1
|
Yao Y, Li X, Yang X, Mou H, Wei L. Indirubin, an Active Component of Indigo Naturalis, Exhibits Inhibitory Effects on Leukemia Cells via Targeting HSP90AA1 and PI3K/Akt Pathway. Anticancer Agents Med Chem 2024; 24:718-727. [PMID: 38347773 DOI: 10.2174/0118715206258293231017063340] [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: 04/17/2023] [Revised: 08/10/2023] [Accepted: 09/14/2023] [Indexed: 06/04/2024]
Abstract
BACKGROUND This research intended to predict the active ingredients and key target genes of Indigo Naturalis in treating human chronic myeloid leukemia (CML) using network pharmacology and conduct the invitro verification. METHODS The active components of Indigo Naturalis and the corresponding targets and leukemia-associated genes were gathered through public databases. The core targets and pathways of Indigo Naturalis were predicted through protein-protein interaction (PPI) network, gene ontology (GO) function, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Next, after intersecting with leukemia-related genes, the direct core target gene of Indigo Naturalis active components was identified. Subsequently, HL-60 cells were stimulated with indirubin (IND) and then examined for cell proliferation using CCK-8 assay and cell cycle, cell apoptosis, and mitochondrial membrane potential using flow cytometry. The content of apoptosis-associated proteins (Cleaved Caspase 9, Cleaved Caspase 7, Cleaved Caspase 3, and Cleaved parp) were detected using Western blot, HSP90AA1 protein, and PI3K/Akt signaling (PI3K, p-PI3K, Akt, and p-Akt) within HL-60 cells. RESULTS A total of 9 active components of Indigo Naturalis were screened. The top 10 core target genes (TNF, PTGS2, RELA, MAPK14, IFNG, PPARG, NOS2, IKBKB, HSP90AA1, and NOS3) of Indigo Naturalis active components within the PPI network were identified. According to the KEGG enrichment analysis, these targets were associated with leukemia-related pathways (such as acute myeloid leukemia and CML). After intersecting with leukemia-related genes, it was found that IND participated in the most pairs of target information and was at the core of the target network; HSP90AA1 was the direct core gene of IND. Furthermore, the in-vitro cell experiments verified that IND could inhibit the proliferation, elicit G2/M-phase cell cycle arrest, enhance the apoptosis of HL-60 cells, reduce mitochondrial membrane potential, and promote apoptosis-related protein levels. Under IND treatment, HSP90AA1 overexpression notably promoted cell proliferation and inhibited apoptosis. Additionally, IND exerted tumor suppressor effects on leukemia cells by inhibiting HSP90AA1 expression. CONCLUSION IND, an active component of Indigo Naturalis, could inhibit CML progression, which may be achieved via inhibiting HSP90AA1 and PI3K/Akt signaling expression levels.
Collapse
MESH Headings
- Humans
- Cell Proliferation/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Apoptosis/drug effects
- Phosphatidylinositol 3-Kinases/metabolism
- Indoles/pharmacology
- Indoles/chemistry
- HSP90 Heat-Shock Proteins/antagonists & inhibitors
- HSP90 Heat-Shock Proteins/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Drug Screening Assays, Antitumor
- HL-60 Cells
- Molecular Structure
- Dose-Response Relationship, Drug
- Structure-Activity Relationship
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia/drug therapy
- Leukemia/pathology
- Leukemia/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Yuanzhi Yao
- College of Biology and Food Engineering, Huaihua University, Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Huaihua 418000, China
| | - Xiaoying Li
- College of Biology and Food Engineering, Huaihua University, Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Huaihua 418000, China
| | - Xiaoqin Yang
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Hai Mou
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Lin Wei
- College of Biology and Food Engineering, Huaihua University, Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Huaihua 418000, China
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| |
Collapse
|
2
|
Pharmacological properties of indirubin and its derivatives. Biomed Pharmacother 2022; 151:113112. [PMID: 35598366 DOI: 10.1016/j.biopha.2022.113112] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/23/2022] [Accepted: 05/10/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Indirubin is the main bioactive component of the traditional Chinese medicine Indigo naturalis and is a bisindole alkaloid. Multiple studies have shown that indirubin exhibits good anticancer, anti-inflammatory and neuroprotective properties. METHODS The purpose of this review is to provide a summary of the pharmacological mechanisms of indirubin and its derivatives. RESULTS Indirubin and its derivatives exert anticancer effects by regulating the expression of cyclin-dependent kinases (CDKs), GSK-3β, Bax, Bcl-2, C-MYC, matrix metalloproteinases (MMPs), and focal adhesion kinase (FAK) through the PI3K/AKT/mTOR, nuclear factor (NF)-κB, mitogen-activated protein kinase (MAPK), JAK/signal transducer and activator of transcription 3 (STAT3) pathways and other signaling pathways. We also reviewed the anti-inflammatory and neuroprotective properties of indirubin and its derivatives. CONCLUSION The findings of recent studies assessing indirubin and its derivatives suggest that these compounds can be used as potential drugs to treat tumors, inflammation, neuropathy and bacterial infection.
Collapse
|
3
|
Jo A, Kwak JH, Woo SY, Kim BY, Son Y, Choi HS, Kim J, Kwon M, Cho HR, Eo SK, Nam JH, Kim HS, Baryawno N, Lee D, Kim K. Oxime derivative TFOBO promotes cell death by modulating reactive oxygen species and regulating NADPH oxidase activity in myeloid leukemia. Sci Rep 2022; 12:7519. [PMID: 35525902 PMCID: PMC9079095 DOI: 10.1038/s41598-022-11543-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/18/2022] [Indexed: 11/09/2022] Open
Abstract
Several derivatives derived from the oxime structure have been reported as potential anticancer agents in various cancers. Here, we first tested a novel oxime-containing derivative of 2-((2,4,5-trifluorobenzyl)oxy)benzaldehyde oxime (TFOBO) to evaluate its anticancer effect in myeloid leukemic cells. Compared to (2-((2,4,5-trifluorobenzyl)oxy)phenyl)methanol (TFOPM), the oxime derivative TFOBO suppresses leukemic cell growth by significantly increasing reactive oxygen species (ROS) levels and cell death. Leukemic cells treated with TFOBO displayed apoptotic cell death, as indicated by nuclear condensation, DNA fragmentation, and annexin V staining. TFOBO increases Bax/Bcl2 levels, caspase9, and caspase3/7 activity and decreases mitochondrial membrane potential. ROS production was reduced by N-acetyl-L-cysteine, a ROS scavenger, diphenyleneiodonium chloride, a nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor, after exogenous TFOBO treatment. ROS inhibitors protect leukemic cells from TFOBO-induced cell death. Thus, our study findings suggest that TFOBO promotes apoptosis by modulating ROS and regulating NADPH oxidase activity. Collectively, the oxime-containing derivative TFOBO is a novel therapeutic drug for myeloid leukemia.
Collapse
Affiliation(s)
- Ahyoung Jo
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Jae-Hwan Kwak
- College of Pharmacy, Kyungsung University, Busan, 48434, Republic of Korea
| | - Soo-Yeon Woo
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Bo-Young Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Yonghae Son
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hee-Seon Choi
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Jayoung Kim
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Munju Kwon
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hyok-Rae Cho
- Department of Neurosurgery, College of Medicine, Kosin University, Busan, 49267, Republic of Korea
| | - Seong-Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Ji Ho Nam
- Department of Radiation Oncology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Hyung-Sik Kim
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
4
|
Chang JM, Wu JY, Chen SH, Chao WY, Chuang HH, Kam KH, Zhao PW, Li YZ, Yen YP, Lee YR. 9-O-Terpenyl-Substituted Berberrubine Derivatives Suppress Tumor Migration and Increase Anti-Human Non-Small-Cell Lung Cancer Activity. Int J Mol Sci 2021; 22:ijms22189864. [PMID: 34576028 PMCID: PMC8469690 DOI: 10.3390/ijms22189864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/06/2021] [Accepted: 09/10/2021] [Indexed: 01/06/2023] Open
Abstract
Lung cancer is one of the most common cancers and the leading cause of death in humans worldwide. Non-small-cell lung cancer (NSCLC) accounts for approximately 85% of lung cancer cases and is often diagnosed at a late stage. Among patients with NSCLC, 50% die within 1 year after diagnosis. Even with clinical intervention, the 5-year survival rate is only approximately 20%. Therefore, the development of an advanced therapeutic strategy or novel agent is urgently required for treating NSCLC. Berberine exerts therapeutic activity toward NSCLC; therefore, its activity as an antitumor agent needs to be explored further. In this study, three terpenylated-bromide derivatives of berberrubine were synthesized and their anti-NSCLC activities were evaluated. Each derivative had higher anti-NSCLCs activity than berberrubine and berberine. Among them, 9-O-gernylberberrubine bromide (B4) and 9-O-farnesylberberrubine bromide (B5) showed greater growth inhibition, cell-cycle regulation, in vitro tumorigenesis suppression, and tumor migration reduction. In addition, some degree of apoptosis and autophagic flux blocking was noted in the cells under B4 and B5 treatments. Our study demonstrates that the berberrubine derivatives, B4 and B5, exhibit impressive anti-NSCLC activities and have potential for use as chemotherapeutic agents against NSCLC.
Collapse
Affiliation(s)
- Jia-Ming Chang
- Department of Surgery, Division of Thoracic Surgery, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 60002, Taiwan; (J.-M.C.); (K.-H.K.)
- Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 60002, Taiwan; (S.-H.C.); (P.-W.Z.); (Y.-Z.L.); (Y.-P.Y.)
| | - Jin-Yi Wu
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi 60004, Taiwan;
| | - Shu-Hsin Chen
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 60002, Taiwan; (S.-H.C.); (P.-W.Z.); (Y.-Z.L.); (Y.-P.Y.)
| | - Wen-Ying Chao
- Department of Nursing, Min-Hwei College of Health Care Management, Tainan 73658, Taiwan;
| | - Hsiang-Hao Chuang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Kam-Hong Kam
- Department of Surgery, Division of Thoracic Surgery, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 60002, Taiwan; (J.-M.C.); (K.-H.K.)
| | - Pei-Wen Zhao
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 60002, Taiwan; (S.-H.C.); (P.-W.Z.); (Y.-Z.L.); (Y.-P.Y.)
| | - Yi-Zhen Li
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 60002, Taiwan; (S.-H.C.); (P.-W.Z.); (Y.-Z.L.); (Y.-P.Y.)
| | - Yu-Pei Yen
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City 60002, Taiwan; (S.-H.C.); (P.-W.Z.); (Y.-Z.L.); (Y.-P.Y.)
| | - Ying-Ray Lee
- Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: ; Tel.: +886-7-3121101
| |
Collapse
|
5
|
Wang H, Wang Z, Wei C, Wang J, Xu Y, Bai G, Yao Q, Zhang L, Chen Y. Anticancer potential of indirubins in medicinal chemistry: Biological activity, structural modification, and structure-activity relationship. Eur J Med Chem 2021; 223:113652. [PMID: 34161865 DOI: 10.1016/j.ejmech.2021.113652] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/13/2021] [Accepted: 06/13/2021] [Indexed: 10/21/2022]
Abstract
Indirubin is the crucial ingredient of Danggui Longhui Wan and Qing-Dai, traditional Chinese medicine herbal formulas used for the therapy of chronic myelocytic leukemia in China for hundreds of years. Although the monomeric indirubin has been used in China for the treatment human chronic myelocytic leukemia. However, due to low water solubility, poor pharmacokinetic properties and low therapeutic effects are the major obstacle, and had significantly limited its clinical application. Consequently, the attractive anticancer profile of indirubin has enthused numerous researchers to discover novel indirubin derivatives with improved pharmacodynamic activity as well as good pharmacokinetic property. In this paper, we comprehensively review the recent progress of anticancer potential of indirubins, structural modification and structure-activity relationship, which may provide useful direction for the further development of novel indirubins with improved pharmacological profiles for the treatment of various types of cancer.
Collapse
Affiliation(s)
- Hezhen Wang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Zhiyuan Wang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Chunyong Wei
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Jing Wang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Yingshu Xu
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China
| | - Guohui Bai
- Key Laboratory of Oral Disease of Higher Schools in Guizhou Province, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China.
| | - Qizheng Yao
- School of Pharmacy, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing, 210009, PR China.
| | - Lei Zhang
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China.
| | - Yongzheng Chen
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xuefu Road, Zunyi, 563000, PR China.
| |
Collapse
|
6
|
Schepetkin IA, Plotnikov MB, Khlebnikov AI, Plotnikova TM, Quinn MT. Oximes: Novel Therapeutics with Anticancer and Anti-Inflammatory Potential. Biomolecules 2021; 11:biom11060777. [PMID: 34067242 PMCID: PMC8224626 DOI: 10.3390/biom11060777] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Oximes have been studied for decades because of their significant roles as acetylcholinesterase reactivators. Over the last twenty years, a large number of oximes have been reported with useful pharmaceutical properties, including compounds with antibacterial, anticancer, anti-arthritis, and anti-stroke activities. Many oximes are kinase inhibitors and have been shown to inhibit over 40 different kinases, including AMP-activated protein kinase (AMPK), phosphatidylinositol 3-kinase (PI3K), cyclin-dependent kinase (CDK), serine/threonine kinases glycogen synthase kinase 3 α/β (GSK-3α/β), Aurora A, B-Raf, Chk1, death-associated protein-kinase-related 2 (DRAK2), phosphorylase kinase (PhK), serum and glucocorticoid-regulated kinase (SGK), Janus tyrosine kinase (JAK), and multiple receptor and non-receptor tyrosine kinases. Some oximes are inhibitors of lipoxygenase 5, human neutrophil elastase, and proteinase 3. The oxime group contains two H-bond acceptors (nitrogen and oxygen atoms) and one H-bond donor (OH group), versus only one H-bond acceptor present in carbonyl groups. This feature, together with the high polarity of oxime groups, may lead to a significantly different mode of interaction with receptor binding sites compared to corresponding carbonyl compounds, despite small changes in the total size and shape of the compound. In addition, oximes can generate nitric oxide. This review is focused on oximes as kinase inhibitors with anticancer and anti-inflammatory activities. Oximes with non-kinase targets or mechanisms of anti-inflammatory activity are also discussed.
Collapse
Affiliation(s)
- Igor A. Schepetkin
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA;
| | - Mark B. Plotnikov
- Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, 634028 Tomsk, Russia;
| | - Andrei I. Khlebnikov
- Kizhner Research Center, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
- Scientific Research Institute of Biological Medicine, Altai State University, 656049 Barnaul, Russia
| | - Tatiana M. Plotnikova
- Department of Pharmacology, Siberian State Medical University, 634050 Tomsk, Russia;
| | - Mark T. Quinn
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA;
- Correspondence: ; Tel.: +1-406-994-4707; Fax: +1-406-994-4303
| |
Collapse
|
7
|
FAS/FAS-L-mediated apoptosis and autophagy of SPC-A-1 cells induced by water-soluble polysaccharide from Polygala tenuifolia. Int J Biol Macromol 2020; 150:449-458. [DOI: 10.1016/j.ijbiomac.2020.02.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 02/02/2020] [Accepted: 02/03/2020] [Indexed: 12/21/2022]
|
8
|
Yang H, Jiang Z, Wang S, Zhao Y, Song X, Xiao Y, Yang S. Long non-coding small nucleolar RNA host genes in digestive cancers. Cancer Med 2019; 8:7693-7704. [PMID: 31691514 PMCID: PMC6912041 DOI: 10.1002/cam4.2622] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 08/21/2019] [Accepted: 09/26/2019] [Indexed: 12/22/2022] Open
Abstract
Although long noncoding RNAs (lncRNAs) do not have protein coding capacities, they are involved in the pathogenesis of many types of cancers, including hepatocellular carcinoma, cervical cancer, and gastric cancer. Notably, the roles of lncRNAs are vital in nearly every aspect of tumor biology. Long non-coding small nucleolar RNA host genes (lnc-SNHGs) are abnormally expressed in multiple cancers, including urologic neoplasms, respiratory tumors, and digestive cancers, and play vital roles in these cancers. These host genes could participate in tumorigenesis by regulating proliferation, migration, invasion and apoptosis of tumor cells. This review focuses on the overview of the roles that lnc-SNHGs play in the formation and progression of digestive cancers.
Collapse
Affiliation(s)
- Huan Yang
- Department of GastroenterologyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Zheng Jiang
- Department of GastroenterologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Shuang Wang
- Department of GastroenterologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of GastroenterologyPeople's Hospital of Changshou ChongqingChongqingChina
| | - Yongbing Zhao
- Department of GastroenterologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of GastroenterologyPeople's Hospital of Changshou ChongqingChongqingChina
| | - Xiaomei Song
- Department of GastroenterologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of GastroenterologyPeople's Hospital of Changshou ChongqingChongqingChina
| | - Yufeng Xiao
- Department of GastroenterologyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Shiming Yang
- Department of GastroenterologyXinqiao HospitalArmy Medical UniversityChongqingChina
| |
Collapse
|