1
|
Li J, Tuo D, Guo G, Gao Y, Gan J. The clinical significance and oncogenic function of LRRFIP1 in pancreatic cancer. Discov Oncol 2024; 15:123. [PMID: 38634978 PMCID: PMC11026317 DOI: 10.1007/s12672-024-00977-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
PURPOSE Pancreatic cancer is a lethal malignancy with a grim prognosis. Previous studies have proven that Leucine Rich Repeat of Flightless-1 Interacting Protein 1 (LRRFIP1) plays a pivotal role in cell biological processes, while its clinical significance and function in pancreatic cancer remain to be elucidated. Hence, we aimed to explore the roles and mechanisms of LRRFIP1 in pancreatic cancer. METHODS The expression of LRRFIP1 in pancreatic cancer tissues and its clinical significance for pancreatic cancer were analyzed by immunohistochemistry assay and bioinformatic analysis. The influences of LRRFIP1 on the proliferation and migration of pancreatic cancer cells were assessed in vitro. The underlying mechanisms of LRRFIP1 in pancreatic cancer progression were explored using gene set enrichment analysis (GSEA) and molecular experiments. RESULTS The results showed that LRRFIP1 expression was significantly upregulated in pancreatic cancer tissues compared to the normal tissues, and such upregulation was associated with poor prognosis of patients with pancreatic cancer. GSEA revealed that LRRFIP1 upregulation was significantly associated with various cancer-associated signaling pathways, including PI3K/AKT signaling pathway and Wnt pathway. Furthermore, LRRFIP1 was found to be associated with the infiltration of various immune cells. Functionally, LRRFIP1 silencing suppressed cell proliferation somewhat and inhibited migration substantially. Further molecular experiments indicated that LRRFIP1 silencing inactivated the AKT/GSK-3β/β-catenin signaling axis. CONCLUSION Taken together, LRRFIP1 is associated with tumorigenesis, immune cell infiltration, and prognosis in pancreatic cancer, which suggests that LRRFIP1 may be a potential biomarker and therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Jinping Li
- Department of Histology and Embryology, School of Preclinical Medicine, Guilin Medical University, Guilin, 541199, Guangxi, People's Republic of China.
| | - Dayun Tuo
- Department of Histology and Embryology, School of Preclinical Medicine, Guilin Medical University, Guilin, 541199, Guangxi, People's Republic of China
- Department of Pathology, Liuzhou People's Hospital, Liuzhou, 545006, Guangxi, People's Republic of China
| | - Gunan Guo
- Department of Histology and Embryology, School of Preclinical Medicine, Guilin Medical University, Guilin, 541199, Guangxi, People's Republic of China
- School of Stomatology, Zhaoqing Medical College, Zhaoqing, 526020, Guangdong, People's Republic of China
| | - Yan Gao
- Department of Histology and Embryology, School of Preclinical Medicine, Guilin Medical University, Guilin, 541199, Guangxi, People's Republic of China
| | - Jinfeng Gan
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, Guangxi, People's Republic of China.
- Department of Gastroenterology, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, People's Republic of China.
| |
Collapse
|
2
|
Li J, Tuo D, Cheng T, Deng Z, Gan J. GCF2 mediates nicotine-induced cancer stemness and progression in hepatocellular carcinoma. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 271:115952. [PMID: 38218109 DOI: 10.1016/j.ecoenv.2024.115952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/10/2023] [Accepted: 01/05/2024] [Indexed: 01/15/2024]
Abstract
Cigarette smoking is one of the most impactful behavior-related risk factors for multiple cancers including hepatocellular carcinoma (HCC). Nicotine, as the principal component of tobacco, is not only responsible for smoking addiction but also a carcinogen; nevertheless, the underlying mechanisms remain unclear. Here we report that nicotine enhances HCC cancer stemness and malignant progression by upregulating the expression of GC-rich binding factor 2 (GCF2), a gene that was revealed to be upregulated in HCC and whose upregulation predicts poor prognosis, and subsequently activating the Wnt/ꞵ-catenin/SOX2 signaling pathway. We found that nicotine significantly increased GCF2 expression and that silencing of GCF2 reduced nicotine-induced cancer stemness and progression. Mechanistically, nicotine could stabilize the protein level of GCF2, and then GCF2 could robustly activate its downstream Wnt/β-catenin signaling pathway. Taken together, our results thus suggest that GCF2 is a potential target for a therapeutic strategy against nicotine-promoted HCC.
Collapse
Affiliation(s)
- Jinping Li
- Department of Histology and Embryology, School of Preclinical Medicine, Guilin Medical University, Guilin, Guangxi, PR China.
| | - Dayun Tuo
- Department of Histology and Embryology, School of Preclinical Medicine, Guilin Medical University, Guilin, Guangxi, PR China; Department of Pathology, Liuzhou People's Hospital, Liuzhou, Guangxi, PR China
| | - Tan Cheng
- Department of Human Anatomy, School of Preclinical Medicine, Guilin Medical University, Guilin, Guangxi, PR China
| | - Zhenyan Deng
- Department of Clinical Laboratory, Guilin Hospital of the Second Xiangya Hospital CSU, Guilin, Guangxi, PR China
| | - Jinfeng Gan
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, PR China; Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi, PR China.
| |
Collapse
|
3
|
Ma W, Bao Z, Qian Z, Zhang K, Fan W, Xu J, Ren C, Zhang Y, Jiang T. LRRFIP1, an epigenetically regulated gene, is a prognostic biomarker and predicts malignant phenotypes of glioma. CNS Neurosci Ther 2022; 28:873-883. [PMID: 35338570 PMCID: PMC9062568 DOI: 10.1111/cns.13817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 11/27/2022] Open
Abstract
Aims Glioblastoma (GBM) is the most common malignant brain tumor with an adverse prognosis in the central nervous system. Traditional histopathological diagnosis accompanied by subjective deviations cannot accurately reflect tumor characteristics for clinical guidance. DNA methylation plays a critical role in GBM genesis. The focus of this project was to identify an effective methylation point for the classification of gliomas, the interactions between DNA methylation and potential epigenetic targeted therapies for clinical treatments. Methods Three online (TCGA, CGGA, and REMBRANDT) databases were employed in this study. T‐test, Venn analysis, univariate cox analysis, and Pearson's correlation analysis were adopted to screen significant prognostic methylation genes. Clinical samples were collected to determine the distributions of LRRFIP1 (Leucine Rich Repeat of Flightless‐1 Interacting Protein) protein by immunohistochemistry assay. Kaplan–Meier survival and Cox analysis were adopted to evaluate the prognostic value of LRRFIP1. Nomogram model was used to construct a prediction model. GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway were performed to explore functions and related mechanisms of LRRFIP1 in gliomas. Results Our results showed that 16 genes were negatively connected with their methylation level and correlated with clinical prognosis of GBM patients. Among them, LRRFIP1 expression showed the highest correlation with its methylation level. LRRFIP1 was highly expressed in WHO IV, mesenchymal, and IDH wild‐type subtype. LRRFIP1 expression was an independent risk factor for OS (overall survival) in gliomas. Conclusion LRRFIP1 is an epigenetically regulated gene and a potential prognostic biomarker for glioma. Our research may be beneficial to evaluate clinical efficacy, assess the prognosis, and provide individualized treatment for gliomas.
Collapse
Affiliation(s)
- Wenping Ma
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Zhaoshi Bao
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Zenghui Qian
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Kenan Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Wenhua Fan
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Jianbao Xu
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Changyuan Ren
- Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Ying Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| |
Collapse
|
4
|
Identification and Validation of Marketing Weight-Related SNP Markers Using SLAF Sequencing in Male Yangzhou Geese. Genes (Basel) 2021; 12:genes12081203. [PMID: 34440377 PMCID: PMC8393582 DOI: 10.3390/genes12081203] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 11/17/2022] Open
Abstract
Growth performance is a complex economic trait for avian production. The swan goose (Anser cygnoides) has never been exploited genetically like chickens or other waterfowl species such as ducks. Traditional phenotypic selection is still the main method for genetic improvement of geese body weight. In this study, specific locus amplified fragment sequencing (SLAF-seq) with bulked segregant analysis (BSA) was conducted for discovering and genotyping single nucleotide polymorphisms (SNPs) associated with marketing weight trait in male geese. A total of 149,045 SNPs were obtained from 427,093 SLAF tags with an average sequencing depth of 44.97-fold and a Q30 value of 93.26%. After SNPs' filtering, a total of 12,917 SNPs were included in the study. The 31 highest significant SNPs-which had different allelic frequencies-were further validated by individual-based AS-PCR genotyping in two populations. The association between 10 novel SNPs and the marketing weight of male geese was confirmed. The 10 significant SNPs were involved in linear regression model analysis, which confirmed single-SNP associations and revealed three types of SNP networks for marketing weight. The 10 significant SNPs were located within or close to 10 novel genes, which were identified. The qPCR analysis showed significant difference between genotypes of each SNP in seven genes. Developed SLAF-seq and identified genes will enrich growth performance studies, promoting molecular breeding applications to boost the marketing weight of Chinese geese.
Collapse
|
5
|
Kawasaki S, Ohtsuka H, Sato Y, Douchi D, Sato M, Ariake K, Masuda K, Fukase K, Mizuma M, Nakagawa K, Hayashi H, Morikawa T, Motoi F, Unno M. Silencing of LRRFIP1 enhances the sensitivity of gemcitabine in pancreatic cancer cells by activating JNK/c-Jun signaling. Pancreatology 2021; 21:771-778. [PMID: 33707114 DOI: 10.1016/j.pan.2021.02.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND The epithelial-mesenchymal transition (EMT) in cancer cells has been shown to closely associate with the survival and drug resistance of cancer cells. We recently provided evidence that Wnt signal activator leucine-rich repeat in flightless-1-interacting protein 1 (LRRFIP1) regulates EMT in pancreatic cancer. LRRFIP1 silencing inhibits the translocation of β-catenin to the nucleus, which led to reverse EMT in cancer cells. It was suggested that LRRFIP1 was implicated in gemcitabine sensitivity by regulating EMT signaling. METHODS Gemcitabine chemosensitivity was investigated in LRRFIP1-knockdown pancreatic cancer cells (PANC-1 and MIA Paca-2). In addition, the effects of LRRFIP1 knockdown on JNK/SAPK (stress activated-protein kinase) signaling and apoptosis were evaluated. RESULTS LRRFIP1 silencing accelerates gemcitabine-induced caspase activity and cell death in pancreatic cancer cells. It was also revealed that gemcitabine-induced phosphorylation of c-Jun N-terminal kinase (JNK) and c-Jun were increased in LRRFIP1 knockdown cells. The activation of JNK/c-Jun in LRRFIP1-knockdown cells was significantly diminished by the inhibition of Rac activity. It was confirmed that the acquisition of gemcitabine sensitivity by LRRFIP1 silencing largely depends on the stimulation of JNK/SAPK (stress activated-protein kinase) signaling. CONCLUSIONS Our findings suggest that reversing EMT and transient activation of JNK might be essential for the gemcitabine sensitivity in LRRFIP1 knockdown pancreatic cancer cells. Our discoveries highlight the potential role of LRRFIP1 in the chemosensitivity related to the regulation of EMT signaling.
Collapse
Affiliation(s)
- Shuhei Kawasaki
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Hideo Ohtsuka
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan.
| | - Yoshihiro Sato
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Daisuke Douchi
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Masaki Sato
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Kyohei Ariake
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Kunihiro Masuda
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Koji Fukase
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Masamichi Mizuma
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Kei Nakagawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Hiroki Hayashi
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Takanori Morikawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Fuyuhiko Motoi
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Japan
| |
Collapse
|
6
|
Yao Y, Xu K, Sun Y, Tian T, Shen W, Sun F, Yuan W, Wu H, Chen G, Yuan L, Zhang W, Lu M, Lei M. MiR-215-5p inhibits the inflammation injury in septic H9c2 by regulating ILF3 and LRRFIP1. Int Immunopharmacol 2019; 78:106000. [PMID: 31806569 DOI: 10.1016/j.intimp.2019.106000] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs (ncRNAs) playing crucial roles in sepsis-induced diseases, including myocardial inflammation. Nevertheless, the expression pattern and role of miR-215-5p in myocardial inflammation are still un-investigated up to now. The purpose of our study is to further inquire the effect of miR-215-5p on lipopolysaccharide (LPS)-activated inflammation injury in H9c2 cells and the possibly associated mechanisms. First of all, LPS-induced H9c2 cells models were constructed and affirmed via detection of pro-inflammatory factors, the viability and apoptosis. MiR-215-5p was overtly down-regulated in LPS-treated H9c2 cells and miR-215-5p overexpression could suppress the inflammation injury. LRRFIP1 was proved to be the target gene of miR-215-5p and meanwhile, miR-215-5p also targeted ILF3 that experimented to bind to and stabilize LRRFIP1. Final rescue assays confirmed that the overexpression of LRRFIP1 or ILF3 rescued the repressive effect of miR-215-5p up-regulation on the inflammation injury in septic H9c2. Totally, miR-215-5p exerted protective function in the inflammation injury in septic H9c2 via targeting ILF3 and LRRFIP1, suggesting an additional treatment method for sepsis-activated myocardial inflammation.
Collapse
Affiliation(s)
- Yulong Yao
- Intensive Care Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Kailiang Xu
- Intensive Care Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Yuxia Sun
- Intensive Care Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Tianyu Tian
- Trauma Emergency Center, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Weihong Shen
- Intensive Care Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Fangyuan Sun
- Intensive Care Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Weifang Yuan
- Intensive Care Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Hao Wu
- Trauma Emergency Center, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Gang Chen
- Intensive Care Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Lin Yuan
- Intensive Care Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Wei Zhang
- Trauma Emergency Center, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China
| | - Ming Lu
- Trauma Emergency Center, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China.
| | - Ming Lei
- Intensive Care Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, NO.358, Datong Road, Pudong New District, Shanghai 200137, China.
| |
Collapse
|
7
|
Ma Y, Ren Y, Guan J. Knockdown of GC binding factor 2 by RNA interference inhibits invasion and migration of vascular smooth muscle cells. Mol Med Rep 2019; 20:1781-1789. [PMID: 31257544 PMCID: PMC6625445 DOI: 10.3892/mmr.2019.10410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 05/17/2019] [Indexed: 11/05/2022] Open
Abstract
GC binding factor 2 (GCF2) is a transcriptional repressor that inhibits the transcription of GC‑rich promoters, thereby regulating biological processes, including proliferation. However, the role of GCF2 in vascular smooth muscle cells (VSMCs) remains unclear. The level of α‑smooth muscle (α‑SM) actin was determined by immunofluorescence. Cell viability, migration and invasion were analyzed using Cell Counting Kit‑8, wound healing and Transwell assays, respectively. Apoptosis and cell cycle progression were determined using flow cytometry. The expressions of Bcl‑2, Bax, cleaved caspase‑3, cyclin E, CDK2 and the CDK inhibitor p21 were determined by reverse transcription‑quantitative (RT‑q)PCR and western blot analysis. RT‑qPCR was performed to analyze the levels of GCF2 and western blot analysis was conducted to determine the phosphorylation levels of PI3K and AKT. α‑SM actin was found to be expressed in VSMCs. Cell viability, migration and invasion were inhibited by small interfering (si)RNA targeting GCF2. Changes in the expression levels of Bcl‑2, Bax and cleaved caspase‑3 showed that the pro‑apoptotic capacity of the cells was increased by siGCF2. Cell cycle arrest in the G0/G1 phase was induced by siGCF2, which was accompanied by changes in the levels of cyclin E, CDK2 and p21. Furthermore, phosphorylation of PI3K and AKT was suppressed by siGCF2. However, the inhibitory effects of siGCF2 on cell viability, migration and invasion were increased by insulin‑like growth factor 1, which is a specific agonist of AKT. The anti‑proliferative activity of siGCF2 may be associated with the PI3K/AKT pathway in VSMCs.
Collapse
Affiliation(s)
- Ying Ma
- Qingdao University, Qingdao, Shandong 266073, P.R. China
| | - Yongqiang Ren
- Department of Cardiology, Qingdao Municipal Hospital (Group), Qingdao, Shandong 266034, P.R. China
| | - Jun Guan
- Department of Cardiology, Qingdao Municipal Hospital (Group), Qingdao, Shandong 266034, P.R. China
| |
Collapse
|
8
|
Takimoto M. Multidisciplinary Roles of LRRFIP1/GCF2 in Human Biological Systems and Diseases. Cells 2019; 8:cells8020108. [PMID: 30709060 PMCID: PMC6406849 DOI: 10.3390/cells8020108] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/21/2019] [Accepted: 01/27/2019] [Indexed: 01/28/2023] Open
Abstract
Leucine Rich Repeat of Flightless-1 Interacting Protein 1/GC-binding factor 2 (LRRFIP1/GCF2) cDNA was cloned for a transcriptional repressor GCF2, which bound sequence-specifically to a GC-rich element of epidermal growth factor receptor (EGFR) gene and repressed its promotor. LRRFIP1/GCF2 was also cloned as a double stranded RNA (dsRNA)-binding protein to trans-activation responsive region (TAR) RNA of Human Immunodeficiency Virus-1 (HIV-1), termed as TAR RNA interacting protein (TRIP), and as a binding protein to the Leucine Rich Repeat (LRR) of Flightless-1(Fli-1), termed as Flightless-1 LRR associated protein 1 (FLAP1) and LRR domain of Flightless-1 interacting Protein 1 (LRRFIP1). Subsequent functional studies have revealed that LRRFIP1/GCF2 played multiple roles in the regulation of diverse biological systems and processes, such as in immune response to microorganisms and auto-immunity, remodeling of cytoskeletal system, signal transduction pathways, and transcriptional regulations of genes. Dysregulations of LRRFIP1/GCF2 have been implicated in the causes of several experimental and clinico-pathological states and the responses to them, such as autoimmune diseases, excitotoxicity after stroke, thrombosis formation, inflammation and obesity, the wound healing process, and in cancers. LRRFIP1/GCF2 is a bioregulator in multidisciplinary systems of the human body and its dysregulation can cause diverse human diseases.
Collapse
Affiliation(s)
- Masato Takimoto
- Institute for Genetic Medicine, Hokkaido University, Hokkaido 060-0815, Japan.
| |
Collapse
|
9
|
Kopecki Z, Stevens NE, Yang GN, Melville E, Cowin AJ. Recombinant Leucine-Rich Repeat Flightless-Interacting Protein-1 Improves Healing of Acute Wounds through Its Effects on Proliferation Inflammation and Collagen Deposition. Int J Mol Sci 2018; 19:ijms19072014. [PMID: 29996558 PMCID: PMC6073877 DOI: 10.3390/ijms19072014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/04/2018] [Accepted: 07/07/2018] [Indexed: 02/07/2023] Open
Abstract
Wound healing is an increasing clinical problem involving substantial morbidity, mortality, and rising health care costs. Leucine-rich repeat flightless-interacting protein-1 (LRRFIP-1) regulates toll-like receptor (TLR)-mediated inflammation, suggesting a potential role in the healing of wounds. We sought to determine the role of LRRFIP-1 in wound repair and whether the exogenous addition of recombinant LRRFIP-1 (rLRRFIP-1) affected healing responses. Using a model of full-thickness incisional acute wounds in BALB/c mice, we investigated the effect of wounding on LRRFIP-1 expression. The effect of rLRRFIP-1 on cellular proliferation, inflammation, and collagen deposition was also investigated. LRRFIP-1 was upregulated in response to wounding, was found to directly associate with flightless I (Flii), and significantly increased cellular proliferation both in vitro and in vivo. rLRRFIP-1 reduced Flii expression in wounds in vivo and resulted in significantly improved healing with a concurrent dampening of TLR4-mediated inflammation and improved collagen deposition. Additionally, decreased levels of TGF-β1 and increased levels of TGF-β3 were observed in rLRRFIP-1-treated wounds suggesting a possible antiscarring effect of rLRRFIP-1. Further studies are required to elucidate if the mechanisms behind LRRFIP-1 action in wound repair are independent of Flii. However, these results identify rLRRFIP-1 as a possible treatment modality for improved healing of acute wounds.
Collapse
Affiliation(s)
- Zlatko Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| | - Natalie E Stevens
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| | - Gink N Yang
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| | - Elizabeth Melville
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| | - Allison J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| |
Collapse
|
10
|
Liu YC, Ma WH, Ge YL, Xue ML, Zhang Z, Zhang JY, Hou L, Mu RH. RNAi-mediated gene silencing of vascular endothelial growth factor C suppresses growth and induces apoptosis in mouse breast cancer in vitro and in vivo. Oncol Lett 2016; 12:3896-3904. [PMID: 27895746 PMCID: PMC5104198 DOI: 10.3892/ol.2016.5158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/19/2016] [Indexed: 12/23/2022] Open
Abstract
Vascular endothelial cell growth factor (VEGF)-C promotes tumorigenesis by allowing lymph node metastasis and lymphangiogenesis, among other actions. RNA interference (RNAi) is a novel technique for suppressing target gene expression and may increase the effectiveness of cancer treatments. The present study assessed the influence of VEGF-C RNAi on the apoptosis and proliferation of mouse breast cancer cells in vitro and in vivo. A total of three pairs of small interfering RNA (siRNA) targeting mouse VEGF-C were designed and synthesized prior to transfection into 4T1 cells via a liposomal approach. Reverse transcription polymerase chain reaction, western blot analysis, a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, Hoechst 33258 staining and flow cytometry were performed in vitro to analyze VEGF-C expression, cleaved caspase-3 protein expression and 4T1 cell proliferation and apoptosis. Experiments were also conducted in vivo on BALB/c mice with breast cancer. Tumor weight and volume were measured and the number of apoptotic cells in tumor tissues was assessed by a TUNEL assay. Immunohistochemical assays and an enzyme-linked immunosorbent assay were used to measure the expression of VEGF-C in tumor tissues. The results demonstrated that the three pairs of siRNA, particularly siV2, significantly reduced VEGF-C mRNA and protein levels in 4T1 cells. siV2 was deemed to be the most efficient siRNA and therefore was selected to be used in subsequent experiments. Furthermore, in vitro studies indicated that VEGF-C RNAi significantly decreased cell growth, induced apoptosis and upregulated the expression of cleaved caspase-3 protein. Tumor weight and volume in breast cancer in vivo models was reduced by the intratumoral injection of siV2. Antitumor efficacy was associated with decreased VEGF-C expression and increased induction of apoptosis. The present study therefore indicated that VEGF-C RNAi inhibited mouse breast cancer growth in vitro and in vivo and that it may be a novel targeted therapy for breast cancer.
Collapse
Affiliation(s)
- Yong-Chao Liu
- Department of Biochemistry and Molecular Biology, Medical College, Qingdao University, Qingdao, Shandong 266021, P.R. China; Department of Immunology, Medical College, Beihua University, Jilin, Jilin 132013, P.R. China
| | - Wen-Hui Ma
- Department of Biochemistry and Molecular Biology, Medical College, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Yin-Lin Ge
- Department of Biochemistry and Molecular Biology, Medical College, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Mei-Lan Xue
- Department of Biochemistry and Molecular Biology, Medical College, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Zheng Zhang
- Department of Biochemistry and Molecular Biology, Medical College, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Jin-Yu Zhang
- Department of Biochemistry and Molecular Biology, Medical College, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Lin Hou
- Department of Biochemistry and Molecular Biology, Medical College, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Run-Hong Mu
- Department of Immunology, Medical College, Beihua University, Jilin, Jilin 132013, P.R. China
| |
Collapse
|
11
|
Farshori NN, Al-Sheddi ES, Al-Oqail MM, Musarrat J, Al-Khedhairy AA, Siddiqui MA. Cytotoxicity assessments of Portulaca oleracea and Petroselinum sativum seed extracts on human hepatocellular carcinoma cells (HepG2). Asian Pac J Cancer Prev 2015; 15:6633-8. [PMID: 25169500 DOI: 10.7314/apjcp.2014.15.16.6633] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The Pharmacological potential, such as antioxidant, anti-inflammatory, and antibacterial activities of Portulaca oleracea (PO) and Petroselinum sativum (PS) extracts are well known. However, the preventive properties against hepatocellular carcinoma cells have not been explored so far. Therefore, the present investigation was designed to study the anticancer activity of seed extracts of PO and PS on the human hepatocellular carcinoma cells (HepG2). The HepG2 cells were exposed with 5-500 μg/ml of PO and PS for 24 h. After the exposure, cell viability by 3-(4,5-dimethylthiazol-2yl)-2,5-biphenyl tetrazolium bromide (MTT) assay, neutral red uptake (NRU) assay, and cellular morphology by phase contrast inverted microscope were studied. The results showed that PO and PS extracts significantly reduced the cell viability of HepG2 in a concentration dependent manner. The cell viability was recorded to be 67%, 31%, 21%, and 17% at 50, 100, 250, and 500 μg/ml of PO, respectively by MTT assay and 91%, 62%, 27%, and 18% at 50, 100, 250, and 500 μg/ml of PO, respectively by NRU assay. PS exposed HepG2 cells with 100 μg/ml and higher concentrations were also found to be cytotoxic. The decrease in the cell viability at 100, 250, and 500 μg/ml of PS was recorded as 70%, 33%, and 15% by MTT assay and 63%, 29%, and 17%, respectively by NRU assay. Results also showed that PO and PS exposed cells reduced the normal morphology and adhesion capacity of HepG2 cells. HepG2 cells exposed with 50 μg/ml and higher concentrations of PO and PS lost their typical morphology, become smaller in size, and appeared in rounded bodies. Our results demonstrated preliminary screening of anticancer activity of Portulaca oleracea and Petroselinum sativum extracts against HepG2 cells, which can be further used for the development of a potential therapeutic anticancer agent.
Collapse
Affiliation(s)
- Nida Nayyar Farshori
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia E-mail :
| | | | | | | | | | | |
Collapse
|
12
|
Shi QQ, Zuo GW, Feng ZQ, Zhao LC, Luo L, You ZM, Li DY, Xia J, Li J, Chen DL. Effect of trichostatin A on anti HepG2 liver carcinoma cells: inhibition of HDAC activity and activation of Wnt/β-Catenin signaling. Asian Pac J Cancer Prev 2014; 15:7849-7855. [PMID: 25292076 DOI: 10.7314/apjcp.2014.15.18.7849] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
PURPOSE To investigate the effect of deacetylase inhibitory trichostatin A (TSA) on anti HepG2 liver carcinoma cells and explore the underlying mechanisms. MATERIALS AND METHODS HepG2 cells exposed to different concentrations of TSA for 24, 48, or 72h were examined for cell growth inhibition using CCK8, changes in cell cycle distribution with flow cytometry, cell apoptosis with annexin V-FTIC/PI double staining, and cell morphology changes under an inverted microscope. Expression of β-catenin, HDAC1, HDAC3, H3K9, CyclinD1 and Bax proteins was tested by Western blotting. Gene expression for β-catenin, HDAC1and HDAC3 was tested by q-PCR. β-Catenin and H3K9 proteins were also tested by immunofluorescence. Activity of Renilla luciferase (pTCF/LEF-luc) was assessed using the Luciferase Reporter Assay system reagent. The activity of total HDACs was detected with a HDACs colorimetric kit. RESULTS Exposure to TSA caused significant dose-and time-dependent inhibition of HepG2 cell proliferation (p<0.05) and resulted in increased cell percentages in G0/ G1 and G2/M phases and decrease in the S phase. The apoptotic index in the control group was 6.22±0.25%, which increased to 7.17±0.20% and 18.1±0.42% in the treatment group. Exposure to 250 and 500nmol/L TSA also caused cell morphology changes with numerous floating cells. Expression of β-catenin, H3K9and Bax proteins was significantly increased, expression levels of CyclinD1, HDAC1, HDAC3 were decreased. Expression of β-catenin at the genetic level was significantly increased, with no significant difference in HDAC1and HDAC3 genes. In the cytoplasm, expression of β-catenin fluorescence protein was not obvious changed and in the nucleus, small amounts of green fluorescence were observed. H3K9 fluorescence protein were increased. Expression levels of the transcription factor TCF werealso increased in HepG2 cells following induction by TSA, whikle the activity of total HDACs was decreased. CONCLUSIONS TSA inhibits HDAC activity, promotes histone acetylation, and activates Wnt/β-catenin signaling to inhibit proliferation of HepG2 cell, arrest cell cycling and induce apoptosis.
Collapse
Affiliation(s)
- Qing-Qiang Shi
- Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, China E-mail : ;
| | | | | | | | | | | | | | | | | | | |
Collapse
|