1
|
Ren D, Zhao F, Li J, Guo X, Ma X, Zheng Y, Shen G, Zhao J. lncRNA TCONS_00251376 promotes the proliferation and migration of gastric cancer cell through upregulating ETV1. CANCER INNOVATION 2025; 4:e156. [PMID: 39668941 PMCID: PMC11636580 DOI: 10.1002/cai2.156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 12/14/2024]
Abstract
Background Although there have been significant advancements in the treatment modalities for gastric cancer (GC) in recent years, the overall prognosis remains poor, particularly for individuals in advanced stages. The absence of a sensitive tumor marker in GC is a crucial factor contributing to this challenge. Methods Our study focused on investigating a newly discovered long noncoding RNA (lncRNA) known as TCONS_00251376, which has been confirmed to exhibit differential expression in GC compared to adjacent tissues. To further validate these expression differences, we collected 22 pairs of GC and adjacent noncancerous tissues. Subsequent cell function experiments and animal studies were conducted to elucidate the role and underlying mechanisms of lncRNA TCONS_00251376 in the development of GC. Results The study revealed a significant upregulation of lncRNA TCONS_00251376 in cancer tissues (p < 0.01) and a consistent upregulation in GC cell lines (AGS, MKN45, BGC-823, and MGC-803). Furthermore, it was observed that lncRNA TCONS_00251376 played a promotive role in the proliferation, migration, and invasion of GC cells. Subsequent analysis indicated that lncRNA TCONS_00251376 could upregulate the expression of ETV1, a factor associated with the prognosis of GC. Conclusions Therefore, our findings suggest that lncRNA TCONS_00251376 functions as an oncogenic lncRNA, promoting tumorigenesis and progression by regulating the expression of ETV1 gene. This highlights its potential as an effective target for treating GC.
Collapse
Affiliation(s)
- Dengfeng Ren
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
| | - Fuxing Zhao
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
| | - Jinming Li
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
- Graduate School, Qinghai UniversityXiningChina
| | - Xinjian Guo
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
- Department of PathologyAffiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
| | - Xinfu Ma
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
- Department of Gastrointestinal Oncology SurgeryAffiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
| | - Yonghui Zheng
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
| | - Guoshuang Shen
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
| |
Collapse
|
2
|
Yuan W, Shi Y, Dai S, Deng M, Zhu K, Xu Y, Chen Z, Xu Z, Zhang T, Liang S. The role of MAPK pathway in gastric cancer: unveiling molecular crosstalk and therapeutic prospects. J Transl Med 2024; 22:1142. [PMID: 39719645 PMCID: PMC11667996 DOI: 10.1186/s12967-024-05998-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/15/2024] [Indexed: 12/26/2024] Open
Abstract
Gastric cancer remains a significant health burden globally, especially prevalent in Asian and European regions. Despite a notable decline in incidence in the United States and Western Europe over recent decades, the disease's persistence underscores the urgency for advanced research in its pathogenesis and treatment strategies. Central to this pursuit is the exploration of the mitogen-activated protein kinase (MAPK) pathway, a pivotal cellular mechanism implicated in the complex processes of gastric cancer development, including cellular proliferation, invasion, migration, and metastasis. The MAPK or extracellular signal-regulated kinase pathway serves as a crucial conduit for transmitting extracellular signals to elicit intracellular responses, with its signaling cascades subject to alterations due to genetic and epigenetic variations across various diseases, prominently cancer. This review delves into the intricate role of the MAPK signaling pathway in the pathogenesis of gastric cancer, drawing upon the most recent and critical studies that shed light on MAPK pathway alterations as a gateway to the disease. It highlights the pathway's involvement in Helicobacter pylori-mediated gastric carcinogenesis and the tumorigenic processes induced by the Epstein-Barr virus, showcasing the substantial influence of miRNAs and lncRNAs in modulating gastric cancer's biological properties through their interaction with the MAPK pathway. Furthermore, the review extends into the therapeutic arena, discussing the promising impacts of herbal medicines, MAPK pathway inhibitors, and immunosuppressants on mitigating gastric cancer's progression. Through an exhaustive examination of the MAPK pathway's multifaceted role in gastric cancer, from molecular crosstalks to therapeutic prospects, this review aspires to contribute to the ongoing efforts in understanding and combating this global health challenge, paving the way for novel therapeutic interventions and improved patient outcomes.
Collapse
Affiliation(s)
- Weiwei Yuan
- Department of Thyroid Surgery, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, China
| | - Yin Shi
- Department of Internal Medicine, Yiwu Maternity and Children Hospital, Yiwu, Zhejiang, China
| | - Shiping Dai
- Department of General Surgery, Wuwei City People's Hospital, No.256, West Street, Wuwei, 238300, China
| | - Mao Deng
- Department of General Surgery, Wuwei City People's Hospital, No.256, West Street, Wuwei, 238300, China
| | - Kai Zhu
- Department of General Surgery, Wuwei City People's Hospital, No.256, West Street, Wuwei, 238300, China
| | - Yuanmin Xu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zhangming Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zhou Xu
- Department of Thyroid Surgery, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, China.
| | - Tianlong Zhang
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China.
| | - Song Liang
- Department of General Surgery, The Lu'an Affiliated Hospital of Anhui Medical University, Lu'an People's Hospital, Lu'an, 237000, China.
| |
Collapse
|
3
|
Shi Y, Men X, Wang F, Li X, Zhang B. Role of long non-coding RNAs (lncRNAs) in gastric cancer metastasis: A comprehensive review. Pathol Res Pract 2024; 262:155484. [PMID: 39180802 DOI: 10.1016/j.prp.2024.155484] [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: 01/15/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024]
Abstract
One of the greatest frequent types of malignancy is gastric cancer (GC). Metastasis, an essential feature of stomach cancer, results in a high rate of mortality and a poor prognosis. However, metastasis biological procedures are not well recognized. Long non-coding RNAs (lncRNAs) have a role in numerous gene regulation pathways via epigenetic modification as well as transcriptional and post-transcriptional control. LncRNAs have a role in a variety of disorders, such as cardiovascular disease, Alzheimer's, and cancer. LncRNAs are substantially related to GC incidence, progression, metastasis and drug resistance. Several research released information on the molecular processes of lncRNAs in GC pathogenesis. By interacting with a gene's promoter or enhancer region to influence gene expression, lncRNAs can operate as an oncogene or a tumor suppressor. This review includes the lncRNAs associated with metastasis of GC, which may give insights into the processes as well as potential clues for GC predicting and tracking.
Collapse
Affiliation(s)
- Yue Shi
- Department of Microbiology and Immunology, Changchun University of Chinese Medicine, Jilin 130117, PR China.
| | - Xiaoping Men
- Department of Clinical Laboratory, The First Affiliated Hospital to Changchun University of Chinese Medicine, Jilin 130021, PR China.
| | - Fang Wang
- Department of Microbiology and Immunology, Changchun University of Chinese Medicine, Jilin 130117, PR China.
| | - Xueting Li
- Experimental Center, Changchun University of Chinese Medicine, Jilin 130021, PR China.
| | - Biao Zhang
- School of Health Management, Changchun University of Chinese Medicine, Jilin 130117, PR China.
| |
Collapse
|
4
|
Elimam H, Abdel Mageed SS, Hatawsh A, Moussa R, Radwan AF, Elfar N, Alhamshry NAA, Abd-Elmawla MA, Mohammed OA, Zaki MB, Doghish AS. Unraveling the influence of LncRNA in gastric cancer pathogenesis: a comprehensive review focus on signaling pathways interplay. Med Oncol 2024; 41:218. [PMID: 39103705 DOI: 10.1007/s12032-024-02455-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/16/2024] [Indexed: 08/07/2024]
Abstract
Gastric cancers (GCs) are among the most common and fatal malignancies in the world. Despite our increasing understanding of the molecular mechanisms underlying GC, further biomarkers are still needed for more in-depth examination, focused prognosis, and treatment. GC is one among the long non-coding RNAs, or lncRNAs, that have emerged as key regulators of the pathophysiology of cancer. This comprehensive review focuses on the diverse functions of long noncoding RNAs (lncRNAs) in the development of GC and their interactions with important intracellular signaling pathways. LncRNAs affect GC-related carcinogenic signaling cascades including pathways for EGFR, PI3K/AKT/mTOR, p53, Wnt/β-catenin, JAK/STAT, Hedgehog, NF-κB, and hypoxia-inducible factor. Dysregulated long non-coding RNA (lncRNA) expression has been associated with multiple characteristics of cancer, such as extended growth, apoptosis resistance, enhanced invasion and metastasis, angiogenesis, and therapy resistance. For instance, lncRNAs such as HOTAIR, MALAT1, and H19 promote the development of GC via altering these pathways. Beyond their main roles, GC lncRNAs exhibit potential as diagnostic and prognostic biomarkers. The overview discusses CRISPR/Cas9 genome-modifying methods, antisense oligonucleotides, small molecules, and RNA interference as potential therapeutic approaches to regulate the expression of long noncoding RNAs (lncRNAs). An in-depth discussion of the intricate functions that lncRNAs play in the development of the majority of stomach malignancies is provided in this review. It provides the groundwork for future translational research in lncRNA-based whole processes toward GC by highlighting their carcinogenic effects, regulatory roles in significant signaling cascades, and practical scientific uses as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt.
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Abdulrahman Hatawsh
- Biotechnology School, Nile University, 26th of July Corridor, Sheikh Zayed City, 12588, Giza, Egypt
| | - Rewan Moussa
- Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | - Abdullah F Radwan
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, 11829, Cairo, Egypt
| | - Nourhan Elfar
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, 11578, Cairo, Egypt
- Egyptian Drug Authority (EDA), Ministry of Health and Population, Cairo, 11567, Egypt
| | - Nora A A Alhamshry
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
5
|
Guo J, Song Z, Muming A, Zhang H, Awut E. Cysteine protease inhibitor S promotes lymph node metastasis of esophageal cancer cells via VEGF-MAPK/ERK-MMP9/2 pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6051-6059. [PMID: 38386044 DOI: 10.1007/s00210-024-03014-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/15/2024] [Indexed: 02/23/2024]
Abstract
Cysteine protease inhibitor S (CST4) plays a pivotal role in the regulation of growth, invasion, and metastasis of a variety of malignancies. However, the potential mechanism behind how CST4 contributes to CST4 in lymph node metastasis (LNM) and tumor-associated lymphangiogenesis of esophageal cancer (EC) cells has not been elucidated previously. Short hairpin RNA technique was utilized to upregulate the CST4 gene expression. Different experiments, including the tubule formation assay and immunofluorescence, were conducted to observe the cellular behavior. Enzyme-linked immunosorbent assay (ELISA) and Western blot analyses were employed to determine the expression levels of relevant proteins. In our study, we discovered that high expression of CST4 in EC cells had multiple effects. It stimulated cell proliferation, invasion, and migration and caused epithelial-mesenchymal transition (EMT). Moreover, it also inhibited the apoptosis of EC cells and caused them to stagnate in the G2/M phase. High expression of CST4 promoted the secretion of lymphangiogenic markers (TGFβ1, VEGF, VEGF-C/D) in EC cells. In addition, high expression of CST4 in EC cells not only enhanced the proliferation and migration of HLECs, but also stimulated the lumen formation and F-actin expression and rearrangement of HLECs. The elevated expression of CST4 also facilitated the secretion of p-ERK1/2, MMP9, and MMP-2 in HLECs. However, various tumor-promoting effects of high expression of CST4 on HLECs could be inhibited by VEGF inhibitors in EC cells. Overall, our findings indicate that CST4 plays a significant role in the accumulation, migration, and EMT of EC cells. CST4 can activate the VEGF-MAPK/ERK-MMP9/2 signaling axis to promote LNM and lymphangiogenesis in EC.
Collapse
Affiliation(s)
- Jiayi Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830054, Xinjiang, China
- The First Clinical Medical College, Xinjiang Medical University, Urumqi, 830054, China
| | - Zhengyu Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830054, Xinjiang, China
- The First Clinical Medical College, Xinjiang Medical University, Urumqi, 830054, China
| | - AlimuJiang Muming
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830054, Xinjiang, China
- The First Clinical Medical College, Xinjiang Medical University, Urumqi, 830054, China
| | - Haiping Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830054, Xinjiang, China
- The First Clinical Medical College, Xinjiang Medical University, Urumqi, 830054, China
| | - Edris Awut
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830054, Xinjiang, China.
- The First Clinical Medical College, Xinjiang Medical University, Urumqi, 830054, China.
| |
Collapse
|
6
|
Saleh RO, Al-Hawary SIS, Hammoud A, Hjazi A, Ayad Abdulrazzaq S, Rajput P, Alawsi T, Alnajar MJ, Alawadi A. The long non-coding RNAs (lncRNA) in the pathogenesis of gastric cancer cells: molecular mechanisms and involvement miRNAs. Mol Biol Rep 2024; 51:615. [PMID: 38704760 DOI: 10.1007/s11033-024-09546-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024]
Abstract
A complex sequence of occurrences, including host genetic vulnerability, Helicobacter pylori infection, and other environmental variables, culminate in gastric cancer (GC). The development of several genetic and epigenetic changes in oncogenes and tumor suppressor genes causes dysregulation of several signaling pathways, which upsets the cell cycle and the equilibrium between cell division and apoptosis, leading to GC. Developments in computational biology and RNA-seq technology enable quick detection and characterization of long non-coding RNAs (lncRNAs). Recent studies have shown that long non-coding RNAs (lncRNAs) have multiple roles in the development of gastric cancer. These lncRNAs interact with molecules of protein, RNA, DNA, and/or combinations. This review article explores several gastric cancer-associated lncRNAs, such as ADAMTS9-AS2, UCA1, XBP-1, and LINC00152. These various lncRNAs could change GC cell apoptosis, migration, and invasion features in the tumor microenvironment. This review provides an overview of the most recent research on lncRNAs and GC cell apoptosis, migration, invasion, and drug resistance, focusing on studies conducted in cancer cells and healthy cells during differentiation.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | | | - Ahmad Hammoud
- Department of Medical and Technical Information Technology, Bauman Moscow State Technical University, Moscow, Russia.
- Department of Mathematics and Natural Sciences, Gulf University for Science and Technology, Mishref Campus, Kuwait City, Kuwait.
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences , Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | | | - Pranchal Rajput
- School of Applied and Life Sciences, Divison of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Taif Alawsi
- Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
- Department of Laser and Optoelectronics Engineering, University of Technology, Baghdad, Iraq
| | | | - Ahmed Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al-Qadisiyyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
7
|
Xu W, Li H, Wang Z, Kang Y, Zheng L, Liu Y, Xu P, Li Z. LINC00152: Potential driver oncogene in pan-cancer. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1851. [PMID: 38702938 DOI: 10.1002/wrna.1851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 05/06/2024]
Abstract
Long noncoding RNAs (lncRNA) are a class of non-coding RNAs greater than 200 bp in length with limited peptide-coding function. The transcription of LINC00152 is derived from chromosome 2p11.2. Many studies prove that LINC00152 influences the progression of various tumors via promoting the tumor cells malignant phenotype, chemoresistance, and immune escape. LINC00152 is regulated by multiple transcription factors and DNA hypomethylation. In addition, LINC00152 participates in the regulation of complex molecular signaling networks through epigenetic regulation, protein interactions, and competitive endogenous RNA (ceRNA). Here, we provide a systematic review of the upstream regulatory factors of LINC00152 expression level in different types of tumors. In addition, we revisit the main functions and mechanisms of LINC00152 as driver oncogene and biomarker in pan-cancer. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Methods > RNA Analyses in Cells RNA Interactions with Proteins and Other Molecules > RNA-Protein Complexes.
Collapse
Affiliation(s)
- Wei Xu
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Huiting Li
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ziyao Wang
- Department of Thoracic Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan Kang
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Luojie Zheng
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yiping Liu
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ping Xu
- Department of Respiratory and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Zheng Li
- NHC Key Laboratory of Carcinogenesis, National Clinical Research Center for Geriatric Disorders, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Department of oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
8
|
Ito Y, Kanda M, Sasahara M, Tanaka C, Shimizu D, Umeda S, Inokawa Y, Hattori N, Hayashi M, Nakayama G, Kodera Y. Killer cell lectin-like receptor G2 facilitates aggressive phenotypes of gastric cancer cells via dual activation of the ERK1/2 and JAK/STAT pathways. Gastric Cancer 2024; 27:506-518. [PMID: 38386237 DOI: 10.1007/s10120-024-01480-y] [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: 11/05/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Advanced gastric cancer (GC) has a poor prognosis. This study aimed to identify novel GC-related genes as potential therapeutic targets. METHODS Killer cell lectin-like receptor G2 (KLRG2) was identified as a candidate gene by transcriptome analysis of metastatic GC tissues. Small interfering RNA-mediated KLRG2 knockdown in human GC cell lines was used to investigate KLRG2 involvement in signaling pathways and functional behaviors in vitro and in vivo. Clinicopathological data were analyzed in patients stratified according to tumor KLRG2 mRNA expression. RESULTS KLRG2 knockdown in GC cells decreased cell proliferation, migration, and invasion; caused cell cycle arrest in G2/M phase; induced apoptosis via caspase activation; suppressed JAK/STAT and MAPK-ERK1/2 pathway activities; and upregulated p53 and p38 MAPK activities. In mouse xenograft models of peritoneal metastasis, the number and weight of disseminated GC nodules were decreased by KLRG2 knockdown. High tumor levels of KLRG2 mRNA were significantly associated with lower 5-year overall survival (OS) and relapse-free survival (RFS) rates in patients with Stage I-III GC (5-year OS rate: 64.4% vs. 80.0%, P = 0.009; 5-year RFS rate: 62.8% vs. 78.1%, P = 0.030). CONCLUSIONS KLRG2 knockdown attenuated the malignant phenotypes of GC cells via downregulation of JAK/STAT and MAPK-ERK1/2 pathway activity and upregulation of p38 MAPK and p53. Targeted suppression of KLRG2 may serve as a new treatment approach for GC.
Collapse
Affiliation(s)
- Yuki Ito
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan.
| | - Masahiro Sasahara
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Dai Shimizu
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Shinichi Umeda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yoshikuni Inokawa
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Norifumi Hattori
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Goro Nakayama
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| |
Collapse
|
9
|
Cheng S, Liu Y, He B, Zhang J, Yang Y, Wang X, Li Z. Chlamydia trachomatis upregulates lncRNA CYTOR to mediate autophagy through miR-206/MAPK1 axis. Pathog Dis 2024; 82:ftae011. [PMID: 38821518 PMCID: PMC11210502 DOI: 10.1093/femspd/ftae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 06/02/2024] Open
Abstract
Chlamydia trachomatis infection can be regulated by autophagy-related genes. LncRNA CYTOR has been proven to be involved in autophagy. In this research, we investigated the role of CYTOR in autophagy induced by C. trachomatis and the potential mechanisms. After C. trachomatis infection, CYTOR and MAPK1 were up-regulated and miR-206 was down-regulated, meanwhile, the autophagy-related protein Beclin1 and LC3-Ⅱ/LC3-Ⅰ ratio were increased. Interference with CYTOR or overexpression with miR-206 downregulated the autophagy-related protein Beclin1 and the number of autophagic spots LC3, decreased the protein ratio of LC3-II/LC3-I, and upregulated the expression of P62 protein. The luciferase reporter assay confirmed that CYTOR acted as a sponge for miR-206 to target MAPK1. In addition, CYTOR promoted autophagy induced by C. trachomatis infection through the MAPK1/ERK signaling pathway activation. Taken together, we have identified a novel molecular mechanism that the CYTOR/miR-206/MAPK1 axis was involved in the regulation of autophagy in C. trachomatis infection. This work provides an experimental basis for elucidating the pathogenesis of C. trachomatis for the treatment, prevention and control of related infectious diseases.
Collapse
Affiliation(s)
- Shan Cheng
- Institute of Pathogenic Biology, School of Nursing, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001 Hunan, China
| | - Yi Liu
- Institute of Pathogenic Biology, School of Nursing, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001 Hunan, China
| | - Bei He
- Institute of Pathogenic Biology, School of Nursing, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001 Hunan, China
| | - Jinrong Zhang
- Institute of Pathogenic Biology, School of Nursing, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001 Hunan, China
| | - Yewei Yang
- Institute of Pathogenic Biology, School of Nursing, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001 Hunan, China
| | - Xinglv Wang
- Institute of Pathogenic Biology, School of Nursing, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001 Hunan, China
| | - Zhongyu Li
- Institute of Pathogenic Biology, School of Nursing, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001 Hunan, China
| |
Collapse
|
10
|
Zabeti Touchaei A, Vahidi S, Samadani AA. Decoding the regulatory landscape of lncRNAs as potential diagnostic and prognostic biomarkers for gastric and colorectal cancers. Clin Exp Med 2024; 24:29. [PMID: 38294554 PMCID: PMC10830721 DOI: 10.1007/s10238-023-01260-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/12/2023] [Indexed: 02/01/2024]
Abstract
Colorectal cancer (CRC) and gastric cancer (GC) are major contributors to cancer-related mortality worldwide. Despite advancements in understanding molecular mechanisms and improved drug treatments, the overall survival rate for patients remains unsatisfactory. Metastasis and drug resistance are major challenges contributing to the high mortality rate in both CRC and GC. Recent research has shed light on the role of long noncoding RNAs (lncRNAs) in the development and progression of these cancers. LncRNAs regulate gene expression through various mechanisms, including epigenetic modifications and interactions with microRNAs (miRNAs) and proteins. They can serve as miRNA precursors or pseudogenes, modulating gene expression at transcriptional and post-transcriptional levels. Additionally, circulating lncRNAs have emerged as non-invasive biomarkers for the diagnosis, prognosis, and prediction of drug therapy response in CRC and GC. This review explores the intricate relationship between lncRNAs and CRC/GC, encompassing their roles in cancer development, progression, and chemoresistance. Furthermore, it discusses the potential of lncRNAs as therapeutic targets in these malignancies. The interplay between lncRNAs, miRNAs, and tumor microenvironment is also highlighted, emphasizing their impact on the complexity of cancer biology. Understanding the regulatory landscape and molecular mechanisms governed by lncRNAs in CRC and GC is crucial for the development of effective diagnostic and prognostic biomarkers, as well as novel therapeutic strategies. This review provides a comprehensive overview of the current knowledge and paves the way for further exploration of lncRNAs as key players in the management of CRC and GC.
Collapse
Affiliation(s)
| | - Sogand Vahidi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
11
|
Shen J, Huang C, Cui L, Zhao Y, Zhu M, Chen Z, Wang M, Zhu W, Shen B. Chemotherapeutic Drugs Endow Gastric Cancer Mesenchymal Stem Cells with Stronger Tumor-Promoting Ability. J Environ Pathol Toxicol Oncol 2024; 43:1-13. [PMID: 37824366 DOI: 10.1615/jenvironpatholtoxicoloncol.2023041847] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023] Open
Abstract
Gastric cancer (GC) is one of the most aggressive tumors and has a poor prognosis. It has been demonstrated that gastric cancer mesenchymal stem cells (GC-MSCs) can promote the progression, metastasis, and chemoresistance of GC through various mechanisms, but the effect of GC-MSCs on GC during chemotherapy is still unknown. In this study, flow cytometry, CCK8 assay, migration assay, colony formation assay, and western blot were conducted. We also analyzed GC patients from the cancer genome atlas (TCGA). Our results showed that GC-MSCs were resistant to 5-FU and Taxol at the IC50 concentration for GC cells, and 5-FU could promote the migration of GC-MSCs at low doses. Furthermore, the conditioned medium of GC-MSCs pretreated with chemotherapeutic drugs was more effective in promoting the proliferation, migration, and stemness of GC cell lines than the conditioned medium of GC-MSCs without chemotherapeutic drugs treatment. These effects were dependent on the activation of phosphorylated AKT (p-AKT) in GC cell lines. Correspondingly, the inhibition of p-AKT reversed the tumor-promoting effect of the conditioned medium of GC-MSCs pretreated with chemotherapeutic drugs. Additionally, the expression of AKT1 was higher in GC tissues than in both paracancerous tissues and normal tissues, and patients resistant to chemotherapy expressed more AKT1 compared to those who were sensitive. Taken together, our data demonstrated that GC-MSCs gained more tumor-promoting abilities during chemotherapy.
Collapse
Affiliation(s)
- Jiaqi Shen
- School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Chao Huang
- School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Linjing Cui
- School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yuanyuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan 430030, China; NHC Key Laboratory of Organ Transplantation, Wuhan 430030, China; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China; School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Miaolin Zhu
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical, Jiangsu University, Nanjing 21000, China
| | - Zhihong Chen
- Department of Gastrointestinal Surgery, Affiliated People's Hospital of Jiangsu, Zhenjiang 212002, China
| | - Mei Wang
- Department of Oncology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai, People's Republic of China, 200433; School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Wei Zhu
- School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Bo Shen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical, Jiangsu University, Nanjing 21000, China
| |
Collapse
|
12
|
Shi J, Li W, Jia Z, Peng Y, Hou J, Li N, Meng R, Fu W, Feng Y, Wu L, Zhou L, Wang D, Shen J, Chang J, Wang Y, Cao J. Synaptotagmin 1 Suppresses Colorectal Cancer Metastasis by Inhibiting ERK/MAPK Signaling-Mediated Tumor Cell Pseudopodial Formation and Migration. Cancers (Basel) 2023; 15:5282. [PMID: 37958455 PMCID: PMC10649299 DOI: 10.3390/cancers15215282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/21/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Although synaptotagmin 1 (SYT1) has been identified participating in a variety of cancers, its role in colorectal cancer (CRC) remains an enigma. This study aimed to demonstrate the effect of SYT1 on CRC metastasis and the underlying mechanism. We first found that SYT1 expressions in CRC tissues were lower than in normal colorectal tissues from the CRC database and collected CRC patients. In addition to this, SYT1 expression was also lower in CRC cell lines than in the normal colorectal cell line. SYT1 expression was downregulated by TGF-β (an EMT mediator) in CRC cell lines. In vitro, SYT1 overexpression repressed pseudopodial formation and reduced cell migration and invasion of CRC cells. SYT1 overexpression also suppressed CRC metastasis in tumor-bearing nude mice in vivo. Moreover, SYT1 overexpression promoted the dephosphorylation of ERK1/2 and downregulated the expressions of Slug and Vimentin, two proteins tightly associated with EMT in tumor metastasis. In conclusion, SYT1 expression is downregulated in CRC. Overexpression of SYT1 suppresses CRC cell migration, invasion, and metastasis by inhibiting ERK/MAPK signaling-mediated CRC cell pseudopodial formation. The study suggests that SYT1 is a suppressor of CRC and may have the potential to be a therapeutic target for CRC.
Collapse
Affiliation(s)
- Jianyun Shi
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| | - Wenjing Li
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| | - Zhenhua Jia
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| | - Ying Peng
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| | - Jiayi Hou
- Department of Clinical Laboratory, Shanxi Provincial Academy of Traditional Chinese Medicine, Taiyuan 030071, China
| | - Ning Li
- Department of Gastrointestinal and Pancreatic Surgery & Hernia and Abdominal Surgery, Shanxi Provincial People’s Hospital, Taiyuan 030045, China
| | - Ruijuan Meng
- Department of Radiology, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030606, China
| | - Wei Fu
- Department of Radiology, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030606, China
| | - Yanlin Feng
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| | - Lifei Wu
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| | - Lan Zhou
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| | - Deping Wang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| | - Jing Shen
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| | - Jiasong Chang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| | - Yanqiang Wang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030606, China
| | - Jimin Cao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan 030606, China
| |
Collapse
|
13
|
Ou C, He X, Liu Y, Zhang X. lncRNA cytoskeleton regulator RNA (CYTOR): Diverse functions in metabolism, inflammation and tumorigenesis, and potential applications in precision oncology. Genes Dis 2023; 10:415-429. [PMID: 37223495 PMCID: PMC10201560 DOI: 10.1016/j.gendis.2021.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 08/20/2021] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are a novel class of non-coding RNA (ncRNA), that have been studied extensively in the field of tumor research in recent years. In the case of tumor-associated lncRNAs, lncRNA cytoskeleton regulator RNA (CYTOR) displays extensive functions in tumorigenesis, including invasion, metastasis, malignant proliferation, glycolysis, and inflammatory response. Moreover, the dysregulation of CYTOR is closely related to clinicopathological characteristics, such as tumor stage, lymph node metastasis and infiltration, and poor prognosis of tumor patients. In this review, we provide a novel strategy to summarize the biological functions and clinical value of CYTOR in tumors through an overview of the literature combined with gene set enrichment analysis. A deeper understanding of the role of CYTOR in tumorigenesis may provide new diagnostic, prognostic and therapeutic markers for human tumors.
Collapse
Affiliation(s)
- Chunlin Ou
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, Changsha, Hunan 410008, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan 410008, China
| | - Xin Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, Changsha, Hunan 410008, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan 410008, China
| |
Collapse
|
14
|
A review on the role of LINC00152 in different disorders. Pathol Res Pract 2023; 241:154274. [PMID: 36563561 DOI: 10.1016/j.prp.2022.154274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
LINC00152 is an important lncRNA in human disorders. It is mainly regarded as a tumor-promoting lncRNA. Mechanistically, LINC00152 serves as a molecular sponge for miR-143a-3p, miR-125a-5p, miR-139, miR-215, miR-193a/b-3p, miR-16-5p, miR-206, miR-195, miR-138, miR-185-5p, miR-103, miR-612, miR-150, miR-107, miR-205-5p and miR-153-3p. In addition, it can regulate activity of mTOR, EGFR/PI3K/AKT, ERK/MAPK, Wnt/β-Catenin, EGFR, NF-κB, HIF-1 and PTEN. In this review, we provide a concise but comprehensive explanation about the role of LINC00152 in tumor development and progression as well as its role in the pathology of non-malignant conditions with the aim of facilitating the clinical implementation of this lncRNA as a diagnostic or prognostic tumor marker and therapeutic target.
Collapse
|
15
|
Fu D, Hu Z, Xu X, Dai X, Liu Z. Key signal transduction pathways and crosstalk in cancer: Biological and therapeutic opportunities. Transl Oncol 2022; 26:101510. [PMID: 36122506 PMCID: PMC9486121 DOI: 10.1016/j.tranon.2022.101510] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/16/2022] [Accepted: 08/01/2022] [Indexed: 12/03/2022] Open
Abstract
Several different signaling pathways and molecular mechanisms have been identified as responsible for controlling critical functions in human cancer cells, such as selective growth and proliferative advantage, altered stress response favoring overall survival, vascularization, invasion and metastasis, metabolic rewiring, an abetting microenvironment, and immune modulation. This concise summary will provide a selective review of recent studies of key signal transduction pathways, including mitogen-activated protein kinase (MAPK) pathway, Phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling, and Wnt/β-catenin signaling pathway, which are altered in cancer cells, as the novel and promising therapeutic targets.
Collapse
Affiliation(s)
- Dongliao Fu
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang 471023, China
| | - Zhigang Hu
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang 471023, China
| | - Xinyang Xu
- Zhengzhou Foreign Language School, Zhengzhou, Henan 450001, China
| | - Xiaoyan Dai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, No.280, Waihuan East Road, Guangzhou, Guangdong 511436, China.
| | - Ziyi Liu
- Laboratory of Physiologic Studies, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20891, United States.
| |
Collapse
|
16
|
Abnormally Expressed lncRNAs as Potential Biomarkers for Gastric Cancer Risk: A Diagnostic Meta-Bioinformatics Analysis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6712625. [PMID: 36389111 PMCID: PMC9652703 DOI: 10.1155/2022/6712625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/10/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Background and Aims Abnormal expression of lncRNAs is relevant to the occurrence and development of gastric cancer (GC), but the significance remains inconclusive. We performed a diagnostic meta-bioinformatics analysis to elucidate the association between lncRNA expression and GC risk. Methods Published datasets were selected from PubMed, Embase, CNKI, and Web of Science, up to 1st December 2021. The pooled sensitivity (SEN), specificity (SPE), positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and area under the curve (AUC) were calculated to evaluate the diagnostic value. RNA sequencing data were downloaded for validation. Results 54 studies with 4671 patients and 4652 matched controls were included in the meta-analysis. The pooled SEN, SPE, PLR, NLR, DOR, and AUC were 0.71, 0.76, 2.9, 0.39, 8, and 0.79, respectively. Subgroup analyses showed that the DOR and AUC of intergenic lncRNAs, circulating lncRNAs, larger sample size (>200), and high-quality (NOS score ≥ 7) groups were superior to antisense lncRNAs, tissue lncRNAs, smaller sample size (≤200), and low-quality (NOS score < 7) groups, respectively. However, only circulating lncRNAs had significantly higher diagnostic utility than that tissue lncRNAs. Nine differentially expressed lncRNAs in the meta-analysis were verified in TCGA-STAD. PVT1 was the most effective single lncRNA, with AUC of 0.949, SEN of 0.808, and SPE of 0.969, while PVT1 and C5orf66-AS1 were the most effective combination, with AUC of 0.972, SEN of 0.941, and SPE of 0.937. Conclusion Abnormally expressed lncRNAs, especially circulating lncRNAs, might be potential diagnostic biomarkers for GC risk. A novel combined model of lncRNAs might achieve better GC diagnosis performance.
Collapse
|
17
|
Li S, Yao W, Liu R, Gao L, Lu Y, Zhang H, Liang X. Long non-coding RNA LINC00152 in cancer: Roles, mechanisms, and chemotherapy and radiotherapy resistance. Front Oncol 2022; 12:960193. [PMID: 36033524 PMCID: PMC9399773 DOI: 10.3389/fonc.2022.960193] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Long non-coding RNA LINC00152 (cytoskeleton regulator, or LINC00152) is an 828-bp lncRNA located on chromosome 2p11.2. LINC00152 was originally discovered during research on hepatocarcinogenesis and has since been regarded as a crucial oncogene that regulates gene expression in many cancer types. LINC00152 is aberrantly expressed in various cancers, including gastric, breast, ovarian, colorectal, hepatocellular, and lung cancer, and glioma. Several studies have indicated that LINC00152 is correlated with cell proliferation, apoptosis, migration, invasion, cell cycle, epithelial-mesenchymal transition (EMT), chemotherapy and radiotherapy resistance, and tumor growth and metastasis. High LINC00152 expression in most tumors is significantly associated with poor patient prognosis. Mechanistic analysis has demonstrated that LINC00152 can serve as a competing endogenous RNA (ceRNA) by sponging miRNA, regulating the abundance of the protein encoded by a particular gene, or modulating gene expression at the epigenetic level. LINC00152 can serve as a diagnostic or prognostic biomarker, as well as a therapeutic target for most cancer types. In the present review, we discuss the roles and mechanisms of LINC00152 in human cancer, focusing on its functions in chemotherapy and radiotherapy resistance.
Collapse
Affiliation(s)
- Shuang Li
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Jinzhou Medical University, Jinzhou, China
| | - Weiping Yao
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Bengbu Medical College, Bengbu, China
| | - Ruiqi Liu
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Bengbu Medical College, Bengbu, China
| | - Liang Gao
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yanwei Lu
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Haibo Zhang
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Xiaodong Liang, ; Haibo Zhang,
| | - Xiaodong Liang
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Jinzhou Medical University, Jinzhou, China
- *Correspondence: Xiaodong Liang, ; Haibo Zhang,
| |
Collapse
|
18
|
Pellegrino R, Castoldi M, Ticconi F, Skawran B, Budczies J, Rose F, Schwab C, Breuhahn K, Neumann UP, Gaisa NT, Loosen SH, Luedde T, Costa IG, Longerich T. LINC00152 Drives a Competing Endogenous RNA Network in Human Hepatocellular Carcinoma. Cells 2022; 11:cells11091528. [PMID: 35563834 PMCID: PMC9103153 DOI: 10.3390/cells11091528] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/29/2022] [Accepted: 05/01/2022] [Indexed: 11/16/2022] Open
Abstract
Genomic and epigenomic studies revealed dysregulation of long non-coding RNAs in many cancer entities, including liver cancer. We identified an epigenetic mechanism leading to upregulation of the long intergenic non-coding RNA 152 (LINC00152) expression in human hepatocellular carcinoma (HCC). Here, we aimed to characterize a potential competing endogenous RNA (ceRNA) network, in which LINC00152 exerts oncogenic functions by sponging miRNAs, thereby affecting their target gene expression. Database and gene expression data of human HCC were integrated to develop a potential LINC00152-driven ceRNA in silico. RNA immunoprecipitation and luciferase assay were used to identify miRNA binding to LINC00152 in human HCC cells. Functionally active players in the ceRNA network were analyzed using gene editing, siRNA or miRNA mimic transfection, and expression vectors in vitro. RNA expression in human HCC in vivo was validated by RNA in situ hybridization. Let-7c-5p, miR-23a-3p, miR-125a-5p, miR-125b-5p, miR-143a-3p, miR-193-3p, and miR-195-5p were detected as new components of the potential LINC00152 ceRNA network in human HCC. LINC00152 was confirmed to sponge miR143a-3p in human HCC cell lines, thereby limiting its binding to their respective target genes, like KLC2. KLC2 was identified as a central mediator promoting pro-tumorigenic effects of LINC00152 overexpression in HCC cells. Furthermore, co-expression of LINC00152 and KLC2 was observed in human HCC cohorts and high KLC2 expression was associated with shorter patient survival. Functional assays demonstrated that KLC2 promoted cell proliferation, clonogenicity and migration in vitro. The LINC00152-miR-143a-3p-KLC2 axis may represent a therapeutic target in human HCC.
Collapse
Affiliation(s)
- Rossella Pellegrino
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
- Correspondence: ; Tel.: +49-(0)6221-56-34094
| | - Mirco Castoldi
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.C.); (S.H.L.); (T.L.)
| | - Fabio Ticconi
- Institute for Computational Genomics, Joint Research Center for Computational Biomedicine, University Hospital RWTH Aachen, 52074 Aachen, Germany; (F.T.); (I.G.C.)
| | - Britta Skawran
- Institute of Human Genetics, Hannover Medical School, 30625 Hannover, Germany;
| | - Jan Budczies
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
| | - Fabian Rose
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
| | - Constantin Schwab
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
| | - Kai Breuhahn
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
| | - Ulf P. Neumann
- Department of General, Visceral and Transplant Surgery, University Hospital RWTH Aachen, 52074 Aachen, Germany;
- Department of Surgery, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Nadine T. Gaisa
- Institute of Pathology, University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Sven H. Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.C.); (S.H.L.); (T.L.)
| | - Tom Luedde
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.C.); (S.H.L.); (T.L.)
| | - Ivan G. Costa
- Institute for Computational Genomics, Joint Research Center for Computational Biomedicine, University Hospital RWTH Aachen, 52074 Aachen, Germany; (F.T.); (I.G.C.)
| | - Thomas Longerich
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
| |
Collapse
|
19
|
Liu Y, Ding W, Yu W, Zhang Y, Ao X, Wang J. Long non-coding RNAs: Biogenesis, functions, and clinical significance in gastric cancer. Mol Ther Oncolytics 2021; 23:458-476. [PMID: 34901389 PMCID: PMC8637188 DOI: 10.1016/j.omto.2021.11.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC) is one of the most prevalent malignant tumor types and the third leading cause of cancer-related death worldwide. Its morbidity and mortality are very high due to a lack of understanding about its pathogenesis and the slow development of novel therapeutic strategies. Long non-coding RNAs (lncRNAs) are a class of non-coding RNAs with a length of more than 200 nt. They play crucial roles in a wide spectrum of physiological and pathological processes by regulating the expression of genes involved in proliferation, differentiation, apoptosis, cell cycle, invasion, metastasis, DNA damage, and carcinogenesis. The aberrant expression of lncRNAs has been found in various cancer types. A growing amount of evidence demonstrates that lncRNAs are involved in many aspects of GC pathogenesis, including its occurrence, metastasis, and recurrence, indicating their potential role as novel biomarkers in the diagnosis, prognosis, and therapeutic targets of GC. This review systematically summarizes the biogenesis, biological properties, and functions of lncRNAs and highlights their critical role and clinical significance in GC. This information may contribute to the development of better diagnostics and treatments for GC.
Collapse
Affiliation(s)
- Ying Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266021, China
| | - Wei Ding
- Department of Comprehensive Internal Medicine, Affiliated Hospital, Qingdao University, Qingdao 266003, China
| | - Wanpeng Yu
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266021, China
| | - Xiang Ao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Jianxun Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| |
Collapse
|
20
|
Wang Y, Zhang J, Liu M, Zhang S, Wang W, Cheng S. Clinical values and potential pathways of miR-183-5p in gastric cancer: a study based on integrational bioinformatics analysis. J Gastrointest Oncol 2021; 12:2123-2131. [PMID: 34790379 DOI: 10.21037/jgo-21-599] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background The clinicopathological value and exploration of the potential molecular mechanism of microRNA-183-5p (miR-183-5p) have been investigated in various cancers. This study further explored the transcriptome profile regulated by miR-183-5p. Methods Messenger RNA (mRNA) expression data, miRNA expression, and clinical information of stomach adenocarcinoma (STAD) were downloaded from The Cancer Genome Atlas (TCGA) database. Differentially expressed genes (DEGs) that related to mir-189-5p expression and cancer proliferation were acquired using bioinformatics analysis. The biological functions of these genes were analyzed in terms of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Hub genes relating to gastric cancer (GC) signal pathway were explored. The results were validated by further experiments. Results A total of 308 genes were found to be regulated by miR-183-5p, and they were related to cancer and GC patients' survival outcome. The biological function of these genes was found to act mainly on biological processes and the involved signal pathways included neuroactive ligand-receptor interaction, cell adhesion molecules, and axon guidance. In addition, miR-183-5p was also shown to regulate the mTOR, Wnt, MAPK, and PI3K-Akt signaling pathways through the genes WNT2B, NGFR, and NTRK2. Conclusions The miRNA miR-183-5p participates in the tumorigenesis and development of GC via certain signaling pathways, in particular the nerve- and immunity-related genes.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Jinku Zhang
- Department of Pathology, No. 1 Central Hospital of Baoding, Baoding, China
| | - Mingkai Liu
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Shun Zhang
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Weina Wang
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Shujie Cheng
- Department of Surgery, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
21
|
The LINC00152/miR-138 Axis Facilitates Gastric Cancer Progression by Mediating SIRT2. JOURNAL OF ONCOLOGY 2021; 2021:1173869. [PMID: 34697541 PMCID: PMC8541877 DOI: 10.1155/2021/1173869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/15/2021] [Accepted: 10/06/2021] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC) is the most common gastrointestinal cancer and the main cause of tumor-related death. Exploring markers for early diagnosis and new therapeutic targets is always on the way. In the last 10 years, long noncoding RNAs (lncRNAs) have been widely proved to be involved in the progress of many tumors and are regarded as potential targets for tumor therapy. We found that LINC00152, a newly identified lncRNA, was significantly upregulated in GC tissues and affected clinicopathological characteristics in GC patients. Furthermore, we observed that LINC00152 knockdown can significantly reduce cell proliferation and promote apoptosis in human gastric cancer cells. Further bioinformatic analysis indicated that LINC00152 competitively bound with miR-138 and regulated the expression of miR-138. Moreover, SIRT2 was further proved to be a downstream target of miR-138. Overall, this study elucidates the molecular mechanism of LINC00152 underlying the malignant phenotype of GC cells by mediating miR-138/SIRT2 axis, which provides a new understanding of the role and molecular mechanism of lncRNA in GC and also provides a new way for the treatment of gastric cancer.
Collapse
|
22
|
Xu Y, Jiang E, Shao Z, Shang Z. Long Noncoding RNAs in the Metastasis of Oral Squamous Cell Carcinoma. Front Oncol 2021; 10:616717. [PMID: 33520725 PMCID: PMC7845733 DOI: 10.3389/fonc.2020.616717] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common malignant tumor worldwide. Metastasis is the main cause of the death of OSCC patients. Long noncoding RNAs (lncRNAs), one of the key factors affecting OSCC metastasis, are a subtype of RNA with a length of more than 200 nucleotides that has little or no coding potential. In recent years, the important role played by lncRNAs in biological processes, such as chromatin modification, transcription regulation, RNA stability regulation, and mRNA translation, has been gradually revealed. More and more studies have shown that lncRNAs can regulate the metastasis of various tumors including OSCC at epigenetic, transcriptional, and post-transcriptional levels. In this review, we mainly discussed the role and possible mechanisms of lncRNAs in OSCC metastasis. Most lncRNAs act as oncogenes and only a few lncRNAs have been shown to inhibit OSCC metastasis. Besides, we briefly introduced the research status of cancer-associated fibroblasts-related lncRNAs in OSCC metastasis. Finally, we discussed the research prospects of lncRNAs-mediated crosstalk between OSCC cells and the tumor microenvironment in OSCC metastasis, especially the potential research value of exosomes and lymphangiogenesis. In general, lncRNAs are expected to be used for screening, treatment, and prognosis monitoring of OSCC metastasis, but more work is still required to better understand the biological function of lncRNAs.
Collapse
Affiliation(s)
- Yuming Xu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Erhui Jiang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhe Shao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengjun Shang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Venom peptides in cancer therapy: An updated review on cellular and molecular aspects. Pharmacol Res 2020; 164:105327. [PMID: 33276098 DOI: 10.1016/j.phrs.2020.105327] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023]
Abstract
Based on the high incidence and mortality rates of cancer, its therapy remains one of the most vital challenges in the field of medicine. Consequently, enhancing the efficacy of currently applied treatments and finding novel strategies are of great importance for cancer treatment. Venoms are important sources of a variety of bioactive compounds including salts, small molecules, macromolecules, proteins, and peptides that are defined as toxins. They can exhibit different pharmacological effects, and in recent years, their anti-tumor activities have gained significant attention. Several different compounds are responsible for the anti-tumor activity of venoms, and peptides are one of them. In the present review, we discuss the possible anti-tumor activities of venom peptides by highlighting molecular pathways and mechanisms through which these molecules can act effectively. Venom peptides can induce cell death in cancer cells and can substantially enhance the efficacy of chemotherapy and radiotherapy. Also, the venom peptides can mitigate the migration of cancer cells via suppression of angiogenesis and epithelial-to-mesenchymal transition. Notably, nanoparticles have been applied in enhancing the bioavailability of venom peptides and providing targeted delivery, thereby leading to their elevated anti-tumor activity and potential application for cancer therapy.
Collapse
|
24
|
Wang X, Song X, Cheng G, Zhang J, Dong L, Bai J, Luo D, Xiong Y, Li S, Liu F, Sun Y, Wang X, Li Y, Huang Y. The Regulatory Mechanism and Biological Significance of Mitochondrial Calcium Uniporter in the Migration, Invasion, Angiogenesis and Growth of Gastric Cancer. Onco Targets Ther 2020; 13:11781-11794. [PMID: 33235465 PMCID: PMC7680189 DOI: 10.2147/ott.s262049] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/27/2020] [Indexed: 12/25/2022] Open
Abstract
Objective Increasing evidences suggest that mitochondrial calcium uniporter (MCU), a selective channel responsible for mitochondrial Ca2+ uptake, is involved in the progression of several cancers. In this study, we aimed to observe the clinical implications and biological functions of MCU in gastric cancer. Methods The expression of MCU in 90 pairs of gastric cancer tissues and adjacent normal tissues was examined using immunohistochemistry and correlation between MCU expression and clinical features was analyzed. After construction of stable MCU knockdown or overexpression gastric cancer cells, mitochondrial membrane potential (MMP), wound healing and transwell assays were performed to examine MMP levels, migration and invasion. Subcutaneous xenograft tumors induced by gastric cancer cells transfected with MCU siRNAs or controls were constructed. Immunofluorescence was used to detect CD34 expression. Western blot was used to detect the expression of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), epithelial-mesenchymal transition (EMT)-related proteins. Results MCU had a higher expression in gastric cancer tissues than normal tissues. Compared to gastric cancer tissues, its expression was significantly higher after omental metastasis. MCU expression was significantly correlated with depth of invasion (p=0.048), lymph metastasis (p=0.027), TNM stage (p=0.036) and distant metastasis (p=0.029). Patients with high MCU expression indicated a worse prognosis than those with its low expression (p=0.0098). MCU significantly increased the MMP levels of gastric cancer cells. Wound healing and transwell assay results showed that MCU promoted migration and invasion of gastric cancer cells. In vivo, MCU knockdown significantly inhibited tumor growth and angiogenesis. Both in vitro and in vivo, silencing MCU suppressed the expression of HIF-1α and VEGF as well as activity of EMT processes. Conclusion Our findings suggested that highly expressed MCU could promote migration, invasion, angiogenesis and growth of gastric cancer, which could become a potential therapeutic marker for gastric cancer.
Collapse
Affiliation(s)
- Xiaofei Wang
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Xudong Song
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Guang Cheng
- Central Laboratory of Clinical Medical College, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Jingwen Zhang
- School of Basic Medical Science, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, NingXia Medical University, Yinchuan 750004, NingXia, People's Republic of China
| | - Liru Dong
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Jie Bai
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Dan Luo
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Yanjie Xiong
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Shuang Li
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Fang Liu
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Yuanyuan Sun
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Xin Wang
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Yuyang Li
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei, People's Republic of China
| | - Yunning Huang
- Department of Gastrointestinal Surgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750001, Ningxia, People's Republic of China
| |
Collapse
|
25
|
Li W, Duan J, Shi W, Lei L, Lv P. Long Non-Coding RNA NCK1-AS1 Serves an Oncogenic Role in Gastric Cancer by Regulating miR-137/NUP43 Axis. Onco Targets Ther 2020; 13:9929-9939. [PMID: 33116577 PMCID: PMC7547806 DOI: 10.2147/ott.s259336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/07/2020] [Indexed: 01/19/2023] Open
Abstract
Introduction Long non-coding RNA (lncRNA) NCK1-AS1 could regulate multiple cancer progression. However, little is known regarding the roles and acting mechanisms of NCK-AS1 in gastric cancer (GC) progression. This work was aimed to explore the relationship between NCK1-AS1 and GC progression to illustrate the mechanisms of NCK1-AS1. Methods NCK1-AS1 expression level in GC tissues and cells was measured with a quantitative real-time PCR method. In vitro experiments including cell counting kit-8 assay, colony formation assay, wound-healing assay, and transwell invasion assay were employed to detect biological roles of NCK1-AS1 in GC progression. In vivo experiments were performed to analyze the roles of NCK1-AS1 on GC malignant phenotype. Moreover, mechanisms behind the biological roles of NCK1-AS1 in GC were investigated using bioinformatic analysis, luciferase activity reporter assay, RNA immunoprecipitation assay, and rescue experiments. Results NCK1-AS1 was found to have elevated expression in GC tissues and cells in comparison with normal counterparts. Loss-of-function experiments showed knockdown of NCK1-AS1 refrained GC cell proliferation, colony formation, migration, and invasion in vitro. Animal experiments showed silence of NCK1-AS1 suppresses tumor growth in vivo. Functionally, NCK1-AS1 serves as a sponge for microRNA-137 (miR-137) to upregulate nucleoporin 43 (NUP43) expression in GC. Rescue experiments proved the carcinogenic role of NCK1-AS1/miR-137/NUP43 axis in GC progression. Discussion In conclusion, the NCK1-AS1/miR-137/NUP43 axis was identified that could contribute to GC malignancy behaviors.
Collapse
Affiliation(s)
- Wenxing Li
- Department of General Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Jiming Duan
- Department of General Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Wenbin Shi
- Department of General Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Liqiang Lei
- Department of General Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Pin Lv
- Department of General Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030001, People's Republic of China
| |
Collapse
|
26
|
Li Y, Li Q, Gu J, Qian D, Qin X, Li D. Exosomal prostate-specific G-protein-coupled receptor induces osteoblast activity to promote the osteoblastic metastasis of prostate cancer. Transl Cancer Res 2020; 9:5857-5867. [PMID: 35117199 PMCID: PMC8798947 DOI: 10.21037/tcr-20-1858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/21/2020] [Indexed: 12/28/2022]
Abstract
Background Prostate cancer (PCa) is the second leading cause of cancer-related deaths worldwide. Prostate-specific G-protein-coupled receptor (PSGR) has been identified as a new potential biomarker and therapeutic target for PCa. However, the influence of exosomal PSGR on PCa metastasis remains unknown. This study aimed to identify the regulatory role of exosomal PSGR in the bone microenvironment, prior to metastasis of PCa and the underlying mechanism. Methods hFOB1.19 cells were co-cultured with PC-3 exosomes exhibiting PSGR overexpression. Alkaline phosphatase (ALP) and von Kossa staining methods were used to measure the osteogenesis of hFOB1.19 cells. RNA sequencing was used to screen the downstream target genes of PSGR and the signaling pathways involved. The expression of the candidate genes was verified using quantitative real-time polymerase chain reaction (qRT-PCR). Results ALP and von Kossa staining results showed that PC-3 exosomes with overexpressed PSGR enhanced osteogenesis of hFOB1.19 cells. A total of 853 mRNAs were differentially expressed in hFOB1.19 cells of the PSGR-overexpressing PC3 cell (PC3PSGR+ exosome) group compared to the negative exosome control (NC) group, among which 182 mRNAs were significantly upregulated and 671 were downregulated. The functional enrichment and pathway analysis showed that differentially expressed mRNAs were mainly involved in cellular responses to interleukin-1 (IL1), chemotaxis, inflammation, transcriptional misregulation in cancer, and MAKP and NF-κB signaling pathways. qRT-PCR showed that levels of intercellular adhesion molecule-1 (ICAM1), RELB proto-oncogene, NF-κB subunit (RELB), and IL1 beta (IL1B) were significantly decreased in hFOB1.19 cells of the PSGR-overexpression group. Conclusions This study suggests that PSGR may regulate the MAKP and NF-κB signaling pathways involved in the process of bony metastases by targeting ICAM1, RELB, and IL1B.
Collapse
Affiliation(s)
- Yao Li
- Department of Urology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Urology, Changzheng Hospital Affiliated to Naval Military Medical University, Shanghai, China
| | - Quan Li
- Department of Urology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jie Gu
- Department of Urology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Duocheng Qian
- Department of Urology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xiaojing Qin
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Dujian Li
- Department of Urology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Du Q, Liu J, Tian D, Zhang X, Zhu J, Qiu W, Wu J. Long Noncoding RNA LINC00173 Promotes NUTF2 Expression Through Sponging miR-765 and Facilitates Tumorigenesis in Glioma. Cancer Manag Res 2020; 12:7211-7217. [PMID: 32848473 PMCID: PMC7429190 DOI: 10.2147/cmar.s262279] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Background Glioma is one of the leading causes of cancer-related deaths. This study aimed to investigate the function and mechanism of long noncoding RNA (lncRNA) LINC00173 in the regulation of glioma progression. Methods LINC00173 expression was measured using qRT-PCR. Survival rate was analyzed through Kaplan–Meier method. CCK8, colony formation and EdU assays were performed to measure cell proliferation while transwell was used to determine cell migration and invasion. Luciferase reporter assay was conducted to test RNA interaction. Results LINC00173 expression was elevated in glioma tissues and cells. LINC00173 high expression predicted poor prognosis. Loss of LINC00173 inhibited proliferation, migration and invasion. LINC00173 interacted with miR-765 to enhance NUTF2 expression. MiR-765 expression was negatively correlated with LINC00173 and NUTF2 in glioma tissues. NUTF2 level was increased in glioma tissues. NUTF2 overexpression rescued the potential of proliferation, migration and invasion in LINC00173-silenced cells. Conclusion Our research demonstrated that LINC00173 promotes glioma progression through targeting miR-765/NUTF2 axis.
Collapse
Affiliation(s)
- Qinghua Du
- Neurosurgery Department, Lishui City People's Hospital, Lishui 323000, People's Republic of China
| | - Jin Liu
- Neurosurgery Department, Lishui City People's Hospital, Lishui 323000, People's Republic of China
| | - Da Tian
- Neurosurgery Department, Lishui City People's Hospital, Lishui 323000, People's Republic of China
| | - Xuelei Zhang
- Neurosurgery Department, Lishui City People's Hospital, Lishui 323000, People's Republic of China
| | - Jinwei Zhu
- Neurosurgery Department, Lishui City People's Hospital, Lishui 323000, People's Republic of China
| | - Weiwen Qiu
- Neurology Department, Lishui City People's Hospital, Lishui 323000, People's Republic of China
| | - Jun Wu
- Pathology Department, Lishui City People's Hospital, Lishui 323000, People's Republic of China
| |
Collapse
|