1
|
Wang X, Zou C, Hou C, Li M, Bian Z, Zhu L. POU Class 2 Homeobox Associating Factor 1, as a Hub Candidate Gene in OP, Relieves Osteoblast Apoptosis. Appl Biochem Biotechnol 2024; 196:6072-6096. [PMID: 38183606 DOI: 10.1007/s12010-023-04833-y] [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] [Accepted: 12/19/2023] [Indexed: 01/08/2024]
Abstract
Increasing evidence suggests that osteoblast apoptosis contributes to the pathogenesis of postmenopausal osteoporosis (PMOP). This study aimed to identify a hub gene associated with osteoporosis (OP) progression and its functions. We utilized the GSE68303 expression dataset from GEO database and conducted weighted gene co-expression network analysis (WGCNA) to investigate changes in co-expressed genes between sham and ovariectomy (OVX) groups. Differentially expressed genes (DEGs) were identified using the "limma" R package on GSE68303 dataset. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using the DAVID database. A protein-protein interaction (PPI) network was constructed using the STRING database, which was visualized by Cytoscape software. The top ten hub genes were screened using the Cytohubba plugin, among which POU class 2 homeobox associating factor 1 (POU2AF1), an OP-related hub gene, showed a significant increase in OVX-induced mouse model based on immunohistochemical staining. Inhibition of POU2AF1 suppressed cell viability, induced cell cycle arrest at the G1 phase, and promoted osteoblast apoptosis as demonstrated by CCK-8 assay, flow cytometry analysis, and TUNEL assay. Moreover, overexpression of POU2AF1 decreased cleaved caspase-3/-8/-9 expression while increasing cyclinD1 and Ki67 expression in MC3T3-E1 and hFOB1.19 cells. Therefore, POU2AF1 may serve as a potential diagnostic biomarker for slowing down the progression of OP.
Collapse
Affiliation(s)
- Xuepeng Wang
- Department of Orthopedics Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261, Huansha Road, Hangzhou, 310006, Zhejiang Province, China
| | - Chunchun Zou
- Department of Obstetrics and Gynecology, Hangzhou Third People's Hospital, Hangzhou, China
| | - Changju Hou
- Department of Orthopedics Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261, Huansha Road, Hangzhou, 310006, Zhejiang Province, China
| | - Maoqiang Li
- Department of Orthopedics Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261, Huansha Road, Hangzhou, 310006, Zhejiang Province, China
| | - Zhenyu Bian
- Department of Orthopedics Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261, Huansha Road, Hangzhou, 310006, Zhejiang Province, China
| | - Liulong Zhu
- Department of Orthopedics Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261, Huansha Road, Hangzhou, 310006, Zhejiang Province, China.
| |
Collapse
|
2
|
Shi Y, Li H, Chu D, Lin W, Wang X, Wu Y, Li K, Wang H, Li D, Xu Z, Gao L, Li B, Chen H. Rescuing Nucleus Pulposus Cells From Senescence via Dual-Functional Greigite Nanozyme to Alleviate Intervertebral Disc Degeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300988. [PMID: 37400370 PMCID: PMC10477883 DOI: 10.1002/advs.202300988] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/23/2023] [Indexed: 07/05/2023]
Abstract
High levels of reactive oxygen species (ROS) lead to progressive deterioration of mitochondrial function, resulting in tissue degeneration. In this study, ROS accumulation induced nucleus pulposus cells (NPCs) senescence is observed in degenerative human and rat intervertebral disc, suggesting senescence as a new therapeutic target to reverse intervertebral disc degeneration (IVDD). By targeting this, dual-functional greigite nanozyme is successfully constructed, which shows the ability to release abundant polysulfides and presents strong superoxide dismutase and catalase activities, both of which function to scavenge ROS and maintain the tissue at physical redox level. By significantly lowering the ROS level, greigite nanozyme rescues damaged mitochondrial function in IVDD models both in vitro and in vivo, rescues NPCs from senescence and alleviated the inflammatory response. Furthermore, RNA-sequencing reveals ROS-p53-p21 axis is responsible for cellular senescence-induced IVDD. Activation of the axis abolishes greigite nanozyme rescued NPCs senescence phenotype, as well as the alleviated inflammatory response to greigite nanozyme, which confirms the role of ROS-p53-p21 axis in greigite nanozyme's function to reverse IVDD. In conclusion, this study demonstrates that ROS-induced NPCs senescence leads to IVDD and the dual-functional greigite nanozyme holds strong potential to reverse this process, providing a novel strategy for IVDD management.
Collapse
Affiliation(s)
- Yu Shi
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Hanwen Li
- Orthopedic InstituteDepartment of Orthopedic SurgeryFirst Affiliated HospitalSuzhou Medical CollegeSoochow UniversityNo. 899 Pinghai RoadSuzhou215000P. R. China
| | - Dongchuan Chu
- Department of RadiologyAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
| | - Wenzheng Lin
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Xinglong Wang
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Yin Wu
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Ke Li
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Huihui Wang
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Dandan Li
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Zhuobin Xu
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
- Institute of Translational MedicineMedical CollegeYangzhou UniversityNo.136 Jiangyang RoadYangzhou215000P. R. China
| | - Lizeng Gao
- CAS Engineering Laboratory for NanozymeInstitute of BiophysicsChinese Academy of SciencesNo. 15 Datun RoadBeijing100101P. R. China
| | - Bin Li
- Orthopedic InstituteDepartment of Orthopedic SurgeryFirst Affiliated HospitalSuzhou Medical CollegeSoochow UniversityNo. 899 Pinghai RoadSuzhou215000P. R. China
| | - Hao Chen
- Department of OrthopedicsAffiliated Hospital of Yangzhou UniversityNo. 368 Hanjiang RoadYangzhou225000P. R. China
| |
Collapse
|
3
|
Tan Q, Chen J, Gao G, Yizhang, Chen X, Yu Y, Zang G, Tang Z. Adenovirus vector encoding TPPII ignites HBV-specific CTL response by activating autophagy in CD8+ T cell. J Viral Hepat 2022; 29:178-188. [PMID: 34902200 DOI: 10.1111/jvh.13638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 11/01/2021] [Accepted: 11/26/2021] [Indexed: 12/19/2022]
Abstract
Early studies have shown that autophagy and TPPII are associated with HBV infection. In this study, adenovirus vector containing TPPII was constructed to immunize HBV transgenic mice in vivo to explore the potential mechanism of autophagy and HBV infection. Our goal is to provide new ideas for immunotherapy of hepatitis B. First, adenovirus vector containing TPPII was constructed. Then, we used adenovirus to immunize HBV transgenic mice and ATG5 knockout HBV transgenic mice. The autophagy of CD8+ T cells was detected by transmission electron microscopy and immunofluorescence electron microscopy, Western blot was used to detect the expression of autophagy LC3 and BECN1, CTL reaction, HBV DNA and HBsAg in serum, HBsAg and HBcAg in liver tissues by immunohistochemistry, to further examine the possible mechanisms involved in autophagy. Adv-HBcAg-TPPII promotes autophagy of CD8+ T lymphocyte, activates CTL response, inhibits HBV DNA replication and HBsAg expression, and PI3K/ Akt /m TOR signalling pathway may be involved in autophagy. This study demonstrates that autophagy of CD8+ T cells was induced by Adv-HBcAg-TPPII and the molecular mechanism may be related to the PI3K/ Akt /m TOR signalling pathway, providing a possible theoretical basis for immunotherapy of hepatitis B.
Collapse
Affiliation(s)
- Quanhui Tan
- Department of infection Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jie Chen
- Department of infection Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Gao Gao
- Department of infection Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yizhang
- Department of infection Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaohua Chen
- Department of infection Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yongsheng Yu
- Department of infection Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Guoqin Zang
- Department of infection Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhenghao Tang
- Department of infection Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
4
|
Ivashkevich A. The role of isoflavones in augmenting the effects of radiotherapy. Front Oncol 2022; 12:800562. [PMID: 36936272 PMCID: PMC10016616 DOI: 10.3389/fonc.2022.800562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 08/31/2022] [Indexed: 03/05/2023] Open
Abstract
Cancer is one of the major health problems and the second cause of death worldwide behind heart disease. The traditional soy diet containing isoflavones, consumed by the Asian population in China and Japan has been identified as a protective factor from hormone-related cancers. Over the years the research focus has shifted from emphasizing the preventive effect of isoflavones from cancer initiation and promotion to their efficacy against established tumors along with chemo- and radiopotentiating effects. Studies performed in mouse models and results of clinical trials emphasize that genistein or a mixture of isoflavones, containing in traditional soy diet, could be utilized to both potentiate the response of cancer cells to radiotherapy and reduce radiation-induced toxicity in normal tissues. Currently ongoing clinical research explores a potential of another significant isoflavone, idronoxil, also known as phenoxodiol, as radiation enhancing agent. In the light of the recent clinical findings, this article reviews the accumulated evidence which support the clinically desirable interactions of soy isoflavones with radiation therapy resulting in improved tumor treatment. This review discusses important aspects of the development of isoflavones as anticancer agents, and mechanisms potentially relevant to their activity in combination with radiation therapy of cancer. It gives a critical overview of studies characterizing isoflavone targets such as topoisomerases, ENOX2/PMET, tyrosine kinases and ER receptor signaling, and cellular effects on the cell cycle, DNA damage, cell death, and immune responses.
Collapse
Affiliation(s)
- Alesia Ivashkevich
- Faculty of Engineering and Information Sciences, University of Wollongong, Wollongong, NSW, Australia
- Noxopharm, Gordon, NSW, Australia
- *Correspondence: Alesia Ivashkevich,
| |
Collapse
|
5
|
Crumbaker M, Pathmanandavel S, Yam AO, Nguyen A, Ho B, Chan L, Ende JA, Rofe C, Kongrak K, Kwan EM, Azad AA, Sharma S, Pugh TJ, Danesh A, Keane J, Eu P, Joshua AM, Emmett L. Phase I/II Trial of the Combination of 177Lutetium Prostate specific Membrane Antigen 617 and Idronoxil (NOX66) in Men with End-stage Metastatic Castration-resistant Prostate Cancer (LuPIN). Eur Urol Oncol 2020; 4:963-970. [PMID: 32758400 DOI: 10.1016/j.euo.2020.07.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/26/2020] [Accepted: 07/09/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Trials of lutetium prostate specific membrane antigen (PSMA) in men with metastatic castration-resistant prostate cancer (mCRPC) have demonstrated good safety and efficacy, but combination strategies may improve outcomes. Idronoxil is a synthetic flavonoid derivative with radiosensitising properties. OBJECTIVE To evaluate the safety and activity of 177Lu PSMA 617 (LuPSMA-617) in combination with idronoxil suppositories (NOX66) in patients with end-stage mCRPC. DESIGN, SETTING, AND PARTICIPANTS Thirty-two men with progressive mCRPC previously treated with taxane-based chemotherapy (91% treated with both docetaxel and cabazitaxel) and abiraterone and/or enzalutamide were enrolled in this phase I dose escalation study with phase II dose expansion. INTERVENTION Screening with 68Ga PSMA and 18F-fludeoxyglucose positron emission tomography (PET)/computed tomography (CT) was performed. Men received up to six cycles of LuPSMA-617 (7.5 GBq) on day 1, with escalating doses of NOX66 on days 1-10 of a 6-wk cycle. Cohort 1 (n = 8) received 400 mg and cohort 2 (n = 24) 800 mg of NOX66. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Adverse events (AEs), pain inventory scores, prostate-specific antigen (PSA) response, progression-free survival, and overall survival were evaluated. RESULTS AND LIMITATIONS Fifty-six men were screened and 32 (57%) were enrolled with a screen failure rate of 21% for PET imaging criteria. Dosing was as follows: 97% (31/32) received two or more doses and 47% (15/32) completed six doses. Common AEs included xerostomia, fatigue, and anaemia. Anal irritation attributable to NOX66 occurred in 28%. PSA responses were as follows: 91% (29/32) had any PSA response (median -74%; 95% confidence interval [CI] 76-97) and 62.5% (20/32) had a PSA fall of >50% (95% CI 45-77). The median PSA progression-free survival was 6.1 mo (95% CI 2.8-9.2) and median overall survival was 17.1 mo (95% CI 6.5-27.1). CONCLUSIONS NOX66 with LuPSMA-617 is a safe and feasible therapeutic strategy in men treated with third-line therapy and beyond for mCRPC. PATIENT SUMMARY Addition of NOX66 to 177Lu prostate-specific membrane antigen 617 is safe, and further studies are needed to assess its potential to augment the anticancer effects of LuPSMA-617.
Collapse
Affiliation(s)
- Megan Crumbaker
- The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, University of New South Wales, Kensington, NSW, Australia; Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | | | - Andrew O Yam
- The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, University of New South Wales, Kensington, NSW, Australia; Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Andrew Nguyen
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Bao Ho
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Lyn Chan
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Jesse A Ende
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Christopher Rofe
- The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Kamonwan Kongrak
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Edmond M Kwan
- Department of Medical Oncology, Monash Health, Clayton, VIC, Australia; Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Arun A Azad
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Shikha Sharma
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Arnavaz Danesh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Joanne Keane
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Peter Eu
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Anthony M Joshua
- The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, University of New South Wales, Kensington, NSW, Australia; Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Louise Emmett
- St. Vincent's Clinical School, University of New South Wales, Kensington, NSW, Australia; Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.
| |
Collapse
|
6
|
Yaylaci E, Onen HI, Yar Saglam AS. Phenoxodiol sensitizes metastatic colorectal cancer cells to 5-fluorouracil- and oxaliplatin-induced apoptosis through intrinsic pathway. EXCLI JOURNAL 2020; 19:936-949. [PMID: 32665777 PMCID: PMC7355152 DOI: 10.17179/excli2020-2042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/25/2020] [Indexed: 11/10/2022]
Abstract
Colorectal cancer (CRC) is one of the most common types of cancer seen in the world. 5-Fluorouracil (5-Fu) plus Oxaliplatin (1-OHP) remains the backbone of CRC chemotherapeutics, but with limited success. Phenoxodiol (Pxd) is an isoflavone analog with antitumor activity against various types of cancers, and sensitizes chemoresistant cancer cells to chemotherapeutics including platinum and taxanes. This study was, therefore, undertaken to examine whether Pxd pre-treatment with conventional chemotherapeutic agent(s) 5-Fu and 1-OHP co-administration be a therapeutic strategy for CRC. Cell viability and cytotoxicity were evaluated using dimethyl-thiazolyl diphenyl tetrazolium bromide (MTT) and lactate dehydrogenase assays. The percentage of apoptotic and necrotic cells were determined by fluorescence microscopy analysis. Besides, active Caspase-3 levels by ELISA and relative mRNA levels of Caspase 3 (CASP3), CASP8 and CASP9 genes were determined by quantitative real-time PCR (qPCR) analysis. The pre-treatment of Pxd followed by 5-Fu and 1-OHP co-administration was more effective at inhibiting cell viability than either chemotherapeutic agents treatment alone. When compared to 5-Fu with 1-OHP alone treatment, Pxd pre-treatment overwhelmingly increased apoptotic Caspase-3 activity levels in CRC cells. Moreover, qPCR analyses showed that CASP3 and CASP9 mRNA levels significantly increased after pre-treatment with Pxd followed by 5-Fu and 1-OHP treatments, compared to 5-Fu with 1-OHP alone. Our results suggested that Pxd enhanced the in vitro antitumor activity of 5-Fu and 1-OHP. Our study also suggested that Pxd may be a potential candidate agent in advanced CRC and inclusion of Pxd to the conventional chemotherapeutic agent(s) could be an effective therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Esra Yaylaci
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| | - Hacer Ilke Onen
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| | - Atiye Seda Yar Saglam
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| |
Collapse
|
7
|
Triggering of eryptosis, the suicidal erythrocyte death, by phenoxodiol. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1311-1318. [PMID: 31280326 DOI: 10.1007/s00210-019-01681-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/19/2019] [Indexed: 12/14/2022]
Abstract
Phenoxodiol is used for the treatment of malignancy. The substance is effective by triggering suicidal tumor cell death or apoptosis. At least in theory, phenoxodiol could similarly stimulate suicidal erythrocyte death or eryptosis. Eryptosis is characterized by cell shrinkage and breakdown of cell membrane asymmetry with phosphatidylserine translocation to the erythrocyte surface. Signaling of eryptosis includes increase of cytosolic Ca2+ activity ([Ca2+]i), formation of reactive oxygen species (ROS), and increase of ceramide abundance at the cell surface. The present study explored whether phenoxodiol induces eryptosis and whether it modifies Ca2+ entry, ROS, and ceramide. Using flow cytometry, phosphatidylserine exposure at the cell surface was quantified from annexin V binding, cell volume from forward scatter, [Ca2+]i from Fluo3 fluorescence, ROS from DCFDA-dependent fluorescence, and ceramide abundance utilizing specific antibodies. A 48-h exposure of human erythrocytes to phenoxodiol (100 μg/ml [416 μM]) significantly increased the percentage of annexin V binding cells, significantly decreased average forward scatter and Fluo3 fluorescence and significantly increased ceramide abundance, but did not significantly modify DCFDA fluorescence. The effect of phenoxodiol on annexin V binding tended to decrease following removal of extracellular Ca2+, an effect, however, not reaching statistical significance. In conclusion, phenoxodiol triggers eryptosis, an effect paralleled by increase of ceramide abundance.
Collapse
|
8
|
Abu-Serie MM, Habashy NH. Two purified proteins from royal jelly with in vitro dual anti-hepatic damage potency: Major royal jelly protein 2 and its novel isoform X1. Int J Biol Macromol 2019; 128:782-795. [PMID: 30711561 DOI: 10.1016/j.ijbiomac.2019.01.210] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 01/01/2023]
Abstract
Liver diseases are serious life-threating conditions that should be controlled. Here, we identify a protein fraction from royal-jelly (RJ) that represents the most effective composite against CCl4-induced hepatotoxicity and HepG2 cell growth. Two closely related proteins were purified from this fraction by a new simple method and identified by MALDI-TOF MS as major RJ protein 2 (MRJP2) and its predicted isoform X1. The in silico assessment (3D structures and functions) of these proteins were performed using Iterative Threading ASSEmbly Refinement (I-TASSER) analysis and RAMPAGE program. These two purified proteins were able to relieve the necrotic hepatocytes (by 60.4%) via reducing tumor necrosis factor (TNF)-α, mixed lineage kinase domain-like protein (MLKL) and intracellular reactive species. The latter effects associated with improving hepatocyte functions. Furthermore, they revealed the potent anticancer effect via induction of caspase-dependent apoptosis and controlling the expression of both Bcl-2 and p53 in HepG2 cells. Thus, MRJP2 and its isoform X1 can be a promising dual strategy for fighting hepatic injury and cancer in future animal and human studies.
Collapse
Affiliation(s)
- Marwa M Abu-Serie
- Department of Medical Biotechnology, Genetic Engineering, and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab 21934, Alexandria, Egypt.
| | - Noha H Habashy
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| |
Collapse
|
9
|
Hashimoto T, Nozawa D, Mukai K, Matsuyama A, Kuramochi K, Furuya T. Monooxygenase-catalyzed regioselective hydroxylation for the synthesis of hydroxyequols. RSC Adv 2019; 9:21826-21830. [PMID: 35518870 PMCID: PMC9066559 DOI: 10.1039/c9ra03913a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/09/2019] [Indexed: 11/21/2022] Open
Abstract
A one-step product-selective approach for synthesizing hydroxyequols from equol using oxidation biocatalysts was developed.
Collapse
Affiliation(s)
- Takafumi Hashimoto
- Department of Applied Biological Science
- Faculty of Science and Technology
- Tokyo University of Science
- Noda
- Japan
| | - Daiki Nozawa
- Department of Applied Biological Science
- Faculty of Science and Technology
- Tokyo University of Science
- Noda
- Japan
| | | | | | - Kouji Kuramochi
- Department of Applied Biological Science
- Faculty of Science and Technology
- Tokyo University of Science
- Noda
- Japan
| | - Toshiki Furuya
- Department of Applied Biological Science
- Faculty of Science and Technology
- Tokyo University of Science
- Noda
- Japan
| |
Collapse
|
10
|
Liang T, Ye X, Liu Y, Qiu X, Li Z, Tian B, Yan D. FAM46B inhibits cell proliferation and cell cycle progression in prostate cancer through ubiquitination of β-catenin. Exp Mol Med 2018; 50:1-12. [PMID: 30532005 PMCID: PMC6288130 DOI: 10.1038/s12276-018-0184-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023] Open
Abstract
FAM46B is a member of the family with sequence similarity 46. Little is known about the expression and functional role(s) of FAM46B in prostate cancer (PC). In this study, the expression of FAM46B expression in The Cancer Genome Atlas, GSE55945, and an independent hospital database was measured by bioinformatics and real-time PCR analysis. After PC cells were transfected with siRNA or a recombinant vector in the absence or presence of a β-catenin signaling inhibitor (XAV-939), the expression levels of FAM46B, C-myc, Cyclin D1, and β-catenin were measured by western blot and real-time PCR. Cell cycle progression and cell proliferation were measured by flow cytometry and the CCK-8 assay. The effects of FAM46B on tumor growth and protein expression in nude mice with PC tumor xenografts were also measured. Our results showed that FAM46B was downregulated but that β-catenin was upregulated in patients with PC. FAM46B silencing promoted cell proliferation and cell cycle progression in PC, which were abrogated by XAV-939. Moreover, FAM46B overexpression inhibited PC cell cycle progression and cell proliferation in vitro and tumor growth in vivo. FAM46B silencing promoted β-catenin protein expression through the inhibition of β-catenin ubiquitination. Our data clearly show that FAM46B inhibits cell proliferation and cell cycle progression in PC through ubiquitination of β-catenin. A little-studied protein may help in early diagnosis and treatment of prostate cancer (PC), one of the most common cancers in men. Because early-stage PC causes few symptoms, many patients are not diagnosed until later stages, when treatment options are limited. New methods for early diagnosis and treatment are actively sought. Proteins in the FAM46 family are known to be involved in many types of cancer. Dongliang Yan at Shanghai Sixth People’s Hospital East and co-workers investigated what role one protein in this family, FAM46B, might play in PC. Analysis of tumor samples showed that FAM46B levels were much lower in PC than in healthy tissues. These changes were linked to another tumor-associated protein, β-catenin. In further tests in mice, artificially increasing FAM46B levels decreased tumor size. These results could improve treatments for PC.
Collapse
Affiliation(s)
- Tao Liang
- Department of Urology, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201306, China
| | - Xuxiao Ye
- Department of Urology, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201306, China
| | - Yuanyuan Liu
- Department of Urology, Affiliated Hospital of Binzhou Medical University, Binzhou, 256603, China
| | - Xinkai Qiu
- Department of Urology, Affiliated Hospital of Binzhou Medical University, Binzhou, 256603, China
| | - Zuowei Li
- Department of Urology, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201306, China
| | - Binqiang Tian
- Department of Urology, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201306, China
| | - Dongliang Yan
- Department of Urology, Shanghai Sixth People's Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201306, China.
| |
Collapse
|
11
|
Akef H, Kotb N, Abo-Elmatty D, Salem S. Anti-proliferative Effects of Androctonus amoreuxi Scorpion and Cerastes cerastes Snake Venoms on Human Prostate Cancer Cells. J Cancer Prev 2017; 22:40-46. [PMID: 28382285 PMCID: PMC5380188 DOI: 10.15430/jcp.2017.22.1.40] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 02/20/2017] [Accepted: 02/27/2017] [Indexed: 11/24/2022] Open
Abstract
The present study evaluated the effects of Androctonus amoreuxi scorpion venom, Cerastes cerastes snake venom and their mixture on prostate cancer cells (PC3). An MTT assay was used to determine the anti-proliferative effect of the venoms, while quantitative real time PCR was used to evaluate the expression of apoptosis-related genes (Bax and Bcl-2). Furthermore, colorimetric assays were used to measure the levels of malondialdehyde (MDA) and antioxidant enzymes. Our results show that the venoms significantly reduced PC3 cell viability in a dose-dependent manner. On the other hand, these venoms significantly decreased Bcl-2 gene expression. Additionally, C. cerastes venom significantly reduced Bax gene expression, while A. amoreuxi venom and a mixture of A. amoreuxi & C. cerastes venoms did not alter Bax expression. Consequently, these venoms significantly increased the Bax/Bcl-2 ratio and the oxidative stress biomarker MDA. Furthermore, these venoms also increased the activity levels of the antioxidant enzymes, catalase, superoxide dismutase, glutathione peroxidase, glutathione reductase, and glutathione-S-transferase. Overall, the venoms have cytotoxic and anti-proliferative effects on PC3 cells.
Collapse
Affiliation(s)
- Hassan Akef
- National Organization for Research and Control of Biologicals (NORCB), Giza, Egypt
| | - Nahla Kotb
- National Organization for Research and Control of Biologicals (NORCB), Giza, Egypt
| | - Dina Abo-Elmatty
- Biochemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Sayed Salem
- Animal Health Research Institute (AHRI), Giza, Egypt
| |
Collapse
|
12
|
Quan Y, Wang N, Chen Q, Xu J, Cheng W, Di M, Xia W, Gao WQ. SIRT3 inhibits prostate cancer by destabilizing oncoprotein c-MYC through regulation of the PI3K/Akt pathway. Oncotarget 2016; 6:26494-507. [PMID: 26317998 PMCID: PMC4694917 DOI: 10.18632/oncotarget.4764] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/25/2015] [Indexed: 12/18/2022] Open
Abstract
SIRT3 is involved in aging-related diseases including cancer, but its role in prostate cancer and detailed regulatory function are not known. We found that SIRT3 was moderately down-regulated in prostate carcinomas. Overexpression of SIRT3 by lentiviral transfection inhibited prostate cancer growth both in vitro and in vivo, whereas knockdown of SIRT3 increased prostate tumor growth. Mechanistically, the tumor suppression effect of SIRT3 was achieved via its inhibition of the PI3K/Akt pathway. Notably, upregulation of SIRT3 suppressed the phosphorylation of Akt, leading to the ubiquitination and degradation of oncoprotein c-MYC; this could be attenuated by constitutive activation of PI3K/Akt signaling. Collectively, our results unveiled SIRT3's tumor suppressive function and the underlying mechanism in prostate cancer, which might provide therapeutic implications for the disease.
Collapse
Affiliation(s)
- Yizhou Quan
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Naitao Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qianqian Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Xu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Cheng
- Department of Urology, First People's Hospital of Xiaoshan, Hangzhou, Zhejiang, China
| | - Meijuan Di
- Department of Pathology, First People's Hospital of Xiaoshan, Hangzhou, Zhejiang, China
| | - Weiliang Xia
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Collaborative Innovation Center of Systems Biomedicine, Shanghai, China
| |
Collapse
|
13
|
Lewandowska U, Fichna J, Gorlach S. Enhancement of anticancer potential of polyphenols by covalent modifications. Biochem Pharmacol 2016; 109:1-13. [PMID: 26776305 DOI: 10.1016/j.bcp.2015.12.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 12/23/2015] [Indexed: 12/23/2022]
Abstract
As evidenced by a growing number of respective clinical trials, a promising and increasingly valued approach to cancer prevention is chemoprevention which is based on using synthetic, semisynthetic, or natural compounds with the aim of preventing, delaying, arresting, or reversing carcinogenesis. Research carried out in the last two decades indicates that natural polyphenols isolated from plants (as well as their derivatives and synthetic analogs) exhibit pleiotropic actions toward cancer cells and therefore they could be used in both cancer prevention and therapy. This review discusses selected covalent modifications of polyphenols as a means for increasing their anticancer potential in relation to the parent compounds. The modifications include hydroxylation, methylation, acylation, and galloylation, among others. They were demonstrated to enhance cytotoxic, pro-oxidant, antiproliferative, proapoptotic, proautophagic, and antimigratory activities of phenolics toward various cancer cell lines in vitro. Importantly, some derivatives proved to suppress tumor growth and metastasis in animal models more strongly than the parent compounds. Some of the above-mentioned covalent modifications were also shown to increase absorption and tissue distribution of tested phenolic compounds in vivo. Anticancer clinical trials with polyphenol derivatives therefore seem warranted.
Collapse
Affiliation(s)
| | - Jakub Fichna
- Department of Biochemistry, Medical University of Lodz, Lodz, Poland
| | - Sylwia Gorlach
- Department of Biochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|