1
|
Kapri A, Singh D, Onteru SK. Deciphering Aflatoxin B1 affected critical molecular pathways governing cancer: A bioinformatics study using CTD and PANTHER databases. Mycotoxin Res 2025; 41:93-111. [PMID: 39417919 DOI: 10.1007/s12550-024-00563-0] [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: 02/02/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
Aflatoxin B1 (AFB1) is a fungal toxin consistently found as a contaminant in food products such as cereals, nuts, spices, and oilseeds. AFB1 exposure can lead to hepatotoxicity, cancer, immune suppression, reproductive deficiency, nutritional dysfunction, and growth impairment. AFB1 has also been listed as one of the most potent human carcinogens by the International Agency for Research on Cancer. Although the correlation between AFB1 exposure and cancer initiation and progression is already reported in the literature, very little information is available about what molecular pathways are affected during cancer development. Considering this, we first selected AFB1-responsive genes involved in five deadliest cancer types including lung, colorectal, liver, stomach, and breast cancers from the Comparative Toxicogenomics Database (CTD). Then, using the PANTHER database, a statistical overrepresentation test was performed to identify the significantly affected pathways in each cancer type. The gonadotropin-releasing hormone receptor (GnRHR) pathway, the CCKR signaling pathway, and angiogenesis were found to be the most affected pathways in lung, breast, liver, and stomach cancers. In addition, AFB1 toxicity majorly impacted apoptosis and Wnt signaling pathways in liver and stomach cancers, respectively. Moreover, the most affected pathways in colorectal cancer were the Wnt, CCKR, and GnRHR pathways. Furthermore, gene analysis was also performed for the most affected pathways associated with each cancer and identified thirteen key genes (e.g., FOS, AKT1) that may serve as biological markers for a particular type of AFB1-induced cancer as well as for in vitro AFB1 toxicological studies using specific cancer cell lines.
Collapse
Affiliation(s)
- Ankita Kapri
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Dheer Singh
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Suneel Kumar Onteru
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India.
| |
Collapse
|
2
|
Li Y, Liu J, Wu S, Xiao J, Zhang Z. Ferroptosis: opening up potential targets for gastric cancer treatment. Mol Cell Biochem 2024; 479:2863-2874. [PMID: 38082184 DOI: 10.1007/s11010-023-04886-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/24/2023] [Indexed: 10/15/2024]
Abstract
The fifth most frequent cancer in the world is gastric cancer. It ranks as the fourth most common reason for cancer-related deaths. Even though surgery is the only curative treatment for stomach cancer, adding adjuvant radiotherapy and chemotherapy is preferable than only surgery. The majority of patients, however, are discovered to be extremely tardy the first time and have a terrible prognosis. Therefore, it is necessary to create more viable therapy modalities. A growing number of studies in recent years have shown that ferroptosis and many cancer types are related. This gives our treatment a fresh viewpoint. We investigated the relationship between different signal pathways and non-coding RNA on ferroptosis in gastric cancer cells. Also discussed the targets cause ferroptosis resistance increased or reduced to the influence of the chemoresistance,proliferation and metastasis.
Collapse
Affiliation(s)
- Yuwei Li
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, 28 Changsheng Road, Hengyang, 421001, Hunan, China
| | - Jiangrong Liu
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, 28 Changsheng Road, Hengyang, 421001, Hunan, China
| | - Shihua Wu
- Department of Pathology, The Second Affiliated Hospital, Shaoyang University, Shaoyang, 422000, Hunan, China
| | - Juan Xiao
- Department of Head and Neck Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Zhiwei Zhang
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, 28 Changsheng Road, Hengyang, 421001, Hunan, China.
| |
Collapse
|
3
|
Ayoup MS, Rabee AR, Abdel-Hamid H, Amer A, Abu-Serie MM, Ashraf S, Ghareeb DA, Ibrahim RS, Hawsawi MB, Negm A, Ismail MMF. Design and Synthesis of Quinoxaline Hybrids as Modulators of HIF-1a, VEGF, and p21 for Halting Colorectal Cancer. ACS OMEGA 2024; 9:24643-24653. [PMID: 38882127 PMCID: PMC11170630 DOI: 10.1021/acsomega.4c01075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 06/18/2024]
Abstract
A library of 16 3-benzyl-N 1-substituted quinoxalin-2-ones was synthesized as N 1-substituted quinoxalines and quinoxaline-triazole hybrids via click reaction. These compounds were tested for their anticancer activity via MTT assay on HCT-116 and normal colonocyte cell lines to assess their cytotoxic potentials and safety profiles. Overall, compounds 6, 9, 14, and 20 were found to be promising anticolorectal cancer agents; they exhibited remarkable cytotoxicity (IC50 0.05-0.07 μM) against HCT-116 cells within their safe doses (EC100) on normal colon cells. Their pronounced anticancer activities were observed as severe morphological alterations and shrinkage of the treated cancer cells. Besides, qRT-PCR analysis was conducted showing the potential of the promising hits to downregulate HIF-1a, VEGF, and BCL-2 as well as their ability to enhance the expression of proapoptotic genes p21, p53, and BAX in HCT-116 cells. In silico prediction revealed that most of our compounds agree with Lipinski and Veber parameters of rules, in addition to remarkable medicinal chemistry and drug-likeness parameters with no CNS side effects. Interestingly, docking studies of the compounds in the VEGFR-2' active site showed significant affinity toward the essential amino acids, which supported the biological results.
Collapse
Affiliation(s)
- Mohammed Salah Ayoup
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria 21321, Egypt
| | - Ahmed R Rabee
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria 21321, Egypt
| | - Hamida Abdel-Hamid
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria 21321, Egypt
| | - Adel Amer
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria 21321, Egypt
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El Arab, Alexandria 21934, Egypt
| | - Samah Ashraf
- Bio-screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, 21511 Alexandria, Egypt
| | - Doaa A Ghareeb
- Bio-screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, 21511 Alexandria, Egypt
- Center of Excellence for Drug Preclinical Studies (CE-DPS), Pharmaceutical and Fermentation Industry Development Center, City of Scientific Research & Technological Applications (SRTA-city), New Borg El Arab, Alexandria 21934, Egypt
| | - Rabab S Ibrahim
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt
| | - Mohammed B Hawsawi
- Department of Chemistry, Faculty of Science, Umm Al-Qura University, Al Taif Road, Makkah 24382, Saudi Arabia
| | - Amr Negm
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Chemistry Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Magda M F Ismail
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt
| |
Collapse
|
4
|
Zhao H, Ding Y, Zhang L. SIRT1/APE1 promotes the viability of gastric cancer cells by inhibiting p53 to suppress ferroptosis. Open Med (Wars) 2023; 18:20220620. [PMID: 36820068 PMCID: PMC9938643 DOI: 10.1515/med-2022-0620] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 02/16/2023] Open
Abstract
Gastric cancer (GC) is a common cancer worldwide with high mortality. Sirtuin 1 (SIRT1) and apurinic/apyrimidinic endodeoxyribonuclease 1 (APE1) are abnormally expressed in GC cells and related to p53, which is involved in ferroptosis. Thus, we explore the mechanism via which SIRT1, APE1, and p53 impact ferroptosis in GC cells. Specifically, GC cells were transfected with small-interfering RNA for SIRT1 (SiSIRT1) or small-interfering RNA for APE1 (SiAPE1) or with short-hairpin RNA for p53, and the cell viability, Fe2+, malondialdehyde (MDA), and glutathione (GSH) contents were detected by cell counting kit-8 assay and enzyme-linked immunosorbent assay. Western blot, immunofluorescence, and quantitative real-time polymerase chain reaction were conducted to quantify SIRT1, APE1, p53, solute carrier family 7 member 11 (SLC7A11), and glutathione peroxidase 4 (GPX4) levels in GC cells. Silencing of SIRT1 decreased viability, GSH content, and expressions of GPX4 and SLC7A11, while increased Fe2+, MDA content, and p53 expression in GC cells. Such aforementioned effects were reversed by APE1 overexpression. Also, SiAPE1 generated the same effects as SiSIRT1 on the above aspects, which was offset by p53 silencing. In short, SIRT1/APE1 promotes the growth of GC cells by targeting p53 to inhibit ferroptosis.
Collapse
Affiliation(s)
- Huijin Zhao
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, 050000, China
| | - Yuanyi Ding
- Department of No. 2 General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, 050000, China
| | - Lan Zhang
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, 050000, China
| |
Collapse
|
5
|
Wu QJ, Zhang TN, Chen HH, Yu XF, Lv JL, Liu YY, Liu YS, Zheng G, Zhao JQ, Wei YF, Guo JY, Liu FH, Chang Q, Zhang YX, Liu CG, Zhao YH. The sirtuin family in health and disease. Signal Transduct Target Ther 2022; 7:402. [PMID: 36581622 PMCID: PMC9797940 DOI: 10.1038/s41392-022-01257-8] [Citation(s) in RCA: 352] [Impact Index Per Article: 117.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 12/30/2022] Open
Abstract
Sirtuins (SIRTs) are nicotine adenine dinucleotide(+)-dependent histone deacetylases regulating critical signaling pathways in prokaryotes and eukaryotes, and are involved in numerous biological processes. Currently, seven mammalian homologs of yeast Sir2 named SIRT1 to SIRT7 have been identified. Increasing evidence has suggested the vital roles of seven members of the SIRT family in health and disease conditions. Notably, this protein family plays a variety of important roles in cellular biology such as inflammation, metabolism, oxidative stress, and apoptosis, etc., thus, it is considered a potential therapeutic target for different kinds of pathologies including cancer, cardiovascular disease, respiratory disease, and other conditions. Moreover, identification of SIRT modulators and exploring the functions of these different modulators have prompted increased efforts to discover new small molecules, which can modify SIRT activity. Furthermore, several randomized controlled trials have indicated that different interventions might affect the expression of SIRT protein in human samples, and supplementation of SIRT modulators might have diverse impact on physiological function in different participants. In this review, we introduce the history and structure of the SIRT protein family, discuss the molecular mechanisms and biological functions of seven members of the SIRT protein family, elaborate on the regulatory roles of SIRTs in human disease, summarize SIRT inhibitors and activators, and review related clinical studies.
Collapse
Affiliation(s)
- Qi-Jun Wu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tie-Ning Zhang
- grid.412467.20000 0004 1806 3501Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Huan-Huan Chen
- grid.412467.20000 0004 1806 3501Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xue-Fei Yu
- grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jia-Le Lv
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Yang Liu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ya-Shu Liu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Gang Zheng
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun-Qi Zhao
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Fan Wei
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing-Yi Guo
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fang-Hua Liu
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qing Chang
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Xiao Zhang
- grid.412467.20000 0004 1806 3501Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cai-Gang Liu
- grid.412467.20000 0004 1806 3501Department of Cancer, Breast Cancer Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Hong Zhao
- grid.412467.20000 0004 1806 3501Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China ,grid.412467.20000 0004 1806 3501Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Ivey A, Pratt H, Boone BA. Molecular pathogenesis and emerging targets of gastric adenocarcinoma. J Surg Oncol 2022; 125:1079-1095. [PMID: 35481910 PMCID: PMC9069999 DOI: 10.1002/jso.26874] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/15/2022] [Accepted: 03/19/2022] [Indexed: 12/24/2022]
Abstract
Gastric adenocarcinoma (GC) is a devastating disease and is the third leading cause of cancer deaths worldwide. This heterogeneous disease has several different classification systems that consider histological appearance and genomic alterations. Understanding the etiology of GC, including infection, hereditary conditions, and environmental factors, is of particular importance and is discussed in this review. To improve survival in GC, we also must improve our therapeutic strategies. Here, we discuss new targets that warrant further exploration.
Collapse
Affiliation(s)
- Abby Ivey
- Department of Cancer Cell Biology, West Virginia University Cancer Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Hillary Pratt
- Department of Cancer Cell Biology, West Virginia University Cancer Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Brian A Boone
- Department of Cancer Cell Biology, West Virginia University Cancer Institute, West Virginia University, Morgantown, West Virginia, USA
- Department of Surgery, Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
7
|
Wang Y, Wang J, Liu C, Li M. Silent Information Regulator 1 Promotes Proliferation, Migration, and Invasion of Cervical Cancer Cells and Is Upregulated by Human Papillomavirus 16 E7 Oncoprotein. Gynecol Obstet Invest 2021; 87:22-29. [PMID: 34808628 DOI: 10.1159/000520642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/01/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Silent information regulator 1 (SIRT1), an NAD+-dependent III class histone deacetylase, plays crucial roles in cell proliferation, apoptosis, senescence, metabolism, and stress responses. Nevertheless, the role of SIRT1 in tumorigenesis remains unclear. METHODS In the present study, we measured expression levels of SIRT1 and HPV16 E7 protein in cervical cancer (CC) tissue and calculated their correlations. We measured the effect of silencing SIRT1 on the proliferation, migration, invasion, and apoptosis in human CC SiHa cells. RESULTS Immunohistochemistry results revealed that the expression of SIRT1 was upregulated with progression from CIN II-III to CC, but was not expressed in normal cervical tissues and CIN I. There was a positive correlation between SIRT1 expression and HPV16 E7 expression in CC tissues, and silencing of HPV16 E7 downregulated the expression of SIRT1. Depletion of SIRT1 downregulated SIRT1 expression, and inhibited proliferation, migration, and invasion of SiHa cells, inducing apoptosis. CONCLUSIONS Taken together, the data suggest that SIRT1 promotes CC carcinogenesis. SIRT1 inhibition is a potential treatment strategy for CC.
Collapse
Affiliation(s)
- Yujing Wang
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China,
| | - Jing Wang
- Qingdao Municipal Hospital, Qingdao, China
| | - Chunmei Liu
- Qingdao Research and Development Center of Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Qingdao, China
| | - Min Li
- Department of Gynecology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
8
|
Sousa F, Costa-Pereira AI, Cruz A, Ferreira FJ, Gouveia M, Bessa J, Sarmento B, Travasso RDM, Mendes Pinto I. Intratumoral VEGF nanotrapper reduces gliobastoma vascularization and tumor cell mass. J Control Release 2021; 339:381-390. [PMID: 34592385 DOI: 10.1016/j.jconrel.2021.09.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 01/05/2023]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and invasive malignant brain cancer. GBM is characterized by a dramatic metabolic imbalance leading to increased secretion of the pro-angiogenic factor VEGF and subsequent abnormal tumor vascularization. In 2009, FDA approved the intravenous administration of bevacizumab, an anti-VEGF monoclonal antibody, as a therapeutic agent for patients with GBM. However, the number of systemic side effects and reduced accessibility of bevacizumab to the central nervous system and consequently to the GBM tumor mass limited its effectiveness in improving patient survival. In this study, we combined experimental and computational modelling to quantitatively characterize the dynamics of VEGF secretion and turnover in GBM and in normal brain cells and simultaneous monitoring of vessel growth. We showed that sequestration of VEGF inside GBM cells, can be used as a novel target for improved bevacizumab-based therapy. We have engineered the VEGF nanotrapper, a cargo system that allows cellular uptake of bevacizumab and inhibits VEGF secretion required for angiogenesis activation and development. Here, we show the therapeutic efficacy of this nanocargo in reducing vascularization and tumor cell mass of GBM in vitro and in vivo cancer models.
Collapse
Affiliation(s)
- Flávia Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, 4150-180 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal; INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, 4715-330 Braga, Portugal
| | | | - Andrea Cruz
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, 4715-330 Braga, Portugal
| | - Fábio Júnio Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal
| | - Marcos Gouveia
- CFisUC - Department of Physics, University of Coimbra, Rua Larga, 3004-516 Coimbra, Portugal
| | - José Bessa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Rui D M Travasso
- CFisUC - Department of Physics, University of Coimbra, Rua Larga, 3004-516 Coimbra, Portugal
| | - Inês Mendes Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, 4715-330 Braga, Portugal.
| |
Collapse
|
9
|
Golubeva TS, Cherenko VA, Orishchenko KE. Recent Advances in the Development of Exogenous dsRNA for the Induction of RNA Interference in Cancer Therapy. Molecules 2021; 26:701. [PMID: 33572762 PMCID: PMC7865971 DOI: 10.3390/molecules26030701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/21/2021] [Accepted: 01/24/2021] [Indexed: 11/17/2022] Open
Abstract
Selective regulation of gene expression by means of RNA interference has revolutionized molecular biology. This approach is not only used in fundamental studies on the roles of particular genes in the functioning of various organisms, but also possesses practical applications. A variety of methods are being developed based on gene silencing using dsRNA-for protecting agricultural plants from various pathogens, controlling insect reproduction, and therapeutic techniques related to the oncological disease treatment. One of the main problems in this research area is the successful delivery of exogenous dsRNA into cells, as this can be greatly affected by the localization or origin of tumor. This overview is dedicated to describing the latest advances in the development of various transport agents for the delivery of dsRNA fragments for gene silencing, with an emphasis on cancer treatment.
Collapse
Affiliation(s)
- Tatiana S. Golubeva
- Department of Genetic Technologies, Novosibirsk State University, Novosibirsk 630090, Russia; (V.A.C.); (K.E.O.)
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Viktoria A. Cherenko
- Department of Genetic Technologies, Novosibirsk State University, Novosibirsk 630090, Russia; (V.A.C.); (K.E.O.)
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Konstantin E. Orishchenko
- Department of Genetic Technologies, Novosibirsk State University, Novosibirsk 630090, Russia; (V.A.C.); (K.E.O.)
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| |
Collapse
|
10
|
Hsieh HL, Tsai MM. Tumor progression-dependent angiogenesis in gastric cancer and its potential application. World J Gastrointest Oncol 2019; 11:686-704. [PMID: 31558974 PMCID: PMC6755109 DOI: 10.4251/wjgo.v11.i9.686] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/05/2019] [Accepted: 08/19/2019] [Indexed: 02/05/2023] Open
Abstract
Despite improvements in the early diagnosis, prognosis and therapeutic strategies for gastric cancer (GC), human GC remains one of the most frequently diagnosed malignant tumors in the world, and the survival rate of GC patients remains very poor. Thus, a suitable therapeutic strategy for GC is important for prolonging survival. Both tumor cells themselves and the tumor microenvironment play an important role in tumorigenesis, including angiogenesis, inflammation, immunosuppression and metastasis. Importantly, these cells contribute to gastric carcinogenesis by altering the angiogenic phenotype switch. The development, relapse and spreading of tumors depend on new vessels that provide the nutrition, growth factors and oxygen required for continuous tumor growth. Therefore, a state of tumor dormancy could be induced by blocking tumor-associated angiogenesis. Recently, several antiangiogenic agents have been identified, and their potential for the clinical management of GC has been tested. Here, we provide an up-to-date summary of angiogenesis and the angiogenic factors associated with tumor progression in GC. We also review antiangiogenic agents with a focus on the anti-vascular endothelial growth factor receptor (VEGFR)-mediated pathway for endothelial cell growth and their angiogenesis ability in GC. However, most antiangiogenic agents have reported no benefit to overall survival (OS) compared to chemotherapy alone in local or advanced GC. In phase III clinical trials, only ramucirumab (anti-VEGFR blocker) and apatinib (VEGFR-TKI blocker) have reported an improved median overall response rate and prolonged OS and progression-free survival outcomes as a 2nd-line agent combined with chemotherapy treatment in advanced GC. By providing insights into the molecular mechanisms of angiogenesis associated with tumor progression in GC, this review will hopefully aid the optimization of antiangiogenesis strategies for GC therapy in combination with chemotherapy and adjuvant treatment.
Collapse
Affiliation(s)
- Hsi-Lung Hsieh
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of Nursing, Division of Basic Medical Sciences, Chang-Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ming-Ming Tsai
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of Nursing, Division of Basic Medical Sciences, Chang-Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| |
Collapse
|
11
|
Nienhüser H, Schmidt T. Angiogenesis and Anti-Angiogenic Therapy in Gastric Cancer. Int J Mol Sci 2017; 19:ijms19010043. [PMID: 29295534 PMCID: PMC5795993 DOI: 10.3390/ijms19010043] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is one of the most frequent malignancies worldwide. Despite improvements in diagnosis and therapy, the overall prognosis remains poor. In the last decade, several anti-angiogenic drugs for cancer treatment have been approved and lately also introduced to gastric cancer treatment. While the initial trials focused only on unresectable or metastatic cancer, anti-angiogenic treatment is now also investigated in the perioperative and neoadjuvant setting. In this review, an overview of the role of angiogenesis and angiogenic factors in gastric cancer as well as anti-angiogenic treatment of gastric cancer is provided. Findings from in vitro and animal studies are summarized and put in a context with translational data on angiogenesis in gastric cancer. The most important angiogenic factors and their effect in gastric cancer are highlighted and clinical trials including anti-angiogenic drugs are discussed. Finally, an outlook of biomarkers for predicting response to anti-angiogenic treatment is presented, the ongoing trials on this topic are discussed and current challenges of anti-angiogenic therapy are outlined.
Collapse
Affiliation(s)
- Henrik Nienhüser
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| |
Collapse
|
12
|
Zou Y, Guo CG, Yang ZG, Sun JH, Zhang MM, Fu CY. A small interfering RNA targeting vascular endothelial growth factor efficiently inhibits growth of VX2 cells and VX2 tumor model of hepatocellular carcinoma in rabbit by transarterial embolization-mediated siRNA delivery. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1243-55. [PMID: 27069355 PMCID: PMC4818046 DOI: 10.2147/dddt.s94122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction Hepatocellular carcinoma is currently the second leading cause of cancer-related deaths worldwide with an increasing incidence. Objective The objective of this study is to investigate the effect of vascular endothelial growth factor small interfering RNA (VEGF-siRNA) on rabbit VX2 carcinoma cell viability in vitro and the effect of transarterial embolization (TAE)-mediated VEGF-siRNA delivery on the growth of rabbit VX2 liver-transplanted model in vivo. Methods Quantitative reverse transcription polymerase chain reaction, enzyme-linked immunosorbent assay, and Western blot technologies were used to detect the expression level of VEGF. TAE and computed tomography scan were used to deliver the VEGF-siRNA and detect the tumor volume in vivo, respectively. Microvessel density was detected by immunohistochemistry with CD34 antibody. A biochemical autoanalyzer was used to evaluate the hepatic and renal toxicity. Results The designed VEGF-siRNAs could effectively decrease the expression levels of VEGF mRNA and protein in vitro and in vivo. In vitro, the viability of rabbit VX2 carcinoma cells was reduced by 38.5%±7.3% (VEGF-siRNA no 1) and 30.0%±5.8% (VEGF-siRNA no 3) at 48 hours after transfection. Moreover, in rabbit VX2 liver-transplanted model, the growth ratios of tumors at 28 days after TAE-mediated siRNA delivery were 155.18%±19.42% in the control group, 79.67%±19.63% in the low-dose group, and 36.09%±15.73% in the high-dose group, with significant differences among these three groups. Microvessel density dropped to 34.22±4.01 and 22.63±4.07 in the low-dose group and high-dose group, respectively, compared with the control group (57.88±5.67), with significant differences among these three groups. Furthermore, inoculation of VX2 tumor into the liver itself at later stage induced significant increase in alanine aminotransferase and aspartate aminotransferase, indicating an obvious damage of liver functions, while treatment of VX2 tumor via TAE-mediated VEGF-siRNA had no toxicity to the livers and kidneys of rabbits, and VEGF-siRNA had the ability to protect liver damage induced by tumor growth. Conclusion This is the first study to demonstrate that targeting VEGF via TAE-mediated siRNA delivery may become a powerful new option for effective treatment of hepatocellular carcinoma in the clinic.
Collapse
Affiliation(s)
- Yu Zou
- Department of Radiology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Department of Radiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Chuan-Gen Guo
- Department of Radiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Zheng-Gang Yang
- Institute of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jun-Hui Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Min-Ming Zhang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Cai-Yun Fu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, People's Republic of China; Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
13
|
Kim SY, Ko YS, Park J, Choi Y, Park JW, Kim Y, Pyo JS, Yoo YB, Lee JS, Lee BL. Forkhead Transcription Factor FOXO1 Inhibits Angiogenesis in Gastric Cancer in Relation to SIRT1. Cancer Res Treat 2015; 48:345-54. [PMID: 25761483 PMCID: PMC4720104 DOI: 10.4143/crt.2014.247] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 12/05/2014] [Indexed: 01/26/2023] Open
Abstract
Purpose We previously reported that forkhead transcription factors of the O class 1 (FOXO1) expression in gastric cancer (GC) was associated with angiogenesis-related molecules. However, there is little experimental evidence for the direct role of FOXO1 in GC. In the present study, we investigated the effect of FOXO1 on the tumorigenesis and angiogenesis in GC and its relationship with SIRT1. Materials and Methods Stable GC cell lines (SNU-638 and SNU-601) infected with a lentivirus containing FOXO1 shRNA were established for animal studies as well as cell culture experiments. We used xenograft tumors in nude mice to evaluate the effect of FOXO1 silencing on tumor growth and angiogenesis. In addition, we examined the association between FOXO1 and SIRT1 by immunohistochemical tissue array analysis of 471 human GC specimens and Western blot analysis of xenografted tumor tissues. Results In cell culture, FOXO1 silencing enhanced hypoxia inducible factor-1α (HIF-1α) expression and GC cell growth under hypoxic conditions, but not under normoxic conditions. The xenograft study showed that FOXO1 downregulation enhanced tumor growth, microvessel areas, HIF-1α activation and vascular endothelial growth factor (VEGF) expression. In addition, inactivated FOXO1 expression was associated with SIRT1 expression in human GC tissues and xenograft tumor tissues. Conclusion Our results indicate that FOXO1 inhibits GC growth and angiogenesis under hypoxic conditions via inactivation of the HIF-1α–VEGF pathway, possibly in association with SIRT1. Thus, development of treatment modalities aiming at this pathway might be useful for treating GC.
Collapse
Affiliation(s)
- Sue Youn Kim
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea
| | - Young San Ko
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea
| | - Jinju Park
- Tumour Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yiseul Choi
- Tumour Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Wan Park
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea ; Ischemic/Hypoxic Disease Institute Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Younghoon Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Jung-Soo Pyo
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Bok Yoo
- Department of Anatomy, Dankook University School of Medicine, Cheonan, Korea
| | - Jae-Seon Lee
- Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Korea
| | - Byung Lan Lee
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea ; Tumour Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea ; Ischemic/Hypoxic Disease Institute Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Park DJ, Thomas NJ, Yoon C, Yoon SS. Vascular endothelial growth factor a inhibition in gastric cancer. Gastric Cancer 2015; 18:33-42. [PMID: 24993497 DOI: 10.1007/s10120-014-0397-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 06/11/2014] [Indexed: 02/07/2023]
Abstract
Angiogenesis is a vital process in the progression and metastasis of solids tumors including gastric adenocarcinoma. Tumors induce angiogenesis by secreting proangiogenic molecules such as vascular endothelial growth factor A (VEGF-A), and VEGF-A inhibition has become a common therapeutic strategy for many cancers. Several drugs targeting the VEGF-A pathway have been approved for clinical use in selected solid tumors, and several anti-VEGF-A strategies have been examined for gastric cancer. Phase II studies suggested that bevacizumab, an anti-VEGF antibody, can increase the efficacy of chemotherapy for advanced gastric cancer, but two international phase III trials failed to show an overall survival benefit. Two more recent international phase III trials have examined ramucirumab, an antibody targeting the primary receptor for VEGF-A, as second-line therapy for advanced gastric cancer and found a survival benefit both as single agent therapy and when combined with chemotherapy. Finally, correlative science studies suggest that the VEGF-A pathway may have varying importance in gastric cancer progression depending on ethnicity or race. This article will review the preclinical and clinical studies on the role of the VEGF-A pathway inhibition in gastric cancer.
Collapse
Affiliation(s)
- Do Joong Park
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | | | | | | |
Collapse
|
15
|
Doan CC, Le LT, Hoang SN, Do SM, Le DV. Simultaneous silencing of VEGF and KSP by siRNA cocktail inhibits proliferation and induces apoptosis of hepatocellular carcinoma Hep3B cells. Biol Res 2014; 47:70. [PMID: 25723613 PMCID: PMC4289546 DOI: 10.1186/0717-6287-47-70] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/06/2014] [Indexed: 01/04/2023] Open
Abstract
Background Vascular endothelial growth factor (VEGF) is involved in the growth of new blood vessels that feed tumors and kinesin spindle protein (KSP) plays a critical role in mitosis involving in cell proliferation. Simultaneous silencing of VEGF and KSP, an attractive and viable approach in cancer, leads on restricting cancer progression. The purpose of this study is to examine the therapeutic potential of dual gene targeted siRNA cocktail on human hepatocellular carcinoma Hep3B cells. Results The predesigned siRNAs could inhibit VEGF and KSP at mRNA level. siRNA cocktail showed a further downregulation on KSP mRNA and protein levels compared to KSP-siRNA or VEGF-siRNA, but not on VEGF expression. It also exhibited greater suppression on cell proliferation as well as cell migration or invasion capabilities and induction of apoptosis in Hep3B cells than single siRNA simultaneously. This could be explained by the significant downregulation of Cyclin D1, Bcl-2 and Survivin. However, no sigificant difference in the mRNA and protein levels of ANG2, involving inhibition of angiogenesis was found in HUVECs cultured with supernatant of Hep3B cells treated with siRNA cocktail, compared to that of VEGF-siRNA. Conclusion Silencing of VEGF and KSP plays a key role in inhibiting cell proliferation, migration, invasion and inducing apoptosis of Hep3B cells. Simultaneous silencing of VEGF and KSP using siRNA cocktail yields promising results for eradicating hepatocellular carcinoma cells, a new direction for liver cancer treatment.
Collapse
Affiliation(s)
- Chung Chinh Doan
- Faculty of Biology, University of Science, Vietnam National University, Ward 4,District 5, 227 Nguyen Van Cu Street, Ho Chi Minh City, Vietnam. .,Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, 9/621 Xa lo Ha Noi Street,Linh Trung Ward, Thu Duc District, Ho Chi Minh City, Vietnam.
| | - Long Thanh Le
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, 9/621 Xa lo Ha Noi Street,Linh Trung Ward, Thu Duc District, Ho Chi Minh City, Vietnam.
| | - Son Nghia Hoang
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, 9/621 Xa lo Ha Noi Street,Linh Trung Ward, Thu Duc District, Ho Chi Minh City, Vietnam.
| | - Si Minh Do
- Faculty of Biology, University of Science, Vietnam National University, Ward 4,District 5, 227 Nguyen Van Cu Street, Ho Chi Minh City, Vietnam.
| | - Dong Van Le
- Department of Immunology, Vietnam Military Medical University, 160 Phung Hung Street,Ha Dong District, Ha Noi City, Vietnam.
| |
Collapse
|
16
|
Gao JZ, DU JL, Wang YL, Li J, Wei LX, Guo MZ. Synergistic effects of curcumin and bevacizumab on cell signaling pathways in hepatocellular carcinoma. Oncol Lett 2014; 9:295-299. [PMID: 25435978 PMCID: PMC4246621 DOI: 10.3892/ol.2014.2694] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 09/26/2014] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to explore the effects of curcumin in combination with bevacizumab on the vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR)/K-ras pathway in hepatocellular carcinoma. A total of 30 Sprague Dawley (SD) rats were randomly divided into five groups: Control, model, curcumin, VEGF blocker, and curcumin + VEGF blocker groups. The mRNA levels of VEGF and VEGFR in all groups were subsequently measured by quantitative reverse transcriptase-polymerase chain reaction and the protein expression of K-ras was detected by western blot analysis. Compared with the control group, the mRNA levels of VEGF and VEGFR were revealed to be significantly increased in the model, curcumin and VEGF blocker groups. The VEGF mRNA levels in the curcumin, VEGF blocker and curcumin + VEGF blocker groups were all decreased when compared with the model group. In addition, the VEGF mRNA levels in the curcumin + VEGF blocker group were significantly lower compared with the curcumin group (P<0.05). The VEGF mRNA levels in the curcumin, VEGF blocker and curcumin + VEGF blocker groups were decreased when compared with the model group (P=0.0001). No significant differences in VEGF mRNA levels were identified between the VEGF blocker and curcumin groups (P=0.863), whereas the VEGF mRNA levels in the curcumin + VEGF blocker group were significantly lower than that of the curcumin group (P=0.025). Curcumin and the VEGF blocker are each capable of inhibiting hepatocellular carcinoma progression by regulating the VEGF/VEGFR/K-ras pathway. The combination of the two compounds has a synergistic effect on the inhibition of the effects of the VEGF signaling pathways in hepatocellular carcinoma progression.
Collapse
Affiliation(s)
- Jian-Zhi Gao
- Department of Pathology, General Hospital of the People's Liberation Army, Beijing 100853, P.R. China ; Basic Medical College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Jing-Li DU
- Department of Gastroenterology and Hepatology, General Hospital of the People's Liberation Army, Beijing 100853, P.R. China ; Department of Gastroenterology, Armed Police Corps Hospital of Qinghai, Xining, Qinghai 810006, P.R. China
| | - Yong-Ling Wang
- Basic Medical College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Jia Li
- Department of Pathology, General Hospital of the People's Liberation Army, Beijing 100853, P.R. China
| | - Li-Xin Wei
- Department of Pathology, General Hospital of the People's Liberation Army, Beijing 100853, P.R. China
| | - Ming-Zhou Guo
- Department of Gastroenterology and Hepatology, General Hospital of the People's Liberation Army, Beijing 100853, P.R. China
| |
Collapse
|
17
|
JIAO FENG, JIN ZILIANG, WANG LEI, WANG LIWEI. Research and clinical applications of molecular biomarkers in gastrointestinal carcinoma (Review). Biomed Rep 2013; 1:819-827. [PMID: 24649035 PMCID: PMC3917016 DOI: 10.3892/br.2013.158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 08/12/2013] [Indexed: 11/06/2022] Open
Abstract
Gastrointestinal (GI) carcinoma is a common malignant disease worldwide. Its development and progression is a multistage process involving a multifactorial etiology. Although the detailed mechanisms of the development of GI carcinoma remain controversial, the elucidation of its molecular biology over the last few years has resulted in a better perspective on its epidemiology, carcinogenesis and pathogenesis. More significantly, it is currently possible to use biological indicators or biomarkers in differential diagnosis, prognostic evaluation and specific clinical interventions. In this review, we aimed to describe the biomarkers of pathogenesis, invasion, metastasis and prognosis of GI carcinoma and discuss their potential clinical applications. The majority of these biomarkers, such as tumor-associated antigens, oncogenes and tumor suppressor genes, metastasis-associated genes, cell adhesion molecules, cytokines, growth factors and microRNAs, are currently broadly applicable.
Collapse
Affiliation(s)
- FENG JIAO
- Department of Oncology, The First People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 201620, P.R. China
| | - ZILIANG JIN
- Department of Oncology, The First People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 201620, P.R. China
| | - LEI WANG
- Department of Oncology, The First People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 201620, P.R. China
| | - LIWEI WANG
- Department of Oncology, The First People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 201620, P.R. China
| |
Collapse
|
18
|
Yang S, Lee KT, Lee JY, Lee JK, Lee KH, Rhee JC. Inhibition of SCAMP1 suppresses cell migration and invasion in human pancreatic and gallbladder cancer cells. Tumour Biol 2013; 34:2731-2739. [PMID: 23653380 DOI: 10.1007/s13277-013-0825-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 04/26/2013] [Indexed: 12/25/2022] Open
Abstract
Lymph node (LN) metastasis is one of the most important risk factors for the prognosis of pancreatic cancer. This study aimed to identify novel LN metastasis-associated markers and therapeutic targets for pancreatic and gallbladder cancers. DNA microarray analysis was carried out to identify genes differentially expressed between 17 pancreatic cancer tissues with LN metastasis and 17 pancreatic cancer tissues without LN metastasis. The expression of LZIC, FXR, SCAMP1, and SULT1E1 is significantly higher in pancreatic cancer tissues with LN metastasis than in pancreatic cancer tissues without LN metastasis. We recently reported that FXR plays an important role in LN metastasis of pancreatic cancer, and in this study, we selected the secretory carrier membrane protein 1 (SCAMP1) gene. To determine that function of the SCAMP1 gene, we examined the effects of SCAMP1 knockdown on pancreatic and gallbladder cancer proliferation, migration, and invasion using SCAMP1 small interfering RNA (siRNA) and the activity of vascular endothelial growth factor (VEGF) was measured by enzyme-linked immunosorbent assay. SCAMP1 overexpression is associated with LN metastasis in pancreatic cancer patients. The siRNA-mediated downregulation of SCAMP1 resulted in a marked reduction in cell migration and invasion, but not proliferation in MIA-PaCa2, PANC-1, TGBC-1, and TGBC-2 cells. In addition, downregulation of SCAMP1 inhibited VEGF levels of conditioned medium from SCAMP1 siRNA-transfected cells. These results suggest that downregulation of SCAMP1 could be a potential therapeutic target for patients with pancreatic and gallbladder cancer.
Collapse
Affiliation(s)
- Sera Yang
- Samsung Biomedical Research Institute, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
19
|
Cao Y, Tu Y, Mei J, Li Z, Jie Z, Xu S, Xu L, Wang S, Xiong Y. RNAi‑mediated knockdown of PRL‑3 inhibits cell invasion and downregulates ERK 1/2 expression in the human gastric cancer cell line, SGC‑7901. Mol Med Rep 2013; 7:1805-11. [PMID: 23589069 DOI: 10.3892/mmr.2013.1420] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 03/26/2013] [Indexed: 12/14/2022] Open
Abstract
The deregulated expression of members of the phophatase of regenerating liver (PRL) family is important in the metastatic progression of multiple human cancers; however, the underlying mechanisms are not well understood. Previous studies have demonstrated that PRLs are able to enhance the activation of extracellular signal‑regulated kinase 1/2 (ERK 1/2) in cancer cells, which may contribute to tumor metastasis. However, the effect of PRL‑3 activation in gastric cancer (GC) remains unclear. The present study aimed to investigate whether the downregulation of PRL‑3 by small interfering RNA (siRNA) was able to inhibit cell motility and affect ERK 1/2 expression in human GC. The results demonstrated that the downregulation of PRL‑3 expression by siRNA in human GC cells significantly inhibited cell invasion and migration in vitro; accordingly, inhibition of PRL‑3 also prevented ERK1/2 protein and mRNA expression, and reduced the mRNA level of matrix metalloproteinase‑7 (MMP‑7), the downstream target of ERK 1/2 signaling. Our data demonstrated that RNAi‑mediated downregulation of PRL‑3 expression leads to potent antitumor activity in human GC. Furthermore, ERK 1/2 and MMP‑7 may contribute to the carcinogenesis and development of human GC in combination with PRL‑3.
Collapse
Affiliation(s)
- Yi Cao
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|