1
|
El-Hammady AM, Marei YM, Mohammed RR, Rahman SMAE, Marei YM, Sarhan RS. PLASMA EXPRESSION LEVELS OF MICRORNA-21 MIGHT HELP IN THE DETECTION OF HCV PATIENTS COMPLICATED BY HEPATOCELLULAR CARCINOMA. ARQUIVOS DE GASTROENTEROLOGIA 2024; 61:e24025. [PMID: 39776121 DOI: 10.1590/s0004-2803.24612024-025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/29/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVE To investigate the ability of the estimated plasma gene-expression levels of microRNA (miR)-21 and 126 to define patients suspected to have hepatocellular carcinoma (HCC) among patients with complicated hepatitis-C virus (HCV) infection. METHODS Patients with uncomplicated (U-HCV) or complicated HCV underwent clinical and ultrasonographic (US) evaluations and assessment for the computerized hepatorenal index, hepatic steatosis index and fibrosis indices. Blood samples were obtained for estimation of serum levels of alpha-fetoprotein (AFP) and tumor necrosis factor-α (TNF-α), and plasma expression levels of miR-21 and miR-126 using the quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR). RESULTS Serum levels of AFP and TNF-α were significantly higher in samples of HCV-HCC patients than controls and other HCV patients. Plasma levels of miR-21 were the highest, while miR-126 levels were the lowest in samples of HCV-HCC patients with significant differences in comparison to samples of controls and other HCV patients. The ROC curve analysis defined high plasma miR-21 levels as specific predictor for HCV infection, and could identify samples of complicated HCV, and samples of HCV-HCC patients, while estimated plasma levels of miR-126 could be applied to screen for HCV and its related complications. CONCLUSION Deregulated plasma expression levels of miR-21 and miR-126 might distinguish cases of HCV complicated by HCC and define cases of HCV-LC, even those that showed low Fib-4 scores.
Collapse
Affiliation(s)
- Amr M El-Hammady
- Department of Internal Medicine, Faculty of Medicine, Benha University, Benha, Egypt
| | - Yasmin M Marei
- Department of Medical Biochemistry, Faculty of Medicine, Benha University, Benha, Egypt
| | - Raafat R Mohammed
- Assistant Consultant of Medical Biochemistry, Hospital Lab, Clinical Pathology Department, Benha University, Benha, Egypt
| | | | - Yomna M Marei
- Department of Internal Medicine, Faculty of Medicine, Benha University, Benha, Egypt
| | - Rizk S Sarhan
- Department of Internal Medicine, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
2
|
Al-Ashmawy GM, El-Sherbeni SAEH, Ali DA, Abo-Saif MA. Chemotherapeutic effect of baicalein/epirubicin combination against liver cell carcinoma in-vitro: Inducing apoptosis and autophagy. Toxicol In Vitro 2024; 95:105744. [PMID: 38040128 DOI: 10.1016/j.tiv.2023.105744] [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: 07/15/2023] [Revised: 10/30/2023] [Accepted: 11/24/2023] [Indexed: 12/03/2023]
Abstract
Flavonoids have a pivotal cytotoxic effect against hepatocellular carcinoma (HCC). The current study aimed to investigate which flavonoid isolated from Physalis pubescens L. leaves has the most cytotoxic effect against Hep-G2 liver cancer cells and if it could ameliorate epirubicin efficacy and safety. Baicalein trimethyl ether (BTME), rutin, quercitrin and myricitrin were isolated from Physalis Pubescens L. leaves. Hep-G2 cells were treated with the isolated flavonoids as well as a combination of BTME and epirubicin. Cell viability and the chromosomal DNA fragmentation in Hep-G2 cells were assessed. BTME showed the best cytotoxic effect against Hep-G2 cells. Combination of epirubicin with (200 μg/mL) BTME significantly decreased the IC50 of epirubicin from 2.79 ± 0.626 μg/mL to 0.76 ± 0.258 μg/mL. Moreover, the same combination significantly increased the IC50 of BTME against WI-38 normal cells. DNA fragmentation as well as the concentration of beclin 1 and Bax were significantly increased in Hep-G2 cells treated with BTME and BTME+epirubicin compared to untreated cells. Besides, BTME and BTME+epirubicin significantly decreased the gene expression of TGFβ1 whereas increased ATG-7 gene expression. Conclusions: BTME (200μg/mL) significantly enhanced epirubicin's cytotoxicity against Hep-G2 cells and ameliorated its safety profile. BTME could exert anti-hepatocarcinoma effect by enhancing apoptosis and autophagy.
Collapse
Affiliation(s)
- Ghada Mohammad Al-Ashmawy
- Biochemistry Department, Faculty of Pharmacy, Tanta University, El-Gharbia, Tanta 31527, Egypt; Biochemistry Department, Faculty of Pharmacy, Al Salam University, El-Gharbia, Kafr Al Zaiyat 6615062, Egypt
| | | | - Dina Adam Ali
- Clinical Pathology Department, Faculty of Medicine, Tanta University, El-Gharbia, Tanta 31527, Egypt
| | - Mariam Ali Abo-Saif
- Biochemistry Department, Faculty of Pharmacy, Tanta University, El-Gharbia, Tanta 31527, Egypt.
| |
Collapse
|
3
|
Gondaliya P, Driscoll J, Yan IK, Ali Sayyed A, Patel T. Therapeutic restoration of miR-126-3p as a multi-targeted strategy to modulate the liver tumor microenvironment. Hepatol Commun 2024; 8:e0373. [PMID: 38358374 PMCID: PMC10871752 DOI: 10.1097/hc9.0000000000000373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/17/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Impaired natural killer (NK) cell-mediated antitumor responses contribute to the growth of liver tumors. Expression of a disintegrin and metalloprotease 9 (ADAM9) increases shedding of membrane-bound major histocompatibility complex class I chain-related protein A and results in evasion from NK cell-mediated cytolysis. ADAM9 is also involved in angiogenesis and tumor progression and is a target of miR-126-3p, a tumor suppressor that is downregulated and alters tumor cell behavior in the liver and other cancers. We evaluated the restoration of miR-126-3p and modulation of the miR-126-3p/ADAM9 axis as a therapeutic approach to simultaneously enhance NK cell-mediated cytolysis while targeting both tumor cells and their microenvironment. METHODS Precursor miRNAs were loaded into milk-derived nanovesicles to generate therapeutic vesicles (therapeutic milk-derived nanovesicles) for the restoration of functional miR-126-3p in recipient cancer cells. RESULTS Administration of therapeutic milk-derived nanovesicles increased miR-126-3p expression and reduced ADAM9 expression in target cells and was associated with an increase in membrane-bound major histocompatibility complex class I chain-related protein A. This enhanced NK cell cytolysis in adherent tumor cells and in multicellular tumor spheroids while also impairing angiogenesis and modulating macrophage chemotaxis. Moreover, IV administration of therapeutic milk-derived nanovesicles with adoptive transfer of NK cells reduced tumor burden in orthotopic hepatocellular cancer xenografts in mice. CONCLUSION A directed RNA therapeutic approach can mitigate NK cell immune evasion, reduce angiogenesis, and alter the tumor cell phenotype through the restoration of miR-126-3p in liver tumor cells. The pleiotropic effects elicited by this multi-targeted approach to modulate the local tumor microenvironment support its use for the treatment of liver cancer.
Collapse
Affiliation(s)
- Piyush Gondaliya
- Department of Transplantation, Mayo Clinic, Jacksonville, Florida, USA
| | - Julia Driscoll
- Department of Transplantation, Mayo Clinic, Jacksonville, Florida, USA
| | - Irene K. Yan
- Department of Transplantation, Mayo Clinic, Jacksonville, Florida, USA
| | - Adil Ali Sayyed
- Department of Transplantation, Mayo Clinic, Jacksonville, Florida, USA
| | - Tushar Patel
- Department of Transplantation, Mayo Clinic, Jacksonville, Florida, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
4
|
Hajizadeh M, Hajizadeh F, Ghaffarei S, Amin Doustvandi M, Hajizadeh K, Yaghoubi SM, Mohammadnejad F, Khiabani NA, Mousavi P, Baradaran B. MicroRNAs and their vital role in apoptosis in hepatocellular carcinoma: miRNA-based diagnostic and treatment methods. Gene 2023; 888:147803. [PMID: 37716587 DOI: 10.1016/j.gene.2023.147803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 08/03/2023] [Accepted: 09/13/2023] [Indexed: 09/18/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies with high invasive and metastatic capability. Although significant advances have been made in the treatment of HCC, the overall survival rate of patients is still low. It is essential to explore accurate biomarkers for early diagnosis and prognosis along with therapeutic procedures to increase the survival rate of these patients. Anticancer therapies can contribute to induce apoptosis for the elimination of cancerous cells. However, dysregulated apoptosis and proliferation signaling pathways lead to treatment resistance, a significant challenge in improving efficient therapies. MiRNAs, short non-coding RNAs, play crucial roles in the progression of HCC, which regulate gene expression through post-transcriptional inhibition and targeting mRNA degradation in cancers. Dysregulated expression of multiple miRNAs is associated with numerous biological processes, including cell proliferation, apoptosis, invasion and metastasis, epithelial-mesenchymal transition (EMT), angiogenesis, and drug resistance in HCC. This review summarizes the role and potential efficacy of miRNAs in promoting and inhibiting cell proliferation and apoptosis in HCC, as well as the role of miRNAs in therapy resistance in HCC.
Collapse
Affiliation(s)
- Masoumeh Hajizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farnaz Hajizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevil Ghaffarei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Khadijeh Hajizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyed Mohammad Yaghoubi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | | | - Pegah Mousavi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Kciuk M, Yahya EB, Mohamed MMI, Abdulsamad MA, Allaq AA, Gielecińska A, Kontek R. Insights into the Role of LncRNAs and miRNAs in Glioma Progression and Their Potential as Novel Therapeutic Targets. Cancers (Basel) 2023; 15:3298. [PMID: 37444408 DOI: 10.3390/cancers15133298] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Accumulating evidence supports that both long non-coding and micro RNAs (lncRNAs and miRNAs) are implicated in glioma tumorigenesis and progression. Poor outcome of gliomas has been linked to late-stage diagnosis and mostly ineffectiveness of conventional treatment due to low knowledge about the early stage of gliomas, which are not possible to observe with conventional diagnostic approaches. The past few years witnessed a revolutionary advance in biotechnology and neuroscience with the understanding of tumor-related molecules, including non-coding RNAs that are involved in the angiogenesis and progression of glioma cells and thus are used as prognostic biomarkers as well as novel therapeutic targets. The emerging research on lncRNAs and miRNAs highlights their crucial role in glioma progression, offering new insights into the disease. These non-coding RNAs hold significant potential as novel therapeutic targets, paving the way for innovative treatment approaches against glioma. This review encompasses a comprehensive discussion about the role of lncRNAs and miRNAs in gene regulation that is responsible for the promotion or the inhibition of glioma progression and collects the existing links between these key cancer-related molecules.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Esam Bashir Yahya
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia
| | | | - Muhanad A Abdulsamad
- Department of Molecular Biology, Faculty of Science, Sabratha University, Sabratha 00218, Libya
| | - Abdulmutalib A Allaq
- Faculty of Applied Science, Universiti Teknologi MARA, Shah Alam 40450, Malaysia
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
| |
Collapse
|
6
|
Kalita A, Sikora-Skrabaka M, Nowakowska-Zajdel E. Role of Some microRNA/ADAM Proteins Axes in Gastrointestinal Cancers as a Novel Biomarkers and Potential Therapeutic Targets—A Review. Curr Issues Mol Biol 2023; 45:2917-2936. [PMID: 37185715 PMCID: PMC10136553 DOI: 10.3390/cimb45040191] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Gastrointestinal (GI) cancers are some of the most common cancers in the world and their number is increasing. Their etiology and pathogenesis are still unclear. ADAM proteins are a family of transmembrane and secreted metalloproteinases that play a role in cancerogenesis, metastasis and neoangiogenesis. MicroRNAs are small single-stranded non-coding RNAs that take part in the post-transcriptional regulation of gene expression. Some ADAM proteins can be targets for microRNAs. In this review, we analyze the impact of microRNA/ADAM protein axes in GI cancers.
Collapse
Affiliation(s)
- Agnieszka Kalita
- Department of Nutrition-Related Disease Prevention, Department of Metabolic Disease Prevention, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
- Department of Clinical Oncology, No. 4 Provincial Specialist Hospital, 41-902 Bytom, Poland
| | - Magdalena Sikora-Skrabaka
- Department of Nutrition-Related Disease Prevention, Department of Metabolic Disease Prevention, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
- Department of Clinical Oncology, No. 4 Provincial Specialist Hospital, 41-902 Bytom, Poland
| | - Ewa Nowakowska-Zajdel
- Department of Nutrition-Related Disease Prevention, Department of Metabolic Disease Prevention, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
- Department of Clinical Oncology, No. 4 Provincial Specialist Hospital, 41-902 Bytom, Poland
| |
Collapse
|
7
|
Moirangthem A, Gondaliya P, Yan IK, Sayyed AA, Driscoll J, Patel T. Extracellular vesicle‑mediated miR‑126‑3p transfer contributes to inter‑cellular communication in the liver tumor microenvironment. Int J Oncol 2023; 62:31. [PMID: 36660950 PMCID: PMC9851126 DOI: 10.3892/ijo.2023.5479] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 10/24/2022] [Indexed: 01/18/2023] Open
Abstract
Extracellular vesicles (EVs) and their contents are gaining recognition as important mediators of intercellular communication through the transfer of bioactive molecules, such as non‑coding RNA. The present study comprehensively assessed the microRNA (miRNA/miR) content within EVs released from HepG2 liver cancer (LC) cells and LX2 hepatic stellate cells (HSCs) and determined the contribution of EV miRNA to intercellular communication. Using both transwell and spheroid co‑cultures of LC cells and HSCs, miR‑126‑3p within EV was established as a mediator of HSC to LC cell communication that influenced tumor cell migration and invasion, as well as the growth of multicellular LC/HSC spheroids. Manipulation of miR‑126‑3p either by enforced expression using pre‑miR‑126‑3p or by inhibition using antimiR‑126‑3p did not alter tumor cell viability, proliferation or sensitivity to either sorafenib or regorafenib. By contrast, enforced expression of miR‑126‑3p decreased tumor‑cell migration. Knockdown of miR‑126‑3p in tumor cells increased disintegrin and metalloproteinase domain‑containing protein 9 (ADAM9) expression and in HSCs increased collagen‑1A1 accumulation with an increase in compactness of multicellular spheroids. Within LC/HSC spheroids, ADAM9 and vascular endothelial growth factor expression was increased by silencing of miR‑126‑3p but diminished with the restoration of miR‑126‑3p. These studies implicate miR‑126‑3p in functional effects on migration, invasion and spheroid growth of tumor cells in the presence of HSCs, and thereby demonstrate functional EV‑RNA‑based intercellular signaling between HSCs and LC cells that is directly relevant to tumor‑cell behavior.
Collapse
Affiliation(s)
| | | | - Irene K. Yan
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Adil Ali Sayyed
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Julia Driscoll
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tushar Patel
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
8
|
Tu H, Wang W, Feng Y, Zhang L, Zhou H, Cheng C, Ji L, Cai Q, Feng Y. β-Patchoulene represses hypoxia-induced proliferation and epithelial-mesenchymal transition of liver cancer cells. Bioengineered 2022; 13:11907-11922. [PMID: 35546067 PMCID: PMC9275994 DOI: 10.1080/21655979.2022.2065945] [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] [Indexed: 12/04/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor originating from liver epithelial cells with a high clinical mortality rate. β-Patchoulene (β-PAE) is a compound extracted from patchouli, which has analgesic, anti-inflammatory and antioxidant effects. This research aims to probe the impacts of β-PAE on hypoxia-induced HCC cell proliferation and epithelial-mesenchymal transition (EMT). Firstly, hypoxic injury models were constructed in HCC Huh-7 and MHCC97 cells, and the hypoxic injury cell models were then treated with different concentrations of β-PAE. The cell viability, proliferation, migration, invasion and apoptosis were checked by the cell counting kit-8 (CCK-8) assay, colony formation assay, Transwell assay, flow cytometry and terminal deoxyribonucleotide transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay. The expression of Survivin protein, EMT markers and the NF-κB/HIF-1α pathway was gauged by Western blot (WB) or cellular immunofluorescence or reverse transcription-polymerase chain reaction (RT-PCR). The in-vivo experiment was conducted to confirm the anti-tumor role of β-PAE. As a result, β-PAE abated hypoxia-induced HCC cell growth, proliferation, migration, invasion and EMT and facilitated apoptosis in vitro and in vivo dose-dependently. Further mechanism studies displayed that β-PAE inactivated the NF-κB/HIF-1α pathway, and HIF-1α activation significantly reversed the β-PAE-mediated tumor inhibition. β-PAE repressed the proliferation and EMT of hypoxia-induced HCC cells by choking the NF-κB/HIF-1α pathway, suggesting that β-PAE was a potential drug for HCC treatment.
Collapse
Affiliation(s)
- Huahua Tu
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Wei Wang
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Yanqing Feng
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Linfei Zhang
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Huadong Zhou
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Caitao Cheng
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Lei Ji
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Qinghe Cai
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Yong Feng
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| |
Collapse
|
9
|
Li Z, Liu H, Zhang Y, Tan H. The effect of propofol on the proliferation and apoptosis of hepatocellular carcinoma cells through TGF-Β1/Smad2 signaling pathway. Bioengineered 2021; 12:4581-4592. [PMID: 34323647 PMCID: PMC8806409 DOI: 10.1080/21655979.2021.1955177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/08/2021] [Indexed: 11/06/2022] Open
Abstract
Malignant tumors are a serious threat to human health. Surgical resection is the most effective treatment for liver cancer. However, liver cancer is mostly found at an advanced stage, is difficult to remove by surgery, and has a very high recurrence rate after surgery. The current liver cancer treatment drugs have serious side effects, and the treatment effect is not ideal, far from meeting the clinical needs. Based on this, this paper studies the effect of propofol on the proliferation and apoptosis of liver cancer cells through the TGF-B1/Smad2 signaling pathway, and explores the proliferation, adhesion and apoptosis of cancer cells in patients with propofol. This paper uses a comparative experiment. With medical imaging method, 80 rats with liver cancer in the same period were cultured. High-precision microscope and radiolocation method were used to observe and record the whole process of propofol regulating Smad2 signal pathway. The results show that propofol can effectively inhibit the proliferation of cancer cells in patients with liver cancer. Propofol can increase the activity and content of transforming growth factor-β1 by 12% and 20%, respectively, and then inhibit the proliferation rate of liver cancer cells by 10% through the Smad2 signaling pathway, and exponentially increase the apoptotic number of liver cancer cells. This shows that propofol has a significant inhibitory effect on the cycle of liver cancer cells. Under the action of propofol, the life cycle of liver cancer cells is shortened, which provides a certain theoretical basis for the treatment of liver cancer.
Collapse
Affiliation(s)
- Zongchao Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Honglei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yunxiao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hongyu Tan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
10
|
Soheilifar MH, Masoudi-Khoram N, Madadi S, Nobari S, Maadi H, Keshmiri Neghab H, Amini R, Pishnamazi M. Angioregulatory microRNAs in breast cancer: Molecular mechanistic basis and implications for therapeutic strategies. J Adv Res 2021; 37:235-253. [PMID: 35499045 PMCID: PMC9039675 DOI: 10.1016/j.jare.2021.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/13/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer-associated angiogenesis is a fundamental process in tumor growth and metastasis. Angioregulatory miRNA–target gene interaction is not only involved in sprouting vessels of breast tumors but also, trans-differentiation of breast cancer cells to endothelial cells in a process termed vasculogenic mimicry. Successful targeting of tumor angiogenesis is still a missing link in the treatment of Breast cancer (BC) due to the low effectiveness of anti-angiogenic therapies in this cancer. Response to anti-angiogenic therapeutics are controlled by a miRNAs, so the identification of interaction networks of miRNAs–targets can be applicable in determining anti-angiogeneic therapy and new biomarkers in BC. Angioregulatory miRNAs in breast cancer cells and their microenvironment have therapeutic potential in cancer treatment.
Background Cancer-associated angiogenesis is a fundamental process in tumor growth and metastasis. A variety of signaling regulators and pathways contribute to establish neovascularization, among them as small endogenous non-coding RNAs, microRNAs (miRNAs) play prominent dual regulatory function in breast cancer (BC) angiogenesis. Aim of Review This review aims at describing the current state-of-the-art in BC angiogenesis-mediated by angioregulatory miRNAs, and an overview of miRNAs dysregulation association with the anti-angiogenic response in addition to potential clinical application of miRNAs-based therapeutics. Key Scientific Concepts of Review Angioregulatory miRNA–target gene interaction is not only involved in sprouting vessels of breast tumors but also, trans-differentiation of BC cells to endothelial cells (ECs) in a process termed vasculogenic mimicry. Using canonical and non-canonical angiogenesis pathways, the tumor cell employs the oncogenic characteristics such as miRNAs dysregulation to increase survival, proliferation, oxygen and nutrient supply, and treatment resistance. Angioregulatory miRNAs in BC cells and their microenvironment have therapeutic potential in cancer treatment. Although, miRNAs dysregulation can serve as tumor biomarker nevertheless, due to the association of miRNAs dysregulation with anti-angiogenic resistant phenotype, clinical benefits of anti-angiogenic therapy might be challenging in BC. Hence, unveiling the molecular mechanism underlying angioregulatory miRNAs sparked a booming interest in finding new treatment strategies such as miRNA-based therapies in BC.
Collapse
Affiliation(s)
- Mohammad Hasan Soheilifar
- Department of Medical Laser, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Corresponding authorsat: Yara Institute, Academic Center for Education, Culture and Research (ACECR), Enghelab St, Tehran 1315795613, Iran (Mohammad Hasan Soheilifar). University of Limerick, Limerick V94 T9PX, Ireland (Mahboubeh Pishnamazi).
| | - Nastaran Masoudi-Khoram
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Soheil Madadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sima Nobari
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Maadi
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Hoda Keshmiri Neghab
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Razieh Amini
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahboubeh Pishnamazi
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Limerick, Ireland
- Corresponding authorsat: Yara Institute, Academic Center for Education, Culture and Research (ACECR), Enghelab St, Tehran 1315795613, Iran (Mohammad Hasan Soheilifar). University of Limerick, Limerick V94 T9PX, Ireland (Mahboubeh Pishnamazi).
| |
Collapse
|
11
|
Razavi ZS, Asgarpour K, Mahjoubin-Tehran M, Rasouli S, Khan H, Shahrzad MK, Hamblin MR, Mirzaei H. Angiogenesis-related non-coding RNAs and gastrointestinal cancer. MOLECULAR THERAPY-ONCOLYTICS 2021; 21:220-241. [PMID: 34095461 PMCID: PMC8141508 DOI: 10.1016/j.omto.2021.04.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gastrointestinal (GI) cancers are among the main reasons for cancer death globally. The deadliest types of GI cancer include colon, stomach, and liver cancers. Multiple lines of evidence have shown that angiogenesis has a key role in the growth and metastasis of all GI tumors. Abnormal angiogenesis also has a critical role in many non-malignant diseases. Therefore, angiogenesis is considered to be an important target for improved cancer treatment. Despite much research, the mechanisms governing angiogenesis are not completely understood. Recently, it has been shown that angiogenesis-related non-coding RNAs (ncRNAs) could affect the development of angiogenesis in cancer cells and tumors. The broad family of ncRNAs, which include long non-coding RNAs, microRNAs, and circular RNAs, are related to the development, promotion, and metastasis of GI cancers, especially in angiogenesis. This review discusses the role of ncRNAs in mediating angiogenesis in various types of GI cancers and looks forward to the introduction of mimetics and antagonists as possible therapeutic agents.
Collapse
Affiliation(s)
| | - Kasra Asgarpour
- Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Maryam Mahjoubin-Tehran
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Susan Rasouli
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Mohammad Karim Shahrzad
- Department of Internal Medicine and Endocrinology, Shohadae Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
12
|
Guo YP, Wang ZF, Li N, Lei QQ, Cheng Q, Shi LG, Zhou SL, Wang XH, Sun Y, Kong LF. Suppression of lncRNA HOTAIR alleviates RCC angiogenesis through regulating miR-126/EGFL7 axis. Am J Physiol Cell Physiol 2021; 320:C880-C891. [PMID: 33502949 DOI: 10.1152/ajpcell.00459.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 11/22/2022]
Abstract
Renal cell carcinoma (RCC) has the highest mortality rate among urological cancers and tumor angiogenesis that plays a critical role in RCC progress. Epidermal growth factor-like domain multiple 7 (EGFL7) has been recently identified as a regulator in RCC tumor angiogenesis and progression. Long noncoding RNA (LncRNA) HOTAIR has been considered as a pro-oncogene in multiple cancers, but its precise mechanism of tumor angiogenesis has rarely been reported. MicroRNA-126 (miR-126) functions as a tumor suppressor in RCC. However, the underlying tumor angiogenesis mechanism of HOTAIR/miR-126 axis in RCC has not been studied. The proliferation, migration, angiogenesis, and expression of EGFL7 and related proteins in extracellular signal-regulated kinase (ERK)/activators of transcription 3 (STAT3) signal pathway were determined to examine the effect and mechanism of HOTAIR and miR-126 on RCC progress. The regulatory relationship of HOTAIR and miR-126, as well as miR-126 and EGFL7 were tested using dual-luciferase reporter assay. Aenograft RCC mice model was used to examine the effect of HOTAIR on RCC tumor growth and metastasis in vivo. HOTAIR knockdown and miR-126 overexpression suppressed the proliferation, migration, and angiogenesis of RCC cells. HOTAIR regulated EGFL7 expression by competitively binding to miR-126. Knockdown of HOTAIR significantly suppressed the RCC tumor progression and lung metastasis in vivo. These findings suggest that lncRNA HOTAIR regulate RCC angiogenesis through miR-126/EGFL7 axis and provide a new perspective on the molecular pathways of angiogenesis in RCC development, which might be potential therapeutic targets for RCC treatment.
Collapse
Affiliation(s)
- Yan-Ping Guo
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, People's Republic of China
| | - Zhi-Feng Wang
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Na Li
- Kidney Disease & Immunology Key Laboratory, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Qian-Qian Lei
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, People's Republic of China
| | - Qiong Cheng
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, People's Republic of China
| | - Li-Gang Shi
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, People's Republic of China
| | - Sheng-Li Zhou
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, People's Republic of China
| | - Xin-Hua Wang
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yang Sun
- Department of Pathology, Fuwai Hospital and Cardiovascular Institute, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Ling-Fei Kong
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, People's Republic of China
| |
Collapse
|
13
|
Zhou B, Wang J. Epidermal growth factor-like domain 7 regulates breast cancer cell proliferation and vascular endothelial growth factor expression via the p38MAPK signaling pathway. Am J Transl Res 2021; 13:2608-2616. [PMID: 34017420 PMCID: PMC8129295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/24/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE We aimed to investigate the effects of epidermal growth factor-like domain 7 (EGFL7) on breast cancer cell proliferation and angiogenesis and its association with the p38 mitogen-activated protein kinase (p38MAPK) signaling pathway. METHODS The vectors for stable overexpression of EGFL7 and the vectors for EGFL7 knockout were constructed. The breast cancer cell line MDA-MB-231 was selected for this study and the cells were divided into four groups: the control group, the empty vector group (transfected with an empty vector), the EGFL7 overexpression group (transfected with the EGFL7 overexpression vector), and the EGFL7 knockout group (transfected with the EGFL7 knockout vector). After 72 h of transfection, the mRNA and protein levels of EGFL7 in the cells were detected by RT-PCR and Western blot, respectively. The cell proliferation rates at 12 h, 24 h, 48 h and 72 h of culture in each group were detected using the MTT method. An in vitro tumor angiogenesis model of tumor-endothelial cells co-culture system was established and the angiogenesis ability at 12 h, 24 h, 48 h and 72 h of culture were compared among the groups using an in vitro angiogenesis assay. The cells in the EGFL7 overexpression group were further divided into three groups and were treated with p38MAPK inhibitor SB203580 at a dose of 0 μmol/L, 5 μmol/L, and 10 μmol/L, respectively. Afterward, the cells were co-cultured with endothelial cells for 48 h. Western blot was performed to detect the protein levels of vascular endothelial growth factor (VEGF), p38MAPK, and p-p38MAPK. RESULTS Compared with the control group, the EGFL7 mRNA level was higher in the EGFL7 overexpression group and lower in the EGFL7 knockout group (both P<0.05). Compared with the control group at 12 h, 24 h, 48 h, and 72 h of culture, the cell proliferation rates were lower in the EGFL knockout group and higher in the EGFL overexpression group, respectively (all P<0.05). Moreover, compared with the control group at these time points, the number of vascular sprouts and the protein levels of VEGF, p38MAPK, and p-p38MAPK were lower in the EGFL7 knockout group and higher in the EGFL7 overexpression group, respectively (all P<0.05). After the cells overexpressing EGFL7 were treated with SB203580, the level of p-p38MAPK was deceased, and the protein expression level of VEGF was inversely related with the SB203580 concentration (F=44.24, P<0.01). CONCLUSION EGFL7 can promote the proliferation of breast cancer cells and angiogenesis, and the mechanism may be associated with the activation of p38MAPK signaling pathway and promotion of VEGF expression.
Collapse
|
14
|
Szafranski P, Stankiewicz P. Long Non-Coding RNA FENDRR: Gene Structure, Expression, and Biological Relevance. Genes (Basel) 2021; 12:177. [PMID: 33513839 PMCID: PMC7911649 DOI: 10.3390/genes12020177] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
The FOXF1 Adjacent Noncoding Developmental Regulatory RNA (Fendrr) plays an important role in the control of gene expression in mammals. It is transcribed in the opposite direction to the neighboring Foxf1 gene with which it shares a region containing promoters. In humans, FENDRR is located on chromosome 16q24.1, and is positively regulated both by the FOXF1 distant lung-specific cis-acting enhancer and by trans-acting FOXF1. Fendrr has been shown to function as a competing endogenous RNA, sponging microRNAs and protein factors that control stability of mRNAs, and as an epigenetic modifier of chromatin structure around gene promoters and other regulatory sites, targeting them with histone methyltrasferase complexes. In mice, Fendrr is essential for development of the heart, lungs, and gastrointestinal system; its homozygous loss causes embryonic or perinatal lethality. Importantly, deregulation of FENDRR expression has been causatively linked also to tumorigenesis, resistance to chemotherapy, fibrosis, and inflammatory diseases. Here, we review the current knowledge on the FENDRR structure, expression, and involvement in development and tissue maintenance.
Collapse
Affiliation(s)
- Przemyslaw Szafranski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA;
| | | |
Collapse
|
15
|
Liu J, Wei E, Wei J, Zhou W, Webster KA, Zhang B, Li D, Zhang G, Wei Y, Long Y, Qi X, Zhang Q, Xu D. MiR-126-HMGB1-HIF-1 Axis Regulates Endothelial Cell Inflammation during Exposure to Hypoxia-Acidosis. DISEASE MARKERS 2021; 2021:4933194. [PMID: 34970357 PMCID: PMC8714334 DOI: 10.1155/2021/4933194] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/20/2021] [Indexed: 02/05/2023]
Abstract
Crosstalk between molecular regulators miR-126, hypoxia-inducible factor 1-alpha (HIF-1-α), and high-mobility group box-1 (HMGB1) contributes to the regulation of inflammation and angiogenesis in multiple physiological and pathophysiological settings. Here, we present evidence of an overriding role for miR-126 in the regulation of HMGB1 and its downstream proinflammatory effectors in endothelial cells subjected to hypoxia with concurrent acidosis (H/A). Methods. Primary mouse endothelial cells (PMEC) were exposed to hypoxia or H/A to simulate short or chronic low-flow ischemia, respectively. RT-qPCR quantified mRNA transcripts, and proteins were measured by western blot. ROS were quantified by fluorogenic ELISA and luciferase reporter assays employed to confirm an active miR-126 target in the HMGB1 3'UTR. Results. Enhanced expression of miR-126 in PMECs cultured under neutral hypoxia was suppressed under H/A, whereas the HMGB1 expression increased sequentially under both conditions. Enhanced expression of HMGB1 and downstream inflammation markers was blocked by the premiR-126 overexpression and optimized by antagomiR. Compared with neutral hypoxia, H/A suppressed the HIF-1α expression independently of miR-126. The results show that HMGB1 and downstream effectors are optimally induced by H/A relative to neutral hypoxia via crosstalk between hypoxia signaling, miR-126, and HIF-1α, whereas B-cell lymphoma 2(Bcl2), a HIF-1α, and miR-126 regulated gene expressed optimally under neutral hypoxia. Conclusion. Inflammatory responses of ECs to H/A are dynamically regulated by the combined actions of hypoxia, miR-126, and HIF-1α on the master regulator HMGB1. The findings may be relevant to vascular diseases including atherosclerotic occlusion and interiors of plaque where coexisting hypoxia and acidosis promote inflammation as a defining etiology.
Collapse
Affiliation(s)
- Jinxue Liu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Eileen Wei
- Gulliver High School, Miami, FL 33156, USA
| | - Jianqin Wei
- Department of Medicine Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Wei Zhou
- Department of Ophthalmology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Keith A. Webster
- Integene International, LLC, Miami, FL 33137, USA
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
- Everglades Biopharma, LLC, Houston, TX 77030, USA
| | - Bin Zhang
- Department of Cardiology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Dong Li
- Department of Intensive Care Unit and Clinical Experimental Center, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Gaoxing Zhang
- Department of Cardiology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Yidong Wei
- Department of Surgery, Youjiang Medical University for Nationalities, Chengxiang Rd, Baise, Guangxi 533000, China
| | - Yusheng Long
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
- Department of Cardiology, Guangdong Cardiovascular Institute and Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiuyu Qi
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
- Department of Cardiology, Guangdong Cardiovascular Institute and Shantou University Medical College, Shantou 515041, China
| | - Qianhuan Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
| | - Dingli Xu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
16
|
Annese T, Tamma R, De Giorgis M, Ribatti D. microRNAs Biogenesis, Functions and Role in Tumor Angiogenesis. Front Oncol 2020; 10:581007. [PMID: 33330058 PMCID: PMC7729128 DOI: 10.3389/fonc.2020.581007] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
microRNAs (miRNAs) are small non-coding RNA molecules, evolutionary conserved. They target more than one mRNAs, thus influencing multiple molecular pathways, but also mRNAs may bind to a variety of miRNAs, either simultaneously or in a context-dependent manner. miRNAs biogenesis, including miRNA transcription, processing by Drosha and Dicer, transportation, RISC biding, and miRNA decay, are finely controlled in space and time. miRNAs are critical regulators in various biological processes, such as differentiation, proliferation, apoptosis, and development in both health and disease. Their dysregulation is involved in tumor initiation and progression. In tumors, they can act as onco-miRNAs or oncosuppressor-miRNA participating in distinct cellular pathways, and the same miRNA can perform both activities depending on the context. In tumor progression, the angiogenic switch is fundamental. miRNAs derived from tumor cells, endothelial cells, and cells of the surrounding microenvironment regulate tumor angiogenesis, acting as pro-angiomiR or anti-angiomiR. In this review, we described miRNA biogenesis and function, and we update the non-classical aspects of them. The most recent role in the nucleus, as transcriptional gene regulators and the different mechanisms by which they could be dysregulated, in tumor initiation and progression, are treated. In particular, we describe the role of miRNAs in sprouting angiogenesis, vessel co-option, and vasculogenic mimicry. The role of miRNAs in lymphoma angiogenesis is also discussed despite the scarcity of data. The information presented in this review reveals the need to do much more to discover the complete miRNA network regulating angiogenesis, not only using high-throughput computational analysis approaches but also morphological ones.
Collapse
Affiliation(s)
- Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Michelina De Giorgis
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| |
Collapse
|
17
|
Wang J, Zhang Y, Liu L, Cui Z, Shi R, Hou J, Liu Z, Yang L, Wang L, Li Y. NFAT2 overexpression suppresses the malignancy of hepatocellular carcinoma through inducing Egr2 expression. BMC Cancer 2020; 20:966. [PMID: 33023539 PMCID: PMC7542386 DOI: 10.1186/s12885-020-07474-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Nuclear factor of activated T cells 2 (NFAT2) has been reported to regulate the development and malignancy of few tumors. In this study, we aimed to explore the effect of NFAT2 expression on cell fate of HepG2 cell and its potential mechanisms. METHODS Firstly, the pcDNA3.1-NFAT2 plasmid was transfected into HepG2 cells to construct NFAT2 overexpressed HepG2 cells. Then, the chemical count kit-8 cell viability assay, Annexin V-FITC apoptosis detection, EdU labeling proliferation detection, transwell and wound healing experiments were performed. The expression of Egr2 and FasL, and the phosphorylation of AKT and ERK, after ionomycin and PMA co-stimulation, was detected, while the Ca2+ mobilization stimulated by K+ solution was determined. At last, the mRNA and protein expression of NFAT2, Egr2, FasL, COX-2 and c-myc in carcinoma and adjacent tissues was investigated. RESULTS The NFAT2 overexpression suppressed the cell viability, invasion and migration capabilities, and promoted apoptosis of HepG2 cells. NFAT2 overexpression induced the expression of Egr2 and FasL and suppressed the phosphorylation of AKT and ERK. The sensitivity and Ca2+ mobilization of HepG2 cells was also inhibited by NFAT2 overexpression. Compared with adjacent tissues, the carcinoma tissues expressed less NFAT2, Egr2, FasL and more COX-2 and c-myc. CONCLUSION The current study firstly suggested that NFAT2 suppressed the aggression and malignancy of HepG2 cells through inducing the expression of Egr2. The absence of NFAT2 and Egr2 in carcinoma tissues reminded us that NFAT2 may be a promising therapeutic target for hepatocellular carcinoma treatment.
Collapse
Affiliation(s)
- Jian Wang
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Yamin Zhang
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China.
| | - Lei Liu
- Department of Transplantation Center, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin, 300192, PR China
| | - Zilin Cui
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Rui Shi
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Jiancun Hou
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Zirong Liu
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Long Yang
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Lianjiang Wang
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Yang Li
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| |
Collapse
|
18
|
Role of ADAM9 and miR-126 in the development of abdominal aortic aneurysm. Atherosclerosis 2020; 297:47-54. [PMID: 32078829 DOI: 10.1016/j.atherosclerosis.2020.01.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/21/2019] [Accepted: 01/16/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease when aortic rupture occurs, especially for elders. There is an urgent need to understand the mechanisms of AAA formation and development at molecular level. Our previous study showed that disintegrin and metalloprotease 10 (ADAM10) played an important role in abdominal aortic aneurysm formation. In this study, we investigated the effects of another ADAM protein (ADMA9) in AAA formation. METHOD AND RESULTS Using AngII treated human aortic smooth muscle cells (HASMCs) and human aortic endothelial cells (hAoECs) as in vitro AAA model and murine AAA model, ADAM9 was overexpressed suggesting that ADAM9 may play important roles in AAA formation. Further investigation showed that ADAM9 induced inflammation leading to increased macrophage infiltration. ADAM9 was also found to induce cell apoptosis. AKT/NF-κB pathway was activated in murine AAA. Bioinformatic analysis showed that the 3' UTR of ADMA9 was a potential target of miR-126. We investigated the potential of using miR-126 to modulate ADAM9 expression. The expression level of miR-126 was decreased and inversely correlated with the expression of ADAM9 in the in vitro AAA model. Further investigation showed that miR-126 negatively regulated gene expression of ADAM9 and suppressed the production of inflammatory cytokines. miR-126 was also found to improve cell survival and significantly reduce AAA formation in murine AAA. CONCLUSIONS Our data revealed a link between ADAM9 and AAA formation, providing an approach to control AAA development using miR-126, possibly through modulation of the expression level of ADAM9.
Collapse
|
19
|
Zeng X, Liu Q, Yang Y, Jia W, Li S, He D, Ma R. Placenta-specific protein 8 promotes the proliferation of lung adenocarcinoma PC-9 cells and their tolerance to an epidermal growth factor receptor tyrosine kinase inhibitor by activating the ERK signaling pathway. Oncol Lett 2019; 18:5621-5627. [PMID: 31620204 DOI: 10.3892/ol.2019.10911] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 08/06/2019] [Indexed: 11/06/2022] Open
Abstract
Placenta-specific protein 8 (PLAC8) is a conserved protein with a molecular weight of 12.5 kDa. The specific function of this protein has not been fully elucidated, however, PLAC8 has been found to play an important tumor regulatory role in certain types of cancer, including colon, pancreatic and liver cancer. PLAC8 also participates in the regulation of the cell cycle, autophagy, epithelial-mesenchymal transition and other cellular functions, indicating its potential as a molecular target worth further investigation. The present study investigated the effect of PLAC8 on the proliferation of lung adenocarcinoma PC-9 cells and their sensitivity to gefitinib, an epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI). It was found that the inhibition of PLAC8 expression in PC-9 cells resulted in significantly decreased proliferation, whereas overexpression of PLAC8 significantly increased the proliferation (P<0.05) of PC-9 cells. Furthermore, inhibition of PLAC8 expression resulted in decreased activity of the ERK signaling pathway, while PLAC8 overexpression increased activity of this pathway. Inhibition of the ERK signaling pathway with U0126 reversed the effects induced by inhibiting or overexpressing PLAC8 on cell proliferation. In addition, overexpression of PLAC8 significantly decreased the sensitivity of PC-9 cells to gefitinib, and this effect was reversed by U0126. Overall, these results suggest that PLAC8 is involved in the regulation of proliferation of lung adenocarcinoma PC-9 cells and impacts their sensitivity to an EGFR-TKI. Thus, PLAC8 is a potential novel target in lung adenocarcinoma for future studies.
Collapse
Affiliation(s)
- Xiaofei Zeng
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Qing Liu
- Department of Cardiothoracic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Yanhui Yang
- Department of Cardiothoracic Surgery, The First People's Hospital of Neijiang, Sichuan 641000, P.R. China
| | - Weikun Jia
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Shuping Li
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Dongsheng He
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Ruidong Ma
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
20
|
The Prognostic Significance of Protein Expression of CASZ1 in Clear Cell Renal Cell Carcinoma. DISEASE MARKERS 2019; 2019:1342161. [PMID: 31481981 PMCID: PMC6701416 DOI: 10.1155/2019/1342161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/11/2019] [Accepted: 07/17/2019] [Indexed: 12/24/2022]
Abstract
Backgrounds Clear cell renal cell carcinoma (ccRCC) is the most common histologic subtype of renal cell carcinoma (RCC) and shows a relatively poor prognosis among RCCs. Castor zinc finger 1 (CASZ1) is a transcription factor, prominently known for its tumor suppression role in neuroblastoma and other cancers. However, there has been no research about the prognostic significance of CASZ1 in ccRCC. In this study, we investigated CASZ1 expression in ccRCC and analyzed its prognostic implications. Methods A total of 896 ccRCC patients, who underwent surgical resection from 1995 to 2008, were included. We prepared tissue microarray blocks, evaluated CASZ1 nuclear expression by immunohistochemistry, and classified the cases into low or high expression categories. Results A low expression of CASZ1 was observed in 320 cases (35.7%) and was significantly associated with large tumor size, high World Health Organization/International Society of Urological Pathology (WHO/ISUP) grade, and high T category and M category. In survival analysis, a low expression of CASZ1 was significantly correlated with unfavorable progression-free survival (PFS) (p < 0.001), overall survival (OS) (p < 0.001), and cancer-specific survival (CSS) (p < 0.001) and was an independent prognostic factor for PFS and CSS in multivariate analysis adjusted for tumor size, WHO/ISUP grade, T category, N category, and M category. Conclusions Our study is the first to show the prognostic significance of CASZ1 expression in ccRCC. Our results revealed that low expression of CASZ1 is associated with poor prognosis and may serve as a new prognostic indicator.
Collapse
|
21
|
Toxopeus E, Lynam-Lennon N, Biermann K, Dickens G, de Ruiter PE, van Lanschot J, Reynolds JV, Wijnhoven B, O'Sullivan J, van der Laan L. Tumor microRNA-126 controls cell viability and associates with poor survival in patients with esophageal adenocarcinoma. Exp Biol Med (Maywood) 2019; 244:1210-1219. [PMID: 31390899 DOI: 10.1177/1535370219868671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Esophageal adenocarcinoma displays a poor prognosis and current treatments are often not curative. Pathological TNM-stage is a prognostic parameter, but a better understanding of the pathophysiology of esophageal adenocarcinoma is needed to better predict survival. Recent work in other malignancies indicated an important role for the regulator microRNA-126 (miR-126) in tumors. The aim of this study was to investigate the function of miR-126 in esophageal adenocarcinoma and to correlate expression of miR-126 with tumor cell behavior and patient survival. Functional assays were performed in esophageal adenocarcinoma cell lines (OE33) in vitro by overexpressing or antagonizing miR-126 and assessing cellular processes linked to the hallmarks of cancer. In vivo pre-treatment biopsies of 58 patients with esophageal adenocarcinoma who underwent neoadjuvant chemoradiotherapy and surgery were analyzed for miR-126 expression in tumor cells by qRT-PCR and patient survival was analyzed by Kaplan–Meier and Cox regression. In OE33 cancer cells, stable overexpression of miR-126 modest though significantly altered expression of genes related to cell death (MEK1) and DNA repair (POLB and TERF1) was observed. Also the secretion of the angiogenic and pro-inflammatory factors, VEGF, IL-1β, and IL-6 were regulated by miR-126 ( P < 0.029). Importantly, miR-126 was found to be a regulator of cell viability in OE33 cells. Overexpressing ( P = 0.043) and antagonizing ( P = 0.035) miR-126 showed reciprocal effects on tumor cell viability and significantly regulated expression of pro- and anti-apoptotic genes, TP53, and GATA6 ( P < 0.031). In patients, high levels of miR-126 expression in pre-treatment tumors were significantly associated with poor survival ( P = 0.031). In multivariable analysis, high miR-126 ( P = 0.038) together with ypN-stage ( P = 0.048) were shown to be independent risk factors for poor survival. In conclusion, high expression of miR-126 in esophageal adenocarcinoma prevents tumor-cell death and is associated with poor patient survival. This study warrants further analysis of miR-126 as biomarker or potential therapeutic target for OAC. Impact statement Esophageal adenocarcinoma is a common form of cancer of the esophagus. It has an increasing health impact as it is associated with very poor patient survival. A better understanding of the pathophysiology of this cancer is needed to identify better treatment strategies and to provide a better prognosis for these patients. MicroRNAs have emerged as important molecular regulators of cancer cell viability and proliferation. The aim of our study was to investigate the role of one very well established microRNA, miR-126, in esophageal adenocarcinoma. Our research shows clear experimental evidence that miR-126 controls cell viability of esophageal adenocarcinoma cells. High (over)expression of miR-126 increased the viability of these cells. Our preclinical data were shown to be clinically relevant for this field of oncology. In an independent validation study of esophageal adenocarcinoma biopsies, we confirmed that high miR-126 expression in tumor cells was an independent risk factor for poor patient survival.
Collapse
Affiliation(s)
- Ela Toxopeus
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, GD Rotterdam 3015, the Netherlands
| | - N Lynam-Lennon
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin St. James's Hospital, Dublin 8, Dublin, Ireland
| | - K Biermann
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, GD Rotterdam 3015, the Netherlands
| | - G Dickens
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, GD Rotterdam 3015, the Netherlands
| | - P E de Ruiter
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, GD Rotterdam 3015, the Netherlands
| | - Jjb van Lanschot
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, GD Rotterdam 3015, the Netherlands
| | - J V Reynolds
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin St. James's Hospital, Dublin 8, Dublin, Ireland
| | - Bpl Wijnhoven
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, GD Rotterdam 3015, the Netherlands
| | - J O'Sullivan
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin St. James's Hospital, Dublin 8, Dublin, Ireland
| | - Ljw van der Laan
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, GD Rotterdam 3015, the Netherlands
| |
Collapse
|
22
|
Faranda T, Grossi I, Manganelli M, Marchina E, Baiocchi G, Portolani N, Crosatti M, De Petro G, Salvi A. Differential expression profiling of long non-coding RNA GAS5 and miR-126-3p in human cancer cells in response to sorafenib. Sci Rep 2019; 9:9118. [PMID: 31235746 PMCID: PMC6591391 DOI: 10.1038/s41598-019-45604-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) and microRNAs are involved in numerous physio-pathological conditions included cancer. To better understand the molecular mechanism of the oral antitumor multikinase inhibitor sorafenib, we profiled the expression of a panel of lncRNAs and miRNAs by qPCR array in a sorafenib-treated hepatocellular carcinoma (HCC) cell line. Among the most affected ncRNAs, we found that sorafenib mediated the dysregulation of the lncRNAs GAS5, HOTTIP and HOXA-AS2 and the miR-126-3p, in a panel of human cancer cell lines (HCC, renal and breast carcinomas). By luciferase gene reporter assay, we discovered that GAS5 may act as a sponge for miR-126-3p in HCC cells. The expression level of GAS5 and miR-126-3p was verified in human liquid and/or solid biopsies from HCC patients. miR-126-3p expression in HCC tissues was decreased respect to their correspondent peritumoral tissues. The levels of plasmatic circulating miR-126-3p and GAS5 were significantly higher and lower in HCC patients compared to healthy subjects, respectively. This study highlighted the capability of sorafenib to modulate the expression of a wide range of ncRNAs and specifically, GAS5 and miR-126-3p were involved in the response to sorafenib of different cancer cell types.
Collapse
Affiliation(s)
- Teresa Faranda
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Ilaria Grossi
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Michele Manganelli
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Eleonora Marchina
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Gianluca Baiocchi
- Department of Clinical and Experimental Sciences, Surgical Clinic, University of Brescia, Brescia, Italy
| | - Nazario Portolani
- Department of Clinical and Experimental Sciences, Surgical Clinic, University of Brescia, Brescia, Italy
| | | | - Giuseppina De Petro
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy
| | - Alessandro Salvi
- Department of Molecular and Translational Medicine, Division of Biology and Genetics, University of Brescia, Brescia, Italy.
| |
Collapse
|
23
|
Tu J, Cheung HH, Lu G, Chan CLK, Chen Z, Chan WY. microRNA-126 Is a Tumor Suppressor of Granulosa Cell Tumor Mediated by Its Host Gene EGFL7. Front Oncol 2019; 9:486. [PMID: 31245291 PMCID: PMC6579899 DOI: 10.3389/fonc.2019.00486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 05/23/2019] [Indexed: 02/03/2023] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at a post-transcriptional level. We examined the role of miR-126 in granulosa cell tumor (GCT) of the ovaries. In tissues from malignant GCT patients miR-126 expression was repressed. We showed that miR-126 could inhibit proliferation, migration, hormone production and promote apoptosis of cancerous granulosa cells (GCs) in vitro. The role of miR-126 as “tumor suppressor” was confirmed by using a tumor formation model in vivo. By RNA-seq, immunohistochemical staining (IHC), Western blot and luciferase reporter assay, we identified and confirmed EGFL7 as a direct functional target of miR-126 in cancer GCs. Furthermore, we found that the AKT signaling pathway was associated with miR-126 and EGFL7 in cancer GCs. Taken together, our results demonstrate a function of miR-126 in the suppression of GCT development via the regulation of EGFL7.
Collapse
Affiliation(s)
- Jiajie Tu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Hoi-Hung Cheung
- CUHK-SDU Joint Laboratory on Reproductive Genetics, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Zijiang Chen
- CUHK-SDU Joint Laboratory on Reproductive Genetics, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,Center for Reproductive Medicine, Shandong University, Jinan, China
| | - Wai-Yee Chan
- CUHK-SDU Joint Laboratory on Reproductive Genetics, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| |
Collapse
|
24
|
Li Q, Cheng K, Wang AY, Xu QG, Fu ZF, He SY, Xu PX. microRNA-126 inhibits tube formation of HUVECs by interacting with EGFL7 and down-regulating PI3K/AKT signaling pathway. Biomed Pharmacother 2019; 116:109007. [PMID: 31170663 DOI: 10.1016/j.biopha.2019.109007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 12/28/2022] Open
Abstract
It's critical for tube formation and angiogenesis to repair ischemic myocardium or stroke. This study aimed to investigate role of microRNA-126 (miR-126) in tube formation in human umbilical vein endothelial cells (HUVECs) and associated mechanisms. Primary neural stem cells (NSCs) and HUVECs were cultured and transfected with microRNA-126 mimics and miR-126 inhibitor. Cell counting kit-8 (CCK-8) and cell cycle assay were conducted for evaluating NSCs viability. Transwell assay was conducted to observe invasive ability of HUVECs. Quantitative real-time PCR (qRT-PCR) assay was used to examine epidermal growth factor like domain 7 (EGFL7) and miR-126 mRNA both in vitro and animal models. Tube forming capability was evaluated in HUVECs. Dual luciferase assay was performed to evaluate interaction between miR-126 and EGFL7 gene. Western blot assay was used to determine phosphoinositide-3-kinase/protein kinase-B (PI3K/AKT) signaling molecules and EGFL7. The results indicated that miR-126 significantly decreased cell viability, inhibited invasive ability and modulated cell cycle of NSCs compared to miR-NC group (p < 0.05). miR-126 significantly inhibited tube formation of HUVECs compared to miR-NC group (p < 0.05). miR-126 significantly down-regulated EGFL7 mRNA and protein expression compared to miR-NC (p < 0.05). Atorvastatin significantly increased CD34 and enhanced EGFL7 expression in traumatic brain injury (TBI) rats brain tissues compared to Model group (p < 0.05). miR-126 significantly down-regulated and atorvastatin up-regulated PI3K/AKT signaling pathway (p < 0.05). Atorvastatin significantly increased EGFL7 and down-regulated miR-126 expression in TBI rats brain tissues compared to Model group (p < 0.05). miR-126 interacted with and negatively correlated with EGFL7 gene both in vitro and in TBI models. In conclusion, microRNA-126 inhibited tube formation of HUVECs by interacting with EGFL7 and down-regulating PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Qiang Li
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China.
| | - Kai Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ai-Yue Wang
- Department of Neurology, Haikou Municipal People's Hospital, Haikou, China
| | - Qiong-Guang Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhou-Feng Fu
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shao-Yu He
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Peng-Xiang Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
25
|
Abstract
Background: Breast cancer is most serious reasons of women death around worldwide result in increasing its morbidity and mortality. MicroRNAs are considered as significant regulators of cancer biological processes. The main aim of this study is restoration of miR-126 could lead to modulate breast cell line and impairs their proliferation by targeting vascular endothelial growth factor gene (VEGF-A). Methods: Breast cancer cell line (MCF7) was transfected by miR-126 lipofectamine and negative miR control for 24 hr. Cytotoxic effects of miR-126 lipofectamine were determined by cell viability assay. Cell proliferation and cell cycle were quantitatively measured using PicoGreen assay and DAPI stain-flow cytometer analysis. For further investigation, Taq-Man real time PCR assay was performed to detect relative VEGF-A and miRNA-126 level. Results: MiR-126 was overexpressed in treated breast cancer cell (MCF7) compared with control cells. miR-126 expression has been associated –with a decrease in cell proliferation and arrested MCF7 cells at G1 phase. The study found that vascular endothelial growth factor is regulated by miR-126. Hence, VEGF-A is considered as functional vital and direct target to miR-126 in breast cancer cell line (MCF7). Conclusions: This study provided that manipulated miR-126 level may suggest a novel therapeutic approach in breast cancer treatment. However, an animal models study is needed to address and prove predictive ability of miR-126 on breast cancer controlling.
Collapse
Affiliation(s)
- Layla Alhasan
- Department of Biology, College Education for Pure Sciences, Thi-Qar University, Nasiriya, Iraq. :
| |
Collapse
|
26
|
Li Y, Zhang T, Qin S, Wang R, Li Y, Zhou Z, Chen Y, Wu Q, Su F. Effects of UPF1 expression on EMT process by targeting E‑cadherin, N‑cadherin, Vimentin and Twist in a hepatocellular carcinoma cell line. Mol Med Rep 2019; 19:2137-2143. [PMID: 30628676 PMCID: PMC6390072 DOI: 10.3892/mmr.2019.9838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 07/30/2018] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. It has been reported that HCC has a poor prognosis. In the majority of cases, once metastatic, HCC is incurable. To identify an effective treatment for HCC, it is important to understand the underlying molecular mechanisms of HCC-associated occurrence, proliferation, metastasis and carcinogenesis. In the present study, the role of Up-frameshift 1 (UPF1), a potential tumor suppressor, was investigated in the HCC cell lines. The expression levels of UPF1 in an HCC cell line were examined by reverse transcription-quantitative polymerase chain reaction. The expression levels of 19 key proteins in numerous signaling pathways were detected via protein array analysis in the presence of UPF1 overexpression. The present study further investigated the effects of UPF1 expression levels on the epithelial-mesenchymal transition (EMT) process by targeting E-cadherin, N-cadherin, Vimentin and Twist-related protein 1 (Twist). The results of the present study revealed that UPF1 was significantly downregulated in an HCC cell line. The majority of the proteins exhibited upregulated expression levels in the presence of UPF1 overexpression in the HCC cell line, Huh-7. Key proteins, including cluster of differentiation (CD)31 (platelet endothelial cell adhesion molecule-1), Vimentin, CD44, PCNA, Ki-67, N-Cadherin, Survivin, P53, Met and retinoblastoma exhibited a significant association with UPF1. Furthermore, western blotting indicated that the expression levels of N-cadherin, Vimentin and Twist were notably upregulated while UPF1 was overexpressed; however, E-cadherin was downregulated and opposing observations were reported with protein array analysis. In summary, E-cadherin expression levels were regulated by the manifold, and UPF1, a potential tumor suppressor, may promote the EMT process in Huh-7 HCC cells. The findings of the present study suggested that UPF1 expression levels affected the EMT process by targeting E-cadherin, N-cadherin, Vimentin and Twist.
Collapse
Affiliation(s)
- Yawei Li
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Tiantian Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Shukui Qin
- Department of Medical Oncology, PLA Cancer Center, Nanjing Bayi Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Rui Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Yumei Li
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Zhengguang Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Yufo Chen
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Qiong Wu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Fang Su
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| |
Collapse
|
27
|
Hu M, Xiong S, Chen Q, Zhu S, Zhou X. Novel role of microRNA-126 in digestive system cancers: From bench to bedside. Oncol Lett 2019; 17:31-41. [PMID: 30655735 PMCID: PMC6313097 DOI: 10.3892/ol.2018.9639] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 09/28/2018] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are ubiquitously expressed, small, non-coding RNAs that regulate the expression of approximately 30% of the human genes at the post-transcriptional level. miRNAs have emerged as crucial modulators in the initiation and progression of various diseases, including numerous cancer types. The high incidence rate of cancer and the large number of cancer-associated cases of mortality are mostly due to a lack of effective treatments and biomarkers for early diagnosis. Therefore there is an urgent requirement to further understand the underlying mechanisms of tumorigenesis. MicroRNA-126 (miR-126) is significantly downregulated in a number of tumor types and is commonly identified as a tumor suppressor in digestive system cancers (DSCs). miR-126 downregulates various oncogenes, including disintegrin and metalloproteinase domain-containing protein 9, v-crk sarcoma virus CT10 oncogene homolog and phosphoinositide-3-kinase regulatory subunit 2. These genes are involved in a number of tumor-associated signaling pathways, including angiogenesis, epithelial-mensenchymal transition and metastasis pathways. The aim of the current review was to summarize the role of miR-126 in DSCs, in terms of its dysregulation, target genes and associated signaling pathways. In addition, the current review has discussed the potential clinical application of miR-126 as a biomarker and therapeutic target for DSCs.
Collapse
Affiliation(s)
- Mingli Hu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Shengwei Xiong
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Qiaofeng Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Shixuan Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Xiaodong Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
28
|
EGFL6 promotes breast cancer by simultaneously enhancing cancer cell metastasis and stimulating tumor angiogenesis. Oncogene 2018; 38:2123-2134. [PMID: 30455428 DOI: 10.1038/s41388-018-0565-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/02/2018] [Accepted: 10/25/2018] [Indexed: 12/19/2022]
Abstract
EGFL6, a member of the EGF-like superfamily, plays an important role during embryonic development and has been implicated in promotion of tumor angiogenesis without affecting wound healing. There is very little known about the function of EGFL6 in cancer cells. Here, we investigated whether EGFL6 plays a direct role in cancer cells in addition to the promotion of tumor angiogenesis. Our study showed that EGFL6 promoted epithelial-mesenchymal transition (EMT) and stemness of breast cancer cells and increased cell migration and invasion in cell culture studies. We also found that EGFL6 reduced apoptotic signaling in cancer cells and promoted tumor growth in vivo. Importantly, expression of EGFL6 in cancer cells and tumor endothelial cells not only increased tumor angiogenesis but also promoted migration of cancer cells. Such dual engagement of cancer and stromal cells suggests crosstalk mediated by EGFL6 in the tumor microenvironment. Blockade of EGFL6 using our novel anti-EGFL6 monoclonal antibody significantly reduced cancer cell migration, tumor angiogenesis, and tumor growth in mouse xenograft tumor models. Silencing EGFL6 mRNA by shRNA transfection of cancer cells also significantly reduced cancer cell migration, tumor angiogenesis, and tumor growth in mouse xenograft tumor models. Taken together, the results of this study indicate that targeting EGFL6 is a unique strategy for inhibiting both cancer cell metastasis and tumor angiogenesis.
Collapse
|
29
|
Dong B, Zhou B, Sun Z, Huang S, Han L, Nie H, Chen G, Liu S, Zhang Y, Bao N, Yang X, Feng H. LncRNA-FENDRR mediates VEGFA to promote the apoptosis of brain microvascular endothelial cells via regulating miR-126 in mice with hypertensive intracerebral hemorrhage. Microcirculation 2018; 25:e12499. [PMID: 30120860 DOI: 10.1111/micc.12499] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/11/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND LncRNA-FENDRR is a kind of endothelial genes critical for vascular development. Moreover, miR-126 and vascular endothelial growth factor A (VEGFA) are also involved in the physiological process of vascular endothelial cells. This study aimed to the underlying mechanism of FENDRR involving miR-126 and VEGFA in hypertensive intracerebral hemorrhage (HICH). METHODS C57BL/6 mice were chosen to establish HICH model. The expression of FENDRR, miR-126, and VEGFA at mRNA level was determined by qRT-PCR. The protein expression of VEGFA was assessed using Western blot. RIP assay and RNA pull-down assay were used to the relationship between FENDRR and miR-126. Flow cytometry was used to analyze cell apoptosis. RESULTS The levels of FENDRR and VEGFA were increased, and miR-126 expression was decreased in vascular endothelial cells (VECs) from the right brain of model mice and human brain microvascular endothelial cells (HBMECs) treated by thrombin. Overexpression of FENDRR promoted the apoptosis of HBMECs. FENDRR regulating VEGFA participated in HBMECs apoptosis through targeting miR-126. Downregulation of FENDRR was indicated to relieve the HICH in mice. CONCLUSIONS FENDRR could promote the apoptosis of HBMECs via miR-126 regulating VEGFA in HICH.
Collapse
Affiliation(s)
- Baizhuo Dong
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Bin Zhou
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Zhigang Sun
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Shengming Huang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Liang Han
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Honghua Nie
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Guohui Chen
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Shibing Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Yanna Zhang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Ning Bao
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Xiaolong Yang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Hongwei Feng
- Department of Neurosurgery, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| |
Collapse
|
30
|
Li Z, Xue T, Yang C, Wang Y, Zhu X, Ni C. EGFL7 promotes hepatocellular carcinoma cell proliferation and inhibits cell apoptosis through increasing CKS2 expression by activating Wnt/β‐catenin signaling. J Cell Biochem 2018; 119:10327-10337. [PMID: 30129142 DOI: 10.1002/jcb.27375] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/02/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Zhi Li
- Department of Interventional Radiology The First Affliated Hospital of Soochow University Suzhou China
| | - Tong‐Qing Xue
- Department of Interventional Radiology The First Affliated Hospital of Soochow University Suzhou China
- Department of Interventional Radiology Huaian Hospital of Huaian City Huaian China
| | - Chao Yang
- Department of Interventional Radiology The First Affliated Hospital of Soochow University Suzhou China
| | - Yun‐Liang Wang
- Department of General Surgery The First Affliated Hospital of Soochow University Suzhou China
| | - Xiao‐Li Zhu
- Department of Interventional Radiology The First Affliated Hospital of Soochow University Suzhou China
| | - Cai‐Fang Ni
- Department of Interventional Radiology The First Affliated Hospital of Soochow University Suzhou China
| |
Collapse
|
31
|
Chen M, Xia Y, Tan Y, Jiang G, Jin H, Chen Y. Downregulation of microRNA-370 in esophageal squamous-cell carcinoma is associated with cancer progression and promotes cancer cell proliferation via upregulating PIN1. Gene 2018; 661:68-77. [PMID: 29605603 DOI: 10.1016/j.gene.2018.03.090] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/23/2018] [Accepted: 03/28/2018] [Indexed: 01/03/2023]
Abstract
PIN1 is a peptidyl-prolyl cis/trans isomerase (PPIase) that controls cell fate by regulating multiple signal transduction pathways and is found to be overexpressed in a variety of malignant tumors. Herein, we found the expression of PIN1 is up-regulated while miRNA-370 (miR-370) down-regulated in both esophageal squamous-cell carcinoma (ESCC) tissues and cells. Transfection of miR-370 can significantly decrease PIN1 expression in targeting ESCC cells. Overexpression of miR-370 can induce decreased cell proliferation and cell cycle arrest, as well as increased apoptosis in ESCC cells, while this function can be significantly prevented by co-transfection of PIN1. Further experimental results demonstrated that β-catenin, cyclin D1, and caspase activation might be involved in miR-370/PIN1 induced growth inhibition and apoptosis. Besides, low miR-370 and high PIN1 expression significantly correlated with tumor diameter, poor differentiation, tumor invasion and lymph node metastasis in patients diagnosed with ESCC. In conclusion, downregulation of miR-370 in ESCC is associated with cancer progression and promotes cancer cell proliferation via upregulating PIN1, which might be a potential therapeutic target and adverse prognostic factor in the clinic.
Collapse
Affiliation(s)
- Mingzhi Chen
- Department of Thoracic and Cardiovascular Surgery, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province 210029, PR China; Department of Thoracic and Cardiovascular Surgery, Yixing People's Hospital affiliated to Jiangsu University, 75 Tongzhen Road, Yixing, Jiangsu Province 214200, PR China
| | - Yang Xia
- Department of Thoracic and Cardiovascular Surgery, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province 210029, PR China
| | - Yongfei Tan
- Department of Thoracic and Cardiovascular Surgery, Yixing People's Hospital affiliated to Jiangsu University, 75 Tongzhen Road, Yixing, Jiangsu Province 214200, PR China
| | - Guojun Jiang
- Department of Thoracic and Cardiovascular Surgery, Yixing People's Hospital affiliated to Jiangsu University, 75 Tongzhen Road, Yixing, Jiangsu Province 214200, PR China
| | - Hai Jin
- Department of Thoracic Surgery, Changhai Hospital affiliated to the Second Military Medical University, 168 Changhai Road, Shanghai 200433, PR China
| | - Yijiang Chen
- Department of Thoracic and Cardiovascular Surgery, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province 210029, PR China.
| |
Collapse
|
32
|
Fang Y, Liu G, Xie C, Qian K, Lei X, Liu Q, Liu G, Cao Z, Fu J, Du H, Liu S, Huang S, Hu J, Xu X. Pharmacological inhibition of TRPV4 channel suppresses malignant biological behavior of hepatocellular carcinoma via modulation of ERK signaling pathway. Biomed Pharmacother 2018; 101:910-919. [PMID: 29635900 DOI: 10.1016/j.biopha.2018.03.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 12/28/2022] Open
Abstract
TRPV4 (transient receptor potential vanilloid 4), a member of the TRP superfamily, has been reported to correlate with several different forms of cancers. However, the role of TRPV4 in human hepatocellular carcinoma (HCC) remains unclear. The present study demonstrated that elevated expression of TRPV4 was shown in HCC tumor tissues when compared with paired non-tumoral livers both in protein and mRNA levels. Furthermore, the enhanced expression of TRPV4 was highly associated with histological grade (P = 0.036) and the number of tumors (P = 0.045). Pharmacological inhibition of TRPV4 channels in HCC cells with the specific antagonist HC-067047 suppressed cell proliferation, induced apoptosis and decreased the migration capability by attenuating the epithelial-mesenchymal transition (EMT) process in vitro. The p-ERK expression was apparently repressed after treatment with the TRPV4 antagonist, further blockade of the ERK pathway with U0126 could significantly aggravate HCC cells apoptosis. In NOD-SCID mouse xenograft models, intraperitoneal injection of HC-067047 could obviously suppress tumor growth and induce apoptosis in vivo. Together, our studies showed that the antitumor effects caused by TRPV4 channel inhibition in HCC cell lines might be attributed to the suppression of EMT process and inactivation of p-ERK which induced subsequent cell apoptosis. Thus, pharmacological inhibition of TRPV4 channel may be an option for HCC treatment.
Collapse
Affiliation(s)
- Yu Fang
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Guoxing Liu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Chengzhi Xie
- Department of General Surgery, The 2nd Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, Hunan 410005, China
| | - Ke Qian
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiaohua Lei
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qiang Liu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Gao Liu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zhenyu Cao
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jie Fu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Huihui Du
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Sushun Liu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Shengfu Huang
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jixiong Hu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xundi Xu
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
33
|
Yu H, Zheng J, Liu X, Xue Y, Shen S, Zhao L, Li Z, Liu Y. Transcription Factor NFAT5 Promotes Glioblastoma Cell-driven Angiogenesis via SBF2-AS1/miR-338-3p-Mediated EGFL7 Expression Change. Front Mol Neurosci 2017; 10:301. [PMID: 28983240 PMCID: PMC5613209 DOI: 10.3389/fnmol.2017.00301] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 09/06/2017] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary intracranial tumor of adults and confers a poor prognosis due to high vascularization. Hence anti-angiogenic therapy has become a promising strategy for GBM treatment. In this study, the transcription factor nuclear factor of activated T-cells 5 (NFAT5) was significantly elevated in glioma samples and GBM cell lines, and positively correlated with glioma WHO grades. Knockdown of NFAT5 inhibited GBM cell-driven angiogenesis. Furthermore, long non-coding RNA SBF2 antisense RNA 1 (SBF2-AS1) was upregulated in glioma samples and knockdown of SBF2-AS1 impaired GBM-induced angiogenesis. Downregulation of NFAT5 decreased SBF2-AS1 expression at transcriptional level. In addition, knockdown of SBF2-AS1 repressed GBM cell-driven angiogenesis via enhancing the inhibitory effect of miR-338-3p on EGF like domain multiple 7 (EGFL7). In vivo study demonstrated that the combination of NFAT5 knockdown and SBF2-AS1 knockdown produced the smallest xenograft volume and the lowest microvessel density. NFAT5/SBF2-AS1/miR-338-3p/EGFL7 pathway may provide novel targets for glioma anti-angiogenic treatment.
Collapse
Affiliation(s)
- Hai Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China.,Liaoning Research Center for Clinical Medicine in Nervous System DiseaseShenyang, China.,Key laboratory of Neuro-oncology in Liaoning ProvinceShenyang, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China.,Liaoning Research Center for Clinical Medicine in Nervous System DiseaseShenyang, China.,Key laboratory of Neuro-oncology in Liaoning ProvinceShenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China.,Liaoning Research Center for Clinical Medicine in Nervous System DiseaseShenyang, China.,Key laboratory of Neuro-oncology in Liaoning ProvinceShenyang, China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of ChinaShenyang, China
| | - Shuyuan Shen
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of ChinaShenyang, China
| | - Lini Zhao
- Department of Neurobiology, College of Basic Medicine, China Medical UniversityShenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of ChinaShenyang, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China.,Liaoning Research Center for Clinical Medicine in Nervous System DiseaseShenyang, China.,Key laboratory of Neuro-oncology in Liaoning ProvinceShenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical UniversityShenyang, China.,Liaoning Research Center for Clinical Medicine in Nervous System DiseaseShenyang, China.,Key laboratory of Neuro-oncology in Liaoning ProvinceShenyang, China
| |
Collapse
|