K.K.); (M.L.); (E.K.); (B.C.)
- The School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Yevgeniy Romin
- Molecular Cytology, The Memorial Sloan Kettering Cancer Center, 415-417 E 68 Street, ZRC 1962, New York, NY 10065, USA; (Y.R.); (S.F.); (K.M.)
| | - Sho Fujisawa
- Molecular Cytology, The Memorial Sloan Kettering Cancer Center, 415-417 E 68 Street, ZRC 1962, New York, NY 10065, USA; (Y.R.); (S.F.); (K.M.)
| | - Katia Manova
- Molecular Cytology, The Memorial Sloan Kettering Cancer Center, 415-417 E 68 Street, ZRC 1962, New York, NY 10065, USA; (Y.R.); (S.F.); (K.M.)
| | - Malcolm A. S. Moore
- Cell Biology, The Memorial Sloan Kettering Cancer Center, 430 E 67th St, RRL 717, New York, NY 10065, USA;
| | - Hans Zoellner
- The Cellular and Molecular Pathology Research Unit, Oral Pathology and Oral Medicine, School of Dentistry, Faculty of Medicine and Health, The University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia; (S.M.); (K.K.); (M.L.); (E.K.); (B.C.)
- Cell Biology, The Memorial Sloan Kettering Cancer Center, 430 E 67th St, RRL 717, New York, NY 10065, USA;
- Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia
- Graduate School of Biomedical Engineering, University of NSW, Kensington, NSW 2052, Australia
- Strongarch Pty Ltd., Pennant Hills, NSW 2120, Australia
- Correspondence: ; Tel.: +61-466400028
| |
Collapse
|
4
|
Abstract
Osteosarcoma is the most common primary bone malignancy in adolescents. Its high propensity to metastasize is the leading cause for treatment failure and poor prognosis. Although the research of osteosarcoma has greatly expanded in the past decades, the knowledge and new therapy strategies targeting metastatic progression remain sparse. The prognosis of patients with metastasis is still unsatisfactory. There is resonating urgency for a thorough and deeper understanding of molecular mechanisms underlying osteosarcoma to develop innovative therapies targeting metastasis. Toward the goal of elaborating the characteristics and biological behavior of metastatic osteosarcoma, it is essential to combine the diverse investigations that are performed at molecular, cellular, and animal levels from basic research to clinical translation spanning chemical, physical sciences, and biology. This review focuses on the metastatic process, regulatory networks involving key molecules and signaling pathways, the role of microenvironment, osteoclast, angiogenesis, metabolism, immunity, and noncoding RNAs in osteosarcoma metastasis. The aim of this review is to provide an overview of current research advances, with the hope to discovery druggable targets and promising therapy strategies for osteosarcoma metastasis and thus to overcome this clinical impasse.
Collapse
Affiliation(s)
- Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
A ligand-insensitive UNC5B splicing isoform regulates angiogenesis by promoting apoptosis. Nat Commun 2021; 12:4872. [PMID: 34381052 PMCID: PMC8358048 DOI: 10.1038/s41467-021-24998-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
The Netrin-1 receptor UNC5B is an axon guidance regulator that is also expressed in endothelial cells (ECs), where it finely controls developmental and tumor angiogenesis. In the absence of Netrin-1, UNC5B induces apoptosis that is blocked upon Netrin-1 binding. Here, we identify an UNC5B splicing isoform (called UNC5B-Δ8) expressed exclusively by ECs and generated through exon skipping by NOVA2, an alternative splicing factor regulating vascular development. We show that UNC5B-Δ8 is a constitutively pro-apoptotic splicing isoform insensitive to Netrin-1 and required for specific blood vessel development in an apoptosis-dependent manner. Like NOVA2, UNC5B-Δ8 is aberrantly expressed in colon cancer vasculature where its expression correlates with tumor angiogenesis and poor patient outcome. Collectively, our data identify a mechanism controlling UNC5B’s necessary apoptotic function in ECs and suggest that the NOVA2/UNC5B circuit represents a post-transcriptional pathway regulating angiogenesis. UNC5B is a Netrin-1 receptor expressed in endothelial cells that in the absence of ligand induces apoptosis. Here the authors identify an UNC5B splicing isoform that is insensitive to the pro-survival ligand Netrin-1 and is required for apoptosis-dependent blood vessel development.
Collapse
|
6
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
7
|
David MS, Kelly E, Cheung I, Xaymardan M, Moore MAS, Zoellner H. SAOS-2 osteosarcoma cells bind fibroblasts via ICAM-1 and this is increased by tumour necrosis factor-α. PLoS One 2014; 9:e101202. [PMID: 24979620 PMCID: PMC4076326 DOI: 10.1371/journal.pone.0101202] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 06/04/2014] [Indexed: 11/22/2022] Open
Abstract
We recently reported exchange of membrane and cytoplasmic markers between SAOS-2 osteosarcoma cells and human gingival fibroblasts (h-GF) without comparable exchange of nuclear markers, while similar h-GF exchange was seen for melanoma and ovarian carcinoma cells. This process of “cellular sipping” changes phenotype such that cells sharing markers of both SAOS-2 and h-GF have morphology intermediate to that of either cell population cultured alone, evidencing increased tumour cell diversity without genetic change. TNF-α increases cellular sipping between h-GF and SAOS-2, and we here study binding of SAOS-2 to TNF-α treated h-GF to determine if increased cellular sipping can be accounted for by cytokine stimulated SAOS-2 binding. More SAOS-2 bound h-GF pe-seeded wells than culture plastic alone (p<0.001), and this was increased by h-GF pre-treatment with TNF-α (p<0.001). TNF-α stimulated binding was dose dependent and maximal at 1.16nM (p<0.05) with no activity below 0.006 nM. SAOS-2 binding to h-GF was independent of serum, while the lipopolysaccharide antagonist Polymyxin B did not affect results, and TNF-α activity was lost on boiling. h-GF binding of SAOS-2 started to increase after 30min TNF-α stimulation and was maximal by 1.5hr pre-treatment (p<0.001). h-GF retained maximal binding up to 6hrs after TNF-α stimulation, but this was lost by 18hrs (p<0.001). FACS analysis demonstrated increased ICAM-1 consistent with the time course of SAOS-2 binding, while antibody against ICAM-1 inhibited SAOS-2 adhesion (p<0.04). Pre-treating SAOS-2 with TNF-α reduced h-GF binding to background levels (p<0.003), and this opposite effect to h-GF cytokine stimulation suggests that the history of cytokine exposure of malignant cells migrating across different microenvironments can influence subsequent interactions with fibroblasts. Since cytokine stimulated binding was comparable in magnitude to earlier reported TNF-α stimulated cellular sipping, we conclude that TNF-α stimulated cellular sipping likely reflects increased SAOS-2 binding as opposed to enhanced exchange mechanisms.
Collapse
Affiliation(s)
- Manu S. David
- The Cellular and Molecular Pathology Research Unit, Department of Oral Pathology and Oral Medicine, Faculty of Dentistry, The University of Sydney, Westmead Centre for Oral Health, Westmead Hospital, Westmead, New South Wales, Australia
| | - Elizabeth Kelly
- The Cellular and Molecular Pathology Research Unit, Department of Oral Pathology and Oral Medicine, Faculty of Dentistry, The University of Sydney, Westmead Centre for Oral Health, Westmead Hospital, Westmead, New South Wales, Australia
| | - Ivan Cheung
- The Cellular and Molecular Pathology Research Unit, Department of Oral Pathology and Oral Medicine, Faculty of Dentistry, The University of Sydney, Westmead Centre for Oral Health, Westmead Hospital, Westmead, New South Wales, Australia
| | - Munira Xaymardan
- The Cellular and Molecular Pathology Research Unit, Department of Oral Pathology and Oral Medicine, Faculty of Dentistry, The University of Sydney, Westmead Centre for Oral Health, Westmead Hospital, Westmead, New South Wales, Australia
| | - Malcolm A. S. Moore
- Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Hans Zoellner
- The Cellular and Molecular Pathology Research Unit, Department of Oral Pathology and Oral Medicine, Faculty of Dentistry, The University of Sydney, Westmead Centre for Oral Health, Westmead Hospital, Westmead, New South Wales, Australia
- Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
8
|
Huynh M, Zoellner H. Mechanisms of tumour cell induced apoptosis in human smooth muscle cells and fibroblasts. Aust Dent J 2014. [DOI: 10.1111/j.1834-7819.2007.tb06127.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
9
|
David MS, Kelly E, Zoellner H. Opposite cytokine synthesis by fibroblasts in contact co-culture with osteosarcoma cells compared with transwell co-cultures. Cytokine 2013; 62:48-51. [DOI: 10.1016/j.cyto.2013.02.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 02/20/2013] [Accepted: 02/23/2013] [Indexed: 10/27/2022]
|
10
|
David MS, Huynh MD, Kelly E, Rizos H, Coleman H, Rogers G, Zoellner H. Membrane and cytoplasmic marker exchange between malignant neoplastic cells and fibroblasts via intermittent contact: increased tumour cell diversity independent of genetic change. J Pathol 2012; 228:495-505. [PMID: 22692803 DOI: 10.1002/path.4063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 05/22/2012] [Accepted: 05/27/2012] [Indexed: 12/29/2022]
Abstract
We previously demonstrated that human osteosarcoma cells (SAOS-2) induce contact-dependent apoptosis in endothelium, and expected similar apoptosis in human gingival fibroblasts (h-GF) using SAOS-2 alkaline phosphatase (AP) to identify cells. However, h-GF apoptosis did not occur, despite reduction in AP-negative h-GF number (p < 0.01) and enhancement of this by h-GF TNFα pretreatment (p < 0.01). We suggest that TNFα-enhanced transfer of membrane AP from SAOS-2 to h-GF would explain these data. This idea was investigated using fluorescence prelabelled cells and confocal laser scanning microscopy. Co-cultures of membrane-labelled h-GF (marker-DiO) and SAOS-2 (marker-DiD) generated dual-labelled cells, primarily at the expense of single labelled h-GF (p < 0.001), suggesting predominant membrane transfer from SAOS-2 to h-GF. However, opposite directional transfer predominated when membrane labels were reversed; SAOS-2 further expressed green fluorescent protein (GFP) in cytoplasm and nuclei, and h-GF additionally bore nuclear label (Syto59) (p < 0.001). Cytoplasmic exchange was investigated using h-GF prelabelled with cytoplasmic DDAO-SE and nuclear Syto59, co-cultured with SAOS-2 expressing GFP in cytoplasm and nuclei, and predominant cytoplasmic marker transferred from h-GF to SAOS-2 (p < 0.05). Pretreating h-GF with TNFα increased exchange of membrane markers (p < 0.04) but did not affect either cell surface area profile or circularity. Dual-labelled cells had a morphological phenotype differing from SAOS-2 and h-GF (p < 0.001). Time-lapse microscopy revealed extensive migration of SAOS-2 and cell process contact with h-GF, with the appearance of SAOS-2 indulging in 'cellular sipping' from h-GF. Similar exchange of membrane was seen between h-GF and with other cell lines (melanoma MeIRMu, NM39, WMM175, MM200-B12; osteosarcoma U20S; ovarian carcinoma cells PE01, PE04 and COLO316), while cytoplasmic sharing was also seen in all cell lines other than U20S. We suggest that in some neoplasms, cellular sipping may contribute to phenotypic change and the generation of diverse tumour cell populations independent of genetic change, raising the possibility of a role in tumour progression.
Collapse
Affiliation(s)
- Manu S David
- Cellular and Molecular Pathology Research Unit, Department of Oral Pathology and Oral Medicine, University of Sydney, Westmead Centre for Oral Health, Westmead Hospital, Westmead, NSW, Australia
| | | | | | | | | | | | | |
Collapse
|
11
|
Phenotypic and proliferative modulation of human mesenchymal stem cells via crosstalk with endothelial cells. Stem Cell Res 2011; 7:186-97. [DOI: 10.1016/j.scr.2011.05.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 05/26/2011] [Accepted: 05/31/2011] [Indexed: 12/14/2022] Open
|
12
|
Lin RZ, Wang TP, Hung RJ, Chuang YJ, Chien CCM, Chang HY. Tumor-induced endothelial cell apoptosis: Roles of NAD(P)H oxidase-derived reactive oxygen species. J Cell Physiol 2011; 226:1750-62. [DOI: 10.1002/jcp.22504] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
13
|
Hsiao PN, Chang MC, Cheng WF, Chen CA, Lin HW, Hsieh CY, Sun WZ. Morphine induces apoptosis of human endothelial cells through nitric oxide and reactive oxygen species pathways. Toxicology 2008; 256:83-91. [PMID: 19070643 DOI: 10.1016/j.tox.2008.11.015] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 11/05/2008] [Accepted: 11/05/2008] [Indexed: 01/23/2023]
Abstract
Morphine has been widely used for pain management. Other than analgesia, it has effects on vascular endothelial cells, including angiogenesis and apoptosis. An in vitro model of human umbilical vein endothelial cells (HUVECs) was made to investigate the effects and comprehensive mechanisms of morphine on vascular endothelial cells. Morphine enhanced apoptosis of HUVECs, increased intracellular reactive oxygen species (ROS), and reduced mitochondrial membrane potentials (MMPs). It also induced the release of NO and activated NF-kappaB in HUVECs. Naloxone, the opioid receptor antagonist, could reverse cell apoptosis and ROS generation, NO production, and MMP loss. Expression levels of Bak and Bax, and the activation of caspases 3 and 7 in HUVECs significantly increased when treated with morphine. Inhibition of NO production by NO synthase inhibitor reduced morphine-induced apoptosis. Morphine could induce apoptosis of HUVECs through both the NO and ROS pathways. Thus, inhibiting NO or ROS may be a potential target in blocking morphine-induced apoptosis of endothelial cells.
Collapse
Affiliation(s)
- Po-Ni Hsiao
- Department of Anesthesiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
14
|
Merkle CJ, Torres BJ, Baruch JM, Stevens K, Munoz C, Schaeffer RC, Montgomery DW. In vitro age-related responses of endothelial cells to breast cancer cell addition. ACTA ACUST UNITED AC 2005; 29:518-27. [PMID: 16289385 DOI: 10.1016/j.cdp.2005.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2005] [Indexed: 11/26/2022]
Abstract
AIM The purpose of this study was to determine if the in vitro age of endothelial cells alters endothelial response(s) to breast cancer cells. METHOD After characterizing lower passage ("young"; passages 10-16) and higher passage ("old"; passages 30-36) bovine pulmonary artery endothelial cells (BPAECs), fluorescently labeled MCF-7 breast cancer cells were added to confluent monolayers of young and old BPAECs. RESULTS Transient gaps that peaked in size by 12 h and closed by 48h occurred between the young BPAECs, while large persistent gaps formed between the old BPAECs. Gap formation did not occur when 184A1 cells, a non-malignant mammary epithelial cell line, were added in place of MCF-7 cells, suggesting that the age-related responses of the endothelial cells to MCF-7 cell addition were specific to the tumor cell addition. Additionally, more MCF-7 cells migrated through old BPAEC monolayers, than young BPAEC monolayers, grown on Matrigel-coated filters. Finally, DNA fragmentation and fluorescent annexin-V binding assays suggested increased MCF-7 cell-induced apoptosis in older BPAECs, though results from a caspase-3 activation assay were equivocal. CONCLUSIONS In sum, our findings support the notion that aged endothelial cells are more susceptible to breast cancer-induced injury, perhaps due to increased apoptosis.
Collapse
Affiliation(s)
- Carrie J Merkle
- College of Nursing, The University of Arizona, Tucson, AZ 85721, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Chen Q, Stone PR, McCowan LME, Chamley LW. Interaction of Jar choriocarcinoma cells with endothelial cell monolayers. Placenta 2004; 26:617-25. [PMID: 16085041 DOI: 10.1016/j.placenta.2004.09.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2004] [Revised: 08/09/2004] [Accepted: 09/18/2004] [Indexed: 11/28/2022]
Abstract
During human pregnancy the uterine spiral arteries are invaded by placental trophoblasts which replace the endothelial cells that line the non-pregnant spiral arteries and transform these vessels into large-bore conduits enabling adequate perfusion of the placenta with maternal blood. Failure of this process may predispose to preeclampsia and fetal growth restriction [Brosens I, Robertson WB, Dixon HG. The physiological response of the vessels of the placental bed to normal pregnancy. Journal of Pathology and Bacteriology 1967;93:569-79; Khong TY, De Wolf F, Robertson WB, Brosens I. Inadequate maternal vascular response to placentation in pregnancies complicated by pre-eclampsia and by small-for-gestational age infants. British Journal of Obstetrics and Gynaecology 1986;93:1049-59]. There is a paucity of data on the role of maternal endothelial cells in this process. In this study we investigated the cellular interactions between trophoblast-derived Jar cells and endothelial cells (HUVECs and HMEC-1). The effect of coculturing Jar cells with endothelial cell monolayers was determined by confocal microscopy, DNA fragmentation assay and flow cytometry. We demonstrated that Jar cells migrate into focal areas in endothelial cell monolayers, where they induce endothelial cell death and, then phagocytose the dead endothelial cells. Our results suggest that endothelial cells may not simply be passive targets for invading trophoblasts during the remodeling of the spiral arteries.
Collapse
Affiliation(s)
- Q Chen
- Department of Obstetrics & Gynaecology, Faculty of Medical & Health Sciences, The University of Auckland, New Zealand.
| | | | | | | |
Collapse
|