101
|
Cao H, Zhang P, Yu H, Xi J. Extracellular Vesicles-Encapsulated miR-153-3p Potentiate the Survival and Invasion of Lung Adenocarcinoma. Mol Cells 2022; 45:376-387. [PMID: 35611687 PMCID: PMC9200658 DOI: 10.14348/molcells.2022.2221] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/29/2021] [Accepted: 01/14/2022] [Indexed: 11/27/2022] Open
Abstract
Extracellular vesicles (EVs) play an essential role in the communication between cells and the tumor micro-environment. However, the effect of tumor-derived EVs on the growth and metastasis of lung adenocarcinoma (LUAD) remains to be explored. This study aimed to elucidate the role of miR-153-3p-EVs in the invasion and migration capabilities of LUAD cells and explore its mechanism through in vivo and in vitro experiments. We found that miR-153-3p was specifically and highly expressed in LUAD and its secreted EVs. Furthermore, the expression of BANCR was negatively regulated by miR-153-3p and identified as a target gene of miR-153-3p using luciferase reporter assays. Through further investigation, we found that the downregulation of BANCR activates the PI3K/AKT pathway and accelerates the process of epithelial-mesenchymal transition (EMT), which ultimately leads to the aggravation of LUAD. The orthotopic xenograft mouse model was established to illustrate the effect of miR-153-3p-EVs on LUAD. Animal studies showed that miR-153-3p-EVs accelerated tumor growth in mice. Besides, we found that miR-153-3p-EVs could damage the respiratory ability of mice and produce a mass of inflammatory cells around the lung tissue of mice. Nevertheless, antagomir-153-3p treatment could inhibit the deterioration of respiratory function and inhibit the growth of lung tumors in mice. In conclusion, our study reveals the potential molecular mechanism of miR-153-3p-EVs in the development of LUAD and provides a potential strategy for the treatment of LUAD.
Collapse
Affiliation(s)
- Hongli Cao
- Department of Respiratory, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Ping Zhang
- Department of Respiratory, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Hong Yu
- Department of Respiratory, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Jianing Xi
- Department of Respiratory, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| |
Collapse
|
102
|
Zhang H, Cai YH, Ding Y, Zhang G, Liu Y, Sun J, Yang Y, Zhan Z, Iliuk A, Gu Z, Gu Y, Tao WA. Proteomics, Phosphoproteomics and Mirna Analysis of Circulating Extracellular Vesicles through Automated and High-Throughput Isolation. Cells 2022; 11:2070. [PMID: 35805153 PMCID: PMC9265938 DOI: 10.3390/cells11132070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/28/2022] Open
Abstract
Extracellular vesicles (EVs) play an important role in the diagnosis and treatment of diseases because of their rich molecular contents involved in intercellular communication, regulation, and other functions. With increasing efforts to move the field of EVs to clinical applications, the lack of a practical EV isolation method from circulating biofluids with high throughput and good reproducibility has become one of the biggest barriers. Here, we introduce a magnetic bead-based EV enrichment approach (EVrich) for automated and high-throughput processing of urine samples. Parallel enrichments can be performed in 96-well plates for downstream cargo analysis, including EV characterization, miRNA, proteomics, and phosphoproteomics analysis. We applied the instrument to a cohort of clinical urine samples to achieve reproducible identification of an average of 17,000 unique EV peptides and an average of 2800 EV proteins in each 1 mL urine sample. Quantitative phosphoproteomics revealed 186 unique phosphopeptides corresponding to 48 proteins that were significantly elevated in prostate cancer patients. Among them, multiple phosphoproteins were previously reported to associate with prostate cancer. Together, EVrich represents a universal, scalable, and simple platform for EV isolation, enabling downstream EV cargo analyses for a broad range of research and clinical applications.
Collapse
Affiliation(s)
- Hao Zhang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
- EVLiXiR Biotech, Nanjing 210032, China
| | - Yu-Han Cai
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
| | - Yajie Ding
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
| | - Guiyuan Zhang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
- Bell Mountain Molecular MedTech Institute, Nanjing 210032, China
| | - Yufeng Liu
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
- Bell Mountain Molecular MedTech Institute, Nanjing 210032, China
| | - Jie Sun
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
| | - Yuchen Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China; (Y.Y.); (Y.G.)
| | - Zhen Zhan
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, IN 47906, USA;
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China; (Y.Y.); (Y.G.)
| | - W. Andy Tao
- Tymora Analytical Operations, West Lafayette, IN 47906, USA;
- Department of Chemistry and Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
103
|
Li R, Zhou J, Wu X, Li H, Pu Y, Liu N, Han Z, Zhou L, Wang Y, Zhu H, Yang L, Li Q, Ji Q. Jianpi Jiedu Recipe inhibits colorectal cancer liver metastasis via regulating ITGBL1-rich extracellular vesicles mediated activation of cancer-associated fibroblasts. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154082. [PMID: 35381565 DOI: 10.1016/j.phymed.2022.154082] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/05/2022] [Accepted: 03/26/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Extracellular vesicles (EVs) contribute greatly to the formation of pre-metastatic niche and tumor metastasis. Our previous study has revealed that tumor-derived ITGBL1 (integrin beta- like 1)-rich EVs activate fibroblasts through the NF-κB signaling to promote colorectal cancer (CRC) metastasis. Targeting ITGBL1-loaded EVs may be a new and effective therapy for treating CRC metastasis. Simultaneously, our preliminary clinical trial has demonstrated that Jianpi Jiedu Recipe (JPJDR) was an ideal alternative traditional Chinese medicine for the prevention and treatment of CRC metastasis. However, the underlying mechanism of JPJDR in the prevention of CRC metastasis is not clear. In this study, we will investigate the regulatory effect of JPJDR on ITGBL1 levels in CRC-derived EVs, and to detect how JPJDR regulate ITGBL1-rich EVs mediated activation of fibroblasts to inhibit CRC metastasis. METHODS EVs derived from CRC cells with/without JPJDR treatment were obtained by ultracentrifugation, following by characterization with electron microscopy, LM10 nanoparticle characterization system and western blot. The migration and growth of CRC cells were tested by transwell assay, wound healing assay and colony formation assay. The effect of JPJDR on the fibroblasts-activation associated inflammatory factors including IL-6, IL-8 and α-SMA was detected by real-time PCR. The levels of IL-6, IL-8 and α-SMA in the cell culture supernatant were detected by ELISA. The protein expressions of TNFAIP3, ITGBL1, p-NF-κB, IκBα and β-actin were detected by western blot. Liver metastasis model in mice was established by injecting MC38 single cell suspension into the spleen of mice to observe the effect of JPJDR on CRC liver metastasis. Immunohistochemistry were applied to detect the expression of ITGBL1 and TNFAIP3 in the liver metastatic tissues. Tissue immunofluorescence detection was performed to observe the regulatory effect of JPJDR on the ITGBL1-NF-κB signaling pathway. Cancer-associated fibroblasts (CAFs) in the liver metastatic tissues were sorted and characterized by platelet-derived growth factor receptor β (PDGFRβ) with flow cytometry, following by the detection of inflammatory factors including IL-6, IL-8 and α-SMA using real-time PCR. RESULTS JPJDR reduced the ITGBL1 levels in CRC cells-derived EVs. JPJDR inhibited the migration and growth of CRC cells via regulating ITGBL1-rich EVs mediated fibroblasts activity. Mechanically, JPJDR decreased fibroblasts activation by regulating ITGBL1-rich EVs mediated TNFAIP3-NF-κB signaling. Further in vivo experiments demonstrated that JPJDR reduced CRC liver metastasis by regulating ITGBL1-rich EVs secretion from CRC and blocked the fibroblasts activation by regulating ITGBL1-TNFAIP3- NF-κB signaling. CONCLUSION Our research demonstrated that JPJDR preventd CRC liver metastasis via down-regulating CRC-derived ITGBL1-loaded EVs mediated activation of CAFs, providing the experimental evidence for the clinical application of JPJDR in the prevention and treatment of CRC metastasis. More importantly, our study confirmed the great benefits of therapeutic targeting the EVs-mediated metastasis and warranted future clinical validation.
Collapse
Affiliation(s)
- Ruixiao Li
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zhou
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinnan Wu
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haoze Li
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunzhou Pu
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ningning Liu
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhifen Han
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihong Zhou
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huirong Zhu
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Yang
- Department of Oncology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qi Li
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qing Ji
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
104
|
Wang Z, Fu L, Zhang J, Ge Y, Guo C, Wang R, Deng M, Wang Q, Wang Z. A comprehensive analysis of potential gastric cancer prognostic biomarker ITGBL1 associated with immune infiltration and epithelial-mesenchymal transition. Biomed Eng Online 2022; 21:30. [PMID: 35596183 PMCID: PMC9123716 DOI: 10.1186/s12938-022-00998-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 04/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background Integrin, beta-like 1 (ITGBL1) is involved in a variety of human malignancies. However, the information on the involvement of ITGBL1 in gastric carcinoma (GC) is limited. Hence, this study aimed further to explore the functions and mechanisms of ITGBL1 in GC. Methods First, multiple bioinformatics databases, including Oncomine, Tumor Immune Estimation Resource, UALCAN, and Kaplan–Meier Plotter, were used to predict the expression level and prognostic value of ITGBL1, as well as its association with immune infiltration and epithelial–mesenchymal transition (EMT) in GC. Quantitative reverse transcription–polymerase chain reaction and immunohistochemical analysis were used to detect the expression of ITGBL1 in both GC tissues and cells. Then, targeted silencing of ITGBL1 in GC cells was further used to examine the biological functions of ITGBL1. Results These databases revealed that ITGBL1 was overexpressed and affected the overall survival in GC. Besides, the expression of ITGBL1 positively correlated with immune-infiltrating cells and EMT-related markers. Subsequently, molecular biology experiments verified these predictions. In GC tissues and cells, ITGBL1 was notably overexpressed. Loss-of-function studies showed that the knockdown of ITGBL1 significantly suppressed migration and invasion but promoted apoptosis in MGC803 GC cells. Furthermore, the inhibition of ITGBL1 resulted in remarkably increased protein expression levels of cadherin 1, while the expression of Vimentin, Snail, and transforming growth factor-β1 was downregulated, indicating the initiation and progression of GC caused by ITGBL1 partly via inducing EMT. Conclusions To sum up, the findings indicated that ITGBL1 acted as a valuable oncogenic factor in GC.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Gastroenterology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Putuo District, Shanghai, 200065, People's Republic of China
| | - Liu Fu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University, Shanghai, 200060, People's Republic of China
| | - Junjie Zhang
- Department of Gastroenterology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Putuo District, Shanghai, 200065, People's Republic of China
| | - Yanli Ge
- Department of Gastroenterology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Putuo District, Shanghai, 200065, People's Republic of China
| | - Cheng Guo
- Department of Gastroenterology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Putuo District, Shanghai, 200065, People's Republic of China
| | - Rui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Putuo District, Shanghai, 200065, People's Republic of China
| | - Min Deng
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People's Republic of China
| | - Qizhi Wang
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, People's Republic of China
| | - Zhirong Wang
- Department of Gastroenterology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Putuo District, Shanghai, 200065, People's Republic of China.
| |
Collapse
|
105
|
Rodrigues-Junior DM, Tsirigoti C, Lim SK, Heldin CH, Moustakas A. Extracellular Vesicles and Transforming Growth Factor β Signaling in Cancer. Front Cell Dev Biol 2022; 10:849938. [PMID: 35493080 PMCID: PMC9043557 DOI: 10.3389/fcell.2022.849938] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Complexity in mechanisms that drive cancer development and progression is exemplified by the transforming growth factor β (TGF-β) signaling pathway, which suppresses early-stage hyperplasia, yet assists aggressive tumors to achieve metastasis. Of note, several molecules, including mRNAs, non-coding RNAs, and proteins known to be associated with the TGF-β pathway have been reported as constituents in the cargo of extracellular vesicles (EVs). EVs are secreted vesicles delimited by a lipid bilayer and play critical functions in intercellular communication, including regulation of the tumor microenvironment and cancer development. Thus, this review aims at summarizing the impact of EVs on TGF-β signaling by focusing on mechanisms by which EV cargo can influence tumorigenesis, metastatic spread, immune evasion and response to anti-cancer treatment. Moreover, we emphasize the potential of TGF-β-related molecules present in circulating EVs as useful biomarkers of prognosis, diagnosis, and prediction of response to treatment in cancer patients.
Collapse
Affiliation(s)
| | - Chrysoula Tsirigoti
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology (A*-STAR), Singapore, Singapore
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- *Correspondence: Aristidis Moustakas,
| |
Collapse
|
106
|
Li Q, Chen JX, Wu Y, Lv LL, Ying HF, Zhu WH, Xu JY, Ruan M, Guo Y, Zhu WR, Zheng L. The mechanism of FZXJJZ decoction suppresses colorectal liver metastasis via the VDR/TGF-β/Snail1 signaling pathways based on network pharmacology-TCGA data-transcriptomics analysis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 287:114904. [PMID: 34952191 DOI: 10.1016/j.jep.2021.114904] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fuzheng Xiaojijinzhan (FZXJJZF) decoction is an effective prescription for treating colorectal cancer liver metastasis (LMCRC). AIM OF THE STUDY To elucidate the pharmacological mechanism of the FZXJJZF decoction therapy on LMCRC. MATERIALS AND METHODS Firstly, a network pharmacological approach was used to characterize the underlying targets of FZXJJZF on LMCRC. Secondly, LMCRC-related genes are obtained from the public database TCGA, and those genes are further screened and clustered through Mfuzz, an R package tool. Then, targets of FZXJJZF predicted by network pharmacology were overlapped with LMCRC related genes screened by Mfuzz. Meanwhile, FZJZXJF intervened in LMCRC model,epithelial-to-mesenchymal transition (EMT), and migration and invasion of HCT-116 cells. Thirdly, the transcriptomics data of FZJZXJF inhibited HCT-116 cells of EMT cells were overlapped with EMT database data to narrow the possible range of targets. Based on this, the potential targets and signal pathways of FZJZXJF were speculated by combining the transcriptomics data with the targets from network pharmacology-TCGA. Finally, the anti-cancer mechanism of FZXJJZF on LMCRC was verified in vitro by Real-Time PCR and Western Blot in vitro. RESULTS By network pharmacological analysis, 282 ingredients and 429 potential targets of FZXJJZF were predicted. The 9268 LMCRC-related genes in the TCGA database were classified into 10 clusters by the Mfuzz. The two clustering genes with the most similar clustering trends were overlapped with 429 potential targets, and 32 genes were found, such as CD34, TRPV3, PGR, VDR, etc. In vivo experiments, FZJZXJF inhibited the tumor size in LMCRC models, and the EMT, migration, and invasion of HCT-116 also be inhibited. Intersecting transcriptomics dates with 32 target genes, it is speculated that the VDR-TGF-β signaling pathway may be an effective mechanism of FZXJJZF. Additionally, it is shown that FZXJJZF up-regulated the expression levels of VDR and E-cadherin and down-regulated the expression levels of TGF-β and Snail1 in vitro. These results confirmed that FZXJJZF plays an effective role in LMCRC mainly by inhibiting EMT phenotype via the VDR-TGF-β signaling pathway. CONCLUSIONS Collectively, this study reveals the anti-LMCRC effect of FZXJJZF and its potential therapeutic mechanism from the perspective of potential targets and potential pathways.
Collapse
Affiliation(s)
- Qiong Li
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| | - Jing-Xian Chen
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| | - Yuan Wu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| | - Ling-Ling Lv
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| | - Hai-Feng Ying
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| | - Wen-Hua Zhu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| | - Jia-Yue Xu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| | - Ming Ruan
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| | - Yuanbiao Guo
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| | - Wei-Rong Zhu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, 200025, China.
| |
Collapse
|
107
|
Jiang J, Li J, Zhou X, Zhao X, Huang B, Qin Y. Exosomes Regulate the Epithelial-Mesenchymal Transition in Cancer. Front Oncol 2022; 12:864980. [PMID: 35359397 PMCID: PMC8964004 DOI: 10.3389/fonc.2022.864980] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Exosomes are important mediators of intercellular communication and participate in complex biological processes by transferring a variety of bioactive molecules between cells. Epithelial–mesenchymal transition (EMT) is a process in which the cell phenotype changes from epithelioid to mesenchymal-like. EMT is also an important process for cancer cells by which they acquire invasive and metastatic capabilities, which aggravates the degree of tumor malignancy. Numerous studies have demonstrated that exosomes encapsulate various components, such as microRNAs and proteins, and transfer information between tumor cells or between tumor cells and the tumor microenvironment, thereby regulating the EMT process. Exosomes can also be used for cancer diagnosis and treatment or as a drug delivery platform. Thus, they can be used as a therapeutic tool to control the occurrence of EMT and affect cancer progression. In this review, we summarize the latest research advancements in the regulation of the EMT process in tumor cells by the contents of exosomes. Furthermore, we discuss the potential and challenges of using exosomes as a tool for cancer treatment.
Collapse
Affiliation(s)
- Jingwen Jiang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiayu Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiumei Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xueqin Zhao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
108
|
Li J, Chen D, Shen M. Tumor Microenvironment Shapes Colorectal Cancer Progression, Metastasis, and Treatment Responses. Front Med (Lausanne) 2022; 9:869010. [PMID: 35402443 PMCID: PMC8984105 DOI: 10.3389/fmed.2022.869010] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most devastating diseases that accounts for numerous deaths worldwide. Tumor cell-autonomous pathways, such as the oncogenic signaling activation, significantly contribute to CRC progression and metastasis. Recent accumulating evidence suggests that the CRC microenvironment also profoundly promotes or represses this process. As the roles of the tumor microenvironment (TME) in CRC progression and metastasis is gradually uncovered, the importance of these non-cell-autonomous signaling pathways is appreciated. However, we are still at the beginning of this TME function exploring process. In this review, we summarize the current understanding of the TME in CRC progression and metastasis by focusing on the gut microbiota and host cellular and non-cellular components. We also briefly discuss TME-remodeling therapies in CRC.
Collapse
Affiliation(s)
- Jun Li
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Dawei Chen
- Wayne State University School of Medicine, Detroit, MI, United States
| | - Minhong Shen
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Oncology, Wayne State University School of Medicine and Tumor Biology and Microenvironment Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, United States
- *Correspondence: Minhong Shen,
| |
Collapse
|
109
|
Bergonzini C, Kroese K, Zweemer AJM, Danen EHJ. Targeting Integrins for Cancer Therapy - Disappointments and Opportunities. Front Cell Dev Biol 2022; 10:863850. [PMID: 35356286 PMCID: PMC8959606 DOI: 10.3389/fcell.2022.863850] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/16/2022] [Indexed: 12/29/2022] Open
Abstract
Integrins mediate adhesive interactions between cells and their environment, including neighboring cells and extracellular matrix (ECM). These heterodimeric transmembrane receptors bind extracellular ligands with their globular head domains and connect to the cytoskeleton through multi-protein interactions at their cytoplasmic tails. Integrin containing cell–matrix adhesions are dynamic force-responsive protein complexes that allow bidirectional mechanical coupling of cells with their environment. This allows cells to sense and modulate tissue mechanics and regulates intracellular signaling impacting on cell faith, survival, proliferation, and differentiation programs. Dysregulation of these functions has been extensively reported in cancer and associated with tumor growth, invasion, angiogenesis, metastasis, and therapy resistance. This central role in multiple hallmarks of cancer and their localization on the cell surface makes integrins attractive targets for cancer therapy. However, despite a wealth of highly encouraging preclinical data, targeting integrin adhesion complexes in clinical trials has thus far failed to meet expectations. Contributing factors to therapeutic failure are 1) variable integrin expression, 2) redundancy in integrin function, 3) distinct roles of integrins at various disease stages, and 4) sequestering of therapeutics by integrin-containing tumor-derived extracellular vesicles. Despite disappointing clinical results, new promising approaches are being investigated that highlight the potential of integrins as targets or prognostic biomarkers. Improvement of therapeutic delivery at the tumor site via integrin binding ligands is emerging as another successful approach that may enhance both efficacy and safety of conventional therapeutics. In this review we provide an overview of recent encouraging preclinical findings, we discuss the apparent disagreement between preclinical and clinical results, and we consider new opportunities to exploit the potential of integrin adhesion complexes as targets for cancer therapy.
Collapse
|
110
|
Tang H, Zhou X, Zhao X, Luo X, Luo T, Chen Y, Liang W, Jiang E, Liu K, Shao Z, Shang Z. HSP90/IKK‐rich small extracellular vesicles activate pro‐angiogenic melanoma‐associated fibroblasts via NF‐κB/CXCL1 axis. Cancer Sci 2022; 113:1168-1181. [PMID: 35043517 PMCID: PMC8990732 DOI: 10.1111/cas.15271] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 11/30/2022] Open
Abstract
Hypoxia is a main feature of most solid tumors, but how melanoma cells under hypoxic conditions exploit tumor microenvironment (TME) to facilitate tumor progression remains poorly understood. In this study, we found that hypoxic melanoma‐derived small extracellular vesicles (sEVs) could improve the proangiogenic capability of cancer‐associated fibroblasts (CAFs). This improvement was due to the activation of the IKK/IκB/NF‐κB signaling pathway and upregulation of CXCL1 expression and secretion in CAFs. By proteomic analysis, we verified that hypoxia could promote enrichment of chaperone HSP90 and client protein phosphorylated IKKα/β (p‐IKKα/β) in melanoma‐derived sEVs. Delivery of the HSP90/p‐IKKα/β complex by sEVs could activate the IKK/IκB/NF‐κB/CXCL1 axis in CAFs and promote angiogenesis in vitro and in vivo. Taken together, these findings deepen the understanding of hypoxic response in melanoma progression and provide potential targets for melanoma treatment.
Collapse
Affiliation(s)
- Hokeung Tang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory for Oral Biomedicine Ministry of Education School and Hospital of Stomatology Wuhan University Wuhan China
| | - Xiaocheng Zhou
- Department of Oral and Maxillofacial Surgery School and Hospital of Stomatology Wuhan University Wuhan China
| | - Xiaoping Zhao
- Center of Stomatology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xinyue Luo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory for Oral Biomedicine Ministry of Education School and Hospital of Stomatology Wuhan University Wuhan China
| | - Tingting Luo
- Shenzhen PKU‐HKUST Medical Center (Peking University Shenzhen Hospital) Shenzhen China
| | - Yang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory for Oral Biomedicine Ministry of Education School and Hospital of Stomatology Wuhan University Wuhan China
| | - Weilian Liang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) & Key Laboratory for Oral Biomedicine Ministry of Education School and Hospital of Stomatology Wuhan University Wuhan China
| | - Erhui Jiang
- Department of Oral and Maxillofacial‐Head and Neck Oncology School and Hospital of Stomatology Wuhan University Wuhan China
| | - Ke Liu
- Department of Oral and Maxillofacial‐Head and Neck Oncology School and Hospital of Stomatology Wuhan University Wuhan China
| | - Zhe Shao
- Department of Oral and Maxillofacial‐Head and Neck Oncology School and Hospital of Stomatology Wuhan University Wuhan China
| | - Zhengjun Shang
- Department of Oral and Maxillofacial‐Head and Neck Oncology School and Hospital of Stomatology Wuhan University Wuhan China
| |
Collapse
|
111
|
Dong P, Wang Y, Liu Y, Zhu C, Lin J, Qian R, Hua L, Lu C. BMAL1 induces colorectal cancer metastasis by stimulating exosome secretion. Mol Biol Rep 2022; 49:373-384. [PMID: 34727291 DOI: 10.1007/s11033-021-06883-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/22/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND To adapt to daily changes in the external environment, organisms have developed circadian rhythm systems with a period of approximately 24 h. Many studies have reported that both circadian rhythms and exosomes play important roles in the development and metastasis of tumors. However, whether circadian clock genes can affect the progression of tumors by regulating exosomes remains unclear. METHODS AND RESULTS In this study, we isolated exosomes from the supernatant of human colorectal cancer (CRC) cells, including SW480, SW620, and HCT116 cells, by differential centrifugation and characterized exosomes by transmission electron microscopy, nanoparticle tracking analysis, and Western blot analysis. Then, we found that exosomes derived from SW480, SW620 and HCT116 cells could promote the migration of HCT116 and human umbilical vein endothelial cells. Exosomes derived from SW620 cells showed increased stimulating effects when we increased the expression of BMAL1, a core circadian protein. In contrast, exosomes derived from SW480 and HCT116 cells showed decreased stimulating effects when we knocked down the expression of BMAL1. Furthermore, we discovered that BMAL1 promotes the release of exosomes by HCT116 and SW620 cells. In addition, by luciferase assay, we confirmed that BMAL1 transcriptionally regulates the expression of Rab27a, a key molecule related to the secretion of exosomes. CONCLUSIONS Our data reveal a new mechanism by which BMAL1 induces CRC metastasis by stimulating exosome secretion. This finding may help further clarify the role of circadian rhythm in the progression of CRC.
Collapse
Affiliation(s)
- Pengjuan Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yaping Wang
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yutong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chunting Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jiaxin Lin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Ruizhe Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Luchun Hua
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Chao Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
112
|
LncRNA functional annotation with improved false discovery rate achieved by disease associations. Comput Struct Biotechnol J 2022; 20:322-332. [PMID: 35035785 PMCID: PMC8724965 DOI: 10.1016/j.csbj.2021.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 12/11/2022] Open
Abstract
The long non‐coding RNAs (lncRNAs) play critical roles in various biological processes and are associated with many diseases. Functional annotation of lncRNAs in diseases attracts great attention in understanding their etiology. However, the traditional co-expression-based analysis usually produces a significant number of false positive function assignments. It is thus crucial to develop a new approach to obtain lower false discovery rate for functional annotation of lncRNAs. Here, a novel strategy termed DAnet which combining disease associations with cis-regulatory network between lncRNAs and neighboring protein-coding genes was developed, and the performance of DAnet was systematically compared with that of the traditional differential expression-based approach. Based on a gold standard analysis of the experimentally validated lncRNAs, the proposed strategy was found to perform better in identifying the experimentally validated lncRNAs compared with the other method. Moreover, the majority of biological pathways (40%∼100%) identified by DAnet were reported to be associated with the studied diseases. In sum, the DAnet is expected to be used to identify the function of specific lncRNAs in a particular disease or multiple diseases.
Collapse
|
113
|
Forder A, Hsing CY, Trejo Vazquez J, Garnis C. Emerging Role of Extracellular Vesicles and Cellular Communication in Metastasis. Cells 2021; 10:cells10123429. [PMID: 34943937 PMCID: PMC8700460 DOI: 10.3390/cells10123429] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/15/2022] Open
Abstract
Communication between cancer cells and the surrounding stromal cells of the tumor microenvironment (TME) plays a key role in promoting metastasis, which is the major cause of cancer death. Small membrane-bound particles called extracellular vesicles (EVs) are released from both cancer and stromal cells and have a key role in mediating this communication through transport of cargo such as various RNA species (mRNA, miRNA, lncRNA), proteins, and lipids. Tumor-secreted EVs have been observed to induce a pro-tumorigenic phenotype in non-malignant cells of the stroma, including fibroblasts, endothelial cells, and local immune cells. These cancer-associated cells then drive metastasis by mechanisms such as increasing the invasiveness of cancer cells, facilitating angiogenesis, and promoting the formation of the pre-metastatic niche. This review will cover the role of EV-mediated signaling in the TME during metastasis and highlight the therapeutic potential of targeting these pathways to develop biomarkers and novel treatment strategies.
Collapse
Affiliation(s)
- Aisling Forder
- Department of Integrative Oncology, British Cancer Research Center, Vancouver, BC V5Z 1L3, Canada; (A.F.); (C.-Y.H.); (J.T.V.)
| | - Chi-Yun Hsing
- Department of Integrative Oncology, British Cancer Research Center, Vancouver, BC V5Z 1L3, Canada; (A.F.); (C.-Y.H.); (J.T.V.)
| | - Jessica Trejo Vazquez
- Department of Integrative Oncology, British Cancer Research Center, Vancouver, BC V5Z 1L3, Canada; (A.F.); (C.-Y.H.); (J.T.V.)
| | - Cathie Garnis
- Department of Integrative Oncology, British Cancer Research Center, Vancouver, BC V5Z 1L3, Canada; (A.F.); (C.-Y.H.); (J.T.V.)
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Correspondence:
| |
Collapse
|
114
|
Lu Z, Gao Y. Screening differentially expressed genes between endometriosis and ovarian cancer to find new biomarkers for endometriosis. Ann Med 2021; 53:1377-1389. [PMID: 34409913 PMCID: PMC8381947 DOI: 10.1080/07853890.2021.1966087] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/04/2021] [Indexed: 11/18/2022] Open
Abstract
AIM Endometriosis is one of the most common reproductive system diseases, but the mechanisms of disease progression are still unclear. Due to its high recurrence rate, searching for potential therapeutic biomarkers involved in the pathogenesis of endometriosis is an urgent issue. METHODS Due to the similarities between endometriosis and ovarian cancer, four endometriosis datasets and one ovarian cancer dataset were downloaded from Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified, followed by gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and protein-protein interaction (PPI) analyses. Then, we validated gene expression and performed survival analysis with ovarian serous cystadenocarcinoma (OV) datasets in TCGA/GTEx database, and searched for potential drugs in the Drug-Gene Interaction Database. Finally, we explored the miRNAs of key genes to find biomarkers associated with the recurrence of endometriosis. RESULTS In total, 104 DEGs were identified in the endometriosis datasets, and the main enriched GO functions included cell adhesion, extracellular exosome and actin binding. Fifty DEGs were identified between endometriosis and ovarian cancer datasets including 11 consistently regulated genes, and nine DEGs with significant expression in TCGA/GTEx. Only IGHM had both significant expression and an association with survival, three module DEGs and two significantly expressed DEGs had drug associations, and 10 DEGs had druggability. CONCLUSIONS ITGA7, ITGBL1 and SORBS1 may help us understand the invasive nature of endometriosis, and IGHM might be related to recurrence; moreover, these genes all may be potential therapeutic targets.KEY MESSAGEThis manuscript used a bioinformatics approach to find target genes for the treatment of endometriosis.This manuscript used a new approach to find target genes by drawing on common characteristics between ovarian cancer and endometriosis.We screened relevant therapeutic agents for target genes in the drug database, and performed histological validation of target genes with both expression and survival analysis difference in cancer databases.
Collapse
Affiliation(s)
- Zhenzhen Lu
- Department of Gynaecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Gao
- Department of Gynaecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
115
|
Extracellular Vesicles Tropism: A Comparative Study between Passive Innate Tropism and the Active Engineered Targeting Capability of Lymphocyte-Derived EVs. MEMBRANES 2021; 11:membranes11110886. [PMID: 34832115 PMCID: PMC8617986 DOI: 10.3390/membranes11110886] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022]
Abstract
Cellular communications take place thanks to a well-connected network of chemical–physical signals, biomolecules, growth factors, and vesicular messengers that travel inside or between cells. A deep knowledge of the extracellular vesicle (EV) system allows for a better understanding of the whole series of phenomena responsible for cell proliferation and death. To this purpose, here, a thorough immuno-phenotypic characterization of B-cell EV membranes is presented. Furthermore, the cellular membrane of B lymphocytes, Burkitt lymphoma, and human myeloid leukemic cells were characterized through cytofluorimetry assays and fluorescent microscopy analysis. Through cytotoxicity and internalization tests, the tropism of B lymphocyte-derived EVs was investigated toward the parental cell line and two different cancer cell lines. In this study, an innate capability of passive targeting of the native EVs was distinguished from the active targeting capability of monoclonal antibody-engineered EVs, able to selectively drive the vesicles, enhancing their internalization into the target cancer cells. In particular, the specific targeting ability of anti-CD20 engineered EVs towards Daudi cells, highly expressing CD20 marker on their cell membrane, was proved, while almost no internalization events were observed in HL60 cells, since they did not express an appreciable amount of the CD20 marker on their plasma membranes.
Collapse
|
116
|
Chai N, Xiong Y, Zhang Y, Cheng Y, Shi W, Yao Y, Sui H, Zhu H. YYFZBJS inhibits colorectal tumorigenesis by remodeling gut microbiota and influence on M2 macrophage polarization in vivo and in vitro. Am J Cancer Res 2021; 11:5338-5357. [PMID: 34873464 PMCID: PMC8640793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/07/2021] [Indexed: 06/13/2023] Open
Abstract
Our previous studies indicated that the extract of Yi-Yi-Fu-Zi-Bai-Jiang-San (YYFZBJS) had potent anticancer activities by significantly inhibiting intestinal tumor development in ApcMin/+ mice. However, knowledge regarding the mechanism and effect of YYFZBJS in the prevention of colorectal cancer is limited. In this study, we aim to investigate the preventive effects of YYFZBJS in enterotoxigenic Bacteroides fragilis (ETBF)-colonized mice with azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced tumorigenesis. First, the colonic tissues of the AOM/DSS mouse models were collected for biomedical analysis, and gut microbiota profiling was detected post YYFZBJS treatment using a 16S rRNA gene sequencing. Then, antibiotic solution (Abx) mice were acclimated with AOM/DSS treatment and then fed with ETBF with or without YYFZBJS for three cycles. As expected, the intragastric administration of YYFZBJS in the AOM/DSS mouse model significantly decreased the tumor load, the severity of disease activity index (DAI) scores, and the level of M2 macrophage markers such as CD206, Arg-1 and IL-10. Notably, the reverse of polarized macrophages induced by YYFZBJS could suppress CRC cell proliferation and infiltration, as demonstrated by the decrease of some tumor proliferation-related proteins in a dose-dependent manner. Importantly, ETBF dysbiosis can contribute to colon tumor development by stimulating p-STAT3 mediated M2 macrophages polarization to promote chronic inflammation and adenoma malignant transformation, which YYFZBJS can effectively limit. Altogether, we demonstrate that ETBF dysbiosis may contribute to M2 macrophages-promoted colon carcinogenesis and progression of CRC cells, while YYFZBJS could be a promising protective agent against ETBF-mediated colorectal cancer.
Collapse
Affiliation(s)
- Ni Chai
- Oncology Department, Yueyang Hospital of Integrated of Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese MedicineShanghai 200437, China
| | - Yibai Xiong
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical SciencesBeijing 100700, China
| | - Yuli Zhang
- Medical Experiment Center, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai 201803, China
- Department of Traditional Chinese Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai 201803, China
| | - Yuelei Cheng
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Wenfei Shi
- Oncology Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese MedicineShanghai 200271, China
| | - Yiqing Yao
- Oncology Department, Yueyang Hospital of Integrated of Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese MedicineShanghai 200437, China
| | - Hua Sui
- Medical Experiment Center, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai 201803, China
| | - Huirong Zhu
- Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| |
Collapse
|
117
|
Dang Y, Zhang S, Wang Y, Zhao G, Chen C, Jiang W. State-of-the-Art: Exosomes in Colorectal Cancer. Curr Cancer Drug Targets 2021; 22:2-17. [PMID: 34758717 DOI: 10.2174/1568009621666211110094442] [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: 06/14/2021] [Revised: 08/06/2021] [Accepted: 09/09/2021] [Indexed: 11/22/2022]
Abstract
Colorectal cancer (CRC) has a high prevalence and mortality rate, globally. To date, the progression mechanisms of CRC are still elusive. Exosomes (~100 nm in diameter) correspond to a subset of extracellular vesicles formed by an array of cancerous cells and stromal cells. These particular nanovesicles carry and transmit bioactive molecules, like proteins, lipids, and genetic materials, which mediate the crosstalk between cancer cells and the microenvironment. Accumulating evidence has shown the decisive functions of exosomes in the development, metastasis, and therapy resistance of CRC. Furthermore, some recent studies have also revealed the abilities of exosomes to function as either biomarkers or therapeutic targets for CRC. This review focuses on the specific mechanisms of exosomes in regulating CRC progression, and summarizes the potential clinical applications of exosomes in the diagnosis and therapy of CRC.
Collapse
Affiliation(s)
- Yan Dang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing. China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing. China
| | - Yongjun Wang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing. China
| | - Guiping Zhao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing. China
| | - Chuyan Chen
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing. China
| | - Wei Jiang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing. China
| |
Collapse
|
118
|
Rimal R, Desai P, Marquez AB, Sieg K, Marquardt Y, Singh S. 3-D vascularized breast cancer model to study the role of osteoblast in formation of a pre-metastatic niche. Sci Rep 2021; 11:21966. [PMID: 34754042 PMCID: PMC8578551 DOI: 10.1038/s41598-021-01513-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Breast cancer cells (BCCs) preferentially metastasize to bone. It is known that BCCs remotely primes the distant bone site prior to metastasis. However, the reciprocal influence of bone cells on the primary tumor is relatively overlooked. Here, to study the bone-tumor paracrine influence, a tri-cellular 3-D vascularized breast cancer tissue (VBCTs) model is engineered which comprised MDA-MB231, a triple-negative breast cancer cells (TNBC), fibroblasts, and endothelial cells. This is indirectly co-cultured with osteoblasts (OBs), thereby constituting a complex quad-cellular tumor progression model. VBCTs alone and in conjunction with OBs led to abnormal vasculature and reduced vessel density but enhanced VEGF production. A total of 1476 significantly upregulated and 775 downregulated genes are identified in the VBCTs exposed to OBs. HSP90N, CYCS, RPS27A, and EGFR are recognized as upregulated hub-genes. Kaplan Meier plot shows HSP90N to have a significant outcome in TNBC patient survivability. Furthermore, compared to cancer tissues without vessels, gene analysis recognized 1278 significantly upregulated and 566 downregulated genes in VBCTs. DKK1, CXCL13, C3 protein and BMP4 are identified to be downregulated hub genes in VBCTs. Together, a multi-cellular breast cancer model and culture protocols are established to study pre-metastatic events in the presence of OBs.
Collapse
Affiliation(s)
- Rahul Rimal
- DWI-Leibniz Institute for Interactive Materials, Forkenbeckstrasse 50, 52074, Aachen, Germany
| | - Prachi Desai
- DWI-Leibniz Institute for Interactive Materials, Forkenbeckstrasse 50, 52074, Aachen, Germany
| | - Andrea Bonnin Marquez
- DWI-Leibniz Institute for Interactive Materials, Forkenbeckstrasse 50, 52074, Aachen, Germany
| | - Karina Sieg
- DWI-Leibniz Institute for Interactive Materials, Forkenbeckstrasse 50, 52074, Aachen, Germany
| | - Yvonne Marquardt
- Department of Dermatology and Allergology, University Hospital, RWTH Aachen University, 52074, Aachen, Germany
| | - Smriti Singh
- DWI-Leibniz Institute for Interactive Materials, Forkenbeckstrasse 50, 52074, Aachen, Germany.
- Max Planck Institute for Medical Research, Jahnstrasse 29, 69120, Heidelberg, Germany.
| |
Collapse
|
119
|
Ghoroghi S, Mary B, Asokan N, Goetz JG, Hyenne V. Tumor extracellular vesicles drive metastasis (it's a long way from home). FASEB Bioadv 2021; 3:930-943. [PMID: 34761175 PMCID: PMC8565230 DOI: 10.1096/fba.2021-00079] [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: 07/03/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Among a plethora of functions, extracellular vesicles released by primary tumors spread in the organism and reach distant organs where they can induce the formation of a premetastatic niche. This constitutes a favorable microenvironment for circulating tumor cells which facilitates their seeding and colonization. In this review, we describe the journey of extracellular vesicles (EVs) from the primary tumor to the future metastatic organ, with a focus on the mechanisms used by EVs to target organs with a specific tropism (i.e., organotropism). We then highlight important tumor EV cargos in the context of premetastatic niche formation and summarize their known effects on extracellular matrix remodeling, angiogenesis, vessel permeabilization, resident cell activation, recruitment of foreign cells, and ultimately the formation of a pro-inflammatory and immuno-tolerant microenvironment. Finally, we discuss current experimental limitations and remaining opened questions in light of metastatic diagnosis and potential therapies targeting PMN formation.
Collapse
Affiliation(s)
- Shima Ghoroghi
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Benjamin Mary
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Nandini Asokan
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Jacky G Goetz
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Vincent Hyenne
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
- CNRS SNC5055 Strasbourg France
| |
Collapse
|
120
|
Dong Q, Liu X, Cheng K, Sheng J, Kong J, Liu T. Pre-metastatic Niche Formation in Different Organs Induced by Tumor Extracellular Vesicles. Front Cell Dev Biol 2021; 9:733627. [PMID: 34616739 PMCID: PMC8489591 DOI: 10.3389/fcell.2021.733627] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/01/2021] [Indexed: 12/11/2022] Open
Abstract
Primary tumors selectively modify the microenvironment of distant organs such as the lung, liver, brain, bone marrow, and lymph nodes to facilitate metastasis. This supportive metastatic microenvironment in distant organs was termed the pre-metastatic niche (PMN) that is characterized by increased vascular permeability, extracellular matrix remodeling, bone marrow-derived cells recruitment, angiogenesis, and immunosuppression. Extracellular vesicles (EVs) are a group of cell-derived membranous structures that carry various functional molecules. EVs play a critical role in PMN formation by delivering their cargos to recipient cells in target organs. We provide an overview of the characteristics of the PMN in different organs promoted by cancer EVs and the underlying mechanisms in this review.
Collapse
Affiliation(s)
- Qi Dong
- Department of Basic Science of Stomatology, Shanghai Stomatological Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China.,Department of Oral Pathology, School of Stomatology, Dalian Medical University, Dalian, China
| | - Xue Liu
- Department of Basic Science of Stomatology, Shanghai Stomatological Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Ke Cheng
- Department of Oral Pathology, School of Stomatology, Dalian Medical University, Dalian, China
| | - Jiahao Sheng
- Department of Oral Pathology, School of Stomatology, Dalian Medical University, Dalian, China
| | - Jing Kong
- Department of Oral Pathology, School of Stomatology, Dalian Medical University, Dalian, China
| | - Tingjiao Liu
- Department of Basic Science of Stomatology, Shanghai Stomatological Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| |
Collapse
|
121
|
Santos NL, Bustos SO, Bhatt D, Chammas R, Andrade LNDS. Tumor-Derived Extracellular Vesicles: Modulation of Cellular Functional Dynamics in Tumor Microenvironment and Its Clinical Implications. Front Cell Dev Biol 2021; 9:737449. [PMID: 34532325 PMCID: PMC8438177 DOI: 10.3389/fcell.2021.737449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/09/2021] [Indexed: 12/29/2022] Open
Abstract
Cancer can be described as a dynamic disease formed by malignant and stromal cells. The cellular interaction between these components in the tumor microenvironment (TME) dictates the development of the disease and can be mediated by extracellular vesicles secreted by tumor cells (TEVs). In this review, we summarize emerging findings about how TEVs modify important aspects of the disease like continuous tumor growth, induction of angiogenesis and metastasis establishment. We also discuss how these nanostructures can educate the immune infiltrating cells to generate an immunosuppressive environment that favors tumor progression. Furthermore, we offer our perspective on the path TEVs interfere in cancer treatment response and promote tumor recurrence, highlighting the need to understand the underlying mechanisms controlling TEVs secretion and cargo sorting. In addition, we discuss the clinical potential of TEVs as markers of cell state transitions including the acquisition of a treatment-resistant phenotype, and their potential as therapeutic targets for interventions such as the use of extracellular vesicle (EV) inhibitors to block their pro-tumoral activities. Some of the technical challenges for TEVs research and clinical use are also presented.
Collapse
Affiliation(s)
- Nathalia Leal Santos
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Silvina Odete Bustos
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Darshak Bhatt
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Roger Chammas
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luciana Nogueira de Sousa Andrade
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
122
|
Durán-Vinet B, Araya-Castro K, Calderón J, Vergara L, Weber H, Retamales J, Araya-Castro P, Leal-Rojas P. CRISPR/Cas13-Based Platforms for a Potential Next-Generation Diagnosis of Colorectal Cancer through Exosomes Micro-RNA Detection: A Review. Cancers (Basel) 2021; 13:4640. [PMID: 34572866 PMCID: PMC8466426 DOI: 10.3390/cancers13184640] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer with the second highest mortality rate worldwide. CRC is a heterogenous disease with multiple risk factors associated, including obesity, smoking, and use of alcohol. Of total CRC cases, 60% are diagnosed in late stages, where survival can drop to about 10%. CRC screening programs are based primarily on colonoscopy, yet this approach is invasive and has low patient adherence. Therefore, there is a strong incentive for developing molecular-based methods that are minimally invasive and have higher patient adherence. Recent reports have highlighted the importance of extracellular vesicles (EVs), specifically exosomes, as intercellular communication vehicles with a broad cargo, including micro-RNAs (miRNAs). These have been syndicated as robust candidates for diagnosis, primarily for their known activities in cancer cells, including immunoevasion, tumor progression, and angiogenesis, whereas miRNAs are dysregulated by cancer cells and delivered by cancer-derived exosomes (CEx). Quantitative polymerase chain reaction (qPCR) has shown good results detecting specific cancer-derived exosome micro-RNAs (CEx-miRNAs) associated with CRC, but qPCR also has several challenges, including portability and sensitivity/specificity issues regarding experiment design and sample quality. CRISPR/Cas-based platforms have been presented as cost-effective, ultrasensitive, specific, and robust clinical detection tools in the presence of potential inhibitors and capable of delivering quantitative and qualitative real-time data for enhanced decision-making to healthcare teams. Thereby, CRISPR/Cas13-based technologies have become a potential strategy for early CRC diagnosis detecting CEx-miRNAs. Moreover, CRISPR/Cas13-based platforms' ease of use, scalability, and portability also showcase them as a potential point-of-care (POC) technology for CRC early diagnosis. This study presents two potential CRISPR/Cas13-based methodologies with a proposed panel consisting of four CEx-miRNAs, including miR-126, miR-1290, miR-23a, and miR-940, to streamline novel applications which may deliver a potential early diagnosis and prognosis of CRC.
Collapse
Affiliation(s)
- Benjamín Durán-Vinet
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (B.D.-V.); (K.A.-C.); (H.W.)
- Center of Excellence in Translational Medicine (CEMT), Biomedicine and Translational Research Laboratory, Universidad de La Frontera, Temuco 4780000, Chile;
| | - Karla Araya-Castro
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (B.D.-V.); (K.A.-C.); (H.W.)
- Innovation and Entrepreneurship Institute (iDEAUFRO), Universidad de La Frontera, Temuco 4780000, Chile
| | - Juan Calderón
- Center for Genetics and Genomics, School of Medicine, Institute of Science and Innovation in Medicine (ICIM), Clínica Alemana, Universidad del Desarrollo, Santiago 8320000, Chile;
| | - Luis Vergara
- Center of Excellence in Translational Medicine (CEMT), Biomedicine and Translational Research Laboratory, Universidad de La Frontera, Temuco 4780000, Chile;
- Doctoral Program in Cell and Applied Molecular Biology, Universidad de La Frontera, Temuco 4780000, Chile
| | - Helga Weber
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (B.D.-V.); (K.A.-C.); (H.W.)
- Center of Excellence in Translational Medicine (CEMT), Biomedicine and Translational Research Laboratory, Universidad de La Frontera, Temuco 4780000, Chile;
| | - Javier Retamales
- Chilean Cooperative Group for Oncologic Research (GOCCHI), Santiago 8320000, Chile;
| | - Paulina Araya-Castro
- School of Medicine, Clínica Alemana, Universidad del Desarrollo, Santiago 8320000, Chile;
| | - Pamela Leal-Rojas
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (B.D.-V.); (K.A.-C.); (H.W.)
- Center of Excellence in Translational Medicine (CEMT), Biomedicine and Translational Research Laboratory, Universidad de La Frontera, Temuco 4780000, Chile;
- Department of Agricultural Sciences and Natural Resources, Faculty of Agricultural and Forestry Science, Universidad de La Frontera, Temuco 4780000, Chile
| |
Collapse
|
123
|
Przygodzka P, Sochacka E, Soboska K, Pacholczyk M, Papiewska-Pająk I, Przygodzki T, Płociński P, Ballet S, De Prins A, Boncela J. Neuromedin U induces an invasive phenotype in CRC cells expressing the NMUR2 receptor. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:283. [PMID: 34493299 PMCID: PMC8422652 DOI: 10.1186/s13046-021-02073-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022]
Abstract
Background Successful colorectal cancer (CRC) therapy often depends on the accurate identification of primary tumours with invasive potential. There is still a lack of identified pathological factors associated with disease recurrence that could help in making treatment decisions. Neuromedin U (NMU) is a secretory neuropeptide that was first isolated from the porcine spinal cord, and it has emerged as a novel factor involved in the tumorigenesis and/or metastasis of many types of cancers. Previously associated with processes leading to CRC cell invasiveness, NMU has the potential to be a marker of poor outcome, but it has not been extensively studied in CRC. Methods Data from The Cancer Genome Atlas (TCGA) were used to analyse NMU and NMU receptor (NMUR1 and NMUR2) expression in CRC tissues vs. normal tissues, and real-time PCR was used for NMU and NMU receptor expression analysis. NMU protein detection was performed by immunoblotting. Secreted NMU was immunoprecipitated from cell culture-conditioned media and analysed by immunoblotting and protein sequencing. DNA demethylation by 5-aza-CdR was used to analyse the regulation of NMUR1 and NMUR2 expression. NMU receptor activity was monitored by detecting calcium mobilisation in cells loaded with fluo-4, and ERK1/2 kinase activation was detected after treatment with NMU or receptor agonist. Cell migration and invasion were investigated using membrane filters. Integrin expression was evaluated by flow cytometry. Results The obtained data revealed elevated expression of NMU and NMUR2 in CRC tissue samples and variable expression in the analysed CRC cell lines. We have shown, for the first time, that NMUR2 activation induces signalling in CRC cells and that NMU increases the motility and invasiveness of NMUR2-positive CRC cells and increases prometastatic integrin receptor subunit expression. Conclusions Our results show the ability of CRC cells to respond to NMU via activation of the NMUR2 receptor, which ultimately leads to a shift in the CRC phenotype towards a more invasive phenotype. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02073-8.
Collapse
Affiliation(s)
- Patrycja Przygodzka
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland.
| | - Ewelina Sochacka
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland.,Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Kamila Soboska
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland.,Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Marcin Pacholczyk
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, 44-100, Gliwice, Poland
| | | | - Tomasz Przygodzki
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Mazowiecka 6/8, 92-235, Lodz, Poland
| | - Przemysław Płociński
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237, Lodz, Poland
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - An De Prins
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Joanna Boncela
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| |
Collapse
|
124
|
Vidács DL, Veréb Z, Bozó R, Flink LB, Polyánka H, Németh IB, Póliska S, Papp BT, Manczinger M, Gáspár R, Mirdamadi S, Kemény L, Bata-Csörgő Z. Phenotypic plasticity of melanocytes derived from human adult skin. Pigment Cell Melanoma Res 2021; 35:38-51. [PMID: 34467641 DOI: 10.1111/pcmr.13012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/15/2021] [Accepted: 08/10/2021] [Indexed: 12/29/2022]
Abstract
We previously described a novel in vitro culture technique for dedifferentiated human adult skin melanocytes. Melanocytes cultured in a defined, cholera toxin and PMA free medium became bipolar, unpigmented, and highly proliferative. Furthermore, TRP-1 and c-Kit expression disappeared and EGFR receptor and nestin expression were induced in the cells. Here, we further characterized the phenotype of these dedifferentiated cells and by comparing them to mature pigmented melanocytes we detected crucial steps in their phenotype change. Our data suggest that normal adult melanocytes easily dedifferentiate into pluripotent stem cells given the right environment. This dedifferentiation process described here for normal melanocyte is very similar to what has been described for melanoma cells, indicating that phenotype switching driven by environmental factors is a general characteristic of melanocytes that can occur independent of malignant transformation.
Collapse
Affiliation(s)
- Dániel László Vidács
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Zoltán Veréb
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Skin Research Group (HCEMM-USZ Skin Research Group), Szeged, Hungary
| | - Renáta Bozó
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Skin Research Group (HCEMM-USZ Skin Research Group), Szeged, Hungary
| | - Lili Borbála Flink
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Hilda Polyánka
- Department of Medical Genetics, University of Szeged, Szeged, Hungary
| | - István Balázs Németh
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Szilárd Póliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Genomic Medicine and Bioinformatics Core Facility, The University of Debrecen, Debrecen, Hungary
| | - Benjamin Tamás Papp
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Máté Manczinger
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Róbert Gáspár
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Seyedmohsen Mirdamadi
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Skin Research Group (HCEMM-USZ Skin Research Group), Szeged, Hungary.,MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, Szeged, Hungary
| | - Zsuzsanna Bata-Csörgő
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Skin Research Group (HCEMM-USZ Skin Research Group), Szeged, Hungary.,MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, Szeged, Hungary
| |
Collapse
|
125
|
Deasy SK, Erez N. A glitch in the matrix: organ-specific matrisomes in metastatic niches. Trends Cell Biol 2021; 32:110-123. [PMID: 34479765 DOI: 10.1016/j.tcb.2021.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
Modification of the extracellular matrix (ECM) is a critical aspect of developing a metastasis-supportive organ niche. Recent work investigating ECM changes that facilitate metastasis has revealed ways in which different metastatic organ niches are similar as well as the distinct characteristics that make them unique. In this review, we present recent findings regarding how ECM modifications support metastasis in four frequent metastatic sites: the lung, liver, bone, and brain. We discuss ways in which these modifications are shared between metastatic organs as well as features specific to each location. We also discuss areas of technical innovation that could be advantageous to future research and areas of inquiry that merit further investigation.
Collapse
Affiliation(s)
- Sarah K Deasy
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
126
|
Kostallari E, Valainathan S, Biquard L, Shah VH, Rautou PE. Role of extracellular vesicles in liver diseases and their therapeutic potential. Adv Drug Deliv Rev 2021; 175:113816. [PMID: 34087329 PMCID: PMC10798367 DOI: 10.1016/j.addr.2021.05.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/17/2021] [Accepted: 05/29/2021] [Indexed: 02/07/2023]
Abstract
More than eight hundred million people worldwide have chronic liver disease, with two million deaths per year. Recurring liver injury results in fibrogenesis, progressing towards cirrhosis, for which there doesn't exists any cure except liver transplantation. Better understanding of the mechanisms leading to cirrhosis and its complications is needed to develop effective therapies. Extracellular vesicles (EVs) are released by cells and are important for cell-to-cell communication. EVs have been reported to be involved in homeostasis maintenance, as well as in liver diseases. In this review, we present current knowledge on the role of EVs in non-alcoholic fatty liver disease and non-alcoholic steatohepatitis, alcohol-associated liver disease, chronic viral hepatitis, primary liver cancers, acute liver injury and liver regeneration. Moreover, therapeutic strategies involving EVs as targets or as tools to treat liver diseases are summarized.
Collapse
Affiliation(s)
- Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States.
| | - Shantha Valainathan
- Université de Paris, AP-HP, Hôpital Beaujon, Service d'Hépatologie, DMU DIGEST, Centre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE-LIVER, Centre de recherche sur l'inflammation, Inserm, UMR 1149, Paris, France
| | - Louise Biquard
- Université de Paris, Centre de recherche sur l'inflammation, Inserm, UMR 1149, Paris, France.
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States.
| | - Pierre-Emmanuel Rautou
- Université de Paris, AP-HP, Hôpital Beaujon, Service d'Hépatologie, DMU DIGEST, Centre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE-LIVER, Centre de recherche sur l'inflammation, Inserm, UMR 1149, Paris, France.
| |
Collapse
|
127
|
Wang H, Zhu Y, Chen H, Yang N, Wang X, Li B, Ying P, He H, Cai Y, Zhang M, Niu S, Li Y, Lu Z, Peng X, Zou D, Zhong R, Chang J, Dai M, Tian J, Miao X. Colorectal cancer risk variant rs7017386 modulates two oncogenic lncRNAs expression via ATF1-mediated long-range chromatin loop. Cancer Lett 2021; 518:140-151. [PMID: 34274452 DOI: 10.1016/j.canlet.2021.07.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/26/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023]
Abstract
The activating transcription factor 1 (ATF1) has been identified as a vital pathogenic factor in the progression of colorectal cancer (CRC), whiles, the precise regulatory mechanisms remain elusive. Here, we comprehensively characterized the ATF1 cistrome by RNA-seq and ChIP-seq assays in CRC cell lines. As the results, we identified 358 genes differentially regulated and 15,029 ATF1 binding sites and demonstrated that ATF1 was widely involved in major signaling pathways in CRC, such as Wnt, TNF, Jak-STAT. Subsequently, by the expression quantitative trait loci (eQTL) analyses, we found that rs7017386 was associated with the expression of CCAT1 and PVT1 in the Wnt pathway. By a two-stage population study with 6,131 CRC cases and 10,022 healthy controls, we identified the variant was associated with CRC risk. Mechanistically, we found rs7017386 allele-specifically enhanced the binding affinity of ATF1 and promoted the expressions of PVT1 and CCAT1, via forming a long-range chromatin loop. Moreover, those two lncRNAs could synergistically facilitate c-Myc expression to activate the Wnt pathway in CRC progression. Our findings not only demonstrated the transcriptomic profiling of ATF1 in CRC, but also provided important clues for the etiology of CRC.
Collapse
Affiliation(s)
- Haoxue Wang
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongda Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Yang
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyang Wang
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Li
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pingting Ying
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng He
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yimin Cai
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Zhang
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siyuan Niu
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Li
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zequn Lu
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiating Peng
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danyi Zou
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Zhong
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiang Chang
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Dai
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, And the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
128
|
Cancer: a mirrored room between tumor bulk and tumor microenvironment. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:217. [PMID: 34183054 PMCID: PMC8240272 DOI: 10.1186/s13046-021-02022-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022]
Abstract
It has been well documented that the tumor microenvironment (TME) plays a key role in the promotion of drug resistance, the support of tumor progression, invasiveness, metastasis, and even the maintenance of a cancer stem-like phenotype. Here, we reviewed TME formation presenting it as a reflection of a tumor’s own organization during the different stages of tumor development. Interestingly, functionally different groups of stromal cells seem to have specific spatial distributions within the TME that change as the tumor evolves into advanced stage progression which correlates with the fact that cancer stem-like cells (CSCs) are located in the edges of solid tumor masses in advanced tumors. We also focus on the continuos feedback that is established between a tumor and its surroundings. The “talk” between tumor mass cells and TME stromal cells, marks the evolution of both interlocuting cell types. For instance, the metabolic and functional transformations that stromal cells undergo due to tumor corrupting activity. Moreover, the molecular basis of metastatic spread is also approached, making special emphasis on the site-specific pre-metastatic niche formation as another reflection of the primary tumor molecular signature. Finally, several therapeutic approaches targeting primary TME and pre-metastatic niche are suggested. For instance, a systematic analysis of the TME just adjacent to the tumor mass to establish the proportion of myofibroblasts-like cancer-associated fibroblasts (CAFs) which may in turn correspond to stemness and metastases-promotion. Or the implementation of “re-education” therapies consisting of switching tumor-supportive stromal cells into tumor-suppressive ones. In summary, to improve our clinical management of cancer, it is crucial to understand and learn how to manage the close interaction between TME and metastasis.
Collapse
|
129
|
Piran M, Sepahi N, Moattari A, Rahimi A, Ghanbariasad A. Systems Biomedicine of Primary and Metastatic Colorectal Cancer Reveals Potential Therapeutic Targets. Front Oncol 2021; 11:597536. [PMID: 34249670 PMCID: PMC8263939 DOI: 10.3389/fonc.2021.597536] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 05/31/2021] [Indexed: 12/23/2022] Open
Abstract
Colorectal cancer (CRC) is one of the major causes of cancer deaths across the world. Patients' survival at time of diagnosis depends mainly on stage of the tumor. Therefore, understanding the molecular mechanisms from low-grade to high-grade stages of cancer that lead to cellular migration from one tissue/organ to another tissue/organ is essential for implementing therapeutic approaches. To this end, we performed a unique meta-analysis flowchart by identifying differentially expressed genes (DEGs) between normal, primary (primary sites), and metastatic samples (Colorectal metastatic lesions in liver and lung) in some Test datasets. DEGs were employed to construct a protein-protein interaction (PPI) network. A smaller network containing 39 DEGs was then extracted from the PPI network whose nodes expression induction or suppression alone or in combination with each other would inhibit tumor progression or metastasis. These DEGs were then verified by gene expression profiling, survival analysis, and multiple Validation datasets. We suggested for the first time that downregulation of mitochondrial genes, including ETHE1, SQOR, TST, and GPX3, would help colorectal cancer cells to produce more energy under hypoxic conditions through mechanisms that are different from "Warburg Effect". Augmentation of given antioxidants and repression of P4HA1 and COL1A2 genes could be a choice of CRC treatment. Moreover, promoting active GSK-3β together with expression control of EIF2B would prevent EMT. We also proposed that OAS1 expression enhancement can induce the anti-cancer effects of interferon-gamma, while suppression of CTSH hinders formation of focal adhesions. ATF5 expression suppression sensitizes cancer cells to anchorage-dependent death signals, while LGALS4 induction recovers cell-cell junctions. These inhibitions and inductions would be another combinatory mechanism that inhibits EMT and cell migration. Furthermore, expression inhibition of TMPO, TOP2A, RFC3, GINS1, and CKS2 genes could prevent tumor growth. Besides, TRIB3 suppression would be a promising target for anti-angiogenic therapy. SORD is a poorly studied enzyme in cancer, found to be upregulated in CRC. Finally, TMEM131 and DARS genes were identified in this study whose roles have never been interrogated in any kind of cancer, neither as a biomarker nor curative target. All the mentioned mechanisms must be further validated by experimental wet-lab techniques.
Collapse
Affiliation(s)
- Mehran Piran
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
- Department of Bacteriology and Virology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Sepahi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Afagh Moattari
- Department of Bacteriology and Virology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Rahimi
- Bioinformatics and Computational Biology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Ghanbariasad
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
130
|
Wang Z, Liu J, Huang H, Ye M, Li X, Wu R, Liu H, Song Y. Metastasis-associated fibroblasts: an emerging target for metastatic cancer. Biomark Res 2021; 9:47. [PMID: 34112258 PMCID: PMC8194104 DOI: 10.1186/s40364-021-00305-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Metastasis suggests a poor prognosis for cancer patients, and treatment strategies for metastatic cancer are still very limited. Numerous studies have shown that cancer-associated fibroblasts (CAFs), a large component of the tumor microenvironment, contribute to tumor metastasis. Stromal fibroblasts at metastatic sites are different from CAFs within primary tumors and can be termed metastasis-associated fibroblasts (MAFs), and they also make great contributions to the establishment of metastatic lesions and the therapeutic resistance of metastatic tumors. MAFs are capable of remodeling the extracellular matrix of metastatic tumors, modulating immune cells in the tumor microenvironment, promoting angiogenesis and enhancing malignant tumor phenotypes. Thus, MAFs can help establish premetastatic niches and mediate resistance to therapeutic strategies, including immunotherapy and antiangiogenic therapy. The results of preclinical studies suggest that targeting MAFs can alleviate the progression of metastatic cancer and mitigate therapeutic resistance, indicating that MAFs are a promising target for metastatic cancer. Here, we comprehensively summarize the existing evidence on MAFs and discuss their origins, generation, functions and related therapeutic strategies in an effort to provide a better understanding of MAFs and offer treatment perspectives for metastatic cancer.
Collapse
Affiliation(s)
- Zimu Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Jiaxin Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Hairong Huang
- Department of Cardiothoracic Surgery, Jinling Hospital, 210002, Nanjing, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Xinying Li
- Department of Respiratory Medicine, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, 210008, Nanjing, Jiangsu, China
| | - Ranpu Wu
- Department of Respiratory Medicine, Jinling Hospital, Southeast University of Medicine, 210009, Nanjing, Jiangsu, China
| | - Hongbing Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| |
Collapse
|
131
|
Huang Q, Hsueh CY, Shen YJ, Guo Y, Huang JM, Zhang YF, Li JY, Gong HL, Zhou L. Small extracellular vesicle-packaged TGFβ1 promotes the reprogramming of normal fibroblasts into cancer-associated fibroblasts by regulating fibronectin in head and neck squamous cell carcinoma. Cancer Lett 2021; 517:1-13. [PMID: 34089808 DOI: 10.1016/j.canlet.2021.05.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 01/07/2023]
Abstract
Tumor development and progression hinge upon ongoing coevolution and crosstalk with the tumor microenvironment. In particular, fibroblasts in the tumor stroma are coopted to support tumor growth and survival through interactions with tumor cells. Despite their significant importance, there is no consensus on the origin of cancer-associated fibroblasts (CAFs) in head and neck squamous cell carcinoma (HNSCC). In this study, we demonstrated that small extracellular vesicle (sEV)-packaged TGFβ1 can reprogram normal fibroblasts (NFs) into CAFs both in vitro and in vivo. Mechanistically, TGFβ1 in sEV activated NFs by regulating fibronectin, rather than modulating the canonical TGFβ-Smad signal pathway. Furthermore, TGFβ1 and fibronectin are related to HNSCC clinicopathologic features. Plasma sEV TGFβ1 may serve as a potential diagnostic biomarker for HNSCC. This hitherto unknown mechanism of reprogramming of NFs into CAFs by a unique pathway has major implications for underlying cancer-recruited stroma responses.
Collapse
Affiliation(s)
- Qiang Huang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Chi-Yao Hsueh
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yu-Jie Shen
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yang Guo
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Jia-Meng Huang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yi-Fan Zhang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Jiao-Yu Li
- Department of Pediatric, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Hong-Li Gong
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China.
| | - Liang Zhou
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
132
|
Abstract
Colorectal cancer has served as a genetic and biological paradigm for the evolution of solid tumors, and these insights have illuminated early detection, risk stratification, prevention, and treatment principles. Employing the hallmarks of cancer framework, we provide a conceptual framework to understand how genetic alterations in colorectal cancer drive cancer cell biology properties and shape the heterotypic interactions across cells in the tumor microenvironment. This review details research advances pertaining to the genetics and biology of colorectal cancer, emerging concepts gleaned from immune and single-cell profiling, and critical advances and remaining knowledge gaps influencing the development of effective therapies for this cancer that remains a major public health burden.
Collapse
Affiliation(s)
- Jiexi Li
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xingdi Ma
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Deepavali Chakravarti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shabnam Shalapour
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
133
|
Bhatta B, Cooks T. Reshaping the tumor microenvironment: extracellular vesicles as messengers of cancer cells. Carcinogenesis 2021; 41:1461-1470. [PMID: 33047121 DOI: 10.1093/carcin/bgaa107] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/29/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME) comprises an assortment of immune and non-immune cells. The interactions between the cancer cells and their surrounding TME are known to be a cardinal factor in all stages of cancer progression, from initiation to metastasis. Tumor-associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs) are considered two of the most abundant TME members associated with poor prognosis in various cancer types. Intercellular communication between the cancer cells and TME cells might occur via direct cell-cell contact or achieved through secreted factors such as cytokines, growth factors and extracellular vesicles (EVs). EVs are released by almost every cell type and by cancer cells in particular. EVs are loaded with unique molecular cargos that might include DNA, proteins, RNA and lipids, commonly reflecting the physiological traits of their donor cells. Once released, EVs are capable of initiating short- and long-distance communication in an autocrine, paracrine and endocrine fashion. The molecular cargos within the EVs are able to impart phenotypic changes at the receiving end thus allowing EV-releasing cancer cells to deliver messages to TME cells and tighten their grasp over the cancerous tissue. In this concise review, we aim to document the bidirectional EV-based communication between cancer cell, TAMs and CAFs, tilting the balance in favor of cancer progression and metastasis.
Collapse
Affiliation(s)
- Bibek Bhatta
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tomer Cooks
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
134
|
Abstract
Tumors are equipped with a highly complex machinery of interrelated events so as to adapt to hazardous conditions, preserve a growing cell mass and thrive at the site of metastasis. Tumor cells display metastatic propensity toward specific organs where the stromal milieu is appropriate for their further colonization. Effective colonization relies on the plasticity of tumor cells in adapting to the conditions of the new area by reshaping their epigenetic landscape. Breast cancer cells, for instance, are able to adopt brain-like or epithelial/osteoid features in order to pursue effective metastasis into brain and bone, respectively. The aim of this review is to discuss recent insights into organ tropism in tumor metastasis, outlining potential strategies to address this driver of tumor aggressiveness.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Cancer & Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, 66177‐13446, Iran
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, 66177‐13446, Iran
| |
Collapse
|
135
|
Ye F, Huang W, Xue Y, Tang E, Wang M, Shi F, Wei D, Han Y, Chen P, Zhang X, Yu D. Serum Levels of ITGBL1 as an Early Diagnostic Biomarker for Hepatocellular Carcinoma with Hepatitis B Virus Infection. J Hepatocell Carcinoma 2021; 8:285-300. [PMID: 33948441 PMCID: PMC8088298 DOI: 10.2147/jhc.s306966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/07/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose Early diagnostic biomarkers of hepatocellular carcinoma (HCC) are needed to distinguish hepatitis B virus (HBV) associated HCC (HBV-HCC) patients from at-risk patients. We assessed the diagnostic values of serum Integrin beta-like 1 (ITGBL1) for early-stage HBV-HCC. Patients and Methods We recruited 716 participators including 299 in the training and 417 in the validation stage, (HBV-HCC, chronic hepatitis B (CHB), HBV‐related liver cirrhosis (HBV-LC), and healthy controls) between 2017 and 2020 from three centers. Serum ITGBL1 was measured by ELISA. Receiver operating characteristic (ROC) was used to calculate diagnostic accuracy. Results The serum levels of ITGBL1 in HBV-HCC patients were significantly lower than those in CHB and HBV-LC patients. This result was confirmed in the follow-up patients who progressed from HBV-LC to HCC. The optimum diagnostic cutoff value of serum ITGBL1 was 47.93ng/mL for detection of early-stage HBV-HCC. The serum ITGBL1 has higher diagnostic accuracy than AFP20 in differentiating the early-stage HBV-HCC from the at-risk patients (area under curve [AUC] 0.787 vs 0.638, p<0.05). For AFP-negative (<20ng/mL) HBV-HCC patients, serum ITGBL1 maintained diagnostic accuracy (training cohort: AUC 0.756, 95% confidence interval [CI] 0.683–0.819, sensitivity 68.18%, and specificity 68.85%; validation cohort: 0.744, 0.686–0.796, 81.13%, and 55.88%). Combination ITGBL1 with AFP20 significantly increased diagnostic accuracy in differentiating the HBV-HCC from at-risk patients (AUC 0.840; 0.868) than ITGBL1 (AUC 0.773, p<0.05; 0.732, p<0.0001) or AFP20 (AUC 0.705, p<0.0001; 0.773, p<0.0001) alone. Conclusion The serum level of ITGBL1 improved identification of AFP-negative HBV-HCC patients, and increased diagnostic accuracy with AFP20 together in the early detection of HBV-HCC.
Collapse
Affiliation(s)
- Fei Ye
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200025, People's Republic of China
| | - Wei Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226000, People's Republic of China
| | - Yuan Xue
- Institute of Hepatology, The Third People's Hospital of Changzhou, Changzhou, 213000, People's Republic of China
| | - Erjiang Tang
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China.,Institute of Gastrointestinal Surgery and Translational Medicine, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China
| | - Mingjie Wang
- Department of Gastroenterology & Hepatology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201821, People's Republic of China
| | - Fengchun Shi
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200025, People's Republic of China
| | - Dong Wei
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200025, People's Republic of China
| | - Yue Han
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200025, People's Republic of China
| | - Peizhan Chen
- Clinical Research Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201821, People's Republic of China
| | - Xinxin Zhang
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200025, People's Republic of China
| | - Demin Yu
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200025, People's Republic of China
| |
Collapse
|
136
|
Yang L, Li T, Shi H, Zhou Z, Huang Z, Lei X. The cellular and molecular components involved in pre-metastatic niche formation in colorectal cancer liver metastasis. Expert Rev Gastroenterol Hepatol 2021; 15:389-399. [PMID: 33174441 DOI: 10.1080/17474124.2021.1848543] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Liver metastasis is the main cause of death in colorectal cancer (CRC). Premetastatic niche (PMN), a favorable microenvironment for cancer cells colonization at the distant organ, plays a pivotal role in CRC liver metastasis (CRCLM). Our understanding of the mechanisms mediating the formation of liver PMN in CRC has been significantly advanced in recent years, there are still many challenges and questions that remain.Areas covered: This review covers cellular and molecular components, and the interaction of theprimary cancer with the resident microenvironment of the distant organ that leads to PMN formation in CRCLM based on the latest literature.Expert Opinion: Various cellular and molecular events are involved in the liver PMN formation in CRC such as bone marrow-derived cells (BMDCs), hepatic stellate cells, Kupffer cells, extracellular matrix, and CRC-derived factors. The formation of the liver PMN depends on a complex interaction of CRC with the liver microenvironment including BMDCs recruitment, vascularization, immunosuppression, inflammatory response, and extracellular matrix remodeling. This review firstly discusses on the cellular and molecular components contributing to the formation of the liver PMN in CRC, so as to provide new ideas for designing effective therapeutic strategies and prognostic markers for CRCLM.
Collapse
Affiliation(s)
- Lingling Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Taiyuan Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Gastrointestinal Surgical Institute, Nanchang University, Nanchang, Jiangxi, China
| | - Haoran Shi
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhen Zhou
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhixiang Huang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiong Lei
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Gastrointestinal Surgical Institute, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
137
|
Li H, Zhou L, Zhou J, Li Q, Ji Q. Underlying mechanisms and drug intervention strategies for the tumour microenvironment. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:97. [PMID: 33722297 PMCID: PMC7962349 DOI: 10.1186/s13046-021-01893-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/24/2021] [Indexed: 02/08/2023]
Abstract
Cancer occurs in a complex tissue environment, and its progression depends largely on the tumour microenvironment (TME). The TME has a highly complex and comprehensive system accompanied by dynamic changes and special biological characteristics, such as hypoxia, nutrient deficiency, inflammation, immunosuppression and cytokine production. In addition, a large number of cancer-associated biomolecules and signalling pathways are involved in the above bioprocesses. This paper reviews our understanding of the TME and describes its biological and molecular characterization in different stages of cancer development. Furthermore, we discuss in detail the intervention strategies for the critical points of the TME, including chemotherapy, targeted therapy, immunotherapy, natural products from traditional Chinese medicine, combined drug therapy, etc., providing a scientific basis for cancer therapy from the perspective of key molecular targets in the TME.
Collapse
Affiliation(s)
- Haoze Li
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lihong Zhou
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zhou
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qi Li
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China. .,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Qing Ji
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China. .,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
138
|
Isolation and characterization of extracellular vesicle subpopulations from tissues. Nat Protoc 2021; 16:1548-1580. [PMID: 33495626 DOI: 10.1038/s41596-020-00466-1] [Citation(s) in RCA: 218] [Impact Index Per Article: 72.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are lipid bilayered membrane structures released by all cells. Most EV studies have been performed by using cell lines or body fluids, but the number of studies on tissue-derived EVs is still limited. Here, we present a protocol to isolate up to six different EV subpopulations directly from tissues. The approach includes enzymatic treatment of dissociated tissues followed by differential ultracentrifugation and density separation. The isolated EV subpopulations are characterized by electron microscopy and RNA profiling. In addition, their protein cargo can be determined with mass spectrometry, western blot and ExoView. Tissue-EV isolation can be performed in 22 h, but a simplified version can be completed in 8 h. Most experiments with the protocol have used human melanoma metastases, but the protocol can be applied to other cancer and non-cancer tissues. The procedure can be adopted by researchers experienced with cell culture and EV isolation.
Collapse
|
139
|
Chen Q, Tan Y, Zhang C, Zhang Z, Pan S, An W, Xu H. A Weighted Gene Co-Expression Network Analysis-Derived Prognostic Model for Predicting Prognosis and Immune Infiltration in Gastric Cancer. Front Oncol 2021; 11:554779. [PMID: 33718128 PMCID: PMC7947930 DOI: 10.3389/fonc.2021.554779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 01/18/2021] [Indexed: 12/20/2022] Open
Abstract
Background Gastric cancer (GC) is a major public health problem worldwide. In recent decades, the treatment of gastric cancer has improved greatly, but basic research and clinical application of gastric cancer remain challenges due to the high heterogeneity. Here, we provide new insights for identifying prognostic models of GC. Methods We obtained the gene expression profiles of GSE62254 containing 300 samples for training. GSE15459 and TCGA-STAD for validation, which contain 200 and 375 samples, respectively. Weighted gene co-expression network analysis (WGCNA) was used to identify gene modules. We performed Lasso regression and Cox regression analyses to identify the most significant five genes to develop a novel prognostic model. And we selected two representative genes within the model for immunohistochemistry staining with 105 GC specimens from our hospital to verify the prediction efficiency. Moreover, we estimated the correlation coefficient between our model and immune infiltration using the CIBERSORT algorithm. The data from GSE15459 and TCGA cohort validated the robustness and predictive accuracy of this prognostic model. Results Of the 12 gene modules identified, 1,198 green-yellow module genes were selected for further analysis. Multivariate Cox analysis was performed on genes from univariate Cox regression and Lasso regression analysis using the Cox proportional hazards regression model. Finally, we constructed a five gene prognostic model: Risk Score = [(-0.7547) * Expression (ARHGAP32)] + [(-0.8272) * Expression (KLF5)] + [1.09 * Expression (MAMLD1)] + [0.5174 * Expression (MATN3)] + [1.66 * Expression (NES)]. The prognosis of samples in the high-risk group was significantly poorer than that of samples in the low-risk group (p = 6.503e-11). The risk model was also regarded as an independent predictor of prognosis (HR, 1.678, p < 0.001). The observed correlation with immune cells suggested that this risk model could potentially predict immune infiltration. Conclusion This study identified a potential risk model for prognosis and immune infiltration prediction in GC using WGCNA and Cox regression analysis.
Collapse
Affiliation(s)
- Qingchuan Chen
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuen Tan
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chao Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhe Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siwei Pan
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wen An
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Huimian Xu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
140
|
Cao L, Wu Y, Wang X, Li X, Tan Z, Guan F. Role of Site-Specific Glycosylation in the I-Like Domain of Integrin β1 in Small Extracellular Vesicle-Mediated Malignant Behavior and FAK Activation. Int J Mol Sci 2021; 22:ijms22041770. [PMID: 33578954 PMCID: PMC7916680 DOI: 10.3390/ijms22041770] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/24/2022] Open
Abstract
Integrin β1 plays an essential role in the crosstalk between tumor cells and their microenvironment. Aberrant N-glycosylation of integrin β1 was documented to alter integrin β1 expression, dimerization, and biological function. However, the biological function of site-specific N-glycosylation of integrin β1 on extracellular vesicles is not fully understood. In this study, we mutated putative N-glycosylation sites in different domains of integrin β1. Removal of the N-glycosylation sites on the I-like domain of integrin β1 (termed the Δ4–6 β1 mutant) suppressed focal adhesion kinase (FAK) signaling, cell migration, and adhesion compared with other β1 mutants. Cell adhesion, migration, and activation of FAK were suppressed in recipient MCF7 cells co-cultured with Δ4–6 mutant cells and treated with small extracellular vesicles (sEVs) from Δ4–6 mutant cells. Notably, the wild-type and β1 mutant were both present in sEVs, and could be transferred to recipient cells via sEVs, resulting in changes of cell behavior. Our findings demonstrate the important roles of N-glycosylation of the I-like domain of integrin β1. Moreover, the vesicular Δ4–6 β1 mutant can regulate integrin-mediated functions in recipient cells via sEVs.
Collapse
|
141
|
Zhang DX, Vu LT, Ismail NN, Le MTN, Grimson A. Landscape of extracellular vesicles in the tumour microenvironment: Interactions with stromal cells and with non-cell components, and impacts on metabolic reprogramming, horizontal transfer of neoplastic traits, and the emergence of therapeutic resistance. Semin Cancer Biol 2021; 74:24-44. [PMID: 33545339 DOI: 10.1016/j.semcancer.2021.01.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 02/09/2023]
Abstract
Extracellular vesicles (EVs) are increasingly recognised as a pivotal player in cell-cell communication, an attribute of EVs that derives from their ability to transport bioactive cargoes between cells, resulting in complex intercellular signalling mediated by EVs, which occurs under both physiological and pathological conditions. In the context of cancer, recent studies have demonstrated the versatile and crucial roles of EVs in the tumour microenvironment (TME). Here, we revisit EV biology, and focus on EV-mediated interactions between cancer cells and stromal cells, including fibroblasts, immune cells, endothelial cells and neurons. In addition, we focus on recent reports indicating interactions between EVs and non-cell constituents within the TME, including the extracellular matrix. We also review and summarise the intricate cancer-associated network modulated by EVs, which promotes metabolic reprogramming, horizontal transfer of neoplastic traits, and therapeutic resistance in the TME. We aim to provide a comprehensive and updated landscape of EVs in the TME, focusing on oncogenesis, cancer progression and therapeutic resistance, together with our future perspectives on the field.
Collapse
Affiliation(s)
- Daniel Xin Zhang
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR; Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA.
| | - Luyen Tien Vu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore
| | - Nur Nadiah Ismail
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Minh T N Le
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; N.1 Institute for Health, National University of Singapore, Singapore.
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
142
|
Wang H, Pan J, Barsky L, Jacob JC, Zheng Y, Gao C, Wang S, Zhu W, Sun H, Lu L, Jia H, Zhao Y, Bruns C, Vago R, Dong Q, Qin L. Characteristics of pre-metastatic niche: the landscape of molecular and cellular pathways. MOLECULAR BIOMEDICINE 2021; 2:3. [PMID: 35006432 PMCID: PMC8607426 DOI: 10.1186/s43556-020-00022-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/30/2020] [Indexed: 02/08/2023] Open
Abstract
Metastasis is a major contributor to cancer-associated deaths. It involves complex interactions between primary tumorigenic sites and future metastatic sites. Accumulation studies have revealed that tumour metastasis is not a disorderly spontaneous incident but the climax of a series of sequential and dynamic events including the development of a pre-metastatic niche (PMN) suitable for a subpopulation of tumour cells to colonize and develop into metastases. A deep understanding of the formation, characteristics and function of the PMN is required for developing new therapeutic strategies to treat tumour patients. It is rapidly becoming evident that therapies targeting PMN may be successful in averting tumour metastasis at an early stage. This review highlights the key components and main characteristics of the PMN and describes potential therapeutic strategies, providing a promising foundation for future studies.
Collapse
Affiliation(s)
- Hao Wang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Junjie Pan
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Livnat Barsky
- Avram and Stella Goldstein-Goren, Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Yan Zheng
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Chao Gao
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Shun Wang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Wenwei Zhu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Haoting Sun
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Lu Lu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Huliang Jia
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Yue Zhao
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Cologne, Germany
| | - Razi Vago
- Avram and Stella Goldstein-Goren, Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Qiongzhu Dong
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China.
| | - Lunxiu Qin
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China.
| |
Collapse
|
143
|
Fibroblast Subsets in Intestinal Homeostasis, Carcinogenesis, Tumor Progression, and Metastasis. Cancers (Basel) 2021; 13:cancers13020183. [PMID: 33430285 PMCID: PMC7825703 DOI: 10.3390/cancers13020183] [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: 12/21/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Colorectal cancer often develops via the adenoma–carcinoma sequence, a process which is accompanied by (epi) genetic alterations in epithelial cells and gradual phenotypic changes in fibroblast populations. Recent studies have made it clear that these fibroblast populations which, in the context of invasive cancers are termed cancer-associated fibroblasts (CAFs), play an important role in intestinal tumor progression. This review provides an overview on the emerging role of fibroblasts in various stages of colorectal cancer development, ranging from adenoma initiation to metastatic spread of tumor cells. As fibroblasts show considerable heterogeneity in subsets and phenotypes during cancer development, a better functional understanding of stage-specific (alterations in) fibroblast/CAF populations is key to increase the effectiveness of fibroblast-based prognosticators and therapies. Abstract In intestinal homeostasis, continuous renewal of the epithelium is crucial to withstand the plethora of stimuli which can damage the structural integrity of the intestines. Fibroblasts contribute to this renewal by facilitating epithelial cell differentiation as well as providing the structural framework in which epithelial cells can regenerate. Upon dysregulation of intestinal homeostasis, (pre-) malignant neoplasms develop, a process which is accompanied by (epi) genetic alterations in epithelial cells as well as phenotypic changes in fibroblast populations. In the context of invasive carcinomas, these fibroblast populations are termed cancer-associated fibroblasts (CAFs). CAFs are the most abundant cell type in the tumor microenvironment of colorectal cancer (CRC) and consist of various functionally heterogeneous subsets which can promote or restrain cancer progression. Although most previous research has focused on the biology of epithelial cells, accumulating evidence shows that certain fibroblast subsets can also importantly contribute to tumor initiation and progression, thereby possibly providing avenues for improvement of clinical care for CRC patients. In this review, we summarized the current literature on the emerging role of fibroblasts in various stages of CRC development, ranging from adenoma initiation to the metastatic spread of cancer cells. In addition, we highlighted translational and therapeutic perspectives of fibroblasts in the different stages of intestinal tumor progression.
Collapse
|
144
|
Mo Z, Cheong JYA, Xiang L, Le MTN, Grimson A, Zhang DX. Extracellular vesicle-associated organotropic metastasis. Cell Prolif 2021; 54:e12948. [PMID: 33145869 PMCID: PMC7791170 DOI: 10.1111/cpr.12948] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/28/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Metastasis refers to the progressive dissemination of primary tumour cells and their colonization of other tissues and is associated with most cancer-related mortalities. The disproportional and systematic distribution pattern of distant metastasis in different cancers has been well documented, as is termed metastatic organotropism, a process orchestrated by a combination of anatomical, pathophysiological, genetic and biochemical factors. Extracellular vesicles (EVs), nanosized cell-derived membrane-bound particles known to mediate intercellular communication, are now considered crucial in organ-specific metastasis. Here, we review and summarize recent findings regarding EV-associated organotropic metastasis as well as some of the general mechanisms by which EVs contribute to this important process in cancer and provide a future perspective on this emerging topic. We highlight studies that demonstrate a role of tumour-derived EVs in organotropic metastasis via pre-metastatic niche modulation. The bioactive cargo carried by EVs is of diagnostic and prognostic values, and counteracting the functions of such EVs may be a novel therapeutic strategy targeting metastasis. Further investigations are warranted to better understand the functions and mechanisms of EVs in organotropic metastasis and accelerate the relevant clinical translation.
Collapse
Affiliation(s)
- Zhenzhen Mo
- Department of PaediatricsPeople's Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Jia Yang Alex Cheong
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Lirong Xiang
- Department of PaediatricsPeople's Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Minh T. N. Le
- Institute for Digital Medicine and Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Andrew Grimson
- Department of Molecular Biology and GeneticsCornell UniversityIthacaNYUSA
| | - Daniel Xin Zhang
- Department of Biomedical SciencesJockey Club College of Veterinary Medicine and Life SciencesCity University of Hong KongKowloonHong Kong SAR
- Department of Molecular Biology and GeneticsCornell UniversityIthacaNYUSA
| |
Collapse
|
145
|
Pro-Inflammatory Cytokines in the Formation of the Pre-Metastatic Niche. Cancers (Basel) 2020; 12:cancers12123752. [PMID: 33322216 PMCID: PMC7764404 DOI: 10.3390/cancers12123752] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The formation of the pre-metastatic niche, a favorable microenvironment in an organ distant from a primary tumor, is critical for tumor metastasis. We review the role of a key player, a class of proteins named pro-inflammatory cytokines secreted from both tumor cells and other cells in tissues, in helping to build the pre-metastatic niche. Various drugs have been developed to target pro-inflammatory cytokines, and their effects on tumor metastases are under investigation. Future clinical studies should focus on combining those drugs and applying them during cancer surgery, a critical moment for the establishment of the pre-metastatic niche. Abstract In the presence of a primary tumor, the pre-metastatic niche is established in secondary organs as a favorable microenvironment for subsequent tumor metastases. This process is orchestrated by bone marrow-derived cells, primary tumor-derived factors, and extracellular matrix. In this review, we summarize the role of pro-inflammatory cytokines including interleukin (IL)-6, IL-1β, CC-chemokine ligand 2 (CCL2), granulocyte-colony stimulating factor (G-CSF), granulocyte–macrophage colony-stimulating factor (GM-CSF), stromal cell-derived factor (SDF)-1, macrophage migration inhibitory factor (MIF), and Chemokine (C–X–C motif) ligand 1 (CXCL1) in the formation of the pre-metastatic niche according to the most recent studies. Pro-inflammatory cytokines released from tumor cells or stromal cells act in both autocrine and paracrine manners to induce phenotype changes in tumor cells, recruit bone marrow-derived cells, and form an inflammatory milieu, all of which prime a secondary organ’s microenvironment for metastatic cell colonization. Considering the active involvement of pro-inflammatory cytokines in niche formation, clinical strategies targeting them offer ways to inhibit the establishment of the pre-metastatic niche and therefore attenuate metastatic progression. We review clinical trials targeting different inflammatory cytokines in patients with metastatic cancers. Due to the pleiotropy and redundancy of pro-inflammatory cytokines, combined therapies should be designed in the future.
Collapse
|
146
|
Sawaisorn P, Atjanasuppat K, Anurathapan U, Chutipongtanate S, Hongeng S. Strategies to Improve Chimeric Antigen Receptor Therapies for Neuroblastoma. Vaccines (Basel) 2020; 8:vaccines8040753. [PMID: 33322408 PMCID: PMC7768386 DOI: 10.3390/vaccines8040753] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/04/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Chimeric antigen receptors (CARs) are among the curative immunotherapeutic approaches that exploit the antigen specificity and cytotoxicity function of potent immune cells against cancers. Neuroblastomas, the most common extracranial pediatric solid tumors with diverse characteristics, could be a promising candidate for using CAR therapies. Several methods harness CAR-modified cells in neuroblastoma to increase therapeutic efficiency, although the assessment has been less successful. Regarding the improvement of CARs, various trials have been launched to overcome insufficient capacity. However, the reasons behind the inadequate response against neuroblastoma of CAR-modified cells are still not well understood. It is essential to update the present state of comprehension of CARs to improve the efficiency of CAR therapies. This review summarizes the crucial features of CARs and their design for neuroblastoma, discusses challenges that impact the outcomes of the immunotherapeutic competence, and focuses on devising strategies currently being investigated to improve the efficacy of CARs for neuroblastoma immunotherapy.
Collapse
Affiliation(s)
- Piamsiri Sawaisorn
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.S.); (K.A.); (U.A.)
| | - Korakot Atjanasuppat
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.S.); (K.A.); (U.A.)
| | - Usanarat Anurathapan
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.S.); (K.A.); (U.A.)
| | - Somchai Chutipongtanate
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
- Correspondence: (S.C.); (S.H.)
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.S.); (K.A.); (U.A.)
- Correspondence: (S.C.); (S.H.)
| |
Collapse
|
147
|
He X, Zhong X, Hu Z, Zhao S, Wei P, Li D. An insight into small extracellular vesicles: Their roles in colorectal cancer progression and potential clinical applications. Clin Transl Med 2020; 10:e249. [PMID: 33377655 PMCID: PMC7733319 DOI: 10.1002/ctm2.249] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers and a leading cause of mortality worldwide. Small extracellular vesicles (sEVs) are nano-sized extracellular vesicles containing a variety of bioactive molecules, such as nucleic acids, proteins, lipids, and metabolites. Recent evidence from CRC has revealed that sEVs contribute to tumorigenesis, progression, and drug resistance, and serve as a tool for "liquid biopsy" and a drug delivery system for therapy. In this review, we summarize information about the roles of sEVs in the proliferation, invasion, migration, epithelial-mesenchymal transition, formation of the premetastatic niche, and drug resistance to elucidate the mechanisms governing sEVs in CRC and to identify novel targets for therapy and prognostic and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuefeng He
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xinyang Zhong
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zijuan Hu
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Senlin Zhao
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Ping Wei
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Dawei Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
148
|
Marvin DL, Heijboer R, ten Dijke P, Ritsma L. TGF-β signaling in liver metastasis. Clin Transl Med 2020; 10:e160. [PMID: 33252863 PMCID: PMC7701955 DOI: 10.1002/ctm2.160] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
The presence of liver metastases drastically worsens the prognosis of cancer patients. The liver is the second most prevalent metastatic site in cancer patients, but systemic therapeutic opportunities that target liver metastases are still limited. To aid the discovery of novel treatment options for metastatic liver disease, we provide insight into the cellular and molecular steps required for liver colonization. For successful colonization in the liver, adaptation of tumor cells and surrounding stroma is essential. This includes the formation of a pre-metastatic niche, the creation of a fibrotic and immune suppressive environment, angiogenesis, and adaptation of tumor cells. We illustrate that transforming growth factor β (TGF-β) is a central cytokine in all these processes. At last, we devise that future research should focus on TGF-β inhibitory strategies, especially in combination with immunotherapy. This promising systemic treatment strategy has potential to eliminate distant metastases as the efficacy of immunotherapy will be enhanced.
Collapse
Affiliation(s)
- Dieuwke L Marvin
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Rosan Heijboer
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Laila Ritsma
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
149
|
Li F, Zhao X, Sun R, Ou J, Huang J, Yang N, Xu T, Li J, He X, Li C, Yang M, Zhang Q. EGFR-rich extracellular vesicles derived from highly metastatic nasopharyngeal carcinoma cells accelerate tumour metastasis through PI3K/AKT pathway-suppressed ROS. J Extracell Vesicles 2020; 10:e12003. [PMID: 33304472 PMCID: PMC7710133 DOI: 10.1002/jev2.12003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/02/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is the most common cancer with high metastatic potential that occurs in the epithelial cells of the nasopharynx. Distant metastases are the primary cause for treatment failure and mortality of NPC patients. However, the underlying mechanism responsible for the initiation of tumour cell dissemination and tumour metastasis in NPC is not well understood. Here, we demonstrated that epidermal growth factor receptor (EGFR) was highly expressed in tumour tissues of NPC patients with distant metastases and was associated with a decrease in reactive oxygen species (ROS). We also revealed that extracellular vesicles (EVs) transfer occurred from highly to poorly metastatic NPC cells, mediating cell-cell communication and enhancing the metastatic potential of poorly metastatic NPC cells. Further experiments indicated that EVs derived from highly metastatic NPC cells induced the up-regulation of EGFR and down-regulation of ROS in low metastatic NPC cells. Mechanistically, EGFR-rich EVs-mediated EGFR overexpression down-regulated intracellular ROS levels through the PI3K/AKT pathway, thus promoting the metastatic potential of poorly metastatic NPC cells. Strikingly, treatment with EVs secreted from highly metastatic NPC cells was significantly associated with rapid NPC progression and shorter survival in xenografted mice. These findings not only improve our understanding of EVs-mediated NPC metastatic mechanism but also have important implications for the detection and treatment of NPC patients accompanied by aberrant EGFR-rich EVs transmission.
Collapse
Affiliation(s)
- Fei Li
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat-sen University Guangzhou China
| | - Xin Zhao
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat-sen University Guangzhou China
| | - Rui Sun
- Department of Nasopharyngeal Carcinoma State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-sen University Cancer Center Guangzhou China.,Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy Guangzhou China
| | - Jinxin Ou
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat-sen University Guangzhou China
| | - Junyu Huang
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat-sen University Guangzhou China
| | - Nanyan Yang
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat-sen University Guangzhou China
| | - Ting Xu
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat-sen University Guangzhou China
| | - Jingyao Li
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat-sen University Guangzhou China
| | - Xiner He
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat-sen University Guangzhou China
| | - Chaoyi Li
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat-sen University Guangzhou China
| | - Mo Yang
- The Seventh Affiliated Hospital Sun Yat-sen University Shenzhen China.,Lianjiang People's Hospital Lianjiang China
| | - Qing Zhang
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat-sen University Guangzhou China.,Institute of Sun Yat-sen University in Shenzhen Shenzhen China
| |
Collapse
|
150
|
Blavier L, Yang RM, DeClerck YA. The Tumor Microenvironment in Neuroblastoma: New Players, New Mechanisms of Interaction and New Perspectives. Cancers (Basel) 2020; 12:cancers12102912. [PMID: 33050533 PMCID: PMC7599920 DOI: 10.3390/cancers12102912] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
The contribution of the tumor microenvironment (TME) to cancer progression has been well recognized in recent decades. As cancer therapeutic strategies are increasingly precise and include immunotherapies, knowledge of the nature and function of the TME in a tumor becomes essential. Our understanding of the TME in neuroblastoma (NB), the second most common solid tumor in children, has significantly progressed from an initial focus on its Schwannian component to a better awareness of its complex nature, which includes not only immune but also non-immune cells such as cancer-associated fibroblasts (CAFs), the contribution of which to inflammation and interaction with tumor-associated macrophages (TAMs) is now recognized. Recent studies on the TME landscape of NB tumors also suggest significant differences between MYCN-amplified (MYCN-A) and non-amplified (MYCN-NA) tumors, in their content in stromal and inflammatory cells and their immunosuppressive activity. Extracellular vesicles (EVs) released by cells in the TME and microRNAs (miRs) present in their cargo could play important roles in the communication between NB cells and the TME. This review article discusses these new aspects of the TME in NB and the impact that information on the TME landscape in NB will have in the design of precise, biomarker-integrated clinical trials.
Collapse
Affiliation(s)
- Laurence Blavier
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (L.B.); (R.-M.Y.)
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ren-Ming Yang
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (L.B.); (R.-M.Y.)
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yves A. DeClerck
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (L.B.); (R.-M.Y.)
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Correspondence: ; Tel.: +1-323-382-5548 or +1-323-361-5648
| |
Collapse
|