1
|
Qu F, Brough SC, Michno W, Madubata CJ, Hartmann GG, Puno A, Drainas AP, Bhattacharya D, Tomasich E, Lee MC, Yang D, Kim J, Peiris-Pagès M, Simpson KL, Dive C, Preusser M, Toland A, Kong C, Das M, Winslow MM, Pasca AM, Sage J. Crosstalk between small-cell lung cancer cells and astrocytes mimics brain development to promote brain metastasis. Nat Cell Biol 2023; 25:1506-1519. [PMID: 37783795 PMCID: PMC11230587 DOI: 10.1038/s41556-023-01241-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/25/2023] [Indexed: 10/04/2023]
Abstract
Brain metastases represent an important clinical problem for patients with small-cell lung cancer (SCLC). However, the mechanisms underlying SCLC growth in the brain remain poorly understood. Here, using intracranial injections in mice and assembloids between SCLC aggregates and human cortical organoids in culture, we found that SCLC cells recruit reactive astrocytes to the tumour microenvironment. This crosstalk between SCLC cells and astrocytes drives the induction of gene expression programmes that are similar to those found during early brain development in neurons and astrocytes. Mechanistically, the brain development factor Reelin, secreted by SCLC cells, recruits astrocytes to brain metastases. These astrocytes in turn promote SCLC growth by secreting neuronal pro-survival factors such as SERPINE1. Thus, SCLC brain metastases grow by co-opting mechanisms involved in reciprocal neuron-astrocyte interactions during brain development. Targeting such developmental programmes activated in this cancer ecosystem may help prevent and treat brain metastases.
Collapse
Affiliation(s)
- Fangfei Qu
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Siqi C Brough
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Wojciech Michno
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Chioma J Madubata
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Griffin G Hartmann
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alyssa Puno
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexandros P Drainas
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Debadrita Bhattacharya
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Erwin Tomasich
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Myung Chang Lee
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Dian Yang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jun Kim
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Maria Peiris-Pagès
- Cancer Research UK Cancer Biomarker Centre, Manchester, UK
- Cancer Research UK Manchester Institute, Manchester, UK
| | - Kathryn L Simpson
- Cancer Research UK Cancer Biomarker Centre, Manchester, UK
- Cancer Research UK Manchester Institute, Manchester, UK
| | - Caroline Dive
- Cancer Research UK Cancer Biomarker Centre, Manchester, UK
- Cancer Research UK Manchester Institute, Manchester, UK
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Angus Toland
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Millie Das
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anca M Pasca
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Julien Sage
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
2
|
Chitosan IR806 dye-based polyelectrolyte complex nanoparticles with mitoxantrone combination for effective chemo-photothermal therapy of metastatic triple-negative breast cancer. Int J Biol Macromol 2022; 216:558-570. [PMID: 35809672 DOI: 10.1016/j.ijbiomac.2022.07.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/29/2022] [Accepted: 07/03/2022] [Indexed: 11/21/2022]
Abstract
Chemo-photothermal therapy is one of the emerging therapies for treating triple-negative breast cancer. In this study, we have used ionotropic gelation method to fabricate chitosan and IR806 dye-based polyelectrolyte complex (CIR-PEx) nanoparticles. These nano-complexes were in size range of 125 ± 20 nm. The complexation of IR 806 dye with chitosan improved photostability, photothermal transduction, and showed excellent biocompatibility. Cancer cells treated with CIR-PEx NPs enhanced intracellular uptake within 5 h of incubation and also displayed mitochondrial localization. With the combination of CIR-PEx NPs and a chemotherapeutic agent (i.e., mitoxantrone, MTX), a significant decline in cancer cell viability was observed in both 2D and 3D cell culture models. The chemo-photothermal effect of CIR-PEx NPs + MTX augmented apoptosis in cancer cells when irradiated with NIR light. Furthermore, when tested in the 4 T1-tumor model, the chemo-photothermal therapy showed a drastic decline in tumor volume and inhibited metastatic lung nodules. The localized hyperthermia caused by photothermal therapy reduced the primary tumor burden, and the chemotherapeutic activity of mitoxantrone further complemented by inhibiting the spread of cancer cells. The proposed chemo-photothermal therapy combination could be a promising strategy for treating triple-negative metastatic breast cancer.
Collapse
|
4
|
Li JM, Yang F, Li J, Yuan WQ, Wang H, Luo YQ. Reelin Promotes Cisplatin Resistance by Induction of Epithelial-Mesenchymal Transition via p38/GSK3β/Snail Signaling in Non-Small Cell Lung Cancer. Med Sci Monit 2020; 26:e925298. [PMID: 32764530 PMCID: PMC7433388 DOI: 10.12659/msm.925298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Emerging evidence suggests the involvement of Reelin in chemoresistance in various cancers. However, its function in cisplatin (DDP) sensitivity of non-small cell lung cancer (NSCLC) needs to be investigated. Material/Methods Reelin expression in cisplatin-sensitive A549 cells and cisplatin-resistant NSCLC (A549/DDP) cells was analyzed by western blot analysis. qRT-PCR, western blotting, immunofluorescence, CCK-8 assays, Annexin V/propidium iodide apoptosis assay, and Transwell migration assays were carried out to determine the function of Reelin on DDP resistance. Results Reelin was markedly increased in A549/DDP cells relative to A549 cells. Knockdown of Reelin enhanced DDP chemosensitivity of A549/DDP cells, whereas overexpression of Reelin enhanced DDP resistance of A549, H1299, and H460 cells. Reelin induced DDP resistance in NSCLC cells via facilitating epithelial-mesenchymal transition (EMT). Furthermore, Reelin modulated p38/GSK3β signal transduction and promoted Snail (EMT-associated transcription factor) expression. Suppression of p38/Snail reversed Reelin-induced EMT and resistance of NSCLC cells to DDP. Conclusions These data indicated that Reelin induces DDP resistance of NSCLC by regulation of the p38/GSK3β/Snail/EMT signaling pathway and provide evidence that Reelin suppression can be an effective strategy to suppress DDP resistance in NSCLC.
Collapse
Affiliation(s)
- Ji-Min Li
- Department of Laboratory Medicine, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Fang Yang
- Department of Laboratory Medicine, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Juan Li
- Department of Blood Transfusion, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Wei-Qi Yuan
- Department of Laboratory Medicine, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Hao Wang
- Department of Laboratory Medicine, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Yi-Qin Luo
- Department of Laboratory Medicine, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| |
Collapse
|
5
|
Li X, Fan W, Yao A, Song H, Ge Y, Yan M, Shan Y, Zhang C, Li P, Jia L. Downregulation of reelin predicts poor prognosis for glioma. Biomark Med 2020; 14:651-663. [PMID: 32613843 DOI: 10.2217/bmm-2019-0609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: In the present study, we studied the relationship between RELN and prognosis in glioma. Materials & methods: Expression profiles and methylation data of RELN were obtained from bioinformatic datasets. Correlations between RELN and clinicopathological features and overall survival were respectively assessed using chi-square test and Kaplan-Meier analysis. Results: RELN was downregulated in glioma, and its downregulation correlated well with glioma malignancy and overall survival. Meanwhile, hypermethylation of RELN was significantly correlated with low RELN expression. Additionally, gene set enrichment analysis demonstrated that low expression of RELN correlated with many key cancer pathways, possibly highlighting the importance of RELN in carcinogenesis of brain. Conclusion: RELN may serve as a potential prognostic marker and promising target molecule for new therapy of glioma.
Collapse
Affiliation(s)
- Xueli Li
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Wange Fan
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Anhui Yao
- Department of Neurosurgery, The General Hospital of PLA, Beijing, China.,Department of Neurosurgery, 988th Hospital of Chinese People's Liberation Army, Zhengzhou, Henan Province, PR China
| | - Huiling Song
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yunxiao Ge
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mengyao Yan
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yubo Shan
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chujie Zhang
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Pu Li
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Liyun Jia
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
6
|
Zhang W, Qian S, Yang G, Zhu L, Zhou B, Qu X, Yan Z, Liu R, Wang J. Establishment and characterization of McA-RH7777 cells using virus-mediated stable overexpression of enhanced green fluorescent protein. Exp Ther Med 2018; 16:3149-3154. [PMID: 30250518 DOI: 10.3892/etm.2018.6580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/29/2018] [Indexed: 12/31/2022] Open
Abstract
Hepatocellular carcinoma (HCC), the most common primary tumor of the liver, has a poor prognosis, rapid progression. The aim of the current study was to establish a stable lentiviral expression vector for enhanced green fluorescent protein (EGFP) and to evaluate biological characteristics on HCC growth and migration following transfection of HCC cells with EGFP. McA-RH7777 cells were transfected with EGFP overexpression lentiviral vector. Cell activity and mobility were monitored with a Cell-IQ Analyzer. Transwell assays were performed to detect invasiveness and flow cytometry was performed for cell cycle analysis. A subcutaneous tumor rat model was established to analyze the stability of fluorescent protein expression. The result suggested no significant differences between wild-type and EGFP-overexpressing McA-RH7777 cells with regards to cell proliferation, activity, mobility, invasiveness and cell cycle. Green fluorescence was detected over 108 days of culturing. The subcutaneous tumor rat model demonstrated that EGFP expression had no influence on tumor growth and long-term expression was stable. The stable EGFP expression of the HCC transplanted tumor rat model may share biological characteristics with human liver cancer. The model established in the current study may be suitable for various applications, including research focusing on liver cancer metastasis and recurrence, interventional therapy, imaging diagnosis and drug screenings.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Sheng Qian
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Guowei Yang
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Liang Zhu
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Bo Zhou
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Xudong Qu
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Zhiping Yan
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Rong Liu
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Jianhua Wang
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|