1
|
Wang Y, Ge H, Zhang Y, Wang P, Zhao H, Wang L, Fan Z. Antitumor effect of polyphyllin I (PPI) on colorectal cancer: Evidence from patient-derived organoids and Notch signaling suppression. Heliyon 2024; 10:e37226. [PMID: 39315206 PMCID: PMC11417558 DOI: 10.1016/j.heliyon.2024.e37226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Colorectal cancer (CRC) is a malignant tumor with a high incidence, ranking first among gastrointestinal malignancies. We investigated the impact of polyphyllin I (PPI), a natural compound found in Paris polyphylla, on CRC. PPI has been documented to exhibit anticancer activity against various tumors. This study aimed to assess the effects of PPI on colorectal cancer and explore its potential mechanisms. Our research demonstrated that PPI inhibited proliferation, promoted apoptosis, and induced G2 cell-cycle arrest in a dose-dependent manner. Additionally, our results indicated that PPI suppressed Notch signaling by downregulating the Notch1 receptor, its ligand Jagged1, and the downstream target Hes1 expression. Furthermore, we confirmed the antitumor effect of PPI on patient-derived organoids. In conclusion, our study indicates that PPI impedes the growth of colon cancer by suppressing the Notch signaling pathway.
Collapse
Affiliation(s)
- Yu Wang
- Department of Anorectal Medicine, Liyang Hospital of Chinese Medicine, Changzhou, 213300, China
| | - Hao Ge
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210038, China
| | - Yi Zhang
- College of Life Sciences, Nanjing Normal University, Nanjing, 210023, Jiangsu, China
| | - Pei Wang
- Jiangsu Clinical Innovation Center for Anorectal Diseases of T.C.M., Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China
| | - Haoran Zhao
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210038, China
| | - Lu Wang
- Jiangsu Clinical Innovation Center for Anorectal Diseases of T.C.M., Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China
| | - Zhimin Fan
- Department of Anorectal Medicine, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210022, China
| |
Collapse
|
2
|
Zha X, Li Y, Zhao H, Tan Y, Zhou S. Pyrrolidine, Piperazine, and Diazinane Alkaloids from the Marine Bacterium Strain Vibrio ruber ZXR-93. Molecules 2024; 29:4446. [PMID: 39339441 PMCID: PMC11433930 DOI: 10.3390/molecules29184446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Four new alkaloids, vibripyrrolidine A (1), vibripiperazine A (2), and vibridiazinane A, B (3, 4), comprising one pyrrolidine, one piperazine, and two diazinane alkaloids, along with two known analogs (5, 6), were isolated from the marine bacterium Vibrio ruber ZXR-93 cultured in ISP2 medium. Their chemical structures were elucidated by analysis of their 1D and 2D NMR, mass spectra, and electronic circular dichroism (ECD) calculations. Compounds 1 and 3-6 showed vigorous antibacterial activity against Staphylococcus aureus, with MIC values ranging from 0.96 to 7.81 μg/mL. Moreover, compound 1 exhibited robust anti-inflammatory activity in vitro using the LPS-induced RAW264.7 macrophage model. All compounds also showed moderate antineoplastic activity against cervical cancer cells (HeLa) and gastric cancer cells (SGC-7901).
Collapse
Affiliation(s)
| | | | | | - Yinfeng Tan
- NHC Key Laboratory of Tropical Disease Control, Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, School of Tropical Medicine, Hainan Medical University, Haikou 571199, China; (X.Z.); (Y.L.); (H.Z.)
| | - Songlin Zhou
- NHC Key Laboratory of Tropical Disease Control, Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, School of Tropical Medicine, Hainan Medical University, Haikou 571199, China; (X.Z.); (Y.L.); (H.Z.)
| |
Collapse
|
3
|
Fu Y, Huang FY, Dai SZ, Wang L, Zhou X, Zheng ZY, Wang CC, Tan GH, Li Q. Penicilazaphilone C alleviates allergic airway inflammation and improves the immune microenvironment by hindering the NLRP3 inflammasome. Biomed Pharmacother 2024; 175:116788. [PMID: 38772153 DOI: 10.1016/j.biopha.2024.116788] [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: 12/13/2023] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 05/23/2024] Open
Abstract
AIMS Penicilazaphilone C (PAC) is hypothesized to potentially serve as a therapeutic treatment for allergic airway inflammation by inhibiting the NLRP3 inflammasome and reducing oxidative stress. METHODS An allergic asthma model was induced in female BALB/c mice of the OVA, OVA+PAC, OVA+PAC+LPS, and OVA+Dex groups by sensitizing and subsequently challenging them with OVA. The OVA+PAC and Normal+PAC groups were treated with PAC, while the OVA+PAC+LPS group also received LPS. The OVA+Dex group was given dexamethasone (Dex). Samples of serum, bronchoalveolar lavage fluid (BALF), and lung tissue were collected for histological and cytological analysis. RESULTS Allergic mice treated with PAC or Dex showed inhibited inflammation and mucus production in the lungs. There was a decrease in the number of inflammatory cells in the BALF, lower levels of inflammatory cytokines in the serum and BALF, and a reduction in the protein expression of NLRP3, ASC, cleaved caspase-1, IL-1β, activated gasdermin D, MPO, Ly6G, and ICAM-1. Additionally, oxidative stress was reduced, as shown by a decrease in MDA and DCF, but an increase in SOD and GSH. Treatment with PAC also resulted in a decrease in pulmonary memory CD4+ T cells and an increase in regulatory T cells. However, the positive effects seen in the PAC-treated mice were reversed when the NLRP3 inflammasome was activated by LPS, almost returning to the levels of the Sham-treated mice. SIGNIFICANCE PAC acts in a similar way to anti-allergic inflammation as Dex, suggesting it may be a viable therapeutic option for managing allergic asthma inflammation.
Collapse
Affiliation(s)
- Yongshu Fu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China
| | - Feng-Ying Huang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, China.
| | - Shu-Zhen Dai
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Lin Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China
| | - Xiangdong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China
| | - Zhen-You Zheng
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Cai-Chun Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China
| | - Guang-Hong Tan
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, China.
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou 570102, China.
| |
Collapse
|
4
|
Jahan I, Wang Y, Li P, Hussain S, Song J, Yan J. Comprehensive Analysis of Penicillium Sclerotiorum: Biology, Secondary Metabolites, and Bioactive Compound Potential─A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:9555-9566. [PMID: 38648511 DOI: 10.1021/acs.jafc.3c09866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
The filamentous fungus Penicillium sclerotiorum is significant in ecological and industrial domains due to its vast supply of secondary metabolites that have a diverse array of biological functions. We have gathered the metabolic potential and biological activities associated with P. sclerotiorum metabolites of various structures, based on extensive research of the latest literature. The review incorporated literature spanning from 2000 to 2023, drawing from reputable databases including Google Scholar, ScienceDirect, Scopus, and PubMed, among others. Ranging from azaphilones, meroterpenoids, polyketides, and peptides group exhibits fascinating potential pharmacological activities such as antimicrobial, anti-inflammatory, and antitumor effects, holding promise in pharmaceutical and industrial sectors. Additionally, P. sclerotiorum showcases biotechnological potential through the production of enzymes like β-xylosidases, β-d-glucosidase, and xylanases, pivotal in various industrial processes. This review underscores the need for further exploration into its genetic foundations and cultivation conditions to optimize the yield of valuable compounds and enzymes, highlighting the unexplored potential of P. sclerotiorum in diverse applications across industries.
Collapse
Affiliation(s)
- Israt Jahan
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Yihan Wang
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Ping Li
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Sarfaraz Hussain
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255049, Shandong, PR China
| | - Jiayi Song
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jian Yan
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| |
Collapse
|
5
|
Guan X, Yang J, Wang W, Zhao B, Hu S, Yu D, Yuan L, Shi Y, Xu J, Dong J, Wang J, Cheng XD, Qin JJ. Dual inhibition of MYC and SLC39A10 by a novel natural product STAT3 inhibitor derived from Chaetomium globosum suppresses tumor growth and metastasis in gastric cancer. Pharmacol Res 2023; 189:106703. [PMID: 36804016 DOI: 10.1016/j.phrs.2023.106703] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/06/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Gastric cancer remains one of the most common deadly diseases and lacks effective targeted therapies. In the present study, we confirmed that the signal transducer and activator of transcription 3 (STAT3) is highly expressed and associated with a poor prognosis in gastric cancer. We further identified a novel natural product inhibitor of STAT3, termed XYA-2, which interacts specifically with the SH2 domain of STAT3 (Kd= 3.29 μM) and inhibits IL-6-induced STAT3 phosphorylation at Tyr705 and nuclear translocation. XYA-2 inhibited the viability of seven human gastric cancer cell lines with 72-h IC50 values ranging from 0.5 to 0.7 μΜ. XYA-2 at 1 μΜ inhibited the colony formation and migration ability of MGC803 (72.6% and 67.6%, respectively) and MKN28 (78.5% and 96.6%, respectively) cells. In the in vivo studies, intraperitoneal administration of XYA-2 (10 mg/kg/day, 7 days/week) significantly suppressed 59.8% and 88.8% tumor growth in the MKN28-derived xenograft mouse model and MGC803-derived orthotopic mouse model, respectively. Similar results were obtained in a patient-derived xenograft (PDX) mouse model. Moreover, XYA-2 treatment extended the survival of mice bearing PDX tumors. The molecular mechanism studies based on transcriptomics and proteomics analyses indicated that XYA-2 might exert its anticancer activity by synergistically inhibiting the expression of MYC and SLC39A10, two downstream genes of STAT3 in vitro and in vivo. Together, these findings suggested that XYA-2 may be a potent STAT3 inhibitor for treating gastric cancer, and dual inhibition of MYC and SLC39A10 may be an effective therapeutic strategy for STAT3-activated cancer.
Collapse
Affiliation(s)
- Xiaoqing Guan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jing Yang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Weiyi Wang
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian, China.
| | - Bing Zhao
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Shiyu Hu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Dehua Yu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Li Yuan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yunfu Shi
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jingli Xu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jinyun Dong
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jinxin Wang
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xiang-Dong Cheng
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Jiang-Jiang Qin
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Epigenetic Regulation of MAP3K8 in EBV-Associated Gastric Carcinoma. Int J Mol Sci 2023; 24:ijms24031964. [PMID: 36768307 PMCID: PMC9916342 DOI: 10.3390/ijms24031964] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
Super-enhancers (SEs) regulate gene expressions, which are critical for cell type-identity and tumorigenesis. Although genome wide H3K27ac profiling have revealed the presence of SE-associated genes in gastric cancer (GC), their roles remain unclear. In this study, ChIP-seq and HiChIP-seq experiments revealed mitogen-activated protein kinase 8 (MAP3K8) to be an SE-associated gene with chromosome interactions in Epstein-Barr virus-associated gastric carcinoma (EBVaGC) cells. CRISPRi mediated repression of the MAP3K8 SEs attenuated MAP3K8 expression and EBVaGC cell proliferation. The results were validated by treating EBVaGC cells with bromodomain and the extra-terminal motif (BET) inhibitor, OTX015. Further, functional analysis of MAP3K8 in EBVaGC revealed that silencing MAP3K8 could inhibit the cell proliferation, colony formation, and migration of EBVaGC cells. RNA-seq and pathway analysis indicated that knocking down MAP3K8 obstructed the notch signaling pathway and epithelial-mesenchymal transition (EMT) in EBVaGC cells. Further, analysis of the cancer genome atlas (TCGA) and GSE51575 databases exhibited augmented MAP3K8 expression in gastric cancer and it was found to be inversely correlated with the disease-free progression of GC. Moreover, Spearman's correlation revealed that MAP3K8 expression was positively correlated with the expressions of notch pathway and EMT related genes, such as, Notch1, Notch2, C-terminal binding protein 2 (CTBP2), alpha smooth muscle actin isotype 2 (ACTA2), transforming growth factor beta receptor 1 (TGFβR1), and snail family transcriptional repressors 1/2 (SNAI1/SNAI2) in GC. Taken together, we are the first to functionally interrogate the mechanism of SE-mediated regulation of MAP3K8 in EBVaGC cell lines.
Collapse
|
7
|
Zhao H, Ji R, Zha X, Xu Z, Lin Y, Zhou S. Investigation of the bactericidal mechanism of Penicilazaphilone C on Escherichia coli based on 4D label-free quantitative proteomic analysis. Eur J Pharm Sci 2022; 179:106299. [PMID: 36179970 DOI: 10.1016/j.ejps.2022.106299] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/16/2022] [Accepted: 08/27/2022] [Indexed: 11/03/2022]
Abstract
There is an urgent need to find new antibiotics to fight against the increasing drug resistance of microorganisms. A novel natural compound, Penicilazaphilone C (PAC), was isolated from a marine-derived fungus. It has displayed broad bactericidal activities against Gram-negative and Gram-positive bacteria. However, its bactericidal mechanism is still unknown. Herein, time-kill assays verified that PAC is a fast and efficient bactericidal agent. Furthermore, data from 4D label-free quantitative proteome assays revealed that PAC significantly influences over 898 proteins in Escherichia coli. Combining the results of biofilm formation, β-galactosidase measurement, TEM observation, soft agar plate swimming, reactive oxygen species measurement, qRT-PCR, and west-blotting, the mode of PAC action against E. coli was to block respiration, inhibit assimilatory nitrate reduction and dissimilar sulfur reduction, facilitate assimilatory sulfate reduction, suppress cysteine and methionine biosynthesis, down-regulate antioxidant protein expression and induced intracellular ROS accumulation, weaken bacterial chemotaxis, destroy flagellar assembly, etc., and finally cause the bacteria's death. Our findings suggest that PAC could have a multi-target regulatory effect on E. coli and could be used as a new antibiotic in medicine.
Collapse
Affiliation(s)
- Huange Zhao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, NHC Key Laboratory of Control of Tropical Disease Control, Hainan Provincial Key Laboratory of Tropical Medicine, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, 571199
| | - Rong Ji
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, NHC Key Laboratory of Control of Tropical Disease Control, Hainan Provincial Key Laboratory of Tropical Medicine, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, 571199
| | - Xiangru Zha
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, NHC Key Laboratory of Control of Tropical Disease Control, Hainan Provincial Key Laboratory of Tropical Medicine, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, 571199
| | - Zhen Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, NHC Key Laboratory of Control of Tropical Disease Control, Hainan Provincial Key Laboratory of Tropical Medicine, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, 571199
| | - Yingying Lin
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, NHC Key Laboratory of Control of Tropical Disease Control, Hainan Provincial Key Laboratory of Tropical Medicine, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, 571199
| | - Songlin Zhou
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, NHC Key Laboratory of Control of Tropical Disease Control, Hainan Provincial Key Laboratory of Tropical Medicine, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, 571199.
| |
Collapse
|
8
|
Li SY, Hou LZ, Gao YX, Zhang NN, Fan B, Wang F. FIP-nha, a fungal immunomodulatory protein from Nectria haematococca, induces apoptosis and autophagy in human gastric cancer cells via blocking the EGFR-mediated STAT3/Akt signaling pathway. FOOD CHEMISTRY: MOLECULAR SCIENCES 2022; 4:100091. [PMID: 35415679 PMCID: PMC8991989 DOI: 10.1016/j.fochms.2022.100091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/07/2022] [Accepted: 02/23/2022] [Indexed: 11/21/2022]
Abstract
FIP-nha, a new FIP discovered beyond Basidiomycota, has been demonstrated a broad spectrum of antitumor activity and cell selectivity against human cancers. FIP-nha inhibited the growth, induced apoptosis and autophagy of gastric cancer cells through competitively binding to EGFR with EGF to blocking the EGFR-mediated STAT3/Akt pathway. FIP-nha may be a potential chemotherapy drug that targeted EGFR to treat human gastric cancer.
FIP-nha, a fungal immunomodulatory protein from Nectria haematococca, has been demonstrated a broad spectrum of antitumor activity and cell selectivity against human cancers in our previous study. However, the effect and mechanism of FIP-nha on gastric cancer remains unclear. In this study, we systematically observed the cytotoxicity, biological effect, regulatory mechanism and interaction target of FIP-nha on human gastric cancer cell lines, AGS and SGC7901. Our results demonstrated that FIP-nha inhibited the growth of AGS and SGC7901 cells in a dose-dependent manner and exerted proapoptotic effects on both cells as confirmed by flow cytometry, DAPI staining and western blot analysis. Additionally, the exposure of AGS and SGC7901 to FIP-nha induced autophagy as indicated by western blot analysis, GFP-LC3 and mCherry-GFP-LC3 transfection and acridine orange staining. Furthermore, we found that FIP-nha decreased the phosphorylation of EGFR, STAT3 and Akt and inhibited activation effect of ligand factor EGF to EGFR and its downstream signal molecule STAT3 and Akt. Finally, we proved that FIP-nha located on the surface of gastric cancer cells and bound directly to the transmembrane protein of EGFR by immunoprecipitation, cellular localization, molecular docking, microscale thermophoresis assay. The above findings indicated that FIP-nha inhibited the growth of gastric cancer and induced apoptosis and autophagy through competitively binding to EGFR with EGF to blocking the EGFR-mediated STAT3/Akt pathway. In summary, our study provided novel insights regarding the activity of FIP-nha against gastric cancer and contributed to the clinical application of FIP-nha as a potential chemotherapy drugs that targeted EGFR for human gastric cancer.
Collapse
|
9
|
Mansoury F, Babaei N, Abdi S, Entezari M, Doosti A. Changes in NOTCH1 gene and its regulatory circRNA, hsa_circ_0005986 expression pattern in human gastric adenocarcinoma and human normal fibroblast cell line following the exposure to extremely low frequency magnetic field. Electromagn Biol Med 2021; 40:375-383. [PMID: 33620018 DOI: 10.1080/15368378.2021.1891092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The effect of an extremely low-frequency magnetic field (ELF-MFs) on the expression levels of NOTCH1 and its regulatory circular RNA (circ-RNA) in gastric cancer has not yet investigated. This study aimed to find the expression changes of NOTCH1 and its regulatory circ-RNA, hsa_circ_0005986, in human gastric adenocarcinoma cell line (AGS) and human normal fibroblast (Hu02) cells fallowing the exposure to discontinuously magnetic flux densities (MFDs) of 0.25, 0.5 ,1 and 2 millitesla (mT) for 18h in comparison to unexposed cells. In addition, the effect of various MFDs on viability of tumor and normal cells was investigated. The cell viability was evaluated by MTT assay. The relative expression of NOTCH1and hsa_circ_0005986 mRNAs was analyzed by quantitative Real-time PCR. The viability of tumor cells was decreased under the exposure of MFs, while the normal cells viability was increased. NOTCH1 was significantly down-regulated in AGS cells and up-regulated in Hu02 cells at all MFDs. The expression changes of NOTCH1 in tumor and normal cells was depended to the MFD of MFs. According to our results, the tumor and normal cells show different behavior at the molecular level in various MFDs in terms of NOTCH1 and hsa_circ_0005986 expression level. Decrease in tumor cell survival following the exposure to ELF-MFs may be the result of decreased in the expression level of NOTCH1 and its Reg-circ-RNA. These magnetic field-reducing effects on cancer cell survival through the change on the expression of genes involved in the proliferation and progression of cancer can be a new key in cancer treatment.
Collapse
Affiliation(s)
- Fereshteh Mansoury
- Department of Molecular Cell Biology and Genetics, Bushehr Branch, Islamic Azad University, Bushehr, Iran
| | - Nahid Babaei
- Department of Molecular Cell Biology and Genetics, Bushehr Branch, Islamic Azad University, Bushehr, Iran
| | - Soheila Abdi
- Department of Physics, Safadasht Branch, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Abbas Doosti
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
10
|
Chen S, Wei Y, Liu H, Gong Y, Zhou Y, Yang H, Tang L. Analysis of Collagen type X alpha 1 (COL10A1) expression and prognostic significance in gastric cancer based on bioinformatics. Bioengineered 2020; 12:127-137. [PMID: 33371777 PMCID: PMC8291830 DOI: 10.1080/21655979.2020.1864912] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Collagen type X alpha 1 (COL10A1) is a member of the collagen family and the main matrix component. However, COL10A1 expression and prognosis relationship remains unclear in gastric cancer (GC). Through the analysis of database of Oncomine, the Cancer Genome Atlas (TCGA) as well as the Gene Expression Omnibus (GEO), in contrast to the tissue of normal gastric, COL10A1 in gastric cancer, had been upregulated. The high expression of COL10A1 was obviously related to T stage (P = 0.025) and lymph node metastasis (P = 0.025). It has been illustrated by the analysis of logistic regression that COL10A1's heightened expression in gastric cancer had been essentially linked with pathological stage, tumor differentiation, and T classification. The Kaplan-Meier curve in the Kaplan-Meier plotter database (P = 0.0371) and GSE84437 (P = 0.002) indicate that patients with high COL10A1 expression possess poor prognosis, specifically GC patients with lymph node metastasis have it. TCGA's Multivariate analysis (P = 0.025) and GSE84437 dataset (P = 0.034) show that high expression COL10A1 is a key independent predictor of poor overall survival. Searching KEGG pathway enrichment by GSEA, the results suggested that 29 pathways were enriched. qRT-PCR technique was used for verification of the COL10A1's high expression in gastric cancer in contrast to the normal gastric tissues. In conclusion, COL10A1 is of great importance in predicting the survival rate of GC patients.
Collapse
Affiliation(s)
- Shuai Chen
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University , Changzhou, China
| | - Yi Wei
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University , Changzhou, China
| | - Hanyang Liu
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University , Changzhou, China
| | - Yu Gong
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University , Changzhou, China
| | - Yan Zhou
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University , Changzhou, China
| | - Haojun Yang
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University , Changzhou, China
| | - Liming Tang
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University , Changzhou, China
| |
Collapse
|
11
|
Zu WY, Tang JW, Hu K, Zhou YF, Gou LL, Su XZ, Lei X, Sun HD, Puno PT. Chaetolactam A, an Azaphilone Derivative from the Endophytic Fungus Chaetomium sp. g1. J Org Chem 2020; 86:475-483. [PMID: 33263391 DOI: 10.1021/acs.joc.0c02214] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Wen-Yu Zu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, Yunnan, People’s Republic of China
| | - Jian-Wei Tang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, Yunnan, People’s Republic of China
| | - Kun Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, Yunnan, People’s Republic of China
| | - Yuan-Fei Zhou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, Yunnan, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People’s Republic of China
| | - Lei-Lei Gou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, Yunnan, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People’s Republic of China
| | - Xiao-Zheng Su
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, Yunnan, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People’s Republic of China
| | - Xinxiang Lei
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, Hubei 430074, People’s Republic of China
| | - Han-Dong Sun
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, Yunnan, People’s Republic of China
| | - Pema-Tenzin Puno
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, Yunnan, People’s Republic of China
| |
Collapse
|
12
|
Xu Z, Feng Q, Wang M, Zhao H, Lin Y, Zhou S. Green Biosynthesized Silver Nanoparticles With Aqueous Extracts of Ginkgo Biloba Induce Apoptosis via Mitochondrial Pathway in Cervical Cancer Cells. Front Oncol 2020; 10:575415. [PMID: 33194686 PMCID: PMC7606942 DOI: 10.3389/fonc.2020.575415] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
Biosynthetic silver nanoparticles (AgNPs), specifically formed using medicinal plant extracts, have recently exhibited a remarkable therapeutic effect due to their anticancer potential. Here, we synthesized AgNPs using an aqueous extract of Ginkgo biloba leaves and evaluated its activity against cervical cancer (CCa) and the related molecular mechanisms. The physiochemical properties of the AgNPs were measured by ultraviolet-visible spectrophotometry, nanometre particle size analyzer and transmission electron microscopy. The AgNPs effects on cell proliferation and apoptosis were investigated through MTT, MTS, and colony formation assay; Hoechst 33258 staining; and flow cytometry. The intracellular ROS and oxidative stress levels were assessed using the appropriate commercial kits. Apoptosis-related protein levels were determined by western blotting. We prepared a series of different sized ginkgo extract synthesized AgNPs (GB-AgNPs), and the smallest mean particle size was 40.2 ± 1.2 nm with low polydispersity (0.091 ± 0.011), zeta potential values showed -34.56 mV. Compared to the controls, the GB-AgNP treatment inhibited the cell proliferation and induced the apoptosis of HeLa and SiHa cells. In addition, GB-AgNP treatment led to markedly increased levels of intracellular ROS, the release of cytochrome c (Cyt C) from mitochondria into the cytosol and the cleavage of caspase -9 and -3 in both CCa cell lines. Moreover, NAC, an ROS scavenger, eliminated the effect of GB-AgNPs on the HeLa and SiHa cells. This study reveals that GB-AgNPs suppresses cancer cell proliferation and induces apoptosis by upregulating intracellular ROS generation and inducing the activation of the caspase-dependent mitochondrial apoptotic pathway in CCa cells. Thus, GB-AgNPs may be a potential alternative drug for CCa therapy.
Collapse
Affiliation(s)
- Zhen Xu
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education and Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, China
| | - Qi Feng
- Jiangsu Provincial Key Laboratory of Veterinary Bio-pharmaceutical High-tech Research, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
| | - Min Wang
- Health and Family Planning Commission of Wanzai County of Jiangxi Province, Yichun, China
| | - Huange Zhao
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education and Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, China
| | - Yingying Lin
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education and Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, China
| | - Songlin Zhou
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education and Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, China
| |
Collapse
|