1
|
Yang L, Lin Z, Mu R, Wu W, Zhi H, Liu X, Yang H, Liu L. Neurons enhance blood-brain barrier function via upregulating claudin-5 and VE-cadherin expression due to glial cell line-derived neurotrophic factor secretion. eLife 2024; 13:RP96161. [PMID: 39475379 PMCID: PMC11524583 DOI: 10.7554/elife.96161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
Blood-brain barrier (BBB) prevents neurotoxins from entering central nervous system. We aimed to establish and characterize an in vitro triple co-culture BBB model consisting of brain endothelial cells hCMEC/D3, astrocytoma U251 cells, and neuroblastoma SH-SY5Y cells. Co-culture of SH-SY5Y and U251 cells markedly enhanced claudin-5 and VE-cadherin expression in hCMEC/D3 cells, accompanied by increased transendothelial electrical resistance and decreased permeability. Conditioned medium (CM) from SH-SY5Y cells (S-CM), U251 cells (U-CM), and co-culture of SH-SY5Y and U251 cells (US-CM) also promoted claudin-5 and VE-cadherin expression. Glial cell line-derived neurotrophic factor (GDNF) levels in S-CM and US-CM were significantly higher than CMs from hCMEC/D3 and U-CM. Both GDNF and US-CM upregulated claudin-5 and VE-cadherin expression, which were attenuated by anti-GDNF antibody and GDNF signaling inhibitors. GDNF increased claudin-5 expression via the PI3K/AKT/FOXO1 and MAPK/ERK pathways. Meanwhile, GDNF promoted VE-cadherin expression by activating PI3K/AKT/ETS1 and MAPK/ERK/ETS1 signaling. The roles of GDNF in BBB integrity were validated using brain-specific Gdnf silencing mice. The developed triple co-culture BBB model was successfully applied to predict BBB permeability. In conclusion, neurons enhance BBB integrity by upregulating claudin-5 and VE-cadherin expression through GDNF secretion and established triple co-culture BBB model may be used to predict drugs' BBB permeability.
Collapse
Affiliation(s)
- Lu Yang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Zijin Lin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Ruijing Mu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Wenhan Wu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Hao Zhi
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Xiaodong Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Hanyu Yang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| | - Li Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical UniversityNanjingChina
| |
Collapse
|
2
|
Zhang Y, Yang Y, Hu X, Wei B, Shen Q, Shi C, Chen P. RAS protein activator-like 2 (RASAL2) initiates peritubular capillary rarefaction in hypoxic renal interstitial fibrosis. Transl Res 2024; 269:14-30. [PMID: 38453052 DOI: 10.1016/j.trsl.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
The progression of chronic kidney disease (CKD) often involves renal interstitial fibrosis (RIF) and subsequent loss of peritubular capillaries (PTCs), which enhances disease severity. Despite advancements in our understanding of fibrosis, effective interventions for reversing capillary loss remain elusive. Notably, RIF exhibits reduced capillary density, whereas renal cell carcinoma (RCC) shows robust angiogenesis under hypoxic conditions. Using RNA sequencing and bioinformatics, we identified differentially expressed genes (DEGs) in hypoxic human renal tubular epithelial cells (HK-2) and renal cancer cells (786-0). Analysis of altered Ras and PI3K/Akt pathways coupled with hub gene investigation revealed RAS protein activator-like 2 (RASAL2) as a key candidate. Subsequent in vitro and in vivo studies confirmed RASAL2's early-stage response in RIF, which reduced with fibrosis progression. RASAL2 suppression in HK-2 cells enhanced angiogenesis, as evidenced by increased proliferation, migration, and branching of human umbilical vein endothelial cells (HUVECs) co-cultured with HK-2 cells. In mice, RASAL2 knockdown improved Vascular endothelial growth factor A (VEGFA) and Proliferating cell nuclear antigen (PCNA) levels in unilateral ureteral occlusion (UUO)-induced fibrosis (compared to wild type). Hypoxia-inducible factor 1 alpha (HIF-1α) emerged as a pivotal mediator, substantiated by chromatin immunoprecipitation (ChIP) sequencing, with its induction linked to activation. Hypoxia increased the production of RASAL2-enriched extracellular vesicles (EVs) derived from tubular cells, which were internalized by endothelial cells, contributing to the exacerbation of PTC loss. These findings underscore RASAL2's role in mediating reduced angiogenesis in RIF and reveal a novel EV-mediated communication between hypoxic tubular- and endothelial cells, demonstrating a complex interplay between angiogenesis and fibrosis in CKD pathogenesis.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Yiqiong Yang
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Xiuxiu Hu
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Bizhen Wei
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Qian Shen
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Chuanbing Shi
- Department of Pathology, Pukou Branch of Jiangsu People's Hospital, Nanjing, China
| | - Pingsheng Chen
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China; Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
3
|
Abstract
PURPOSE OF THE REVIEW Angiogenesis plays a key role in bladder cancer (BC) pathogenesis. In the last two decades, an increasing number of publications depicting a multitude of novel angiogenic molecules and pathways have emerged. The growing complexity necessitates an evaluation of the breadth of current knowledge to highlight key findings and guide future research. RECENT FINDINGS Angiogenesis is a dynamic biologic process that is inherently difficult to assess. Clinical assessment of angiogenesis in BCs is advancing with the integration of image analysis systems and dynamic contrast-enhanced and magnetic resonance imaging (DCE-MRI). Tumour-associated macrophages (TAMs) significantly influence the angiogenic process, and further research is needed to assess their potential as therapeutic targets. A rapidly growing list of non-coding RNAs affect angiogenesis in BCs, partly through modulation of vascular endothelial growth factor (VEGF) activity. Vascular mimicry (VM) has been repeatedly associated with increased tumour aggressiveness in BCs. Standardised assays are needed for appropriate identification and quantification of VM channels. This article demonstrates the dynamic and complex nature of the angiogenic process and asserts the need for further studies to deepen our understanding.
Collapse
Affiliation(s)
- Ghada Elayat
- Department of Natural Science, Middlesex University, London, UK
- Department of Histopathology, Tanta University, Tanta, Egypt
| | - Ivan Punev
- Department of Natural Science, Middlesex University, London, UK
| | - Abdel Selim
- Histopathology Department, King’s Health Partners, King’s College Hospital, London, UK
| |
Collapse
|
4
|
He F, Wang QF, Li L, Yu C, Liu CZ, Wei WC, Chen LP, Li HY. Melatonin Protects Against Hyperoxia-Induced Apoptosis in Alveolar Epithelial type II Cells by Activating the MT2/PI3K/AKT/ETS1 Signaling Pathway. Lung 2023; 201:225-234. [PMID: 36928143 DOI: 10.1007/s00408-023-00610-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE Hyperoxia-induced apoptosis in alveolar epithelial type II cells (AECIIs) plays a critical role in the development of bronchopulmonary dysplasia (BPD). Melatonin has been shown to improve BPD. However, the protective effect of melatonin on hyperoxia-induced apoptosis in AECIIs and the precise mechanisms involved remain unclear. METHODS Human alveolar epithelial type II A549 cells were treated with hyperoxia as an in vitro model to investigate the antiapoptotic mechanism of melatonin. CCK-8 assays were performed to investigate the viability of A549 cells. Hoechst 33,258 staining was carried out to quantify apoptosis in A549 cells. The protein expression levels of E26 oncogene homolog 1 (ETS1), Bcl-2, Bax, Bim, Wnt, β-catenin, AKT and phosphorylated AKT were measured by western blotting. LY294002, SC79 and the downregulation of ETS1, melatonin receptor 1 (MT1) and MT2 with specific siRNAs were used to investigate the role of the PI3K/AKT pathway, ETS1, MT1 and MT2 in hyperoxia-induced apoptosis in A549 cells. RESULTS Melatonin prevented hyperoxia-induced apoptosis in A549 cells, and the upregulation of E26 oncogene homolog 1 (ETS1) contributed to the antiapoptotic effect of melatonin. Melatonin activated the PI3K/AKT axis, which led to ETS1 upregulation and inhibited apoptosis in hyperoxia-exposed A549 cells. Furthermore, melatonin-induced activation of the PI3K/AKT axis, upregulation of ETS1 and inhibition of apoptosis were reversed by melatonin receptor 2 (MT2) siRNA in hyperoxia-exposed A549 cells. CONCLUSION Melatonin prevents hyperoxia-induced apoptosis by activating the MT2/PI3K/AKT/ETS1 axis in alveolar epithelial cells.
Collapse
Affiliation(s)
- Fan He
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Qiao-Fang Wang
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Lin Li
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Cong Yu
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Chun-Zi Liu
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Wen-Chao Wei
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Li-Ping Chen
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China.
| | - Huan-Yu Li
- Second Department of Respiratory Disease, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No. 152, Aiguo Road, DongHu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
5
|
Yue Y, Wu K, Qian W, Zhu Z, Zhang S, Zhang W, Zhang W, Wu S, Li L, Wu Z, Ma Q, Xie K, Wang Z. RASAL2 mediated the enhancement of YAP1/TIAM1 signaling promotes malignant phenotypes of pancreatic ductal adenocarcinoma. Int J Biol Sci 2022; 18:4245-4259. [PMID: 35844783 PMCID: PMC9274491 DOI: 10.7150/ijbs.72204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/14/2022] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a high incidence of metastasis and dismal prognosis. As a member of Gas-Gap gene, RASAL2 is involved in the hydrolysis of RAS-GTP to RAS-GDP and abnormal expression in human cancers. Here we firstly described the function of RASAL2 on PDAC to enrich the knowledge of RAS family.We interestingly observed that RASAL2 expression was upregulated in PDAC at both mRNA and protein levels, and high expression of RASAL2 predicted a poor prognosis in PDAC patients. Additionally, RASAL2 promoted malignant behaviors of PDAC in vitro and in vivo. To determine the mechanistic roles of RASAL2 signaling and its potential as a therapeutic target in PDAC, we clarified that RASAL2 could accumulate the TIAM1 expression in different level through inhibiting YAP1 phosphorylation, increased TIAM1 mRNA expression and suppressed ubiquitination of TIAM1 protein. In conclusion, RASAL2 enhances YAP1/TIAM1 signaling and promotes PDAC development and progression.
Collapse
Affiliation(s)
- Yangyang Yue
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.,Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Kaijie Wu
- Department of Urology Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zeen Zhu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Simei Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wunai Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Weifan Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Shuai Wu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Li Li
- Department of Ophthalmology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Keping Xie
- Center for Pancreatic Cancer Research, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
6
|
Wang Q, Wu S, Gu Y, Liang H, He F, Wang X, He D, Wu K. RASAL2 regulates the cell cycle and cyclin D1 expression through PI3K/AKT signalling in prostate tumorigenesis. Cell Death Dis 2022; 8:275. [PMID: 35668070 PMCID: PMC9170709 DOI: 10.1038/s41420-022-01069-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/30/2022] [Accepted: 05/25/2022] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PCa) growth and progression are uniquely dependent on androgens, making the androgen receptor pathway a prime target for therapy; however, cancer progression to androgen independence leads to treatment failure and poor prognosis. In recent years, alternative therapeutic pathways for PCa have been extensively explored, such as the PTEN/PI3K/AKT pathway, cell cycle, and DNA repair. In the present study, we discovered that RASAL2, a RAS-GTPase-activating protein, acted as an oncogene to regulate cancer cell proliferation and the cell cycle and contributed to tumorigenesis via the PI3K/AKT/cyclin D1 pathway. First, RASAL2 expression was higher in PCa tumour and metastatic lymph node tissues than in matched adjacent nontumor tissues and was associated with higher PCa tumour stage, Gleason score and poorer prognosis. Mechanistically, we found that RASAL2 promoted tumour cell proliferation, the transition from G1 to S phase in vitro and tumour growth in vivo. Furthermore, we demonstrated that RASAL2 facilitated phosphorylation of AKT, which in turn increased the expression of cyclin D1 encoded by the CCND1 gene. In addition, there was a positive correlation between the expression of RASAL2 and cyclin D1 in subcutaneous xenografts and clinical specimens. Taken together, these findings indicate that RASAL2 plays an oncogenic role in prostate cancer and may promote PCa tumorigenesis through PI3K/AKT signalling and cyclin D1 expression.
Collapse
Affiliation(s)
- Qi Wang
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Shiqi Wu
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Yanan Gu
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Hua Liang
- Department of Pathology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Fei He
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Xinyang Wang
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Dalin He
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Kaijie Wu
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China.
| |
Collapse
|
7
|
Hu Y, Zhang P, Shi Y, Dong X, Wu Y, Dong D, Li E, Fan Y. Inhibition of Ras protein activator like 2 produces antitumor effects in gastric cancer via the suppression of YAP1 activation. ENVIRONMENTAL TOXICOLOGY 2022; 37:527-538. [PMID: 34826200 DOI: 10.1002/tox.23418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/28/2021] [Accepted: 11/14/2021] [Indexed: 06/13/2023]
Abstract
Ras protein activator like 2 (RASAL2) has a cancer-related function, but plays inconsistent roles in different malignancies. This project was designed to determine the role of RASAL2 in carcinogenesis in gastric cancer. The Cancer Genome Atlas data revealed high levels of RASAL2 in gastric cancer tissue, which was confirmed in clinical specimens of gastric cancer via real-time quantitative PCR and western blotting assays. High RASAL2 was correlated with a reduced survival rate in gastric cancer patients. In gastric cancer cell lines, the silencing of RASAL2 restrained cellular proliferation, invasion and epithelial-to-mesenchymal transition, while enhancing chemosensitivity to cisplatin. Mechanistically, the silencing of RASAL2 was found to inhibit the activation of Yes-associated protein 1 (YAP1), a pro-oncogenic protein in gastric cancer, and decrease the expression of YAP1 target genes. The re-expression of constitutively active YAP1 substantially reversed RASAL2-silencing-produced antitumor effects. Moreover, treatment with YAP1 inhibitors could diminish RASAL2-overexpression-evoked oncogenic effects in gastric cancer cells. Additionally, gastric cancer cells with RASAL2 silencing exhibited a reduced ability to form xenograft tumors in nude mice. Collectively, our data demonstrate that the silencing of RASAL2 has noteworthy antitumor effects in gastric cancer cells via the suppression of YAP1 activation. This project underscores a vital role of the RASAL2/YAP1 axis in gastric progression and indicates that targeting this oncogenic axis may be applied as a potential therapeutic option for gastric cancer.
Collapse
Affiliation(s)
- Yuan Hu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Pengchuang Zhang
- Department of Gynecologic Cancer, Shaanxi Provincial Tumor Hospital, China
| | - Yu Shi
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Xuyuan Dong
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yinying Wu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Danfeng Dong
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yangwei Fan
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| |
Collapse
|
8
|
Tailor K, Paul J, Ghosh S, Kumari N, Kwabi-Addo B. RASAL2 suppresses the proliferative and invasive ability of PC3 prostate cancer cells. Oncotarget 2021; 12:2489-2499. [PMID: 34966481 PMCID: PMC8711570 DOI: 10.18632/oncotarget.28158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 12/08/2021] [Indexed: 11/25/2022] Open
Abstract
The RAS protein activator like 2 (RASAL2) negatively regulates RAS proto-oncogene which is activated by high mutation rate in cancer. Thus, RASAL2 expression could potentially limit the function of RAS in prostate cancer (PCa). Genome-wide DNA methylation analysis demonstrated that RASAL2 is differentially hypermethylated in PCa tissues compared to benign prostate tissues. The PCR analysis of RASAL2 mRNA transcript showed differential expression in a panel of prostate cell lines with most PCa showing lower RASAL2 expression compared to benign prostatic epithelial cells. In PCa PC3 cells, the ectopic expression of RASAL2 significantly inhibited cell proliferation and invasion and induced an S phase plus G2/M phase cell cycle arrest. Ingenuity Pathway Analysis (IPA) demonstrated a cross talk between RASAL2 and TNFα, a key cytokine in immune signaling pathway that is relevant in PCa. Over-expression of RASAL2 downregulated TNFα expression whereas the knockdown of RASAL2 caused increased expression of TNFα. Taken together, our data demonstrates tumor suppressor role for RASAL2 in human PCa cells, despite increased RAS oncogenic activity. Our observation provides a new mechanistic insight of RASAL2 expression in aberrant Ras expression and immune signaling in PCa cells suggesting a potential novel therapeutic target for PCa.
Collapse
Affiliation(s)
- Krishma Tailor
- 1Department of Biochemistry and Molecular Biology, Howard University, Washington, DC 20059, USA
| | - Joseph Paul
- 1Department of Biochemistry and Molecular Biology, Howard University, Washington, DC 20059, USA
| | - Somiranjan Ghosh
- 2Department of Biology, Howard University, Washington, DC 20059, USA
| | - Namita Kumari
- 3Center for Sickle Cell Disease, Howard University, Washington, DC 20059, USA
| | - Bernard Kwabi-Addo
- 1Department of Biochemistry and Molecular Biology, Howard University, Washington, DC 20059, USA,Correspondence to:Bernard Kwabi-Addo, email:
| |
Collapse
|
9
|
Zhu W, Gao F, Zhou H, Jin K, Shao J, Xu Z. Knockdown of MCM8 inhibits development and progression of bladder cancer in vitro and in vivo. Cancer Cell Int 2021; 21:242. [PMID: 33931059 PMCID: PMC8086360 DOI: 10.1186/s12935-021-01948-2] [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: 01/15/2021] [Accepted: 04/22/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Bladder cancer is a frequently diagnosed urinary system tumor, whose mortality remains rising. Minichromosome maintenance eight homologous recombination repair factor (MCM8), a newly discovered MCM family member, has been shown to be required for DNA replication. Unfortunately, little is known concerning the roles of MCM8 in bladder cancer. METHODS The present study, we aimed at probing into the impacts and detailed mechanisms of MCM8 in bladder cancer progression. In this study, MCM8 expression level was detected through immunohistochemistry staining (IHC), qRT-PCR and Western blot assay. Silenced MCM8 cell models were constructed by lentivirus transfection. In vitro, the cell proliferation was evaluated by the MTT assay. The wound-healing assay and the transwell assay were utilized to assess the cell migration. Also, the cell apoptosis and the cell cycle were determined by flow cytometry. Moreover, the Human Apoptosis Antibody Array assay was performed to analyze the alterations of apoptosis-related proteins. The in vivo experiments were conducted to verify the effects of MCM8 knockdown on the tumor growth of bladder cancer. RESULTS The results demonstrated that compared with normal adjacent tissues, MCM8 expression in bladder cancer tissues was strongly up-regulated. The up-regulation of MCM8 expression in bladder cancer may be a valuable independent prognostic indicator. Of note, MCM8 inhibition modulated the malignant phenotypes of bladder cancer cells. In terms of mechanism, it was validated that MCM8 knockdown made Akt, P-Akt, CCND1 and CDK6 levels down-regulated, as well as MAPK9 up-regulated. CONCLUSIONS Taken together, our study demonstrated an important role of MCM8 in bladder cancer and created a rationale for the therapeutic potential of MCM8 inhibition in human bladder cancer therapy.
Collapse
Affiliation(s)
- Wei Zhu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.,Department of Urology, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Fei Gao
- Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qingyang Rd, Wuxi, 214023, China
| | - Hongyi Zhou
- Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qingyang Rd, Wuxi, 214023, China
| | - Ke Jin
- Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qingyang Rd, Wuxi, 214023, China
| | - Jianfeng Shao
- Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qingyang Rd, Wuxi, 214023, China.
| | - Zhuoqun Xu
- Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qingyang Rd, Wuxi, 214023, China.
| |
Collapse
|
10
|
Bao Y, Qian C, Liu MY, Jiang F, Jiang X, Liu H, Zhang Z, Sun F, Fu N, Hou Z, Ke Y, Li Y, Qian ZM. PRKAA/AMPKα phosphorylation switches the role of RASAL2 from a suppressor to an activator of autophagy. Autophagy 2021; 17:3607-3621. [PMID: 33563064 DOI: 10.1080/15548627.2021.1886767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RASAL2 (RAS protein activator like 2), a RASGTPase activating protein, can catalyze the hydrolysis of RAS-GTP into RAS-GDP to inactivate the RAS pathway in various types of cancer cells. However, the cellular function of RASAL2 remains elusive. Here we showed that RASAL2 can attenuate PRKAA/AMPKα phosphorylation by recruiting phosphatase PPM1B/pp2cβ, thus inhibiting the initiation of basal autophagy under normal conditions. In addition, we found that glucose starvation could induce dissociation of PPM1B from RASAL2 and then RASAL2 at S351 be phosphorylated by PRKAA, followed by the binding of phosphorylated-RASAL2 with to PIK3C3/VPS34-ATG14-BECN1/Beclin1 complex to increase PIK3C3 activity and autophagy. Furthermore, RASAL2 S351 phosphorylation facilitated breast tumor growth and correlated to poor clinical outcomes in breast cancer patients. Our study demonstrated that the phosphorylation status of RASAL2 S351 can function as a molecular switch to either suppress or promote AMPK-mediated autophagy. Inhibition of RASAL2 S351 phosphorylation might be a potential therapeutic strategy to overcome the resistance of AMPK-activation agents.
Collapse
Affiliation(s)
- Yong Bao
- Institute of Translational and Precision Medicine, Nantong University, Nantong, China.,Department of Pharmacology and Biochemistry, Fudan University School of Pharmacy, Shanghai, China
| | - Christopher Qian
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Meng-Yue Liu
- Institute of Translational and Precision Medicine, Nantong University, Nantong, China
| | - Fei Jiang
- Institute of Translational and Precision Medicine, Nantong University, Nantong, China
| | - Xiaoxiao Jiang
- Department of Pharmacology and Biochemistry, Fudan University School of Pharmacy, Shanghai, China
| | - Huijuan Liu
- Department of Pharmacology and Biochemistry, Fudan University School of Pharmacy, Shanghai, China
| | - Zhuqing Zhang
- Department of Pharmacology and Biochemistry, Fudan University School of Pharmacy, Shanghai, China
| | - Fanghui Sun
- Department of Pharmacology and Biochemistry, Fudan University School of Pharmacy, Shanghai, China
| | - Ningwei Fu
- Department of Anatomy and Physiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhaoyuan Hou
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry & Molecular Cellular Biology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yan Li
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry & Molecular Cellular Biology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhong-Ming Qian
- Institute of Translational and Precision Medicine, Nantong University, Nantong, China
| |
Collapse
|
11
|
Ashrafizadeh M, Hushmandi K, Hashemi M, Akbari ME, Kubatka P, Raei M, Koklesova L, Shahinozzaman M, Mohammadinejad R, Najafi M, Sethi G, Kumar AP, Zarrabi A. Role of microRNA/Epithelial-to-Mesenchymal Transition Axis in the Metastasis of Bladder Cancer. Biomolecules 2020; 10:E1159. [PMID: 32784711 PMCID: PMC7464913 DOI: 10.3390/biom10081159] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
Bladder cancer (BC) is the 11th most common diagnosed cancer, and a number of factors including environmental and genetic ones participate in BC development. Metastasis of BC cells into neighboring and distant tissues significantly reduces overall survival of patients with this life-threatening disorder. Recently, studies have focused on revealing molecular pathways involved in metastasis of BC cells, and in this review, we focus on microRNAs (miRNAs) and their regulatory effect on epithelial-to-mesenchymal transition (EMT) mechanisms that can regulate metastasis. EMT is a vital process for migration of BC cells, and inhibition of this mechanism restricts invasion of BC cells. MiRNAs are endogenous non-coding RNAs with 19-24 nucleotides capable of regulating different cellular events, and EMT is one of them. In BC cells, miRNAs are able to both induce and/or inhibit EMT. For regulation of EMT, miRNAs affect different molecular pathways such as transforming growth factor-beta (TGF-β), Snail, Slug, ZEB1/2, CD44, NSBP1, which are, discussed in detail this review. Besides, miRNA/EMT axis can also be regulated by upstream mediators such as lncRNAs, circRNAs and targeted by diverse anti-tumor agents. These topics are also discussed here to reveal diverse molecular pathways involved in migration of BC cells and strategies to target them to develop effective therapeutics.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran;
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran;
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran;
| | - Mohammad Esmaeil Akbari
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran 1989934148, Iran;
| | - Peter Kubatka
- Department of Medical Biology and Division of Oncology—Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran;
| | - Lenka Koklesova
- Department of Obstetrics and Gynecology, Martin University Hospital and Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Md Shahinozzaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA;
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 55877577, Iran;
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran;
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
- Cancer Science Institute of Singapore, Centre for Translational Medicine, 14 Medical Drive, #11-01M, Singapore 117599, Singapore
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Turkey
| |
Collapse
|
12
|
Li X, Li Z, Zhu Y, Li Z, Yao L, Zhang L, Yuan L, Shang Y, Liu J, Li C. miR-524-5p inhibits angiogenesis through targeting WNK1 in colon cancer cells. Am J Physiol Gastrointest Liver Physiol 2020; 318:G827-G839. [PMID: 32174132 DOI: 10.1152/ajpgi.00369.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
There is increasing evidence that microRNA (miRNA) abnormity is involved in the occurrence and the development of various malignancies, including colon cancer. MiRNA-524-5p has been reported to possess anticancer activity in various tumors, which function is seldom investigated in colon cancer cells. The aim of this study was to explore the effect of the miRNA-524-5p/with-no-lysine kinase 1 (WNK1) system on angiogenesis in a colon cancer cell line (HT-29 and COLO205 cells) and further investigate the potential mechanisms. We found miRNA-524-5p expression was relatively high in COLO205 cells and relatively low in HT-29 cells. Elevating miRNA-524-5p expression inhibited proliferation, induced cycle arrest, diminished vascular endothelial growth factor production, and thereby suppressed angiogenesis in HT-29 cells. WNK1 silencing exerted the ability of antiangiogenesis in HT-29 cells. Besides, miRNA-524-5p deficiency-induced angiogenesis was impeded by WNK1 silence in COLO205 cells. In a murine tumor model, miRNA-524-5p agomir treatment significantly suppressed colon cancer tumorigenicity with the downregulation of WNK1 expression. In summary, our results indicated that miRNA-524-5p inhibited angiogenesis in colon cancer cells via targeting WNK1.NEW & NOTEWORTHY MiRNA-524-5p inhibited angiogenesis in colon cancer cells via targeting with-no-lysine kinase 1.
Collapse
Affiliation(s)
- Xiang Li
- Department of Ultrasonic Diagnosis, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Zitao Li
- Department of Orthopedic Surgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Ye Zhu
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, People's Republic of China
| | - Zhu Li
- Image Equipment and Technology Laboratory, College of Medical Imaging, Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Lihong Yao
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Lamei Zhang
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Lin Yuan
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Yang Shang
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Jianting Liu
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Caijuan Li
- Department of Ultrasound, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China.,Key Laboratory of Tumor Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| |
Collapse
|
13
|
Harrell Stewart DR, Clark GJ. Pumping the brakes on RAS - negative regulators and death effectors of RAS. J Cell Sci 2020; 133:133/3/jcs238865. [PMID: 32041893 DOI: 10.1242/jcs.238865] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mutations that activate the RAS oncoproteins are common in cancer. However, aberrant upregulation of RAS activity often occurs in the absence of activating mutations in the RAS genes due to defects in RAS regulators. It is now clear that loss of function of Ras GTPase-activating proteins (RasGAPs) is common in tumors, and germline mutations in certain RasGAP genes are responsible for some clinical syndromes. Although regulation of RAS is central to their activity, RasGAPs exhibit great diversity in their binding partners and therefore affect signaling by multiple mechanisms that are independent of RAS. The RASSF family of tumor suppressors are essential to RAS-induced apoptosis and senescence, and constitute a barrier to RAS-mediated transformation. Suppression of RASSF protein expression can also promote the development of excessive RAS signaling by uncoupling RAS from growth inhibitory pathways. Here, we will examine how these effectors of RAS contribute to tumor suppression, through both RAS-dependent and RAS-independent mechanisms.
Collapse
Affiliation(s)
- Desmond R Harrell Stewart
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY 40222, USA
| | - Geoffrey J Clark
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY 40222, USA
| |
Collapse
|
14
|
Wang X, Qian C, Yang Y, Liu MY, Ke Y, Qian ZM. Phosphorylated Rasal2 facilitates breast cancer progression. EBioMedicine 2019; 50:144-155. [PMID: 31759919 PMCID: PMC6921363 DOI: 10.1016/j.ebiom.2019.11.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 10/24/2019] [Accepted: 11/11/2019] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Rasal2 has diametric effects on progression of oestrogen receptor-positive (ER+) and -negative (ER-) breast cancers. The relevant causes are unknown. It is also unknown whether the effects of Rasal2 are mediated by an exosome-transport process. METHODS Exosomes were purified from breast cancer cells and identified by transmission electron microscopy and flow cytometry analysis. In vivo and in vitro experiments were conducted to investigate the role of Rasal2 in exosome-mediated breast cancer progression. Western blot analysis was performed to detect Rasal2 and p-Rasal2 (phosphorylated Rasal2) expression in ER+/ER- breast cancer cells and in exosomes, cancer tissues and blood of patients with ER+ or ER- breast cancer. FINDINGS Phosphorylation of Rasal2 at Serine 237 promoted tumour growth in both ER+ and ER- tumour cells and tissues. The functions of both p-Rasal2 and non-p-Rasal2 (non-phosphorylated-Rasal2) in the modulation of breast cancer progression are exosome-mediated. p-Rasal2 expression in ER+ breast cancer cells and exosomes, cancer tissues and blood was significantly lower than in ER- tumour cells and patients. INTERPRETATION p-Rasal2 facilitates tumour progression in both ER+ and ER- breast cancers. The ratio of p-Rasal2/non-p-Rasal2 in ER+ and ER- breast cancers is one of the factors deciding the role of Rasal2 (or total Rasal2) as a suppressor in ER+ breast cancers or as a promoter in ER- breast cancers. Targeting the phosphorylation of Rasal2 machinery may therefore be useful as a therapy to restrain breast cancer progression by reducing p-Rasal2/non-p-Rasal2 ratio, especially in ER- breast cancers. FUND: NSFC and Hong Kong Research Grants Council.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Pharmacology and Biochemistry, Fudan University School of Pharmacy, 826 Zhangheng Road, Pu Dong, Shanghai 201203, China
| | - Christopher Qian
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Yinlong Yang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200000, China
| | - Meng-Yue Liu
- Institute of Translational & Precision Medicine, Nantong University, Nantong, JS 226019, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Zhong-Ming Qian
- Department of Pharmacology and Biochemistry, Fudan University School of Pharmacy, 826 Zhangheng Road, Pu Dong, Shanghai 201203, China; Institute of Translational & Precision Medicine, Nantong University, Nantong, JS 226019, China.
| |
Collapse
|
15
|
Zhou B, Zhu W, Jiang X, Ren C. RASAL2 Plays Inconsistent Roles in Different Cancers. Front Oncol 2019; 9:1235. [PMID: 31799194 PMCID: PMC6863963 DOI: 10.3389/fonc.2019.01235] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/28/2019] [Indexed: 11/18/2022] Open
Abstract
RAS protein activator like 2 (RASAL2) belongs to the RAS GTPase-activating protein family and plays an important role in several cancers, including ovarian cancer, nasopharyngeal carcinoma, malignant astrocytoma, renal cell carcinoma, bladder cancer, colorectal cancer, liver cancer, triple-negative breast cancer, lung adenocarcinoma, and pancreatic ductal adenocarcinoma. Traditionally, RASAL2 has been regarded as a tumor suppressor but recent studies have found that it is an oncogene in specific types of cancer, such as colorectal cancer, liver cancer, triple-negative breast cancer, triple-negative/estrogen receptor-negative breast cancer. In this review, we summarize the latest findings regarding RASAL2 in cancers, which may be important and useful in clinical practice. We discussed the specific functions and mechanisms of RASAL2 in different kinds of cancer cells (including its inhibition of invasion, metastasis and angiogenesis and its opposite effects), which may provide new directions for cancer research and treatments. RASAL2 exhibits different relationship with clinical cancer stage, histological grade, prognosis and overall survival in different kinds of tumor. RASAL2 is a potential prognostic factor and a new therapeutic target for diagnosis and treatment.
Collapse
Affiliation(s)
- Bolun Zhou
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Zhu
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xingjun Jiang
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Caiping Ren
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Yin L, Xiao X, Georgikou C, Luo Y, Liu L, Gladkich J, Gross W, Herr I. Sulforaphane Induces miR135b-5p and Its Target Gene, RASAL2, thereby Inhibiting the Progression of Pancreatic Cancer. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:74-81. [PMID: 31044154 PMCID: PMC6479751 DOI: 10.1016/j.omto.2019.03.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/27/2019] [Indexed: 12/21/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal tumors, with poor therapeutic options in the advanced state. The broccoli-derived anti-inflammatory agent sulforaphane was shown to inhibit the progression of pancreatic cancer and other tumor entities. We examined the involvement of pancreatic cancer cell lines were evaluated by microRNA and gene expression arrays, bioinformatics, in silico analysis, qRT-PCR, western blotting, immunohistochemistry, in situ hybridization, self-renewal and differentiation assays, and in vivo xenograft studies. We selected the top nine differentially expressed microRNAs, and miR135b-5p was chosen as the most important candidate for the sulforaphane-induced upregulation of the tumor suppressor gene RASAL2. The expression of miR135b-5p and RASAL2 was almost absent in malignant pancreatic tissues and cell lines, but not in their normal counterparts. Lipofection of miR135b-5p enhanced RASAL2 expression and inhibited ERK1/2 signaling, viability, self-renewal capacity, and tumor growth. These results indicate that miR135b-5p acts as a tumor suppressor via the induction of RASAL2 in PDA.
Collapse
Affiliation(s)
- Libo Yin
- Molecular OncoSurgery, Section of Surgical Research, Department of General, Visceral, & Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| | - Xi Xiao
- Molecular OncoSurgery, Section of Surgical Research, Department of General, Visceral, & Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| | - Christina Georgikou
- Molecular OncoSurgery, Section of Surgical Research, Department of General, Visceral, & Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| | - Yiqiao Luo
- Molecular OncoSurgery, Section of Surgical Research, Department of General, Visceral, & Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| | - Li Liu
- Molecular OncoSurgery, Section of Surgical Research, Department of General, Visceral, & Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jury Gladkich
- Molecular OncoSurgery, Section of Surgical Research, Department of General, Visceral, & Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| | - Wolfgang Gross
- Molecular OncoSurgery, Section of Surgical Research, Department of General, Visceral, & Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| | - Ingrid Herr
- Molecular OncoSurgery, Section of Surgical Research, Department of General, Visceral, & Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
17
|
Zhu M, Jiang L, Yuan Y, Chen L, Liu X, Liang J, Zhu Q, Ding D, Song E. Intravitreal Ets1 siRNA alleviates choroidal neovascularization in a mouse model of age-related macular degeneration. Cell Tissue Res 2019; 376:341-351. [PMID: 30834976 DOI: 10.1007/s00441-019-03001-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 01/29/2019] [Indexed: 12/16/2022]
Abstract
Choroidal neovascularization (CNV) is the basic feature of neovascular age-related macular degeneration (AMD), the leading cause of blindness in elders. Macrophages and microglia promote CNV via producing pro-angiogenic factors and inflammatory cytokines. Transcription factor E26 transformation specific-1 (Ets1) plays a pro-angiogenic role via its pro-inflammatory function. In this study, Ets1 increased and localized in the macrophages and microglia of a mouse laser-induced CNV region. Ets1 siRNA intravitreal injection ameliorated the leakage and area of CNV, as well as inhibiting the dysfunction of retinal pigment epithelium (RPE) cells and the activation of macrophages/microglia. Taken together, we provide a new insight into the molecular mechanism of CNV progression, in which Ets1 can be a new therapeutic target.
Collapse
Affiliation(s)
- Manhui Zhu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Li Jiang
- Department of Ophthalmology, Laizhou City People's Hospital, Yantai, Shandong, China
| | - You Yuan
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lili Chen
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Juan Liang
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiujian Zhu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Dongmei Ding
- Department of Ophthalmology, Laizhou City People's Hospital, Yantai, Shandong, China.
| | - E Song
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
18
|
Raza Q, Choi JY, Li Y, O’Dowd RM, Watkins SC, Chikina M, Hong Y, Clark NL, Kwiatkowski AV. Evolutionary rate covariation analysis of E-cadherin identifies Raskol as a regulator of cell adhesion and actin dynamics in Drosophila. PLoS Genet 2019; 15:e1007720. [PMID: 30763317 PMCID: PMC6375579 DOI: 10.1371/journal.pgen.1007720] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/07/2018] [Indexed: 12/17/2022] Open
Abstract
The adherens junction couples the actin cytoskeletons of neighboring cells to provide the foundation for multicellular organization. The core of the adherens junction is the cadherin-catenin complex that arose early in the evolution of multicellularity to link actin to intercellular adhesions. Over time, evolutionary pressures have shaped the signaling and mechanical functions of the adherens junction to meet specific developmental and physiological demands. Evolutionary rate covariation (ERC) identifies proteins with correlated fluctuations in evolutionary rate that can reflect shared selective pressures and functions. Here we use ERC to identify proteins with evolutionary histories similar to the Drosophila E-cadherin (DE-cad) ortholog. Core adherens junction components α-catenin and p120-catenin displayed positive ERC correlations with DE-cad, indicating that they evolved under similar selective pressures during evolution between Drosophila species. Further analysis of the DE-cad ERC profile revealed a collection of proteins not previously associated with DE-cad function or cadherin-mediated adhesion. We then analyzed the function of a subset of ERC-identified candidates by RNAi during border cell (BC) migration and identified novel genes that function to regulate DE-cad. Among these, we found that the gene CG42684, which encodes a putative GTPase activating protein (GAP), regulates BC migration and adhesion. We named CG42684 raskol (“to split” in Russian) and show that it regulates DE-cad levels and actin protrusions in BCs. We propose that Raskol functions with DE-cad to restrict Ras/Rho signaling and help guide BC migration. Our results demonstrate that a coordinated selective pressure has shaped the adherens junction and this can be leveraged to identify novel components of the complexes and signaling pathways that regulate cadherin-mediated adhesion. The establishment of intercellular adhesions facilitated the genesis of multicellular organisms. The adherens junction, which links the actin cytoskeletons of neighboring cells, arose early in the evolution of multicellularity and selective pressures have shaped its function and molecular composition over time. In this study, we used evolutionary rate covariation (ERC) analysis to examine the evolutionary history of the adherens junction and to identify proteins that coevolved with the core adherens junction protein Drosophila E-cadherin (DE-cad). ERC analysis of DE-cad revealed a collection of proteins with similar evolutionary histories. We then tested the role of ERC-identified candidates in border cell migration in the fly egg chamber, a process that requires the coordinated regulation of cell-cell adhesion and cell motility. Among these, we found that a previously uncharacterized gene CG42684, which encodes a putative GTPase activating protein (GAP), regulates the collective cell migration of border cells, stabilizes cell-cell adhesions and regulates the actin dynamics. Our results demonstrate that components of the adherens junction share an evolutionary history and that ERC analysis is a powerful method to identify novel components of cell adhesion complexes in Drosophila.
Collapse
Affiliation(s)
- Qanber Raza
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jae Young Choi
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, New York, United States of America
| | - Yang Li
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Roisin M. O’Dowd
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Simon C. Watkins
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Maria Chikina
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Yang Hong
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Nathan L. Clark
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Adam V. Kwiatkowski
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
19
|
Tong H, Yin H, Hossain MA, Wang Y, Wu F, Dong X, Gao S, Zhan K, He W. Starvation-induced autophagy promotes the invasion and migration of human bladder cancer cells via TGF-β1/Smad3-mediated epithelial-mesenchymal transition activation. J Cell Biochem 2018; 120:5118-5127. [PMID: 30320898 DOI: 10.1002/jcb.27788] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/10/2018] [Indexed: 12/21/2022]
Abstract
The biological characteristics of bladder cancer include enhanced invasion and migration, which are the main causes of death in patients. Starvation is a typical feature of the bladder cancer microenvironment and can induce autophagy. Autophagy has an important relationship with the invasion and migration of tumors. However, the role of autophagy in the invasion and migration of bladder cancer cells remains unclear. Hence, the aim of the current study was to clarify this role and underlying mechanism. In this study, we found that starvation enhanced the epithelial-mesenchymal transition (EMT)-mediated invasion and migration of T24 and 5637 cells while inducing autophagy. The inhibition of autophagy with chloroquine (CQ) or 3-methyladenine (3MA) decreased EMT-mediated invasion and migration. In addition, the expression of transforming growth factor 1 (TGF-β1) and phosphorylated Smad3 (p-Smad3) increased after starvation. The inhibition of autophagy with CQ or 3MA also decreased the expression of TGF-β1 and p-Smad3. The inhibitor of TGF-β receptor sb431542 also inhibited the invasion, migration, and EMT of T24 and 5637 cells during starvation. Furthermore, recombinant TGF-β1 induced autophagy and inhibition of the TGF-β/Smad signaling pathway with sb431542 suppressed autophagy. In summary, our results suggested that autophagy promotes the invasion and migration of bladder cancer cells by inducing EMT through the TGF-β1/Smad3 signaling pathway. Moreover, autophagy and TGF-β1 can form a positive feedback loop to synergistically promote invasion and migration. Thus, our findings may provide a theoretical basis for the prevention of invasion and migration in bladder cancer.
Collapse
Affiliation(s)
- Hang Tong
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hubin Yin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mohammad Arman Hossain
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiyang Wang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feixiang Wu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyong Dong
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shun Gao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kai Zhan
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|