1
|
Xie R, You N, Chen WY, Zhu P, Wang P, Lv YP, Yue GY, Xu XL, Wu JB, Xu JY, Liu SX, Lü MH, Yang SQ, Cheng P, Mao FY, Teng YS, Peng LS, Zhang JY, Liao YL, Yang SM, Zhao YL, Chen W, Zou QM, Zhuang Y. Helicobacter pylori-Induced Angiopoietin-Like 4 Promotes Gastric Bacterial Colonization and Gastritis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0409. [PMID: 39022746 PMCID: PMC11254415 DOI: 10.34133/research.0409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/22/2024] [Indexed: 07/20/2024]
Abstract
Helicobacter pylori infection is characterized as progressive processes of bacterial persistence and chronic gastritis with features of infiltration of mononuclear cells more than granulocytes in gastric mucosa. Angiopoietin-like 4 (ANGPTL4) is considered a double-edged sword in inflammation-associated diseases, but its function and clinical relevance in H. pylori-associated pathology are unknown. Here, we demonstrate both pro-colonization and pro-inflammation roles of ANGPTL4 in H. pylori infection. Increased ANGPTL4 in the infected gastric mucosa was produced from gastric epithelial cells (GECs) synergistically induced by H. pylori and IL-17A in a cagA-dependent manner. Human gastric ANGPTL4 correlated with H. pylori colonization and the severity of gastritis, and mouse ANGPTL4 from non-bone marrow-derived cells promoted bacteria colonization and inflammation. Importantly, H. pylori colonization and inflammation were attenuated in Il17a -/-, Angptl4 -/-, and Il17a -/- Angptl4 -/- mice. Mechanistically, ANGPTL4 bound to integrin αV (ITGAV) on GECs to suppress CXCL1 production by inhibiting ERK, leading to decreased gastric influx of neutrophils, thereby promoting H. pylori colonization; ANGPTL4 also bound to ITGAV on monocytes to promote CCL5 production by activating PI3K-AKT-NF-κB, resulting in increased gastric influx of regulatory CD4+ T cells (Tregs) via CCL5-CCR4-dependent migration. In turn, ANGPTL4 induced Treg proliferation by binding to ITGAV to activate PI3K-AKT-NF-κB, promoting H. pylori-associated gastritis. Overall, we propose a model in which ANGPTL4 collectively ensures H. pylori persistence and promotes gastritis. Efforts to inhibit ANGPTL4-associated pathway may prove valuable strategies in treating H. pylori infection.
Collapse
Affiliation(s)
- Rui Xie
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Nan You
- Department of Hepatobiliary Surgery, XinQiao Hospital,
Third Military Medical University, Chongqing, China
| | - Wan-Yan Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Peng Zhu
- Department of Gastroenterology, Suining First People’s Hospital, Suining, Sichuan, China
| | - Pan Wang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Yi-Pin Lv
- Department of Infection, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Geng-Yu Yue
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xiao-Lin Xu
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jiang-Bo Wu
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jing-Yu Xu
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Si-Xu Liu
- Department of Gastroenterology,
Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Mu-Han Lü
- Department of Gastroenterology,
Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Sheng-Qian Yang
- Chongqing Engineering Research Center for Pharmacodynamics Evaluation, Department of Pharmaceutics, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Fang-Yuan Mao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Yong-Sheng Teng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Liu-Sheng Peng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Jin-Yu Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Ya-Ling Liao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Shi-Ming Yang
- Department of Gastroenterology, XinQiao Hospital,
Third Military Medical University, Chongqing, China
| | - Yong-Liang Zhao
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital,
Third Military Medical University, Chongqing, China
| | - Weisan Chen
- La Trobe Institute of Molecular Science,
La Trobe University, Bundoora, Victoria 3085, Australia
| | - Quan-Ming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Yuan Zhuang
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
- Department of Gastroenterology,
Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| |
Collapse
|
2
|
Liao W, Chen Y, Shan S, Chen Z, Wen Y, Chen W, Zhao C. Marine algae-derived characterized bioactive compounds as therapy for cancer: A review on their classification, mechanism of action, and future perspectives. Phytother Res 2024. [PMID: 38895929 DOI: 10.1002/ptr.8240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 06/21/2024]
Abstract
In 2022, there were around 20 million new cases and over 9.7 million cancer-related deaths worldwide. An increasing number of metabolites with anticancer activity in algae had been isolated and identified, which were promising candidates for cancer therapy. Red algae are well-known for the production of brominated metabolites, including terpenoids and phenols, which have the capacity to induce cell toxicity. Some non-toxic biological macromolecules, including polysaccharides, are distinct secondary metabolites found in many algae, particularly green algae. They possess anticancer activities by inhibiting tumor angiogenesis, stimulating the immune response, and inducing apoptosis. However, the structure-activity relationship between these components and antitumor activity, as well as certain taxa within the algae, remains relatively unstudied. This work is based on the reports published from 2003 to 2024 in PubMed and ISI Web of Science databases. A comprehensive review of the characterized algal anticancer active compounds, together with their structure and mechanism of action was performed. Also, their structure-activity relationship was preliminarily summarized to better assess their potential properties as a natural, safe bioactive product to be used as an alternative for the treatment of cancers, leading to new opportunities for drug discovery.
Collapse
Affiliation(s)
- Wei Liao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yaobin Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Shuo Shan
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
- Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, Ourense, Spain
| | - Zhengxin Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yuxi Wen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
- Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, Ourense, Spain
| | - Weichao Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Chao Zhao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
3
|
Nuñez-Ríos JD, Reyna-Jeldes M, Mata-Martínez E, Campos-Contreras ADR, Lazcano-Sánchez I, González-Gallardo A, Díaz-Muñoz M, Coddou C, Vázquez-Cuevas FG. Extracellular ATP/P2X7 receptor, a regulatory axis of migration in ovarian carcinoma-derived cells. PLoS One 2024; 19:e0304062. [PMID: 38870128 PMCID: PMC11175443 DOI: 10.1371/journal.pone.0304062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/06/2024] [Indexed: 06/15/2024] Open
Abstract
ATP is actively maintained at high concentrations in cancerous tissues, where it promotes a malignant phenotype through P2 receptors. In this study, we first evaluated the effect of extracellular ATP depletion with apyrase in SKOV-3, a cell line derived from metastatic ovarian carcinoma. We observed a decrease in cell migration and an increase in transepithelial electrical resistance and cell markers, suggesting a role in maintaining a mesenchymal phenotype. To identify the P2 receptor that mediated the effects of ATP, we compared the transcript levels of some P2 receptors and found that P2RX7 is three-fold higher in SKOV-3 cells than in a healthy cell line, namely HOSE6-3 (from human ovarian surface epithelium). Through bioinformatic analysis, we identified a higher expression of the P2RX7 transcript in metastatic tissues than in primary tumors; thus, P2X7 seems to be a promising effector for the malignant phenotype. Subsequently, we demonstrated the presence and functionality of the P2X7 receptor in SKOV-3 cells and showed through pharmacological approaches that its activity promotes cell migration and contributes to maintaining a mesenchymal phenotype. P2X7 activation using BzATP increased cell migration and abolished E-cadherin expression. On the other hand, a series of P2X7 receptor antagonists (A438079, BBG and OxATP) decreased cell migration. We used a CRISPR-based knock-out system directed to P2RX7. According to the results of our wound-healing assay, SKOV3-P2X7KO cells lacked receptor-mediated calcium mobilization and decreased migration. Altogether, these data let us propose that P2X7 receptor is a regulator for cancer cell migration and thus a potential drug target.
Collapse
Affiliation(s)
- José David Nuñez-Ríos
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Mauricio Reyna-Jeldes
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
- Núcleo Para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Coquimbo, Chile
| | - Esperanza Mata-Martínez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Anaí del Rocío Campos-Contreras
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Iván Lazcano-Sánchez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Adriana González-Gallardo
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Claudio Coddou
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
- Núcleo Para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Coquimbo, Chile
| | - Francisco G. Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| |
Collapse
|
4
|
Glycoproteins of Capsosiphon fulvescens modulate synaptic clustering of PSD95 and prevent social isolation-induced cognitive decline in aged male rats. J Nutr Biochem 2022; 107:109054. [DOI: 10.1016/j.jnutbio.2022.109054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 03/27/2022] [Accepted: 04/06/2022] [Indexed: 11/18/2022]
|
5
|
Purinergic P2Y2 and P2X4 Receptors Are Involved in the Epithelial-Mesenchymal Transition and Metastatic Potential of Gastric Cancer Derived Cell Lines. Pharmaceutics 2021; 13:pharmaceutics13081234. [PMID: 34452195 PMCID: PMC8398939 DOI: 10.3390/pharmaceutics13081234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC) is a major health concern worldwide, presenting a complex pathophysiology that has hindered many therapeutic efforts so far. In this context, purinergic signaling emerges as a promising pathway for intervention due to its known role in cancer cell proliferation and migration. In this work, we explored in more detail the role of purinergic signaling in GC with several experimental approaches. First, we measured extracellular ATP concentrations on GC-derived cell lines (AGS, MKN-45, and MKN-74), finding higher levels of extracellular ATP than those obtained for the non-tumoral gastric cell line GES-1. Next, we established the P2Y2 and P2X4 receptors (P2Y2R and P2X4R) expression profile on these cells and evaluated their role on cell proliferation and migration after applying overexpression and knockdown strategies. In general, a P2Y2R overexpression and P2X4R downregulation pattern were observed on GC cell lines, and when these patterns were modified, concomitant changes in cell viability were observed. These modifications on gene expression also modified transepithelial electrical resistance (TEER), showing that higher P2Y2R levels decreased TEER, and high P2X4R expression had the opposite effect, suggesting that P2Y2R and P2X4R activation could promote and suppress epithelial-mesenchymal transition (EMT), respectively. These effects were confirmed after treating AGS cells with UTP, a P2Y2R-agonist that modified the expression patterns towards mesenchymal markers. To further characterize the effects of P2Y2R activation on EMT, we used cDNA microarrays and observed that UTP induced important transcriptional changes on several cell processes like cell proliferation induction, apoptosis inhibition, cell differentiation induction, and cell adhesion reduction. These results suggest that purinergic signaling plays a complex role in GC pathophysiology, and changes in purinergic balance can trigger tumorigenesis in non-tumoral gastric cells.
Collapse
|
6
|
Ulagesan S, Nam TJ, Choi YH. Cytotoxicity against human breast carcinoma cells of silver nanoparticles biosynthesized using Capsosiphon fulvescens extract. Bioprocess Biosyst Eng 2021; 44:901-911. [PMID: 33486577 DOI: 10.1007/s00449-020-02498-z] [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: 10/09/2020] [Accepted: 12/10/2020] [Indexed: 10/22/2022]
Abstract
Targeting cancer cells with small nanoparticles is a novel and promising approach to cancer therapy. Breast cancer is the most common cancer afflicting women worldwide. In the present study, silver nanoparticles (AgNPs) were synthesized using the aqueous extract of the marine alga Capsosiphon (C.) fulvescens, and the cytotoxicity and anti-cancer activities of the nanoparticles against MCF-7 breast cancer cells were analyzed. Nanoparticle formation was confirmed by solution color change and UV-Vis spectroscopy. The size and distribution of the C. fulvescens-biosynthesized silver nanoparticles (CfAgNPs) were then examined using various analytical methods; the particle size was around 20-22 nm and spherical in shape with no agglomeration. Cytotoxicity analysis revealed that the inhibitory concentration (IC50) of CfAgNPs was 50 μg/ml. MCF-7 cell viability decreased with increasing concentrations of CfAgNPs. MCF-7 cells were evaluated for morphological changes before and after treatment with the CfAgNPs; cells treated with C. fulvescens aqueous algal extract (without CfAgNPs) showed irregular confluent aggregates with round polygonal cells, similar to the untreated control. Tamoxifen- (TMX) and CfAgNPs-treated cells show significant morphological changes. An apoptosis study was conducted using 4',6-diamidino-2-phenylindole (DAPI) staining, in which CfAgNP-treated MCF-7 cells generated bright blue fluorescence and shortened, disjointed chromatin was evident; control cells displayed less bright fluorescence. Flow cytometry analysis revealed that the percentage of cells in late apoptosis was high following treatment with TMX (77.2%) and CfAgNP (74.6%). A novel anti-cancer agent, developed by generating silver nanoparticles from C. fulvescens extract, showed strong cytotoxic activity against MCF-7 cells.
Collapse
Affiliation(s)
- Selvakumari Ulagesan
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041, Republic of Korea
| | - Taek-Jeong Nam
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041, Republic of Korea.
| | - Youn-Hee Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan, 46041, Republic of Korea.
- Department of Marine Bio-Materials and Aquaculture, Pukyong National University, 45, Yongso-ro, Nam-Gu, Busan, 48513, Republic of Korea.
| |
Collapse
|
7
|
Activation of BDNF-mediated PKA signaling in the ventral hippocampus by Capsosiphon fulvescens glycoproteins alleviates depressive-like behavior in aged rats. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
8
|
Oh JH, Nam TJ, Choi YH. Capsosiphon fulvescens Glycoproteins Enhance Probiotics-Induced Cognitive Improvement in Aged Rats. Nutrients 2020; 12:E837. [PMID: 32245093 PMCID: PMC7146536 DOI: 10.3390/nu12030837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 03/18/2020] [Accepted: 03/18/2020] [Indexed: 12/23/2022] Open
Abstract
Aging-induced cognitive dysfunction can be regulated by probiotics through bidirectional communication with the brain. This study aimed to investigate whether Capsosiphon fulvescens glycoproteins (Cf-hGP) enhanced probiotic-induced improvement of memory in aged rats and the underlying mechanism in the dorsal hippocampus. Cf-hGP were isolated using lectin resin. Cf-hGP (15 mg/kg/day) and/or Lactobacillus plantarum (L. plantarum) (109 CFU/rat/day) were orally administered once a day for 4 weeks. Co-treatment with Cf-hGP and L. plantarum synergistically improved spatial memory in aged rats, which was overturned by functional blocks of brain-derived neurotrophic factor (BDNF) signaling. Increases in BDNF expression and nuclear factor erythroid 2-related factor 2 (Nrf2) phosphorylation were accompanied by mono- and/or co-administration in the dorsal hippocampus, while c-Jun N-terminal kinase (JNK) phosphorylation and glucose-regulated protein 78 expression were decreased. These synergistic effects were downregulated by blocks of BDNF/Nrf2-mediated signaling. In particular, co-treatment, not mono-treatment, reduced phosphorylation of eukaryotic elongation factor 2 (eEF2) regulated by eEF2 kinase and protein phosphatase 2A. Additionally, co-treatment downregulated the interaction between eEF2 kinase and JNK. These data demonstrated that cognitive impairment in aged rats was synergistically diminished by co-treatment with Cf-hGP and L. plantarum through BDNF-mediated regulation of Nrf2 and eEF2 signaling pathways in the dorsal hippocampus.
Collapse
Affiliation(s)
- Jeong Hwan Oh
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Korea; (J.H.O.); (T.-J.N.)
| | - Taek-Jeong Nam
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Korea; (J.H.O.); (T.-J.N.)
| | - Youn Hee Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Korea; (J.H.O.); (T.-J.N.)
- Department of Marine Bio-Materials & Aquaculture, Pukyong National University, Busan 48513, Korea
| |
Collapse
|
9
|
Cellulose nanocrystals/nanofibrils loaded astaxanthin nanoemulsion for the induction of apoptosis via ROS-dependent mitochondrial dysfunction in cancer cells under photobiomodulation. Int J Biol Macromol 2020; 149:165-177. [PMID: 31987944 DOI: 10.1016/j.ijbiomac.2020.01.243] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/13/2020] [Accepted: 01/24/2020] [Indexed: 12/29/2022]
Abstract
The present study investigated effects of low-level laser therapy with cellulose nanocrystals/cellulose nanofibrils loaded in nanoemulsion (NE) against skin cancer cells on apoptosis. The nanoemulsion was fabricated and characterized by the standard methods. The toxicity level by cytotoxicity assays, generation of reactive singlet oxygen (ROS) and antioxidant potential, cell proliferation and migration were confirmed by using standard assays. The cellular uptake efficacy was evaluated by differential staining. The protein levels of EGFR, PI3K, AKT, ERK, GAPDH, and β-actin were detected by western blot. The samples showed a spherical shaped structure with the average size confirmed strong and stable hydrogen bonding forces with high degradation temperature and endothermic transition peaks. The fabricated samples showed no toxicity and high cell proliferation by generating more singlet oxygen levels and antioxidants. The intracellular signaling pathways was regulated with high protein expression levels, which was stimulated by specific molecules for cell proliferation, migration, and differentiation in cancer cells. The results proved that combined treatment regulated the intracellular signaling pathways in cancer cells. The current study showed a novel strategy for improving therapeutic efficacy of nanoemulsion by using low-level laser therapy. Further, the current favorable outcomes will be evaluated in in vivo animal models.
Collapse
|
10
|
Anticancer Effect of the Ethyl Acetate Fraction from Orostachys japonicus on MDA-MB-231 Human Breast Cancer Cells through Extensive Induction of Apoptosis, Cell Cycle Arrest, and Antimetastasis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:8951510. [PMID: 31781282 PMCID: PMC6874866 DOI: 10.1155/2019/8951510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/05/2019] [Accepted: 09/18/2019] [Indexed: 01/24/2023]
Abstract
The antibreast cancer activities of the ethyl acetate fraction from Orostachys japonicus (OJEF) were investigated in MDA-MB-231 human breast cancer cells through WST assay, DAPI staining, flow cytometry analysis, and western blotting. OJEF effectively inhibited MDA-MB-231 cells by inducing apoptosis via intrinsic, extrinsic, and endoplasmic reticulum (ER) stress response pathways, cell cycle arrest at the G1/S phase, and antimetastasis including inhibition of tight junction, adherens junction, invasion, and migration. The MAPK family-mediated upstream signal transduction through p-p38 and p-ERK was considered to affect the downstream signal transduction including induction of apoptosis, cell cycle arrest, and antimetastasis. In conclusion, we executed an integrated study on the anticancer activities of OJEF, which extensively induced apoptosis, cell cycle arrest, and antimetastasis in estrogen-independent MDA-MB-231 human breast cancer cells known to be liable to metastasize.
Collapse
|
11
|
Oh JH, Nam TJ. Hydrophilic Glycoproteins of an Edible Green Alga Capsosiphon fulvescens Prevent Aging-Induced Spatial Memory Impairment by Suppressing GSK-3β-Mediated ER Stress in Dorsal Hippocampus. Mar Drugs 2019; 17:E168. [PMID: 30875947 PMCID: PMC6470841 DOI: 10.3390/md17030168] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 02/27/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is involved in various neurodegenerative disorders. We previously found that Capsosiphon fulvescens (C. fulvescens) crude proteins enhance spatial memory by increasing the expression of brain-derived neurotrophic factor (BDNF) in rat dorsal hippocampus. The present study investigated whether the chronic oral administration of hydrophilic C. fulvescens glycoproteins (Cf-hGP) reduces aging-induced cognitive dysfunction by regulating ER stress in the dorsal hippocampus. The oral administration of Cf-hGP (15 mg/kg/day) for four weeks attenuated the aging-induced increase in ER stress response protein glucose-regulated protein 78 (GRP78) in the synaptosome of the dorsal hippocampus; this was attenuated by the function-blocking anti-BDNF antibody (1 μg/μL) and a matrix metallopeptidase 9 inhibitor 1 (5 μM). Aging-induced GRP78 expression was associated with glycogen synthase kinase-3 beta (GSK-3β) (Tyr216)-mediated c-Jun N-terminal kinase phosphorylation, which was downregulated upon Cf-hGP administration. The Cf-hGP-induced increase in GSK-3β (Ser9) phosphorylation was downregulated by inhibiting tyrosine receptor kinase B and extracellular signal-regulated kinase (ERK)1/2 with cyclotraxin-B (200 nM) and SL327 (10 μM), respectively. Cf-hGP administration or the inhibition of ER stress with salubrinal (1 mg/kg, i.p.) significantly decreased aging-induced spatial memory impairment. These findings suggest that the activation of the synaptosomal BDNF-ERK1/2 signaling in the dorsal hippocampus by Cf-hGP attenuates age-dependent ER stress-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Jeong Hwan Oh
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Korea.
| | - Taek-Jeong Nam
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Korea.
| |
Collapse
|
12
|
Hydrophilic compartments of Capsosiphon fulvescens protein alleviate impaired spatial memory by regulating BDNF-mediated ER stress against chronic ethanol exposure. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
13
|
Lin CL, Hsieh SL, Leung W, Jeng JH, Huang GC, Lee CT, Wu CC. 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucoside suppresses human colorectal cancer cell metastasis through inhibiting NF-κB activation. Int J Oncol 2016; 49:629-38. [PMID: 27278328 DOI: 10.3892/ijo.2016.3574] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 06/03/2016] [Indexed: 11/05/2022] Open
Abstract
2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucoside (THSG), a major component of Polygonum multiflorum Thunb (He-Shou-Wu), has been reported to exhibit antioxidant and anti-inflammatory effects. However, its anti-metastatic effect against colorectal cancer is still unclear. In this study, cell migration, invasion and adhesion abilities as well as metastasis-associated protein and NF-κB pathway signaling factor expression were analyzed after treating HT-29 cells with THSG. According to the results, the migration and invasiveness of HT-29 cells were reduced after treatment with 5 or 10 mM THSG (p<0.05). Additionally, the levels of matrix metalloproteinase-2 (MMP-2) and phosphorylated VE-cadherin in HT-29 cells were reduced and the transepithelial electrical resistance (TEER) of EA.hy926 endothelial cell monolayers was increased after incubation in THSG for 24 h (p<0.05). Cell adhesion ability and the E-selectin and intercellular adhesion molecule-1 (ICAM-1) protein levels were reduced when EA.hy926 endothelial cells were treated with THSG (p<0.05). In addition, the cytoplasmic phosphorylation of IκB, the nuclear p65 level and the DNA-binding activity of NF-κB were reduced after treating HT-29 or EA.hy926 cells with 5 or 10 mM THSG (p<0.05). These results suggest that THSG inhibits HT-29 cell metastasis by suppressing cell migration, invasion and adhesion. Furthermore, THSG inhibits metastasis-associated protein expression by suppressing NF-κB pathway activation.
Collapse
Affiliation(s)
- Chien-Liang Lin
- Department of Pharmacy, Yuan's General Hospital, Kaohsiung 802, Taiwan, R.O.C
| | - Shu-Ling Hsieh
- Department of Seafood Science, National Kaohsiung Marine University, Kaohsiung 811, Taiwan, R.O.C
| | - Wan Leung
- Department of Radiology and Nuclear Medicine, Yuan's General Hospital, Kaohsiung 802, Taiwan, R.O.C
| | - Jiiang-Huei Jeng
- Department of Dentistry, National Taiwan University Hospital, Taipei 100, Taiwan, R.O.C
| | - Guan-Cheng Huang
- Department of Health-Business Administration, School of Nursing, Fooyin University, Kaohsiung 831, Taiwan, R.O.C
| | - Chining-Ting Lee
- Department of Seafood Science, National Kaohsiung Marine University, Kaohsiung 811, Taiwan, R.O.C
| | - Chih-Chung Wu
- Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan 711, Taiwan, R.O.C
| |
Collapse
|
14
|
Wu H, Wang W, Tong S, Wu C. Nucleostemin regulates proliferation and migration of gastric cancer and correlates with its malignancy. Int J Clin Exp Med 2015; 8:17634-17643. [PMID: 26770353 PMCID: PMC4694253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/15/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the effects of nucleostemin (NS) knocking down in SGC- 7901 gastric cancer cell line and investigates its correlation with the metastasis and TNM stage ingastric cancer (GC) patients. METHODS NS expression was assessed using immunohistochemistry in 421 patients with GC. The correlation between NS expression, clinicopathological features and prognosis was analyzed. NS gene silencing was performed using a specific small interfering RNA (NS-siRNA). The gene expression level of NS was evaluated by PCR. The viability and growth rate of SGC-7901 cells were determined by trypan blue exclusion test. Cell cycle distribution of the cells was analyzed by flow cytometry. RESULTS High NS expression was correlated with node metastasis, distant metastasis and TNM stage. Kaplan-Meier survival analysis revealed that patients with low NS expression had significantly longer survival than those with high NS expression. Moreover, our results showed that NS knocking down inhibited proliferation and viability of SGC-7901 cells in a time-dependent manner. Cell cycle studies revealed that NS depletion resulted in G1 cell cycle arrest at short times of transfection (24 h) followed with apoptosis at longer times (48 and 72 h), suggest that post-G1 arrest apoptosis is occurred in SGC-7901 cells. CONCLUSION Overall, these results point to essential role of NS in SGC-7901 cells, thus, this gene might be considered as a promising target for treatment of GC.
Collapse
Affiliation(s)
- Hongxue Wu
- Department of Gastrointestinal, Renmin Hospital of Wuhan University Wuhan, Hubei Province, P. R. China
| | - Weixing Wang
- Department of Gastrointestinal, Renmin Hospital of Wuhan University Wuhan, Hubei Province, P. R. China
| | - Shilun Tong
- Department of Gastrointestinal, Renmin Hospital of Wuhan University Wuhan, Hubei Province, P. R. China
| | - Chong Wu
- Department of Gastrointestinal, Renmin Hospital of Wuhan University Wuhan, Hubei Province, P. R. China
| |
Collapse
|
15
|
Yang Z, Wang R, Zhang T, Dong X. MicroRNA-126 regulates migration and invasion of gastric cancer by targeting CADM1. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:8869-8880. [PMID: 26464628 PMCID: PMC4583860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 07/29/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE The aberrant expression of microRNAs has been demonstrated to play a crucial role in the initiation and progression of gastric cancer (GC). We here aimed to investigate the mechanism of microRNAs in the regulation of GC pathogenesis. METHODS Transwell chambers (8-μM pore size; Costar) were used in the in vitro migration and in vision assay. Dual luciferase reporter gene construct and dual luciferase reporter assay to identify the target of miR-126. CADM1 expression was evaluated by immunohistochemical staining. The clinical manifestations, treatments and survival were collected for statistical analysis. RESULTS Inhibition of miR-126 effectively reduced migration and invasion of gastric cancer cell lines. Bioinformatics and luciferase reporter assay revealed that miR-126 specifically targeted the 3'UTR of cell adhesion molecule 1 (CADM1) and regulated its expression. Down-regulation of CADM1 enhanced migration and invasion of GC cell lines. Furthermore, in tumor tissues obtained from gastric cancer patients, the expression of miR-126 was negatively correlated with CADM1 and the high expression of miR-126 combined with low expression of CADM1 might serve as a risk factor for stage1 gastric cancer patients. CONCLUSIONS Our study showed that miR-126, by down-regulation CADM1, enhances migration and invasion in GC cells.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Zhengzhou UniversityChina
| | - Ruoming Wang
- Department of General Surgery, First Renmin Hospital of ShangqiuHenan Province, China
| | - Tengteng Zhang
- Department of Cancer, First Renmin Hospital of ShangqiuHenan Province, China
| | - Xinhua Dong
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Zhengzhou UniversityChina
| |
Collapse
|
16
|
Kim EY, Choi YH, Lee JI, Kim IH, Nam TJ. Antioxidant Activity of Oxygen Evolving Enhancer Protein 1 Purified from Capsosiphon fulvescens. J Food Sci 2015; 80:H1412-7. [PMID: 25944160 DOI: 10.1111/1750-3841.12883] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 03/18/2015] [Indexed: 12/17/2022]
Abstract
This study was conducted to determine the antioxidant activity of a protein purified from Capsosiphon fulvescens. The purification steps included sodium acetate (pH 6) extraction and diethylaminoethyl-cellulose, reversed phase Shodex C4P-50 column chromatography. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis indicated that the molecular weight of the purified protein was 33 kDa. The N-terminus and partial peptide amino acid sequence of this protein was identical to the sequence of oxygen evolving enhancer (OEE) 1 protein. The antioxidant activity of the OEE 1 was determined in vitro using a scavenging test with 4 types of reactive oxygen species (ROS), including the 2,2-diphenyl-1-picrylhydrazyl radical, hydroxyl radical, superoxide anion, and hydrogen peroxide (H2 O2 ). OEE 1 had higher H2 O2 scavenging activity, which proved to be the result of enzymatic antioxidants rather than nonenzymatic antioxidants. In addition, OEE 1 showed less H2 O2 -mediated ROS formation in HepG2 cells. In conclusion, this study demonstrates that OEE 1 purified from C. fulvescens is an excellent antioxidant.
Collapse
Affiliation(s)
- Eun-Young Kim
- Inst. of Fisheries Sciences, Pukyong Natl. Univ., Ilgwang-ro, Ilgwang-myeon, Gijang-gun, Busan, 619-911, Republic of Korea
| | - Youn Hee Choi
- Inst. of Fisheries Sciences, Pukyong Natl. Univ., Ilgwang-ro, Ilgwang-myeon, Gijang-gun, Busan, 619-911, Republic of Korea
| | - Jung Im Lee
- Inst. of Fisheries Sciences, Pukyong Natl. Univ., Ilgwang-ro, Ilgwang-myeon, Gijang-gun, Busan, 619-911, Republic of Korea
| | - In-Hye Kim
- Inst. of Fisheries Sciences, Pukyong Natl. Univ., Ilgwang-ro, Ilgwang-myeon, Gijang-gun, Busan, 619-911, Republic of Korea
| | - Taek-Jeong Nam
- Inst. of Fisheries Sciences, Pukyong Natl. Univ., Ilgwang-ro, Ilgwang-myeon, Gijang-gun, Busan, 619-911, Republic of Korea
| |
Collapse
|
17
|
Chen F, Tian Y, Pang EJ, Wang Y, Li L. MALAT2-activated long noncoding RNA indicates a biomarker of poor prognosis in gastric cancer. Cancer Gene Ther 2015:cgt20156. [PMID: 25721209 DOI: 10.1038/cgt.2015.6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 11/15/2014] [Accepted: 11/17/2014] [Indexed: 12/12/2022]
Abstract
The purpose of this study is to investigate the role of the long noncoding RNA metastasis associated lung adenocarcinoma transcript 2 (MALAT2) in the prognosis of stage II/III gastric cancer (GC) patients. The expression of MALAT2 was evaluated in cancer tissues from 146 stage II/III GC patients undergoing radical resection and 60 paired normal samples using quantitative real-time reverse transcriptase PCR. Differences in the expression of MALAT2 between 23 GC and paired normal colonic mucosa samples were analysed with the Wilcoxon test. Relationships between the expression level of MALAT2, patient clinico-pathological parameters and disease-free survival and overall survival were analysed using the uni-variate Kaplan-Meier method and the multivariate COX regression model. The quantitative polymerase chain reaction results showed that MALAT2 was frequently over-expressed in cancer tissues and this over-expression was found to significantly correlate with lymph node metastasis and tumor stage. The ectopic expression of MALAT2 contributed to the migration of human GC SGC-7901 cells, whereas knockdown of MALAT2 inhibited the migration of the SGC-7901 cells in vitro. Further investigation into the underlying mechanisms responsible for the migratory effects revealed that MALAT2 induced the epithelial-mesenchymal transition (EMT) through an MEK/extracellular signal-regulated kinase-dependent mechanism as treatment with the MEK inhibitor, U0126, decreased migration and reversed the EMT in the MALAT2 over-expressed SGC-7901 cells. The expression of MALAT2 is upregulated in GC tissues, and a higher expression level of MALAT2 might serve as a negative prognostic marker in stage II/III GC patients which implies the potential application of MALAT2 in the therapeutic treatment of GC.Cancer Gene Therapy advance online publication, 27 February 2015; doi:10.1038/cgt.2015.6.
Collapse
Affiliation(s)
- F Chen
- Division of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Y Tian
- Division of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - E-J Pang
- Division of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Y Wang
- Division of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - L Li
- Division of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
18
|
Shan YQ, Ying RC, Zhou CH, Zhu AK, Ye J, Zhu W, Ju TF, Jin HC. MMP-9 is increased in the pathogenesis of gastric cancer by the mediation of HER2. Cancer Gene Ther 2015; 22:101-7. [PMID: 25633484 DOI: 10.1038/cgt.2014.61] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 08/28/2014] [Accepted: 08/30/2014] [Indexed: 12/12/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression is not only closely associated with the tumor growth, but is also related to tumor invasion. We here aimed to investigate the mechanism of HER2 mediation in the pathogenesis of gastric cancer. The human gastric cancer cell lines SGC-7901, MKN-45, AGS, the immortalized cell line GES-1 derived from normal gastric mucosa. Cell transfection and selection of stable cell lines and the gene and protein levels of HER2 and Matrix metalloproteinase-9 (MMP-9) were examined to determine the molecular relationship between them in the pathogenesis of gastric cancer. The human gastric cancer cell lines SGC-7901, MKN-45, AGS, the immortalized cell line GES-1 derived from normal gastric mucosa. Cell transfection and selection of stable cell lines and the gene and protein levels of HER2 and MMP-9 were examined to determine the molecular relationship between them in the pathogenesis of gastric cancer. We demonstrated that vector-based shRNA significantly knocked down the expression of HER2 and considerably inhibited both the migration and invasion of gastric cancer cells. HER2 knockdown resulted in the downregulation of the expression of MMP-9, whereas HER2 overexpression improved the transcription of MMP-9 through the activation of an MMP-9 promoter. The promoter region of MMP-9 between -2500 and -2000 bp was found to be crucial for the upregulation of HER2-mediated transcription. Furthermore, a truncated promoter (-70 to +63) did not display any transcriptional activity. Cell invasion activity was almost completely inhibited when MMP-9 was knocked down. Conversely, the overexpression of MMP-9 partly rescued the invasion ability of cell strains with knockdown HER2. These findings help further understanding of the molecular mechanisms through which HER2 promotes malignancy, and suggest that targeting both HER2 and MMP-9 may be required to effectively block HER2 signaling in gastric cancer therapy.
Collapse
Affiliation(s)
- Y-Q Shan
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - R-C Ying
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - C-H Zhou
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - A-K Zhu
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - J Ye
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - W Zhu
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - T-F Ju
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| | - H-C Jin
- 1] Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, China [2] Affiliated Hangzhou Hospital, Nanjing Medical University, Hangzhou, China
| |
Collapse
|
19
|
Fu YF, Gui R, Liu J. HER-2-induced PI3K signaling pathway was involved in the pathogenesis of gastric cancer. Cancer Gene Ther 2015; 22:145-53. [PMID: 25613482 DOI: 10.1038/cgt.2014.80] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/09/2014] [Accepted: 12/09/2014] [Indexed: 01/02/2023]
Abstract
Human epidermal growth factor receptor-2 (HER-2) overexpression was closely associated with the tumor growth and invasion, we here aimed to investigate the mechanism of HER-2 mediation in the pathogenesis of gastric cancer (GC). We first detected the expression of HER-2 in GC cell line SGC-7901 and then examined the levels of nuclear factor-κB (NF-κB), matrix metalloproteinase-9 (MMP-9) and intercellular adhesion molecule-1 (ICAM-1) and the association between them by molecular methods. Statistical analysis was used to compare the significance. We further detected the possible molecular mechanism involved in their relationship in the SGC-7901 genesis. The MMP-9, NF-κB and secretory type (s-ICAM-1) levels were significantly greater in peripheral blood serum from SGC-7901 than healthy control GES-1 (P<0.01). ICAM-1, MMP-9 and NF-κB mRNA and protein levels were more highly expressed in SGC-7901 than healthy control GES-1. The expression levels of NF-κB, MMP-9 and ICAM-1 were positively related in GC cell line SGC-7901, which was HER-2 positive. The HER-2 positive SGC-7901 secreted more transforming growth factor beta 1 (TGF-β1) and resultantly activated MMP-9 to enhance s-ICAM-1 secretion and further studies showed that phosphatidylinositol-3 kinase (PI3K)/Akt/NF-κB signaling pathway was involved in GC pathogenesis. The GC cells that express the HER-2 oncogene spur the activation of NF-κB that can upregulate the expression of ICAM-1 and induce the expression of MMP-9, which hydrolyzes ICAM-1 into s-ICAM-1 to promote tumor immune escape. TGF-β1-induced PI3K/Akt/NF-κB signaling pathway was involved in the pathogenesis of GC and they could be a new target for cancer therapy. The GC cells that express the HER-2 oncogene spur the activation of NF-κB that can upregulate the expression of ICAM-1 and induce the expression of MMP-9, which hydrolyzes ICAM-1 into s-ICAM-1 to promote tumor immune escape. TGF-β1-induced PI3K/Akt/NF-κB signaling pathway was involved in the pathogenesis of GC and they could be a new target for cancer therapy.
Collapse
Affiliation(s)
- Y F Fu
- The Third Xiang-Ya Hospital, Central South University, Changsha, China
| | - R Gui
- The Third Xiang-Ya Hospital, Central South University, Changsha, China
| | - J Liu
- The Third Xiang-Ya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
MicroRNA let-7b suppresses human gastric cancer malignancy by targeting ING1. Cancer Gene Ther 2015; 22:122-9. [PMID: 25613480 DOI: 10.1038/cgt.2014.75] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 11/23/2014] [Accepted: 11/24/2014] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are important regulators that play key roles in tumorigenesis and tumor progression. In this study, we investigate whether let-7b acts as a tumor suppressor to inhibit invasion and metastasis in gastric cancers. We analyzed the expression of let-7b in 60 pair-matched gastric neoplastic and adjacent non-neoplastic tissues by quantitative real-time polymerase chain reaction. Functional analysis of let-7b expression was assessed in vitro in gastric cancer cell lines with let-7b precursor and inhibitor. The roles of let-7b in tumorigenesis and tumor metastasis were analyzed using a stable let-7b expression plasmid in nude mice. A luciferase reporter assay was used to assess the effect of let-7b on inhibitor of growth family, member 1 (ING1) expression. Real-time PCR showed decreased levels of let-7b expression in metastatic gastric cancer tissues and cell lines that are potentially highly metastatic. Cell invasion and migration were significantly impaired in GC9811-P and SGC7901-M cell lines after transfection with let-7b mimics. Nude mice with xenograft models of gastric cancer confirmed that let-7b could inhibit gastric cancer metastasis in vivo after transfection by the lentivirus pGCsil-GFP- let-7b. Luciferase reporter assays demonstrated that let-7b directly binds to the 3'-UTR of ING1, and real-time PCR and western blotting further indicated that let-7b downregulated the expression of ING1 at the mRNA and protein levels. Our study demonstrates that overexpression of let-7b in gastric cancer can inhibit invasion and migration of gastric cancer cells through directly targeting the tumor metastasis-associated gene ING1. These findings help clarify the molecular mechanisms involved in gastric cancer metastasis and indicate that let-7b modulation may be a bona fide treatment of gastric cancer.
Collapse
|
21
|
Peng J, Chen X, Hu Q, Yang M, Liu H, Wei W, Liu S, Wang H. 1‑calcium phosphate‑uracil, a synthesized pyrimidine derivative agent, has anti‑proliferative, pro‑apoptotic and anti‑invasion effects on multiple tumor cell lines. Mol Med Rep 2014; 10:2271-8. [PMID: 25118659 PMCID: PMC4214340 DOI: 10.3892/mmr.2014.2489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 06/17/2014] [Indexed: 01/01/2023] Open
Abstract
1-calcium phosphate-uracil (1-CP-U), a synthetic pyrimidine derivative, has been documented to demonstrate a variety of different biological activities. However, the potency and mechanisms of this agent’s anti-cancer activity have not been elucidated to date. In the present study, the anti-cancer effects of 1-CP-U were examined in a range of in vitro assays. Different cell lines were treated with 1-CP-U at varied concentrations (0.7, 1.0, 1.4 μmol/l) for indicated durations. The cell proliferation was then examined by MTT assay. The cellular apoptotic effects were detected by Hoechst 33342 and Annexin V/propidium iodide staining, while the capacity of 1-CP-U on invasion and migration were examined by cell invasion and wound healing assays. The expression of matrix metalloproteinase proteins, as well as pro- and antiapoptotic proteins was detected by western blotting analysis. The results identified that 1-CP-U was able to inhibit the viability of SKOV3, HeLa, SMMC-7721 and A549 cell lines in a dose- and time-dependent manner, while it exerted only marginal toxic effects on non-cancerous cells. The IC50 concentration of 1-CP-U for tumor cell lines was ~1.0 μmol/l. The growth inhibition induced by 1-CP-U was accompanied by a broad spectrum of pro-apoptotic activities, in which different cell lines varied in their sensitivity to 1-CP-U. Meanwhile, the increased expression of the pro-apoptotic protein B-cell lymphoma-2 (Bcl-2)-associated X and a marked reduction of Bcl-2 levels were associated with increased 1-CP-U concentrations. Additionally, anti-migration and anti-invasion effects of 1-CP-U were evidently associated with the downregulation of matrix metalloproteinase proteins. Of note, it was observed that 1-CP-U significantly inhibited both the migration and invasion at a lower concentration, as compared with the dose required to achieve significant inhibition of apoptosis. These results indicated that 1-CP-U appeared to be a more effective inhibitor of cell migration and invasion, rather than of apoptosis. In conclusion, the present study was the first, to the best of our knowledge, to demonstrate the function of 1-CP-U in tumor proliferation, apoptosis and invasion with specific effects against cancer cells in vitro, suggesting 1-CP-U as a potential novel anticancer agent.
Collapse
Affiliation(s)
- Jing Peng
- Laboratory of Genetics, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xinlian Chen
- Laboratory of Genetics, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qian Hu
- Laboratory of Genetics, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Mei Yang
- Laboratory of Genetics, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hongqian Liu
- Laboratory of Genetics, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Wei Wei
- Laboratory of Genetics, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shanling Liu
- Laboratory of Cell and Gene Therapy, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - He Wang
- Laboratory of Genetics, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
22
|
Kim YM, Kim IH, Nam TJ. Capsosiphon fulvescens glycoprotein reduces AGS gastric cancer cell migration by downregulating transforming growth factor-β1 and integrin expression. Int J Oncol 2013; 43:1059-65. [PMID: 23934170 PMCID: PMC3829798 DOI: 10.3892/ijo.2013.2055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/19/2013] [Indexed: 12/28/2022] Open
Abstract
Seaweeds are commonly used as functional foods and drugs. A glycoprotein (GP) from the green alga Capsosiphon fulvescens (Cf) has been reported to have antitumor activity toward various cancer cells. We previously observed that Cf-GP induced different pathways of apoptosis in AGS human gastric cancer cells. Transforming growth factor (TGF)-β1 plays an important role in cancer cell migration. Increased TGF-β1 levels increase the expression of the small GTPases and activate the FAK/PI3K/AKT pathways, resulting in the upregulation of integrin receptor proteins, which mediate the attachment of cells to surrounding tissues, cells or extracellular matrix. Thus, the inhibition of TGF-β1 signaling would downregulate integrin expression and thereby effectively decrease cell growth and migration. In the present study, we determined the effect of Cf-GP treatment on the proliferation, migration and apoptosis of AGS human gastric cancer cells. To investigate the mechanism by which Cf-GP exerts its anticancer actions, we examined the effect of Cf-GP on the expression levels of TGF-β1, FAK, PI3K, AKT, the small GTPases and integrins in AGS cells. Our findings indicate that Cf-GP inhibits AGS cell proliferation and migration by downregulating integrin expression via the TGF-β1-activated FAK/PI3K/AKT pathways. These results suggest that Cf-GP may be an important factor in the development of functional foods and therapeutic agents.
Collapse
Affiliation(s)
- Young-Min Kim
- Institute of Fisheries Sciences, Pukyong National University, Gijang-gun, Busan 619-911, Republic of Korea
| | | | | |
Collapse
|