1
|
Song W, Yu X, Yu X, Zhang H, Zhang K, Guo L, Wang JD, Tian DL, Yu Q, Wang X, Zhao J, Xiang W. Antifungal Activity and Potential Mechanisms of Two Bafilomycin Analogues Isolated from Streptomyces sp. NEAU-Y11 against Colletotrichum orbiculare. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11814-11828. [PMID: 40313176 DOI: 10.1021/acs.jafc.5c03728] [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: 05/03/2025]
Abstract
Cucumber anthracnose, caused by Colletotrichum orbiculare, severely affects the cucumber yield and quality. In this study, two active compounds, bafilomycin C1 and JBIR-100, were isolated from strain NEAU-Y11 and exhibited strong antifungal activity against C. orbiculare, with EC50 values of 0.0491 and 0.1042 μg/mL, respectively, significantly lower than those of the commercial fungicide (4.42 μg/mL). Pot experiments demonstrated effective control of cucumber anthracnose at 0.2 μg/mL for bafilomycin C1 and 0.4 μg/mL for JBIR-100, with efficacies reaching 78.5 and 67.7%, respectively. Microscopy and biochemical analyses indicated that both compounds disrupted the fungal cell wall, membrane, and redox homeostasis, leading to cell death. Transcriptome analysis further revealed the effects of bafilomycin C1's on amino acid metabolism, cell structure, redox homeostasis, and DNA double-strand break repair. These findings suggest that bafilomycin C1 and JBIR-100 are promising candidates for use as agrochemical fungicides to control C. orbiculares and may serve as a basis for developing next-generation antifungal agents.
Collapse
Affiliation(s)
- Wenshuai Song
- Key Laboratory of Agricultural Microbiology of Heilongjiang Province, College of Plant Protection, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoyan Yu
- Key Laboratory of Agricultural Microbiology of Heilongjiang Province, College of Plant Protection, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoxin Yu
- Key Laboratory of Agricultural Microbiology of Heilongjiang Province, College of Plant Protection, Northeast Agricultural University, Harbin 150030, China
| | - Haifeng Zhang
- Key Laboratory of Agricultural Microbiology of Heilongjiang Province, College of Plant Protection, Northeast Agricultural University, Harbin 150030, China
| | - Kuan Zhang
- Key Laboratory of Agricultural Microbiology of Heilongjiang Province, College of Plant Protection, Northeast Agricultural University, Harbin 150030, China
| | - Lifeng Guo
- Key Laboratory of Agricultural Microbiology of Heilongjiang Province, College of Plant Protection, Northeast Agricultural University, Harbin 150030, China
| | - Ji-Dong Wang
- Key Laboratory Vector Biology & Pathogen Control Zhejiang Province, College Life Science, Huzhou University, Huzhou 313000, China
| | - Dong-Lan Tian
- Crop Protection, Syngenta China, Bocheng Lu 567, Shanghai 200120, China
| | - Qingtao Yu
- Harbin Academy of Agricultural Sciences, Harbin 150030, China
| | - Xiangjing Wang
- Key Laboratory of Agricultural Microbiology of Heilongjiang Province, College of Plant Protection, Northeast Agricultural University, Harbin 150030, China
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junwei Zhao
- Key Laboratory of Agricultural Microbiology of Heilongjiang Province, College of Plant Protection, Northeast Agricultural University, Harbin 150030, China
| | - Wensheng Xiang
- Key Laboratory of Agricultural Microbiology of Heilongjiang Province, College of Plant Protection, Northeast Agricultural University, Harbin 150030, China
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
2
|
Rui S, Fengrui G, Yining Z, Hong S, Xuewen Y, Changping W, Chunjia Y. Biological activity of secondary metabolites of actinomycetes and their potential sources as antineoplastic drugs: a review. Front Microbiol 2025; 16:1550516. [PMID: 40406345 PMCID: PMC12095151 DOI: 10.3389/fmicb.2025.1550516] [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: 12/23/2024] [Accepted: 04/17/2025] [Indexed: 05/26/2025] Open
Abstract
Actinomycetes are an important group of Gram-positive bacteria, renowned for their ability to produce a wide array of structurally diverse and biologically active secondary metabolites. These secondary metabolites have significant applications in fields such as antimicrobial and antifungal treatments and show tremendous potential in cancer research. To comprehensively review the antitumor potential of actinomycetes-derived secondary metabolites, we conducted a systematic literature search across PubMed, Web of Science, and Scopus databases, covering the period from January 2019 to January 2024. The search used keywords including "actinomycetes," "secondary metabolites," "antitumor," "cancer therapy," "bioactivity," and "clinical application." A total of 95 relevant articles were identified through database searches. After applying inclusion and exclusion criteria, 87 articles were deemed eligible and fully reviewed in this article. These studies highlighted diverse structural classes of actinomycetes-derived antitumor compounds, including polyketides, non-ribosomal peptides, alkaloids, and terpenoids. Many of these metabolites exhibit potent anticancer properties through mechanisms such as inducing apoptosis, inhibiting proliferation, disrupting tumor microenvironment, and targeting key oncogenic signaling pathways. This review underscores the crucial role of actinomycetes secondary metabolites as an invaluable resource for antitumor drug discovery, offering new scientific insights into natural product-based cancer therapies, expanding the molecular toolbox for clinical oncology, and ultimately contributing to public health by advancing effective and innovative treatment options for cancer patients.
Collapse
Affiliation(s)
- Sun Rui
- College of Biology and Agriculture, Jiamusi University, Jiamusi, China
| | - Guo Fengrui
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Zhang Yining
- College of Biology and Agriculture, Jiamusi University, Jiamusi, China
| | - Shao Hong
- College of Biology and Agriculture, Jiamusi University, Jiamusi, China
| | - Yang Xuewen
- College of Biology and Agriculture, Jiamusi University, Jiamusi, China
| | - Wang Changping
- College of Biology and Agriculture, Jiamusi University, Jiamusi, China
| | - Yang Chunjia
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| |
Collapse
|
3
|
Sethi Y, Vora V, Anyagwa OE, Turabi N, Abdelwahab M, Kaiwan O, Chopra H, Attia MS, Yahya G, Emran TB, Padda I. Streptomyces Paradigm in Anticancer Therapy: A State-of-the Art Review. CURRENT CANCER THERAPY REVIEWS 2024; 20:386-401. [DOI: 10.2174/0115733947254550230920170230] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/09/2023] [Accepted: 08/16/2023] [Indexed: 01/12/2025]
Abstract
Abstract:
Cancer is one of the biggest threats to human health with a global incidence of 23.6 million,
mortality of 10 million, and an estimated 250 million lost in disability-adjusted life years
(DALYs) each year. Moreover, the incidence, mortality, and DALYs have increased over the past
decade by 26.3%, 20.9%, and 16.0%, respectively. Despite significant evolutions in medical therapy
and advances in the DNA microarray, proteomics technology, and targeted therapies, anticancer drug
resistance continues to be a growing concern and invites regular discovery of potent agents. One such
agent is the microbe-producing bioactive compounds like Streptomyces, which are proving increasingly
resourceful in anticancer therapy of the future. Streptomyces, especially the species living in
extreme conditions, produce bioactive compounds with cytolytic and anti-oxidative activity which
can be utilized for producing anticancer and chemo-preventive agents. The efficacy of the derived
compounds has been proven on cell lines and some of these have already established clinical results.
These compounds can potentially be utilized in the treatment of a variety of cancers including but not
limited to colon, lung, breast, GI tract, cervix, and skin cancer. The Streptomyces, thus possess the
armory to fuel the anticancer agents of the future and help address the problem of rising resistance to
currently available anti-cancer drugs. We conducted a state-of-art review using electronic databases
of PubMed, Scopus, and Google scholar with an objective to appraise the currently available literature
on Streptomyces as a source of anti-cancer agents and to compile the clinically significant literature
to update the clinicians.
Collapse
Affiliation(s)
- Yashendra Sethi
- PearResearch, Dehradun 248001, India
- Department of Medicine, Government Doon Medical College, HNB Uttarakhand
Medical Education University, Dehradun, Uttarakhand, India
| | - Vidhi Vora
- Department of Medicine, Government Doon Medical College, HNB Uttarakhand
Medical Education University, Dehradun, Uttarakhand, India
- Department of Medicine, Lokmanya Tilak Municipal
Medical College and Sion Hospital, Maharashtra University of Health Sciences, Mumbai, Maharashtra, India
| | | | | | | | - Oroshay Kaiwan
- Department of Medicine, Government Doon Medical College, HNB Uttarakhand
Medical Education University, Dehradun, Uttarakhand, India
- Department of Medicine, Northeast Ohio Medical University, Ohio,
USA
| | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences,
Chennai- 602105, Tamil Nadu, India
| | - Mohamed Shah Attia
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University,
Zagazig 44519, Egypt
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig
44519, Egypt
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Inderbir Padda
- Department of Medicine, Richmond University Medical Centre, Staten Island, NY, USA
| |
Collapse
|
4
|
Alzain AA, Elbadwi FA, Mohamed SGA, Kushk KSA, Bafarhan RI, Alswiri SA, Khushaim SN, Hussein HGA, Abuhajras MYA, Mohamed GA, Ibrahim SRM. Exploring marine-derived compounds for MET signalling pathway inhibition in cancer: integrating virtual screening, ADME profiling and molecular dynamics investigations. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2023; 34:1003-1021. [PMID: 38014514 DOI: 10.1080/1062936x.2023.2284917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023]
Abstract
The MET signalling pathway regulates fundamental cellular processes such as growth, division, and survival. While essential for normal cell function, dysregulation of this pathway can contribute to cancer by triggering uncontrolled proliferation and metastasis. Targeting MET activity holds promise as an effective strategy for cancer therapy. Among potential sources of anti-cancer agents, marine organisms have gained attention. In this study, we screened 47,450 natural compounds derived from marine sources within the CMNPD database against the Met crystal structure. By employing HTVS, SP, and XP docking modes, we identified three compounds (CMNPD17595, CMNPD14026, and CMNPD19696) that outperformed a reference molecule in binding affinity to the Met structure. These compounds demonstrated desirable ADME properties. Molecular Dynamics (MD) simulations for 200 ns confirmed the stability of their interactions with Met. Our findings highlight CMNPD17595, CMNPD14026, and CMNPD19696 as potential inhibitors against Met-dependent cancers. Additionally, these compounds offer new avenues for drug development, leveraging their inhibitory effects on Met to combat carcinogenesis.
Collapse
Affiliation(s)
- A A Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - F A Elbadwi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - S G A Mohamed
- Faculty of Dentistry, British University, El Sherouk City, Egypt
| | - K S A Kushk
- Operations Sales Department, United Pharmaceuticals & Medical Supply Co. Ltd, Al Madinah Al-Munawwarah, Saudi Arabia
| | - R I Bafarhan
- Pharmaceutical Care Services, Medical Department, Private Sector, Tabuk, Saudi Arabia
| | - S A Alswiri
- Pharmaceutical Company, Medical Department, Private Sector, Al Madinah Al-Munawwarah, Saudi Arabia
| | - S N Khushaim
- College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - H G A Hussein
- Preparatory Year Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - M Y A Abuhajras
- Medical Claims Department, Bupa Arabia, Prince Saud AlFaisal, Jeddah, Saudi Arabia
| | - G A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - S R M Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
5
|
Ai B, Mei Y, Liang D, Wang T, Cai H, Yu D. Uncovering the special microbiota associated with occurrence and progression of gastric cancer by using RNA-sequencing. Sci Rep 2023; 13:5722. [PMID: 37029259 PMCID: PMC10082026 DOI: 10.1038/s41598-023-32809-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
Gastric cancer (GC) has been identified as the third deadly cancer in the world. Accumulating researches suggest a potential role of microorganisms in tumorigenesis. However, the composition of microbiota in GC tissues is not clear and it changes throughout the different stages of GC remain mostly elusive. Our study integrated RNA-Seq data of 727 samples derived from gastric tissues across four datasets and revealed its microbial composition. In order to remove the false positive results, core taxa were defined and characterized. Based on it, we analyzed the influence of biological factors on its composition. The pan-microbiome of gastric tissues was estimated to be over than 1400 genera. Seventeen core genera were identified. Among them, Helicobacter, Lysobacter were significantly enriched in normal tissues, while Pseudomonas was enriched in tumor tissues. Interestingly, Acinetobacter, Pasteurella, Streptomyces, Chlamydia, and Lysobacter, showed a significant increase trend during tumor development and formed strong intra/inter-correlations among them or with other genera. Furthermore, we found that tumor stage played an important role in altering the microbial composition of GC tissues. This study provides support for the in-depth study of tumor microbiome, and the specific microbiome excavated provides a possibility for the subsequent identification of potential biomarkers for GC.
Collapse
Affiliation(s)
- Bin Ai
- Department of Precision Medicine, Translational Medicine Research Center, Naval Medical University, Shanghai, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai, China
| | - Yue Mei
- Department of Precision Medicine, Translational Medicine Research Center, Naval Medical University, Shanghai, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai, China
| | - Dong Liang
- Department of Precision Medicine, Translational Medicine Research Center, Naval Medical University, Shanghai, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai, China
| | - Tengjiao Wang
- Department of Precision Medicine, Translational Medicine Research Center, Naval Medical University, Shanghai, China
- Shanghai Key Laboratory of Cell Engineering, Shanghai, China
| | - Hui Cai
- Department of Gastrointestinal Surgery, Changhai Hospital, Shanghai, China.
| | - Dong Yu
- Department of Precision Medicine, Translational Medicine Research Center, Naval Medical University, Shanghai, China.
- Shanghai Key Laboratory of Cell Engineering, Shanghai, China.
| |
Collapse
|
6
|
Jia Y, Yang H, Yu J, Li Z, Jia G, Ding B, Lv C. Crocin suppresses breast cancer cell proliferation by down-regulating tumor promoter miR-122-5p and up-regulating tumor suppressors FOXP2 and SPRY2. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 36988377 DOI: 10.1002/tox.23789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 03/13/2023] [Accepted: 03/19/2023] [Indexed: 06/19/2023]
Abstract
Crocin has been reported to have antitumor activity in several tumors including breast cancer. Nevertheless, the mechanism of action of crocin on breast cancer remains unclear. The cytotoxicity of crocin was evaluated by CCK-8 assay. Cell proliferation was assessed using EdU incorporation assay and western blot analysis. Breast cancer-related genes were extracted from GEPIA. miR-122-5p targets were predicted using Targetscan, starbase, and miRDB softwares. Luciferase reporter assay was employed to confirm whether miR-122-5p targeted sprouty2 (SPRY2) and forkhead box P2 (FOXP2). Results showed that crocin exhibited cytotoxicity and suppressed the proliferation in breast cancer cells. miR-122-5p was upregulated in breast cancer tissues and cells. Crocin suppressed miR-122-5p to block the proliferation of breast cancer cells. Seven targets of miR-122-5p were identified in breast cancer. SPRY2 and FOXP2 were selected for further experiments due to their involvement in breast cancer. miR-122-5p targeted SPRY2 and FOXP2 to inhibit their expression. miR-122-5p knockdown restrained breast cancer cell proliferation by targeting SPRY2 and FOXP2. Additionally, crocin increased SPRY2 and FOXP2 expression by inhibiting miR-122-5p expression. Together, our results suggested that crocin inhibited proliferation of breast cancer cells through decreasing miR-122-5p expression and the subsequent increase of SPRY2 and FOXP2 expression.
Collapse
Affiliation(s)
- Yunhao Jia
- Department of General Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, Henan, 473004, China
| | - Han Yang
- Department of Endocrinology, Nanshi Hospital Affiliated to Henan University, Nanyang, Henan, 473065, China
| | - Jinsong Yu
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, Henan, 473004, China
- Key Laboratory of Thyroid Tumor Prevention and Treatment of Nanyang, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, Henan, 473004, China
| | - Zhong Li
- Department of General Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, Henan, 473004, China
| | - Guangwei Jia
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, Henan, 473004, China
| | - Bo Ding
- Department of General Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, Henan, 473004, China
| | - Chunliu Lv
- Department of Breast Tumor Plastic Surgery (Department of Head and Neck Surgery), Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| |
Collapse
|
7
|
Hashemi M, Paskeh MDA, Orouei S, Abbasi P, Khorrami R, Dehghanpour A, Esmaeili N, Ghahremanzade A, Zandieh MA, Peymani M, Salimimoghadam S, Rashidi M, Taheriazam A, Entezari M, Hushmandi K. Towards dual function of autophagy in breast cancer: A potent regulator of tumor progression and therapy response. Biomed Pharmacother 2023; 161:114546. [PMID: 36958191 DOI: 10.1016/j.biopha.2023.114546] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 03/25/2023] Open
Abstract
As a devastating disease, breast cancer has been responsible for decrease in life expectancy of females and its morbidity and mortality are high. Breast cancer is the most common tumor in females and its treatment has been based on employment of surgical resection, chemotherapy and radiotherapy. The changes in biological behavior of breast tumor relies on genomic and epigenetic mutations and depletions as well as dysregulation of molecular mechanisms that autophagy is among them. Autophagy function can be oncogenic in increasing tumorigenesis, and when it has pro-death function, it causes reduction in viability of tumor cells. The carcinogenic function of autophagy in breast tumor is an impediment towards effective therapy of patients, as it can cause drug resistance and radio-resistance. The important hallmarks of breast tumor such as glucose metabolism, proliferation, apoptosis and metastasis can be regulated by autophagy. Oncogenic autophagy can inhibit apoptosis, while it promotes stemness of breast tumor. Moreover, autophagy demonstrates interaction with tumor microenvironment components such as macrophages and its level can be regulated by anti-tumor compounds in breast tumor therapy. The reasons of considering autophagy in breast cancer therapy is its pleiotropic function, dual role (pro-survival and pro-death) and crosstalk with important molecular mechanisms such as apoptosis. Moreover, current review provides a pre-clinical and clinical evaluation of autophagy in breast tumor.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sima Orouei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Pegah Abbasi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amir Dehghanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negin Esmaeili
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Azin Ghahremanzade
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 4815733971, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari 4815733971, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
8
|
Bai LY, Wu KLH, Chiu CF, Chao HC, Lin WY, Hu JL, Peng BR, Weng JR. Extract of Ficus septica modulates apoptosis and migration in human oral squamous cell carcinoma cells. ENVIRONMENTAL TOXICOLOGY 2023; 38:666-675. [PMID: 36436203 DOI: 10.1002/tox.23716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/08/2022] [Accepted: 11/20/2022] [Indexed: 06/16/2023]
Abstract
According to the alarming statistical analysis of global cancer, there are over 19 million new diagnoses and more than 10 million deaths each year. One such cancer is the oral squamous cell carcinoma (OSCC), which requires new therapeutic strategies. Ficus septica extract has been used in traditional medicine to treat infectious diseases. In this study, we examined the anti-proliferative effects of an extract of F. septica bark (FSB) in OSCC cells. Our results showed that FSB caused a concentration-dependent reduction in the viability of SCC2095 OSCC cells, as determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays, and was less sensitive to fibroblasts. In addition, FSB induced apoptosis by activating caspases, accompanied by the modulation of Akt/mTOR/NF-κB and mitogen-activated protein kinase signaling. Moreover, FSB increased reactive oxygen species generation in a concentration-dependent manner in SCC2095 cells. Furthermore, FSB inhibited cell migration and modulated the levels of the cell adhesion molecules including E-cadherin, N-cadherin, and Snail in SCC2095 cells. Pinoresinol, a lignan isolated from FSB, showed antitumor effects in SCC2095 cells, implying that this compound might play an important role in FSB-induced OSCC cell death. Taken together, FSB is a potential anti-tumor agent against OSCC cells.
Collapse
Affiliation(s)
- Li-Yuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Kay Li-Hui Wu
- Institute of Translational Research in Biomedicine, Kaohsiung Chang-Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chang-Fang Chiu
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- Cancer Center, China Medical University Hospital, Taichung, Taiwan
| | - Hong-Chu Chao
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Wei-Yu Lin
- Department of Pharmacy, Kinmen Hospital, Ministry of Health and Welfare, Kinmen, Taiwan
| | - Jing-Lan Hu
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Bo-Rong Peng
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan
- National Museum of Marine Biology & Aquarium, Pingtung, Taiwan
| | - Jing-Ru Weng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
9
|
Wei J, Liu Y, Teng F, Li L, Zhong S, Luo H, Huang Z. Anticancer effects of marine compounds blocking the nuclear factor kappa B signaling pathway. Mol Biol Rep 2022; 49:9975-9995. [PMID: 35674876 DOI: 10.1007/s11033-022-07556-1] [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: 11/10/2021] [Accepted: 05/03/2022] [Indexed: 11/26/2022]
Abstract
The abnormal expression of nuclear factor kappa B (NF-κB) target genes is closely related to the occurrence, metastasis, and invasion of tumor cells and is an inhibitor of their apoptosis. In recent years, the unique biodiversity in the marine environment has aroused great interest. Many studies indicate that some marine compounds exert anticancer effects on most common human tumors by modulating the NF-κB signaling pathway. In this study, 26 marine compounds that reduce cancer cell survival by suppressing the NF-κB signaling pathway were reviewed. They were derived from a wide range of sources, including sponges, fungi, algae and their derivatives or metabolites. These marine compounds exert antitumor effects through the canonical, noncanonical and atypical NF-κB signaling pathways; however, most of their anticancer targets and mechanisms remain unclear, and more research is needed in the future. Our article provides comprehensive information for researchers investigating the bioactivities of marine compounds and developing marine-derived anticancer drugs.
Collapse
Affiliation(s)
- Jiaen Wei
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, Guangdong, China
| | - Yaqi Liu
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, Guangdong, China
| | - Fei Teng
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, Guangdong, China
| | - Linshan Li
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, Guangdong, China
| | - Shanhong Zhong
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, Guangdong, China
| | - Hui Luo
- Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, 524023, Guangdong, China.
| | - Zunnan Huang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, Guangdong, China.
- Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, 524023, Guangdong, China.
| |
Collapse
|
10
|
Chu PC, Dokla EME, Hu JL, Weng JR. Induction of apoptosis using ATN as a novel Yes-associated protein inhibitor in human oral squamous cell carcinoma cells. ENVIRONMENTAL TOXICOLOGY 2022; 37:1404-1412. [PMID: 35212453 DOI: 10.1002/tox.23493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/13/2022] [Accepted: 02/13/2022] [Indexed: 06/14/2023]
Abstract
Oral squamous cell carcinoma (OSCC) represents a clinical challenge due to the lack of effective therapy to improve prognosis. Hippo/Yes-associated protein (YAP) signaling has emerged as a promising therapeutic target for squamous cell carcinoma treatment. In this study, we investigated the antitumor activity and underlying mechanisms of {[N-(4-(5-(3-(3-(4-acetamido-3-(trifluoromethyl)phenyl)ureido)phenyl)-1,2,4-oxadiazol-3-yl)-3-chlorophenyl)-nicotinamide]} (ATN), a novel YAP inhibitor, in OSCC cells. ATN exhibited differential antiproliferative efficacy against OSCC cells (IC50 as low as 0.29 μM) versus nontumorigenic human fibroblast cells (IC50 = 1.9 μM). Moreover, ATN effectively suppressed the expression of YAP and YAP-related or downstream targets, including Akt, p-AMPK, c-Myc, and cyclin D1, which paralleled the antiproliferative efficacy of ATN. Supporting the roles of YAP in regulating cancer cell survival and migration, ATN not only induced caspase-dependent apoptosis, but also suppressed migration activity in OSCC. Mechanistically, the antitumor activity of ATN in OSCC was attributed, in part, to its ability to regulate Mcl-1 expression. Together, these findings suggest a translational potential of YAP inhibitors, represented by ATN as anticancer therapy for OSCC.
Collapse
Affiliation(s)
- Po-Chen Chu
- Department of Cosmeceutics and Graduate Institute of Cosmeceutics, China Medical University, Taichung, Taiwan
| | - Eman M E Dokla
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Jing-Lan Hu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Jing-Ru Weng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
11
|
Synthetic Tryptanthrin Derivatives Induce Cell Cycle Arrest and Apoptosis via Akt and MAPKs in Human Hepatocellular Carcinoma Cells. Biomedicines 2021; 9:biomedicines9111527. [PMID: 34829756 PMCID: PMC8615277 DOI: 10.3390/biomedicines9111527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/12/2021] [Accepted: 10/20/2021] [Indexed: 12/20/2022] Open
Abstract
Trytanthrin, found in Ban-Lan-Gen, is a natural product containing an indoloquinazoline moiety and has been shown to possess anti-inflammatory and anti-viral activities. Chronic inflammation and hepatitis B are known to be associated with the progression of hepatocellular carcinoma (HCC). In this study, a series of tryptanthrin derivatives were synthesized to generate potent anti-tumor agents against HCC. This effort yielded two compounds, A1 and A6, that exhibited multi-fold higher cytotoxicity in HCC cells than the parent compound. Flow cytometric analysis demonstrated that A1 and A6 caused S-phase arrest and downregulated the expression of cyclin A1, B1, CDK2, and p-CDC2. In addition to inducing caspase-dependent apoptosis, A1 and A6 exhibited similar regulation of the phosphorylation or expression of multiple signaling targets, including Akt, NF-κB, and mitogen-activated protein kinases. The anti-tumor activities of A1 and A6 were also attributable to the generation of reactive oxygen species, accompanied by an increase in p-p53 levels. Therefore, A1 and A6 have potential clinical applications since they target diverse aspects of cancer cell growth in HCC.
Collapse
|
12
|
Zhang N, Xue M, Wang Q, Liang H, Yang J, Pei Z, Qin K. Inhibition of fucoidan on breast cancer cells and potential enhancement of their sensitivity to chemotherapy by regulating autophagy. Phytother Res 2021; 35:6904-6917. [PMID: 34687482 DOI: 10.1002/ptr.7303] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/24/2022]
Abstract
Fucoidan is a marine-origin sulfated polysaccharide that has gained attention for its anticancer activities. However, the inhibitory effect of fucoidan on breast cancers by regulating autophagy and its mechanism are not clear, and the chemotherapeutic sensitization of fucoidan is largely unknown. In the present study, the anticancer potential of fucoidan was revealed in MCF-7 and MDA-MB-231 cells. Additionally, we also studied the chemotherapeutic sensitization of fucoidan by combining chemotherapeutic drugs doxorubicin (ADM) and cisplatin (DDP) with fucoidan on breast cancer cells. In the two kinds of human breast cancer cells, cell viability was determined by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. Apoptosis was examined with flow cytometry. Transfection assay was used to examine autophagy flow. Western blot was used to examine the expressions of related proteins. Results suggested that fucoidan could induce autophagy and might enhance the sensitivity of breast cancer cells to chemotherapeutic drugs. Mechanistically, fucoidan induced autophagy in breast cancer cells by down-regulating m-TOR/p70S6K/TFEB pathway. In conclusion, our research revealed that fucoidan could induce autophagy of breast cancer cells by mediating m-TOR/p70S6K/TFEB pathway, thus inhibiting tumor development. Furthermore, fucoidan might enhance the sensitivity of breast cancer cells to ADM and DDP, and this enhancement was related to autophagy.
Collapse
Affiliation(s)
- Nan Zhang
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Qing Wang
- Department of Ophthalmology, Affiliated Hospital of Qingdao, Qingdao, China
| | - Hui Liang
- The Institute of Human Nutrition, Qingdao University of Medicine, Qingdao, China
| | - Jia Yang
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Zhongqian Pei
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Kunpeng Qin
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| |
Collapse
|