1
|
Jin SK, Baek KH. Unraveling the role of deubiquitinating enzymes on cisplatin resistance in several cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189297. [PMID: 40058507 DOI: 10.1016/j.bbcan.2025.189297] [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: 01/07/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
The use of platinum-based drugs in cancer treatment is one of the most common methods in chemotherapy. Especially, cisplatin induces cell death by interrupting DNA synthesis by binding to the DNA bases, thereby leading to the apoptosis via multiple pathways. However, the major hurdle in chemotherapy is drug resistance. To overcome drug resistance, the ubiquitin-proteasome system (UPS) has emerged as a potential therapeutic target. The UPS is a pivotal signaling pathway that regulates the majority of cellular proteins by attaching ubiquitin to substrates, leading to proteasomal degradation. Conversely, deubiquitinating enzymes (DUBs) remove tagged ubiquitin from the substrate and inhibit degradation, thereby maintaining proteostasis. Recently, studies have been conducted to identify the substrates of DUBs and investigated the cellular mechanisms, and now the development of therapeutics using DUB inhibitors is in clinical trials. However, the mechanism of the DUB response to cisplatin remains still unclear. In this review, we summarize the research reported on the function of DUBs responding to cisplatin.
Collapse
Affiliation(s)
- Sun-Kyu Jin
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea.
| |
Collapse
|
2
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
3
|
Gedik D, Eraslan G. Evaluation of the efficacy of diosmin and chrysin against tau-fluvalinate exposure in rats. Food Chem Toxicol 2025; 195:115097. [PMID: 39522795 DOI: 10.1016/j.fct.2024.115097] [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: 08/20/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Tau-fluvalinate is a type 2 pyrethroid insecticide. Diosmin and chrysin are flavonoids with antioxidant and anti-apoptotic effects. Role of diosmin and chrysin against infavorable toxic effects caused by tau-fluvalinate and the underlying mechanisms of these effects were investigated. Six groups were formed and diosmin, chrysin, tau-fluvalinate, tau-fluvalinate + diosmin and tau-fluvalinate + chrysin were administered orally to rats at a dose of 20 mg/kg.bw except for the control group, once a day for 21 days, respectively. Tau-fluvalinate elevated MDA and NO levels while diminishing the activities of antioxidant enzymes (SOD, CAT, GSH-Px, GR, GST, G6PD) and GSH levels in the majority of the analyzed blood and tissues, statistically significant. Serum triglyceride, cholesterol, total protein and albumin levels as well as LDH and PChE activities decreased. Conversely, serum creatinine, AST, ALT and ALP levels/activities increased. Elevated protein levels of caspase 3, caspase 9, p53 and Bax and decreased protein levels of Bcl-2 were observed in the liver. There were negative changes in body/some organ weights. Diosmin and chrysin administration resulted in a marked recovery in tau-fluvalinate-induced toxic effects, but this improvement was not complete. These flavonoids may be considered as promising potential therapeutic options to alleviate the adverse effects associated with tau-fluvalinate intoxication.
Collapse
Affiliation(s)
- Didem Gedik
- Department of Veterinary Pharmacology and Toxicology, Institute of Health Science, Erciyes University, Kayseri, Turkey
| | - Gökhan Eraslan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey.
| |
Collapse
|
4
|
Sampada MP, David M. Mercuric chloride induced reproductive toxicity associated with oxidative damage in male Wistar albino rat, Rattus norvegicus. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03585-8. [PMID: 39738833 DOI: 10.1007/s00210-024-03585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/28/2024] [Indexed: 01/02/2025]
Abstract
In the field of toxicology, male reproductive hazards attributed to metal exposure is a fast-developing issue. Mercury has been identified as an environmental pollutant that causes potential adverse impacts on organisms. This study aimed to assess the reprotoxic consequences of mercuric chloride (HgCl2). Five groups of sexually mature albino rats were given oral mercuric chloride (HgCl2) treatment. (G1) control group received saline treatment; (G2) (5.25 mg/kg of HgCl2 for 30 days); (G3) (5.25 mg/kg of HgCl2 for 60 days); (G4) (10.5 mg/kg of HgCl2 for 30 days); (G5) (10.5 mg/kg of HgCl2 for 60 days). The hormonal levels, sperm count, sperm motility, sperm viability, and reproductive organ weight, including body weight, were substantially reduced, whereas the sperm abnormality rate was enhanced in rat groups treated with HgCl2. The analysis revealed that the effect size (Cohen's d) for sperm parameters, including sperm count, motility and viability, were extremely high across all groups, except for sperm abnormality in group 2 (d = 0.59) and group 3 (d = 0.18), where moderate and small effect sizes were observed respectively, and this suggests a significant impact of the intervention on sperm parameters. The administration of HgCl2 resulted in the induction of oxidative stress in testis that is manifested by substantially enhanced lipid peroxidation (MDA) with a substantial decrease in activity of antioxidant enzymes like catalase (CAT), superoxide dismutase (SOD), reduced glutathione (GSH), and glutathione peroxidase (GPx) in testes of mercury-treated groups. Concomitantly, there was downregulation in the mRNA levels of the genes involved in spermatogenesis, namely Hsp-70, insulin-like growth factor (IGF), glutathione-S-transferase, and p53 in the testis. The expression of antiapoptotic protein B cell lymphoma (Bcl-2) was decreased, and conversely, the expression of cell proliferative protein Ki-67 was increased in a dose- and duration-dependent manner. Histopathological studies showed degenerative changes in the testis, epididymis, prostate gland, and seminal vesicle, compared to the control group. All the evidence suggests that after mercury exposure, there may be an imbalance between the body's defenses against free radicals and antioxidants, making the testis more susceptible to oxidative damage. This imbalance could potentially have a detrimental effect on the function of the male reproductive system.
Collapse
Affiliation(s)
- M P Sampada
- Zoology, Karnataka University, Dharwad, Karnataka, 580003, India
| | - Muniswamy David
- Zoology, Karnataka University, Dharwad, Karnataka, 580003, India.
| |
Collapse
|
5
|
Tu W, Guo M, Zhang Z, Li C. Pathogen-induced apoptosis in echinoderms: A review. FISH & SHELLFISH IMMUNOLOGY 2024; 155:109990. [PMID: 39481501 DOI: 10.1016/j.fsi.2024.109990] [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: 08/04/2024] [Revised: 10/16/2024] [Accepted: 10/26/2024] [Indexed: 11/02/2024]
Abstract
Echinoderms possess unique biological traits that make them valuable models in immunology, regeneration, and developmental biology studies. As a class rich in active substances with significant nutritional and medicinal value, echinoderms face threats from marine pathogens, including bacteria, viruses, fungi, protozoa, and parasites, which have caused substantial economic losses in echinoderm aquaculture. Echinoderms counteract pathogen invasion through innate immunity and programmed cell death, in particular, with apoptosis being essential for eliminating infected or damaged cells and maintaining homeostasis in many echinoderm cell types. Despite the importance of this process, there is a lack of comprehensive and updated reviews on this topic. This review underscores that echinoderm apoptotic pathways exhibit a complexity comparable to that of vertebrates, featuring proteins with unique domains that may indicate the presence of novel signaling mechanisms. We synthesize current knowledge on how echinoderms utilize diverse transcriptional and post-transcriptional mechanisms to regulate apoptosis in response to pathogen infections and explore how pathogens have evolved strategies to manipulate echinoderm apoptosis, either by inhibiting it to create survival niches or by inducing excessive apoptosis to weaken the host. By elucidating the primary apoptotic pathways in echinoderms and the host-pathogen interactions that modulate these pathways, this review aims to reveal new mechanisms of apoptosis in animal immune defense and provide insights into the evolutionary arms race between hosts and pathogens.
Collapse
Affiliation(s)
- Weitao Tu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Ming Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China.
| | - Zhen Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China.
| |
Collapse
|
6
|
Zhao Q, Zhang S, Feng W, Zhou A, Shi L, Zhang J. Deoxynivalenol-mediated kidney injury via endoplasmic reticulum stress in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117243. [PMID: 39447294 DOI: 10.1016/j.ecoenv.2024.117243] [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: 03/22/2024] [Revised: 10/12/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVE Deoxynivalenol (DON) is a common fungal toxin that poses significant health risks to humans and animals. The present study aimed to investigate the adverse effects and molecular mechanisms of DON-induced kidney injury. METHODS Male C57BL/6 mice aged 5-6 weeks were used to establish a DON-induced acute kidney injury model. Histological analysis, biochemical assays, molecular techniques, Western blot, RNA sequencing, and transmission electron microscopy were employed to analyze kidney damage, inflammation, oxidative stress, apoptosis, and endoplasmic reticulum (ER) stress. RESULTS DON disrupted kidney morphology, induced inflammatory cell infiltration, and triggered inflammatory responses. DON increased MDA content while decreasing antioxidant enzyme activity (SOD and CAT). It also triggered apoptosis, evidenced by elevated levels of caspase-12, cleaved caspase-3, and BAX, and reduced levels of Bcl-2. Transcriptomic analysis identified distinct expression patterns in 1756 genes in DON-exposed mouse kidneys, notably upregulating ER stress-related genes. Further investigation revealed ultrastructural changes in the ER and mitochondrial damage induced by DON, along with increased levels of p-IRE1, p-PERK, and their downstream targets, indicating unfolded protein response (UPR) activation in the kidney. The ER stress inhibitor 4-Phenylbutyric acid (4-PBA) significantly mitigated DON-induced ER stress, oxidative damage, apoptosis, tissue injury, ER expansion, and mitochondrial damage. CONCLUSION Our findings highlight the role of ER stress in DON-induced kidney injury and the protective effect of 4-PBA against these adverse effects.
Collapse
Affiliation(s)
- Qingbo Zhao
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Siyi Zhang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Weili Feng
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Ao Zhou
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Liangyu Shi
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jing Zhang
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming & Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
7
|
Wang Y, Lao Y, Li R, You C, Qing L, Xiao X, Liu S, Wang W, Zhao Y, Dong Z. Network pharmacological analysis and experimental study of melatonin in chronic prostatitis/chronic pelvic pain syndrome. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8691-8706. [PMID: 38822120 DOI: 10.1007/s00210-024-03183-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 05/22/2024] [Indexed: 06/02/2024]
Abstract
This study is aimed at exploring the potential mechanisms of melatonin (MT) in treating chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) using network pharmacology and experimental study. The target genes of MT were acquired from the Swiss Target Prediction, SuperPred, SEA, and PharmMapper databases, and the CP/CPPS targets were collected based on OMIM, DisGeNET, and GeneCards databases. The intersection of MT and CP/CPPS target genes was analyzed. A PPI network was constructed using Cytoscape to identify core targets. The shared targets underwent GO and KEGG enrichment analyses by Using R software. Molecular docking of MT with core targets was performed using AutoDock and PyMOL. GROMACS software was used for molecular dynamics simulation. And using cell experiments to verify the potential effect of MT in CP/CPPS. Network pharmacology analysis reveals 284 shared targets between MT and CP/CPPS, with AKT1, SRC, HSP90AA1, PTGS2, BCL2L1, ALB, CASP3, NFKB1, HIF1A, and ESR1 identified as key targets. Enrichment analysis indicates that MT affects CP/CPPS through various biological processes, and pathway analysis emphasizes the significance of PI3K-Akt, MAPK, Ras, FoxO, HIF-1, EGFR, and apoptosis pathways. Molecular docking confirms strong binding between MT and core targets. It is worth noting that the molecular dynamics simulation showed that the average binding free energy of AKT1, PTGS2, ALB, HSP90AA1 proteins, and MT was - 26.15, - 29.48, - 18.59, and - 20.09 kcal/mol, respectively. These results indicated that AKT1, PTGS2, ALB, and HSP90AA1 proteins were strongly bound to MT. Cell experiments demonstrate that MT can inhibit the secretion of IL-1β, IL-6, and TNF-α in LPS-induced RWPE-1 cells, alleviate inflammation, and suppress cell apoptosis and oxidative stress. Network pharmacology, molecular docking, molecular dynamics simulation, and cell experiments showed that MT could play a role in CP/CPPS by regulating multiple targets and pathways. These findings provide an important scientific basis for further exploration of the molecular mechanism and clinical application of MT in CP/CPPS treatment and are expected to provide new ideas and directions for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
- Institute of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu, China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, 730030, Gansu, China
| | - Yongfeng Lao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
- Institute of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu, China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, 730030, Gansu, China
| | - Rongxin Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
- Institute of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu, China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, 730030, Gansu, China
| | - Chengyu You
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
- Institute of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu, China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, 730030, Gansu, China
| | - Liangliang Qing
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
- Institute of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu, China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, 730030, Gansu, China
| | - Xi Xiao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
- Institute of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu, China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, 730030, Gansu, China
| | - Shuai Liu
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
- Institute of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu, China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, 730030, Gansu, China
| | - Wenyun Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
- Institute of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu, China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, 730030, Gansu, China
| | - Yu Zhao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
- Institute of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu, China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, 730030, Gansu, China
| | - Zhilong Dong
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China.
- Institute of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu, China.
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
8
|
Rahmani F, Ajoudanifar H, Arbab Soleimani N, Imani Fooladi AA. Targeted therapies in HER2-positive breast cancer with receptor-redirected Arazyme-linker-Herceptin as a novel fusion protein. Breast Cancer 2024; 31:1101-1113. [PMID: 39122876 DOI: 10.1007/s12282-024-01625-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] [Received: 12/15/2023] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Targeted treatment of different types of cancers through highly expressed cancer cell surface receptors by fusion proteins is an efficient method for cancer therapy. The HER2 receptor is a member of the tyrosine kinase receptors family, which plays a notable role in breast cancer tumor development. About 25-30% of breast cancers overexpress human epidermal growth factor receptor 2 (HER2). METHODS In this study, we evaluated the particulars of a designed recombinant protein formed by HER2-specific Mab Herceptin linked with Arazyme on a HER2-overexpressing breast cancer cell line (SKBR3). Arazyme, a metalloprotease produced by Serratia proteamaculans was fused to the variable area of light and heavy chains of the Herceptin. The cytotoxic assay of the Arazyme-linker-Herceptin in the SKBR3 and MDA-MB-468 cells was evaluated by the MTT and flow cytometry techniques. The Caspase‑3 activity determination and adhesion assay were performed to evaluate the antitumor activity of the Arazyme-linker-Herceptin against SKBR3 cells. Furthermore, RT-PCR was used to measure the expression levels of the Bcl-2, Bax, MMP2, MMP9, and RIP3 genes. RESULTS The Arazyme-linker-Herceptin showed higher cytotoxicity in SKBR3 cells compared to MDA-MB-468 cells. In addition, flow cytometry results revealed that the Arazyme-linker-Herceptin can significantly induce apoptosis in the HER2-overexpressing breast cancer cell line (SKBR3), which was confirmed by Bax upregulation and the decrease in adhesion of tumor cells and MMP2/MMP9. CONCLUSION The findings of this study demonstrated that the Arazyme-linker-Herceptin induced apoptosis and decreased metastatic genes in SKBR3 cells; however, further research is required to confirm the effectiveness of the fusion protein.
Collapse
Affiliation(s)
- Farideh Rahmani
- Department of Microbiology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Hatef Ajoudanifar
- Department of Microbiology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | | | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Wang W, Wang H, Luo Y, Li Z, Li J. Discovery of petroleum ether extract of eclipta targeting p53/Fas pathway for the treatment of chemotherapy-induced alopecia: Network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118405. [PMID: 38844249 DOI: 10.1016/j.jep.2024.118405] [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: 03/29/2024] [Revised: 05/19/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ecliptea herba, a traditional Chinese herbal medicine for hair loss, was first recorded in the Tang Dynasty's 'Qian Jin Yue Ling', of which the active ingredients and mechanisms of action in the treatment of chemotherapy-induced hair loss remain poorly investigated. AIM OF THE STUDY To investigate the effects of the petroleum ether extract of Eclipta (PEE) on alopecia and follicle damage and elucidate its potential therapeutic mechanisms using the integration of network pharmacology, bioinformatics, and experimental validation. MATERIALS AND METHODS UPLC-MS was used to analyse the chemical composition of PEE. A network pharmacology approach was employed to establish the 'components-targets-pathways' network of PEE to explore potential therapeutic pathways and targets. Molecular docking was used for validation, and the mechanism of PEE in treating chemotherapy-induced alopecia (CIA) was elucidated using in vitro and in vivo on CIA models. RESULTS UPLC-MS analysis of PEE revealed 185 components, while network pharmacology and molecular docking analyses revealed potential active compounds and their target molecules, suggesting the involvement of core genes, such as TP53, ESR1, AKT1, IL6, TNF, and EGFR. The key components included wedelolactone, dimethyl-wedelolactone, luteoloside, linarin, and hispidulin. In vivo, PEE promoted hair growth, restored the number of hair follicles, and reduced follicle apoptosis. Conversely, in vitro, PEE enhanced cell viability, reduced apoptosis, and protected HaCaT cells from damage induced by 4-hydroperoxycyclophosphamide (4-HC). CONCLUSIONS PEE alleviated hair follicle damage in CIA mice by inhibiting the P53/Fas pathway, which may be associated with inhibiting hair follicle cell apoptosis. This study provides a novel therapeutic strategy for treating cyclophosphamide-induced hair loss.
Collapse
Affiliation(s)
- Wuji Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China; Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, 563006, China; Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, 563006, China.
| | - Honglan Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China; Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, 563006, China; Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, 563006, China
| | - Yang Luo
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China; Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, 563006, China; Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, 563006, China
| | - Zheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China; Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, 563006, China; Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, 563006, China
| | - Jingjie Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563006, China; Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, 563006, China; Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, 563006, China.
| |
Collapse
|
10
|
Huwaimel B, Mohammed HA, Elkashlan AM, Alnajjar R, Altamimi OA, Alorainan MM, Altuwayhir MK, Algharby SF, Almahmoud SA, Abouzied AS. Unraveling the therapeutic potential of Satureja nabateorum extract: inducing apoptosis and cell cycle arrest through p53, Bax/Bcl-2, and caspase-3 pathways in human malignant cell lines, with in silico insights. J Biomol Struct Dyn 2024:1-18. [PMID: 39460490 DOI: 10.1080/07391102.2024.2419863] [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: 11/04/2023] [Accepted: 04/14/2024] [Indexed: 10/28/2024]
Abstract
Satureja nabateorum, known as Nabatean savory is a Lamiaceae plant native to the Arabian Peninsula, specifically in the mountainous regions of Saudi Arabia. The study aims to investigate the phytochemical components of the S. nabateorum leaves (SNL) and stems (SNS) extract and to assess their antioxidant, antimicrobial, and antiproliferative properties. Methanol extracts from leaves and stems were analyzed for chemical constituents using the GC-MS technique. Antioxidant capacities were measured using hydrogen peroxide and ABTS radical-scavenging methods, and antimicrobial activity was tested against various microorganisms. Cytotoxic activity on four human malignant cell lines was assessed using MTT and flow cytometry. Molecular docking and molecular dynamics studies were conducted to understand the interactions and binding modes of the extracted compounds at a molecular level. GC-MS analysis of SNL extract revealed thymol, carvacrol, and p-cymen-8-ol as major constituents. SNS extract contained β-sitosterol, stigmasterol, lupeol, and lup-20(29)-ene-3β,28-diol. SNS extract exhibited more potent antioxidant, antimicrobial, and anticancer effects than SNL extract. The extract, SNS, exhibited potential toxicity in A549 cells with an IC50 value of 3.62 µg/mL and induced marked apoptotic effects with S phase-cell cycle arrest. SNS extract also showed higher levels of Caspase 3, Bax, p53, and the Bax/Bcl2 ratio and lower levels of Bcl-2. Molecular docking and dynamic simulation supported these findings, targeting the EGFR TK domain. The study suggests that the S. nabateorum stem extract holds promise as a potent antimicrobial, antioxidant, and anticancer agent. It provides valuable insights for considering the extract as a substitute for chemotherapy and/or protective agents.
Collapse
Affiliation(s)
- Bader Huwaimel
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Hail, Hail, Saudi Arabia
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Akram M Elkashlan
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Radwan Alnajjar
- CADD Uunit, PharmD, Faculty of Pharmacy, Libyan International Medical University, Benghazi, Libya
- Department of Chemistry, University of Cape Town, Rondebosch, South Africa
| | - Osama A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Meshal M Alorainan
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Meshari K Altuwayhir
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Salman F Algharby
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Suliman A Almahmoud
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Amr S Abouzied
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Hail, Hail, Saudi Arabia
- Department of Pharmaceutical Chemistry, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| |
Collapse
|
11
|
Jogdeo CM, Panja S, Kumari N, Tang W, Kapoor E, Siddhanta K, Das A, Boesen EI, Foster KW, Oupický D. Inulin-based nanoparticles for targeted siRNA delivery in acute kidney injury. J Control Release 2024; 376:577-592. [PMID: 39419450 DOI: 10.1016/j.jconrel.2024.10.027] [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: 06/14/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
RNA interference has emerged as a promising therapeutic strategy to tackle acute kidney injury (AKI). Development of targeted delivery systems is highly desired for selective renal delivery of RNA and improved therapeutic outcomes in AKI. Inulin is a plant polysaccharide traditionally employed to measure glomerular filtration rate. Here, we describe the synthesis of inulin modified with α-cyclam-p-toluic acid (CPTA) to form a novel renal-targeted polymer, Inulin-CPTA (IC), which is capable of selective siRNA delivery to the injured kidneys. We show that conjugating CPTA to inulin imparts IC with targeting properties for cells that overexpress the C-X-C chemokine receptor 4 (CXCR4). Self-assembled IC/siRNA nanoparticles (polyplexes) demonstrated rapid accumulation in the injured kidneys with selective uptake and prolonged retention in injured renal tubules overexpressing the CXCR4 receptor. Tumor-suppressor protein p53 contributes significantly to the pathogenesis of AKI. siRNA-induced silencing of p53 has shown therapeutic potential in several preclinical studies, making it an important target in the treatment of AKI. Systemically administered nanoparticles formulated using IC and siRNA against p53 selectively accumulated in the injured kidneys and potently silenced p53 expression. Selective p53 knockdown led to positive therapeutic outcomes in mice with cisplatin-induced AKI, as seen by reduced tubular cell death, renal injury, inflammation, and overall improved renal function. These findings indicate that IC is a promising new carrier for renal-targeted delivery of RNA for the treatment of AKI.
Collapse
Affiliation(s)
- Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Neha Kumari
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ekta Kapoor
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kasturi Siddhanta
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ashish Das
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Erika I Boesen
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kirk W Foster
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
12
|
Zhang Y, Yang T, Zhao X, Xin H, Liu D, Wang Q, Zhou A, Wen Z, Zhang K, Xie J. Comprehensive Analysis of Phycoerythrin 545 Stability and the Apoptotic Impact of Its Degradation Products on HT29 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:22105-22114. [PMID: 39316102 DOI: 10.1021/acs.jafc.4c03808] [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: 09/25/2024]
Abstract
This study investigates the properties and potential applications of phycoerythrin 545, a naturally occurring light-harvesting pigment protein from Rhodomonas salina. Phycoerythrin 545, characterized by its bright red color and maximum absorption wavelength at 545 nm, was extracted using freeze-thawing methods, further purified, and analyzed using chromatographic, spectroscopic techniques, and sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Phycoerythrin 545 consists of two subunits, primarily α and β, but lacks the γ subunit, and is stable at 4 °C within a pH range of 3-10. To further characterize it, its susceptibility to degradation by trypsin was assessed. The biological activity of phycoerythrin 545 and its degradation products were investigated in HT29 human colon cancer cells. The results showed that the degradation products, particularly those within 3-10 kDa, significantly decreased the viability of HT29 cells by inducing apoptosis. Mechanistic studies indicated these effects were mediated through the activation of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinases and MAPK/c-Jun N-terminal Kinase signaling pathways and involved the regulation of key apoptotic proteins such as p53, Bim, Bad, Bak, and Bax, leading to the activation of the Caspase-3 apoptotic pathway. These findings contribute to understanding the structural and functional properties of phycoerythrin 545, laying a foundation for its exploration in food industry applications and cancer therapy supplementation.
Collapse
Affiliation(s)
- Yanqing Zhang
- College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Tan Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaotong Zhao
- Department of Chemistry, Cleveland State University, Cleveland, Ohio 44115, United States
| | - Huijie Xin
- College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Danting Liu
- Department of Chemistry, Cleveland State University, Cleveland, Ohio 44115, United States
| | - Qing Wang
- College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Aimin Zhou
- Department of Chemistry, Cleveland State University, Cleveland, Ohio 44115, United States
| | - Zhiyou Wen
- Department of Food Science and Human Nutrition, Iowa State University, Ames, Iowa 50011, United States
| | - Kunsheng Zhang
- College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Junbo Xie
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
13
|
Shirani Asl V, Rafieemehr H, Tamaddon G. The impact of Trifolium pratense extract on apoptosis and autophagy in NALM-6 cells: implications for B-ALL intervention. Med Oncol 2024; 41:257. [PMID: 39352436 DOI: 10.1007/s12032-024-02485-4] [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: 07/21/2024] [Accepted: 08/21/2024] [Indexed: 11/14/2024]
Abstract
B-cell acute lymphoblastic leukemia (B-ALL), a prevalent malignancy predominantly affecting children, poses challenges such as drug resistance and cytotoxicity despite available treatment methods. The persistence of these challenges underscores the necessity for innovative therapeutic approaches to enhance efficacy. Natural compounds derived from plants, recognized for their potential to inhibit cancer cell growth, have drawn attention. Trifolium pratense extract, known for its significant anticancer properties in previous studies, was the focus of this investigation. This experimental study aimed to explore the impact of T. pratense extract on apoptosis and autophagy in NALM-6 cells. The cells were exposed to varying concentrations of the extract at specific time intervals, with viability and metabolic activity assessed using Trypan blue exclusion and MTT assays. Flow cytometry was employed to evaluate apoptosis using Annexin V/PI staining and ROS production using DCFH-DA staining. Real-time PCR was used to quantify gene expression related to apoptosis, autophagy, and oxidative stress, with data analysis performed using GraphPad PRISM software. Trifolium pratense extract demonstrated the capacity to induce apoptosis, autophagy, and significantly increase ROS production in NALM-6 cells. These effects were facilitated by the upregulation of corresponding genes. The MTT assay revealed an IC50 of 231 μg/mL at 48 h, and Flow cytometry analysis showed a 51.8% increase in apoptosis in this cell line. Overall, this study emphasizes the effectiveness of T. pratense extract in inducing autophagy and apoptosis pathways in NALM-6 cells derived from B-cell acute lymphoblastic leukemia, suggesting its potential as a candidate for further investigation as a supplement in ALL treatment.
Collapse
Affiliation(s)
- Vida Shirani Asl
- Division of Hematology and Blood Bank, Department of Laboratory Science, School of Paramedical Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Rafieemehr
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Gholamhossein Tamaddon
- Division of Hematology and Blood Bank, Department of Laboratory Science, School of Paramedical Science, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
14
|
Mandal B, Pramanik A, Sarkar D, Haldar A, Das D, Saha R, Mandal D, Bhattacharyya S. Novel Octahedral Nickel (II) Complex with Flexible Piperazinyl Moiety Exhibits Potent Cytotoxic Effect Along with Anti-Migratory and Anti-Metastatic Effect on Human Cancer Cells. ChemMedChem 2024; 19:e202300728. [PMID: 38757641 DOI: 10.1002/cmdc.202300728] [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: 12/29/2023] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 05/18/2024]
Abstract
Synthesis of non-platinum transition metal complexes with N,O donor chelating ligand for application against pathogenesis of cancer with higher efficacy and selectivity is currently an important field of research. We assessed the anti-cancer effect of a mixed ligand Ni(II) complex on human breast and lung cancer cell lines in this investigation. Mononuclear mixed ligand octahedral Ni(II) complex [NiIIL(NO3)(MeOH)] complex (1), with tri-dentate phenol-based ligand 2,4-dichloro-6-((4-methylpiperazin-1-yl) methyl) phenol (HL) along with methanol and nitrate as ancillary ligand was prepared. Piperazine moiety of the ligand exists as boat conformation in this complex as revealed from single crystal X-ray study. UV-visible spectrum of complex (1) exhibits three distinct d-d bands due to spin-allowed 3 A2 g→3T1 g (P), 3 A2 g→3T1 g(F) and 3 A2 g→3T2 g(F) transitions as expected in an octahedral d8 system. Our study revealed that Complex (1) induces apoptotic cell death in mouse and human cancer cells such as mcf-7, A549 and MDA-MB-231 through transactivation of p53 and its pro-apoptotic downstream targets in a dose dependent manner. Furthermore, complex (1) was able to slow the migratory rate of MDA-MB-231 cells' in vitro as well as epithelia -mesenchymal transition (EMT), the key step for metastatic transition and malignancy. Over all our results suggest complex (1) as a potential agent in anti-tumor treatment regimen showing both cytotoxic and anti-metastatic activity against malignant neoplasia.
Collapse
Affiliation(s)
- Bikramaditya Mandal
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Sector-81, Knowledge City, Manauli, -140306, India
| | - Anik Pramanik
- Immunobiology and translational medicine laboratory, Department of Zoology, Sidho Kanho Birsha University, Purulia, 723104, West Bengal, India
| | - Debanjan Sarkar
- Immunobiology and translational medicine laboratory, Department of Zoology, Sidho Kanho Birsha University, Purulia, 723104, West Bengal, India
| | - Anwesha Haldar
- Department of Chemistry, Sidho-Kanho-Birsha University, Purulia, 723104, West Bengal, India
| | - Dona Das
- Immunobiology and translational medicine laboratory, Department of Zoology, Sidho Kanho Birsha University, Purulia, 723104, West Bengal, India
| | - Rajat Saha
- Department of Chemistry, Kazi Nazrul University, Asansol, Paschim Bardhaman, -713340, West Bengal, India
| | - Debdas Mandal
- Department of Chemistry, Sidho-Kanho-Birsha University, Purulia, 723104, West Bengal, India
| | - Sankar Bhattacharyya
- Immunobiology and translational medicine laboratory, Department of Zoology, Sidho Kanho Birsha University, Purulia, 723104, West Bengal, India
| |
Collapse
|
15
|
Qi J, Li Q, Xin T, Lu Q, Lin J, Zhang Y, Luo H, Zhang F, Xing Y, Wang W, Cui D, Wang M. MCOLN1/TRPML1 in the lysosome: a promising target for autophagy modulation in diverse diseases. Autophagy 2024; 20:1712-1722. [PMID: 38522082 PMCID: PMC11262240 DOI: 10.1080/15548627.2024.2333715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024] Open
Abstract
MCOLN1/TRPML1 is a nonselective cationic channel specifically localized to the late endosome and lysosome. With its property of mediating the release of several divalent cations such as Ca2+, Zn2+ and Fe2+ from the lysosome to the cytosol, MCOLN1 plays a pivotal role in regulating a variety of cellular events including endocytosis, exocytosis, lysosomal biogenesis, lysosome reformation, and especially in Macroautophagy/autophagy. Autophagy is a highly conserved catabolic process that maintains cytoplasmic integrity by removing superfluous proteins and damaged organelles. Acting as the terminal compartments, lysosomes are crucial for the completion of the autophagy process. This review delves into the emerging role of MCOLN1 in controlling the autophagic process by regulating lysosomal ionic homeostasis, thereby governing the fundamental functions of lysosomes. Furthermore, this review summarizes the physiological relevance as well as molecular mechanisms through which MCOLN1 orchestrates autophagy, consequently influencing mitochondria turnover, cell apoptosis and migration. In addition, we have illustrated the implications of MCOLN1-regulated autophagy in the pathological process of cancer and myocardial ischemia-reperfusion (I/R) injury. In summary, given the involvement of MCOLN1-mediated autophagy in the pathogenesis of cancer and myocardial I/R injury, targeting MCOLN1 May provide clues for developing new therapeutic strategies for the treatment of these diseases. Exploring the regulation of MCOLN1-mediated autophagy in diverse diseases contexts will surely broaden our understanding of this pathway and offer its potential as a promising drug target.Abbreviation: CCCP:carbonyl cyanide3-chlorophenylhydrazone; CQ:chloroquine; HCQ: hydroxychloroquine;I/R: ischemia-reperfusion; MAP1LC3/LC3:microtubule associated protein 1 light chain 3; MCOLN1/TRPML1:mucolipin TRP cation channel 1; MLIV: mucolipidosis type IV; MTORC1:MTOR complex 1; ROS: reactive oxygenspecies; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Jiansong Qi
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qingqing Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tianli Xin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qixia Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jinyi Lin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Haiting Luo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Feifei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanhong Xing
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Derong Cui
- Department of Anesthesiology, The Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengmeng Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital, China of Medical University, Shenyang, LiaoningChina
| |
Collapse
|
16
|
Yang L, Liu D, Jiang S, Li H, Chen L, Wu Y, Essien AE, Opoku M, Naranmandakh S, Liu S, Ru Q, Li Y. SIRT1 signaling pathways in sarcopenia: Novel mechanisms and potential therapeutic targets. Biomed Pharmacother 2024; 177:116917. [PMID: 38908209 DOI: 10.1016/j.biopha.2024.116917] [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: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/09/2024] [Indexed: 06/24/2024] Open
Abstract
Sarcopenia is an aging-related skeletal disease characterized by decreased muscle mass, strength, and physical function, severely affecting the quality of life (QoL) of the elderly population. Sirtuin 1 (SIRT1), as a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases, has been reported to participate in various aging-related signaling pathways and exert protective effect on many human diseases. SIRT1 functioned as an important role in the occurrence and progression of sarcopenia through regulating key pathways related to protein homeostasis, apoptosis, mitochondrial dysfunction, insulin resistance and autophagy in skeletal muscle, including SIRT1/Forkhead Box O (FoxO), AMP-activated protein kinase (AMPK)/SIRT1/nuclear factor κB (NF-κB), SIRT1/p53, AMPK/SIRT1/peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), and SIRT1/live kinase B1 (LKB1)/AMPK pathways. However, the specific mechanisms of these processes have not been fully illuminated. Currently, several SIRT1-mediated interventions on sarcopenia have been preliminarily developed, such as SIRT1 activator polyphenolic compounds, exercising and calorie restriction. In this review, we summarized the predominant mechanisms of SIRT1 involved in sarcopenia and therapeutic modalities targeting the SIRT1 signaling pathways for the prevention and prognosis of sarcopenia.
Collapse
Affiliation(s)
- Luning Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shide Jiang
- Department of Orthopedics, The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Anko Elijah Essien
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Michael Opoku
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shinen Naranmandakh
- Department of chemistry, School of Arts and Sciences, National University of Mongolia, Ulaanbaatar 14201, Mongolia
| | - ShuGuang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
17
|
Lin J, Liu H, Huang X, Deng Y. Toxicological effects of Honokiol on zebrafish and its underlying mechanism. J Biochem Mol Toxicol 2024; 38:e23789. [PMID: 39097765 DOI: 10.1002/jbt.23789] [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: 02/28/2024] [Revised: 06/25/2024] [Accepted: 07/18/2024] [Indexed: 08/05/2024]
Abstract
The compound Honokiol, derived from the bark of Magnolia officinalis, possesses the ability to induce apoptosis and inhibit cellular damage caused by reactive oxygen species. The objective of this study was to investigate the toxicological and histopathological effects of Honokiol on zebrafish (Danio rerio) through conducting a semistatic acute toxicity test involving immersion in an Honokiol-containing solution. The results showed that the toxic effects of Honokiol on zebrafish were primarily manifested in the liver and gills. When exposed to 0.6 mg/L of Honokiol, it could lead to liver hemorrhage as well as swelling and necrosis of gill tissues, and high concentrations of Honokiol could trigger inflammatory responses. Additionally, research found that Honokiol could induce apoptosis in liver and gill tissues through the P53 pathway and possessed the ability to enhance antioxidation. The present findings significantly contribute to a more profound understanding of the toxic impact of Honokiol and its underlying mechanism, thereby providing a valuable reference for the future safe utilization of Honokiol and related pharmaceutical advancements.
Collapse
Affiliation(s)
- Jue Lin
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, Sichuan, China
| | - Hongli Liu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaoli Huang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yongqiang Deng
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, Sichuan, China
| |
Collapse
|
18
|
Samia S, Sandeep Chary P, Khan O, Kumar Mehra N. Recent trends and advances in novel formulations as an armament in Bcl-2/Bax targeted breast cancer. Int J Pharm 2024; 653:123889. [PMID: 38346605 DOI: 10.1016/j.ijpharm.2024.123889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/19/2024]
Abstract
Breast cancer (BC) remains a significant health burden worldwide, necessitating the development of innovative therapeutic strategies. The B-cell lymphoma 2 (Bcl-2) family proteins, Bcl-2 and Bax, play a crucial role in regulating apoptosis and thus are promising targets for BC therapy. We focus on the recent advancements in novel formulations that specifically target Bcl-2/Bax pathway to combat BC. It provides an overview on biological functions of Bcl-2/Bax in apoptosis regulation, emphasizing their significance in pathogenesis and progression of the disease while covering the numerous therapeutic approaches aimed at modulating the Bcl-2/Bax pathway, including small-molecule inhibitors, peptides, gene-based therapies and other repurposed drugs harboured onto cutting-edge technologies and nanocarrier systems employed to enhance the targeted delivery of Bcl-2/Bax inhibitors tumor cells. These advanced formulations aim to improve therapeutic efficacy, minimize off-target effects, and overcome drug resistance, offering promising prospects in its treatment. In conclusion, it illuminates the diverse and evolving landscape of novel formulations as an essential armament in targeting these proteins while bridging and unravelling the obscurity of Bcl-2/Bax pathway-targeted drug delivery systems which are presently in their nascent stages of exploration for BC therapy which can benefit researchers, clinicians, and pharmaceutical scientists.
Collapse
Affiliation(s)
- Shaikh Samia
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Padakanti Sandeep Chary
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Omar Khan
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India.
| |
Collapse
|
19
|
Skou LD, Johansen SK, Okarmus J, Meyer M. Pathogenesis of DJ-1/PARK7-Mediated Parkinson's Disease. Cells 2024; 13:296. [PMID: 38391909 PMCID: PMC10887164 DOI: 10.3390/cells13040296] [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: 12/22/2023] [Revised: 01/28/2024] [Accepted: 02/03/2024] [Indexed: 02/24/2024] Open
Abstract
Parkinson's disease (PD) is a common movement disorder associated with the degeneration of dopaminergic neurons in the substantia nigra pars compacta. Mutations in the PD-associated gene PARK7 alter the structure and function of the encoded protein DJ-1, and the resulting autosomal recessively inherited disease increases the risk of developing PD. DJ-1 was first discovered in 1997 as an oncogene and was associated with early-onset PD in 2003. Mutations in DJ-1 account for approximately 1% of all recessively inherited early-onset PD occurrences, and the functions of the protein have been studied extensively. In healthy subjects, DJ-1 acts as an antioxidant and oxidative stress sensor in several neuroprotective mechanisms. It is also involved in mitochondrial homeostasis, regulation of apoptosis, chaperone-mediated autophagy (CMA), and dopamine homeostasis by regulating various signaling pathways, transcription factors, and molecular chaperone functions. While DJ-1 protects neurons against damaging reactive oxygen species, neurotoxins, and mutant α-synuclein, mutations in the protein may lead to inefficient neuroprotection and the progression of PD. As current therapies treat only the symptoms of PD, the development of therapies that directly inhibit oxidative stress-induced neuronal cell death is critical. DJ-1 has been proposed as a potential therapeutic target, while oxidized DJ-1 could operate as a biomarker for PD. In this paper, we review the role of DJ-1 in the pathogenesis of PD by highlighting some of its key neuroprotective functions and the consequences of its dysfunction.
Collapse
Affiliation(s)
- Line Duborg Skou
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark; (L.D.S.); (S.K.J.); (J.O.)
| | - Steffi Krudt Johansen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark; (L.D.S.); (S.K.J.); (J.O.)
| | - Justyna Okarmus
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark; (L.D.S.); (S.K.J.); (J.O.)
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark; (L.D.S.); (S.K.J.); (J.O.)
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark
- BRIDGE—Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
20
|
Ou L, Wu Z, Hu X, Huang J, Yi Z, Gong Z, Li H, Peng K, Shu C, Koole LH. A tissue-adhesive F127 hydrogel delivers antioxidative copper-selenide nanoparticles for the treatment of dry eye disease. Acta Biomater 2024; 175:353-368. [PMID: 38110136 DOI: 10.1016/j.actbio.2023.12.021] [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: 09/04/2023] [Revised: 12/10/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023]
Abstract
Dry eye disease (DED) is currently the most prevalent condition seen in ophthalmology outpatient clinics, representing a significant public health issue. The onset and progression of DED are closely associated with oxidative stress-induced inflammation and damage. To address this, an aldehyde-functionalized F127 (AF127) hydrogel eye drop delivering multifunctional antioxidant Cu2-xSe nanoparticles (Cu2-xSe NPs) was designed. The research findings revealed that the Cu2-xSe nanoparticles exhibit unexpected capabilities in acting as superoxide dismutase and glutathione peroxidase. Additionally, Cu2-xSe NPs possess remarkable efficacy in scavenging reactive oxygen species (ROS) and mitigating oxidative damage. Cu2-xSe NPs displayed promising therapeutic effects in a mouse model of dry eye. Detailed investigation revealed that the nanoparticles exert antioxidant, anti-apoptotic, and inflammation-mitigating effects by modulating the NRF2 and p38 MAPK signalling pathways. The AF127 hydrogel eye drops exhibit good adherence to the ocular surface through the formation of Schiff-base bonds. These findings suggest that incorporating antioxidant Cu2-xSe nanoparticles into a tissue-adhesive hydrogel could present a highly effective therapeutic strategy for treating dry eye disease and other disorders associated with reactive oxygen species. STATEMENT OF SIGNIFICANCE: A new formulation for therapeutic eye drops to be used in the treatment of dry eye disease (DED) was developed. The formulation combines copper-selenium nanoparticles (Cu2-xSe NPs) with aldehyde-functionalized Pluronic F127 (AF127). This is the first study to directly examine the effects of Cu2-xSe NPs in ophthalmology. The NPs exhibited antioxidant capabilities and enzyme-like properties. They effectively eliminated reactive oxygen species (ROS) and inhibited apoptosis through the NRF2 and p38 MAPK signalling pathways. Additionally, the AF127 hydrogel enhanced tissue adhesion by forming Schiff-base links. In mouse model of DED, the Cu2-xSe NPs@AF127 eye drops demonstrated remarkable efficacy in alleviating symptoms of DED. These findings indicate the potential of Cu2-xSe NPs as a readily available and user-friendly medication for the management of DED.
Collapse
Affiliation(s)
- Liling Ou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zixia Wu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiao Hu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jinyi Huang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhiqi Yi
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zehua Gong
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Huaqiong Li
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Ke Peng
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Chang Shu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| | - Leo H Koole
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
21
|
Gupta P, Singh S, Rai N, Verma A, Tiwari H, Kamble SC, Gautam HK, Gautam V. Unveiling the cytotoxic and anti-proliferative potential of green-synthesized silver nanoparticles mediated by Colletotrichum gloeosporioides. RSC Adv 2024; 14:4074-4088. [PMID: 38292267 PMCID: PMC10825743 DOI: 10.1039/d3ra06145k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/19/2023] [Indexed: 02/01/2024] Open
Abstract
Fungal endophytes are a putative source of bioactive metabolites that have found significant applications in nanomedicine due to their metabolic versatility. In the present study, an aqueous extract of the fungal endophyte, Colletotrichum gloeosporioides associated with a medicinal plant Oroxylum indicum, has been used for the fabrication of green silver nanoparticles (CgAgNPs) and further evaluated their cytotoxic and anti-proliferative activity. Bioanalytical techniques including UV-Vis spectral analysis revealed a sharp band at 435 nm and functional molecules from the aqueous extract involved in the synthesis of CgAgNPs were evidenced through FTIR. Further, the crystalline nature of CgAgNPs was determined through XRD analysis and microscopy techniques including AFM, TEM and FESEM demonstrated the spherical shape of CgAgNPs exhibiting a crystalline hexagonal lattice and the size was found to be in the range of 9-29 nm. The significant cytotoxic potential of CgAgNPs was observed against breast cancer cells, MDA-MB-231 and MCF-7 with IC50 values of 18.398 ± 0.376 and 38.587 ± 1.828 μg mL-1, respectively. The biochemical study revealed that the treatment of MDA-MB-231 and MCF-7 cells with CgAgNPs reduces glucose uptake, suppresses cell proliferation, and enhances LDH release, indicating reduced cell viability and progression. Moreover, our research revealed differential expression of genes associated with apoptosis, cell cycle inhibition and metastasis suppression, evidencing anti-proliferative activity of CgAgNPs. The main objective of the present study is to harness anti-breast cancer activity of novel biogenic nanoparticles synthesized using the aqueous extract of O. indicum associated C. gloeosporioides and study the underlying mechanistic pathway exerted by these mycogenic nanoparticles.
Collapse
Affiliation(s)
- Priyamvada Gupta
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| | - Swati Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| | - Nilesh Rai
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| | - Ashish Verma
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| | - Harshita Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| | - Swapnil C Kamble
- Department of Technology, Savitribai Phule Pune University Ganeshkhind Pune 411007 India
| | - Hemant Kumar Gautam
- Department of Immunology and Infectious Disease Biology, CSIR-Institute of Genomics and Integrative Biology Sukhdev Vihar New Delhi 110025 India
| | - Vibhav Gautam
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India +918860182113
| |
Collapse
|
22
|
Li M, Yang J, Li J, Zhou Y, Li X, Ma Z, Li X, Ma H, Ye X. Epiberberine induced p53/p21-dependent G2/M cell cycle arrest and cell apoptosis in gastric cancer cells by activating γ-aminobutyric acid receptor- β3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155198. [PMID: 38006806 DOI: 10.1016/j.phymed.2023.155198] [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: 03/23/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND AND PURPOSE Epiberberine (EPI) is one of the most important bioalkaloid found in the rhizome of Coptis chinensis, which has been observed to exhibit pharmaceutical effects against gastric cancer (GC). Nevertheless, the potential mechanism of EPI against GC cells still remains unclear. This study aimed to identify the core receptor on GC cells through which EPI inhibited the growth of GC cells and to explore the underlying inhibitory mechanisms. METHODS To identify hub receptor targets that respond to EPI treatment, RNA sequencing (RNA-Seq) data from a tumor-bearing mouse model were analyzed using bioinformatics method and molecular docking. The binding interaction between EPI and GABRB3 was validated through western blotting based-cellular thermal shift assay (WB-CETSA). To further verify the binding region between EPI and GABRB3 through circular dichroism (CD) chromatography, fragments of the extracellular and transmembrane domains of the GABRB3 protein were expressed and purified in vitro. Stable cell lines with the overexpression or knockdown of GABRB3 were established using the recombinant lentivirus system. MTT ((3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide)) assay, colony formation assay, invasion and migration experiments, and flow cytometry were conducted to validate the inhibitory effect of EPI on the GC cells via GABRB3. Additionally, western blotting was utilized to explore the potential inhibitory mechanisms. RESULTS Through the combination of multiple bioinformatics methods and molecular docking, we found that the γ-aminobutyric acid type A receptor subunit -β3 (GABRB3) might be the critical receptor target in response to EPI treatment. The results of WB-CETSA analysis indicated that EPI significantly promoted the thermostability of the GABRB3 protein. Importantly, EPI could directly bind to GABRB3 and alter the secondary structure of GABRB3 fragments similar to the natural agonist, γ-aminobutyric acid (GABA). The EPI-induced suppression of the malignant phenotype of GC cells was dependent on the presence of GABRB3. GABRB3 expression was positively correlated with TP53 in patients with GC. The binding of EPI to GABRB3 stimulated p53 accumulation in GC cells. This activated the p21/CDK1/cyclinB1 pathway, resulting in G2/M cell cycle arrest, and induced the Bcl-2/BAX/Caspase axis-dependent cell apoptosis. CONCLUSION This study revealed the target receptor for EPI in GC cells and provided new insights into its anticancer mechanisms.
Collapse
Affiliation(s)
- Mengmeng Li
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jiaye Yang
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Juan Li
- School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Yuan Zhou
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xiaoduo Li
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Zhengcai Ma
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xuegang Li
- School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Hang Ma
- School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
23
|
Esmaeili F, Abolhasani M, Zabihi-Mahmoudabadi H, Seyyed Ebrahimi SS, Emamgholipour S, Paknejad M. Metabolically healthy/unhealthy obesity and breast cancer: A possible role of plasma-derived extracellular vesicles on the cancerous behavior of triple-negative breast cancer. Biochem Biophys Res Commun 2024; 690:149242. [PMID: 37992524 DOI: 10.1016/j.bbrc.2023.149242] [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: 08/03/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/24/2023]
Abstract
PURPOSE Obesity has known detrimental effects on breast cancer (BC) development and progression. However, it's essential to consider the obesity phenotype based on metabolic health. This study aims to evaluate the impact of circulating extracellular vesicles (EVs) from women with metabolically healthy or unhealthy normal weight, overweight, and obesity on MDA-MB-231 cell migration, invasion, and apoptosis. METHODS Plasma EVs were isolated from different obesity phenotypes in women. EVs were characterized and EVs uptake by MDA-MB-231 cells was assessed. MDA-MB-231 cell lines were treated with EVs obtained from various studied groups, and migration, invasion, MMP-2 and MMP-9 activity, Bax and Bcl-2 mRNA expression, p-53 and Thr55 p-p53 protein expression, and apoptosis were assessed. RESULTS EVs from obese individuals, regardless of phenotype, increased invasion and MMP-2 activity compared to healthy normal-weight EVs. Normal-weight EVs led to higher invasion under unhealthy conditions. BC cell migration was enhanced by EVs from healthy obese individuals compared to healthy normal-weight EVs. EVs from unhealthy obese women exhibited significantly lower p53/p-p53 levels and reduced apoptosis compared to healthy obese groups. CONCLUSION It appears that EVs from both normal-weight women with unhealthy conditions and those with obesity or overweight, irrespective of metabolic status, worsened the cancerous behavior of TNBC cells. Therefore, considering metabolic health, in addition to BMI, is crucial for understanding obesity-related disorders.
Collapse
Affiliation(s)
- Fataneh Esmaeili
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Abolhasani
- Cardiac Primary Prevention Research Center, Tehran University of Medical Sciences, Tehran, Iran; Cardiovascular Diseases Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Zabihi-Mahmoudabadi
- Department of General Surgery, School of Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shadi Sadat Seyyed Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Solaleh Emamgholipour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maliheh Paknejad
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Enemark MBH, Wolter K, Campbell AJ, Andersen MD, Sørensen EF, Hybel TE, Madsen C, Lauridsen KL, Plesner TL, Hamilton-Dutoit SJ, Honoré B, Ludvigsen M. Proteomics identifies apoptotic markers as predictors of histological transformation in patients with follicular lymphoma. Blood Adv 2023; 7:7418-7432. [PMID: 37824846 PMCID: PMC10758743 DOI: 10.1182/bloodadvances.2023011299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/03/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
Follicular lymphoma (FL) is an indolent lymphoma with a generally favorable prognosis. However, histological transformation (HT) to a more aggressive disease leads to markedly inferior outcomes. This study aims to identify biological differences predictive of HT at the time of initial FL diagnosis. We show differential protein expression between diagnostic lymphoma samples from patients with subsequent HT (subsequently-transforming FL [st-FL]; n = 20) and patients without HT (nontransforming FL [nt-FL]; n = 34) by label-free quantification nano liquid chromatography-tandem mass spectrometry analysis. Protein profiles identified patients with high risk of HT. This was accompanied by disturbances in cellular pathways influencing apoptosis, the cytoskeleton, cell cycle, and immune processes. Comparisons between diagnostic st-FL samples and paired transformed FL (n = 20) samples demonstrated differential protein profiles and disrupted cellular pathways, indicating striking biological differences from the time of diagnosis up to HT. Immunohistochemical analysis of apoptotic proteins, CASP3, MCL1, BAX, BCL-xL, and BCL-rambo, confirmed higher expression levels in st-FL than in nt-FL samples (P < .001, P = .015, P = .003, P = .025, and P = .057, respectively). Moreover, all 5 markers were associated with shorter transformation-free survival (TFS; P < .001, P = .002, P < .001, P = .069, and P = .010, respectively). Notably, combining the expression of these proteins in a risk score revealed increasingly inferior TFS with an increasing number of positive markers. In conclusion, proteomics identified altered protein expression profiles (particularly apoptotic proteins) at the time of FL diagnosis, which predicted later transformation.
Collapse
Affiliation(s)
- Marie Beck Hairing Enemark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Katharina Wolter
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Maja Dam Andersen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Trine Engelbrecht Hybel
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Charlotte Madsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | - Bent Honoré
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Maja Ludvigsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
25
|
Karagöz A, Beler M, Altun BD, Ünal İ, Cansız D, Gündüz H, Alturfan AA, Emekli-Alturfan E, Erçalık Yalçınkaya Ş. Panoramic dental X-ray exposure leads to oxidative stress, inflammation and apoptosis-mediated developmental defects in zebrafish embryos. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2023; 124:101661. [PMID: 37866507 DOI: 10.1016/j.jormas.2023.101661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Panoramic x-ray units are widely used in dental radiodiagnostics. Patients are exposed to relatively low radiation doses with panoramic imaging, but considering lifetime frequency of exposure, even a small risk can have serious health consequences. Our aim was to assess the effects of panoramic x-rays at two different exposure times on developing zebrafish embryos, focusing on oxidative stress, inflammation, apoptotic pathways, and development. Zebrafish embryos were divided into three groups: control, standard panoramic (SPE, 5.5 s exposure time) and pedodontic panoramic x-ray group (PPE, 4.8 s exposure time). Optically stimulated luminescence dosimeters were used to measure absorbed doses. Mean radiation doses for SPE and PPE were 7.83 mSv and 5.83 mSv respectively. At the end of 96 h post-fertilization, lipid peroxidation (LPO), nitric oxide (NO), reduced glutathione (GSH), glutathione S-transferase and superoxide dismutase were measured in the embryos. Expressions of genes related with inflammation (tnfα, il6, ill15, il21), immunoregulation (ifng) and apoptosis (p53, bax, casp2, casp3, casp8) were determined by RT-PCR. Even at reduced doses at high-speed mode, developmental toxicity was observed in both groups as evidenced by decreased pigmentation, yolk sac oedema, and spinal curvature. While deterioration of oxidant-antioxidant balance, suppression of immune response, induction of inflammation and apoptosis were observed through increased LPO, NO, decreased GSH, ifng, and increased expressions of genes related with inflammation and apoptosis, these effects were more pronounced in the SPE group. These results demonstrate the influence of exposure time and indicate the need for further consideration of optimal panoramic modes from a radiation-induced damage perspective.
Collapse
Affiliation(s)
- Atakan Karagöz
- Department Biochemistry, Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - Merih Beler
- Department Biochemistry, Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - Büşra Dilara Altun
- Department of Oral and Maxillofacial Radiology, Faculty of Dentistry, Marmara University, Istanbul, Turkey
| | - İsmail Ünal
- Department Biochemistry, Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - Derya Cansız
- Department of Biochemistry, Faculty of Medicine, Istanbul Medipol University, Kavacık, Istanbul, Turkey
| | - Hüseyin Gündüz
- Epsilon Landauer Dosimeter Technologies, Istanbul, Turkey
| | - Ahmet Ata Alturfan
- Department of Biochemistry, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Fatih, Istanbul, Turkey
| | - Ebru Emekli-Alturfan
- Department of Biochemistry, Faculty of Dentistry, Marmara University, Istanbul, Turkey.
| | - Şebnem Erçalık Yalçınkaya
- Department of Oral and Maxillofacial Radiology, Faculty of Dentistry, Marmara University, Istanbul, Turkey
| |
Collapse
|
26
|
Kuo CS, Chen SY, Tsai JC. Effects of the Supercritical Fluid Extract of Magnolia figo on Inducing the Apoptosis of Human Non-Small-Cell Lung Cancer Cells. Molecules 2023; 28:7445. [PMID: 37959865 PMCID: PMC10648894 DOI: 10.3390/molecules28217445] [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: 09/14/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Lung cancer has a high incidence rate worldwide, necessitating the development of new drugs. Although Magnolia figo (Lour.) DC. is known for its medicinal properties, studies on its efficacy against lung cancer are lacking. This study investigated whether the supercritical fluid extract of M. figo (FMO) can induce apoptosis in A549, a human non-small-cell lung cancer cell line. The cell viability was assessed using an MTT assay. A terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) analysis and flow cytometry analysis were conducted. The expression of factors was assessed through Western blotting analyses. Gas chromatography-mass spectrometry (GC-MS) was performed. The results revealed that FMO treatment exhibited cytotoxicity, demonstrating dose-dependent effects. The TUNEL analysis and flow cytometry analysis revealed that FMO induced apoptosis in A549 cells. The Western blotting analysis revealed that FMO upregulated the expression of p53 and Bax protein, and downregulated the expression of Bcl-2 protein. The GC-MS analysis revealed eight components identified in FMO. These findings indicate that FMO can induce A549 apoptosis through the p53/Bcl-2/Bax pathways, confirming the apoptotic effects of M. figo on lung cancer cells. These results highlight the potential, for the first time, of M. figo as a source for developing novel drugs for lung cancer treatment.
Collapse
Affiliation(s)
- Chun-Sheng Kuo
- Fethiann Molecule Applied Co., Ltd., Yilan 260011, Taiwan;
| | - Shih-Yun Chen
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua 515006, Taiwan;
| | - Jen-Chieh Tsai
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua 515006, Taiwan;
| |
Collapse
|
27
|
Wang L, Liu Y, Li S, Zha Z, Chen Y, Wang Q, Zhou S, Huang X, Xu M. Capsaicin alleviates doxorubicin-induced acute myocardial injury by regulating iron homeostasis and PI3K-Akt signaling pathway. Aging (Albany NY) 2023; 15:11845-11859. [PMID: 37916995 PMCID: PMC10683596 DOI: 10.18632/aging.205138] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Capsaicin (CAP), a frequently occurring alkaloid component found in spicy peppers, has demonstrated therapeutic potential against tumors, metabolic disease, and cardiovascular disorders. Doxorubicin (DOX), a widely used anthracycline drug in chemotherapy, is notorious for its cardiotoxicity. This study aimed to investigate the potential of CAP in mitigating DOX toxicity in mouse hearts and H9C2 cells, as well as to explore the underlying mechanisms. METHODS In our study, we conducted experiments on both mice and H9C2 cells. The mice were divided into four groups and treated with different substances: normal saline, CAP, DOX and CAP+DOX. We evaluated the induction of ferroptosis by DOX and the remission of ferroptosis by CAP using various methods, including echocardiography, Hematoxylin and Eosin (H&E) staining, Masson's trichrome staining, and determination of ferroptosis metabolites, genes and proteins. Additionally, we employed RNA-seq to identify the inhibitory effect of CAP on DOX-induced myocardial apoptosis, which was further confirmed through western blotting. Similar approaches were applied to H9C2 cells, yielding reliable results. RESULTS Our study demonstrated that treatment with CAP improved the survival rate of DOX-treated mice and reduced myocardial injury. Mechanistically, CAP downregulated transferrin (Trf) and upregulated solute carrier family 40 member 1 (SLC40A1), which helped maintain iron levels in the cells and prevent ferroptosis. Furthermore, CAP inhibited DOX-induced apoptosis by modulating the phosphoinositide 3-kinase (PI3K)- protein kinase B (Akt) signaling pathway. Specifically, CAP activated the PI3K-Akt pathway and regulated downstream BCL2 and BAX to mitigate DOX-induced apoptosis. Therefore, our results suggest that CAP effectively alleviates acute myocardial injury induced by DOX. CONCLUSION Our findings demonstrate that CAP has the potential to alleviate DOX-induced ferroptosis by regulating iron homeostasis. Additionally, it can inhibit DOX-induced apoptosis by activating PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Longbin Wang
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ying Liu
- Department of Cardiology, Sixth Medical Center, PLA General Hospital, Beijing, China
| | - Si Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhijian Zha
- Chinese Internal Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yu Chen
- School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Shujing Zhou
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary, China
| | - Xufeng Huang
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary, China
| | - Ming Xu
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
28
|
Alateyah N, Alsafran M, Usman K, Ouhtit A. Molecular Evidence of Breast Cancer Cell Proliferation Inhibition by a Combination of Selected Qatari Medicinal Plants Crude Extracts. Nutrients 2023; 15:4276. [PMID: 37836560 PMCID: PMC10574548 DOI: 10.3390/nu15194276] [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: 09/06/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy, and conventional medicine has failed to establish efficient treatment modalities. Conventional medicine failed due to lack of knowledge of the mechanisms that underpin the onset and metastasis of tumors, as well as resistance to treatment regimen. However, Complementary and Alternative medicine (CAM) modalities are currently drawing the attention of both the public and health professionals. Our study examined the effect of a super-combination (SC) of crude extracts, which were isolated from three selected Qatari medicinal plants, on the proliferation, motility and death of BC cells. Our results revealed that SC attenuated cell growth and caused the cell death of MDA-MB-231 cancer cells when compared to human normal neonatal fibroblast cells. On the other hand, functional assays showed that SC reduced BC cell migration and invasion, respectively. SC-inhibited cell cycle and SC-regulated apoptosis was most likely mediated by p53/p21 pathway and p53-regulated Bax/BCL-2/Caspace-3 pathway. Our ongoing experiments aim to validate these in vitro findings in vivo using a BC-Xenograft mouse model. These findings support our hypothesis that SC inhibited BC cell proliferation and induced apoptosis. These findings lay the foundation for further experiments, aiming to validate SC as an effective chemoprevention and/or chemotherapeutic strategy that can ultimately pave the way towards translational research/clinical trials for the eradication of BC.
Collapse
Affiliation(s)
- Nouralhuda Alateyah
- Biological Sciences Program, Department of Biological & Environmental Sciences, College of Arts and Science, Qatar University, Doha P.O. Box 2713, Qatar; (N.A.); (M.A.)
| | - Mohammed Alsafran
- Biological Sciences Program, Department of Biological & Environmental Sciences, College of Arts and Science, Qatar University, Doha P.O. Box 2713, Qatar; (N.A.); (M.A.)
- Agricultural Research Station (ARS), Office of VP for Research & Graduate Studies, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Kamal Usman
- Agricultural Research Station (ARS), Office of VP for Research & Graduate Studies, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Allal Ouhtit
- Biological Sciences Program, Department of Biological & Environmental Sciences, College of Arts and Science, Qatar University, Doha P.O. Box 2713, Qatar; (N.A.); (M.A.)
| |
Collapse
|
29
|
Zhang J, Zhao Q, Xue Z, Zhang S, Ren Z, Chen S, Zhou A, Chen H, Liu Y. Deoxynivalenol induces endoplasmic reticulum stress-associated apoptosis via the IRE1/JNK/CHOP pathway in porcine alveolar macrophage 3D4/21 cells. Food Chem Toxicol 2023; 180:114033. [PMID: 37739053 DOI: 10.1016/j.fct.2023.114033] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/31/2023] [Accepted: 09/09/2023] [Indexed: 09/24/2023]
Abstract
The interplay between cell apoptosis and endoplasmic reticulum (ER) stress has garnered increasing attention. Nevertheless, the precise involvement of the unfolded protein response (UPR) signaling in the apoptosis of porcine macrophage cells induced by Deoxynivalenol (DON) remains enigmatic. In this study, we revealed that exposure to 2 μM DON resulted in a substantial decline in cell viability, concomitant with the initiation of cell apoptosis and the halting of the G1 phase cell cycle in the porcine alveolar macrophage line 3D4/21. Transcriptomic analysis of DON-exposed cells showed distinct expression patterns in 3104 genes, with notable upregulation of ER stress-related genes, including IRE1, CHOP, XBP1 and JNK. Our subsequent validation via qPCR and Western blot analyses confirmed the attenuation of GRP78 and BCL-2, coupled with the upregulation of IRE1, CHOP, JNK, p-JNK, and Bax in DON-induced cells, indicating the instigation of ER stress-associated apoptosis by DON. The addition of 5 mM 4-phenylbutyric acid (4-PBA), an ER stress inhibitor, decreased levels of CHOP, IRE1, JNK, p-JNK, and Bax, while increasing levels of GRP78 and Bcl-2, suggesting that 4-PBA alleviated DON-induced ER stress and apoptosis. Overall, our findings provide new insights into DON-induced ER stress via the IRE1/JNK/CHOP pathway, leading to subsequent cellular apoptosis.
Collapse
Affiliation(s)
- Jing Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Qingbo Zhao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Zhihui Xue
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Siyi Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Zeyu Ren
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Shaokui Chen
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Ao Zhou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Hongbo Chen
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan, 430023, China.
| |
Collapse
|
30
|
Salie R, Lopes J, Kotze L, van Aarde R. The cardioprotective effect of S. africana caerulea/Blue Sage in ischaemia and reperfusion induced oxidative stress. Front Pharmacol 2023; 14:1254561. [PMID: 37818190 PMCID: PMC10561252 DOI: 10.3389/fphar.2023.1254561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/01/2023] [Indexed: 10/12/2023] Open
Abstract
Background: Since antiquity, alternative herbal remedies, such as S. africana caerulea/Blue Sage (BLS) water infusion extract (WIE) has been used by traditional healers, for the effective treatment of various chronic inflammatory disorders associated with reduced cellular antioxidant defense mechanisms and free radical cellular damage. In the heart, ischaemia-reperfusion (I/R) induced oxidative stress becomes an early crucial event in the pathogenesis of ischaemia-reperfusion injury (I/RI) and subsequent heart failure. Purpose/Aim: To investigate whether BLS WIE treatment during ischaemia and/or reperfusion may be cardioprotective. Study design: Isolated perfused rat hearts were exposed to 35 min regional ischaemia (RI) and 60 min reperfusion. The BLS WIE was applied: i) for the last 10 min of RI (PerT) or ii) from onset of reperfusion (PostT) or iii) both (PerT) + (PostT). Methods: Endpoints were functional recovery and infarct size (IS). In another set of experiments, left ventricles were freeze-clamped after RI and 10 min reperfusion for detection of total and phosphorylated p-ERK p44/p42, p-Akt, p-p38-MAPK, p-JNK, Nrf-2, NF-kB, Bax, Bcl-2, Caspase-3, and PGC-1α by Western blot analysis. Results: BLS (PostT) significantly increased ERK p44, p-Akt, Nrf-2, and Bcl-2 levels; significantly decreased p-p38-MAPK as well as p-JNK p46 phosphorylation; did not affect Bax levels and significantly decreased Bax/Bcl-2 ratios. This was associated with significantly reduced Caspase-3 levels and increased PGC-1α phosphorylation, particlarly when BLS WIE was administered as PostT. Conclusion: The administration of polyphenol-rich BLS WIE at different stages of ischaemia and/or reperfusion, activate/inhibit several signaling events simultaneously and mediate cardioprotection in a multitarget manner.
Collapse
Affiliation(s)
- Ruduwaan Salie
- South African Medical Research Council, Biomedical Research and Innovation Platform, Cape Town, Western Cape, South Africa
| | - John Lopes
- Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Western Cape, South Africa
| | - Leon Kotze
- Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Western Cape, South Africa
| | - Ruzayda van Aarde
- South African Medical Research Council, Biomedical Research and Innovation Platform, Cape Town, Western Cape, South Africa
| |
Collapse
|
31
|
Al-Awaida W, Al-Ameer HJ, Sharab A, Akasheh RT. Modulation of wheatgrass ( Triticum aestivum Linn) toxicity against breast cancer cell lines by simulated microgravity. Curr Res Toxicol 2023; 5:100127. [PMID: 37767028 PMCID: PMC10520342 DOI: 10.1016/j.crtox.2023.100127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/11/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
This study scrutinizes the effects of simulated microgravity on the antioxidant and cytotoxic potential, along with the phytochemical content of wheatgrass (Triticum aestivum Linn). To imitate microgravity, wheatgrass seeds were germinated in a 3D-clinostat at different rotations per minute (5, 10, 15, and 20 rpm), together with terrestrial gravity control, over 10 days. After germination, the methanolic extracts were analyzed using UPLC-Triple Quad LCMS for their phytochemical composition and tested for their hydrogen peroxide, nitric oxide, and DPPH scavenging activities. The cytotoxic effects of these extracts were evaluated against normal skin fibroblasts, normal breast cells (MCF-10), and breast cancer cells (MCF-7 and MDA-231). The findings showed an extended root growth in wheatgrass germinated under microgravity (WGM) compared to under gravity (WGG). Additionally, WGM extracts demonstrated increased H2O2-, NO-, and DPPH-scavenging activities and a higher content of polyphenols and flavonoids than WGG extracts. These effects were amplified with an increase in clinostat rotations. Moreover, WGM extracts were found to contain a unique set of bioactive compounds (compounds that were detected in the microgravity-germinated wheatgrass but were either absent or present in lower concentrations in wheatgrass germinated under standard gravity conditions.), including pyridoxine, apigenin, and tocopherol, among others, which were absent in WGG. The UPLC-Triple Quad LCMS analysis revealed these unique bioactive compounds in WGM. Notably, WGM extracts showed enhanced cytotoxic effects against normal skin fibroblasts, normal MCF-10, MCF-7, and breast cancer MDA-231 cell lines, with increased cytotoxicity correlating with the number of clinostat rotations. Particularly, WGM extract (at 20 rpm) demonstrated significantly stronger cytotoxicity against MCF-7 breast cancer cells. Further in-depth gene expression analysis of MCF-7 cells exposed to WGM revealed a significant downregulation of genes integral to breast cancer pathways, tyrosine kinase signaling, and DNA repair, complemented by upregulation of certain cell survival and cytotoxic genes. These alterations in genetic pathways associated with cell survival, hormone responses, and cancer progression may elucidate the enhanced cytotoxicity observed in WGM extracts. Our findings underscore the potential of microgravity as a tool to enhance the cytotoxic capabilities of wheatgrass against cancer cell lines, presenting a promising direction for future research in the field of space biology and its implications for terrestrial health.
Collapse
Affiliation(s)
- Wajdy Al-Awaida
- Department of Biology and Biotechnology, Faculty of Science, American University of Madaba, P.O. Box: 99, Madaba 17110, Jordan
| | - Hamzeh J. Al-Ameer
- Department of Biology and Biotechnology, Faculty of Science, American University of Madaba, P.O. Box: 99, Madaba 17110, Jordan
- Department of Pharmaceutical Biotechnology, Faculty of Allied Medical Sciences, Al-AhliyyaAmman University (AAU), Amman, 19328, Jordan
| | - Ahmad Sharab
- Department of Biology and Biotechnology, Faculty of Science, American University of Madaba, P.O. Box: 99, Madaba 17110, Jordan
| | - Rand T. Akasheh
- Department of Nutrition and Dietetics, American University of Madaba, P.O. Box: 99, Madaba 17110, Jordan
| |
Collapse
|
32
|
Sanniyasi E, Gopal RK, Damodharan R, Arumugam A, Sampath Kumar M, Senthilkumar N, Anbalagan M. In vitro anticancer potential of laminarin and fucoidan from Brown seaweeds. Sci Rep 2023; 13:14452. [PMID: 37660108 PMCID: PMC10475116 DOI: 10.1038/s41598-023-41327-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 08/24/2023] [Indexed: 09/04/2023] Open
Abstract
Marine seaweeds are rich source of polysaccharides present in their cell wall and are cultivated and consumed in China, Japan, Korea, and South Asian countries. Brown seaweeds (Phaeophyta) are rich source of polysaccharides such as Laminarin and Fucoidan. In present study, both the laminarin and fucoidan were isolated was yielded higher in PP (Padina pavonica) (4.36%) and STM (Stoechospermum marginatum) (2.32%), respectively. The carbohydrate content in laminarin and fucoidan was 86.91% and 87.36%, whereas the sulphate content in fucoidan was 20.68%. Glucose and mannose were the major monosaccharide units in laminarin (PP), however, fucose, galactose, and xylose in fucoidan (STM). FT-IR down peaks represent the carbohydrate of laminarin and fucoidan except, for 1219 cm-1, and 843 cm-1, illustrating the sulphate groups of fucoidan. The molecular weight of laminarin was 3-5 kDa, and the same for fucoidan was 2-6 kDa, respectively. Both the Fucoidan and Laminarin showed null cytotoxicity on Vero cells. Contrastingly, the fucoidan possess cytotoxic activity on human liver cancer cells (HepG2) (IC50-24.4 ± 1.5 µg/mL). Simultaneously, laminarin also shown cytotoxicity on human colon cancer cells (HT-29) (IC50-57 ± 1.2 µg/mL). The AO/EB (Acriding Orange/Ethidium Bromide) assay significantly resulted in apoptosis and necrosis upon laminarin and fucoidan treatments, respectively. The DNA fragmentation results support necrotic cancer cell death. Therefore, laminarin and fucoidan from PP and STM were potential bioactive compounds for anticancer therapy.
Collapse
Affiliation(s)
- Elumalai Sanniyasi
- Department of Biotechnology, University of Madras, Guindy Campus, Chennai, 600025, India.
| | - Rajesh Kanna Gopal
- Department of Microbiology, Saveetha Dental College and Hospitals, SIMATS, Chennai, 600077, India
| | - Rajesh Damodharan
- Department of Biotechnology, University of Madras, Guindy Campus, Chennai, 600025, India
| | - Arthi Arumugam
- Department of Biotechnology, Rajalakshmi Engineering College, Chennai, 602105, India
| | | | | | - Monisha Anbalagan
- Department of Biotechnology, Jeppiar Engineering College, Chennai, 600119, India
| |
Collapse
|
33
|
Wei W, Qin F, Gao J, Chang J, Pan X, Jiang X, Che L, Zhuo Y, Wu D, Xu S. The effect of maternal consumption of high-fat diet on ovarian development in offspring. Anim Reprod Sci 2023; 255:107294. [PMID: 37421833 DOI: 10.1016/j.anireprosci.2023.107294] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/10/2023]
Abstract
The environment encountered by the fetus during its development exerts a profound influence on its physiological function and disease risk in adulthood. Women's intake of high-fat diet during pregnancy and lactation has gradually become an issue of widespread concern. Maternal high-fat diet will not only cause abnormal neurological development and metabolic syndrome symptoms in the offspring, but also affect the fertility of female offspring. Maternal high-fat diet affects the expression of genes related to follicle growth in offspring, such as AAT, AFP and GDF-9, which reduces the number of follicles and impairs follicle development. Additionally, maternal high-fat diet also affects ovarian health by inducing ovarian oxidative stress and cell apoptosis, which collectively can impair the reproductive potential of female offspring. Reproductive potential carries significant importance for both humans and animals. Therefore, this review aims to describe the effect of maternal exposure to high-fat diet on the ovarian development of offspring and to discuss possible mechanisms by which maternal diet affects the growth and metabolism of offspring.
Collapse
Affiliation(s)
- Wenyan Wei
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Feng Qin
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Junjie Gao
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Junlei Chang
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Xujing Pan
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Xuemei Jiang
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Lianqiang Che
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Yong Zhuo
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - De Wu
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Shengyu Xu
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China.
| |
Collapse
|
34
|
He XN, Wu P, Jiang WD, Liu Y, Kuang SY, Tang L, Ren HM, Li H, Feng L, Zhou XQ. Aflatoxin B1 exposure induced developmental toxicity and inhibited muscle development in zebrafish embryos and larvae. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 878:163170. [PMID: 37003331 DOI: 10.1016/j.scitotenv.2023.163170] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/02/2023] [Accepted: 03/26/2023] [Indexed: 05/13/2023]
Abstract
The prevalence of aflatoxin B1 (AFB1), one of the most toxic mycotoxins that contaminates feedstock and food is increasing worldwide. AFB1 can cause various health problems in humans and animals, as well as direct embryotoxicity. However, the direct toxicity of AFB1 on embryonic development, especially foetal foetus muscle development, has not been studied in depth. In the present study, we used zebrafish embryos as a model to study the mechanism of the direct toxicity of AFB1 to the foetus, including muscle development and developmental toxicity. Our results showed that AFB1 caused motor dysfunction in zebrafish embryos. In addition, AFB1 induces abnormalities in muscle tissue architecture, which in turn causes abnormal muscle development in larvae. Further studies found that AFB1 destroyed the antioxidant capacity and tight junction complexes (TJs), causing apoptosis in zebrafish larvae. In summary, AFB1 may induce developmental toxicity and inhibit muscle development through oxidative damage, apoptosis and disruption of TJs in zebrafish larvae. Our results revealed the direct toxicity effects of AFB1 on the development of embryos and larvae, including inhibition of muscle development and triggering neurotoxicity, induction of oxidative damage, apoptosis and disruption of TJs, and fills the gap in the toxicity mechanism of AFB1 on foetal development.
Collapse
Affiliation(s)
- Xiang-Ning He
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Hua Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China.
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China.
| |
Collapse
|
35
|
Kumbhar N, Nimal S, Patil D, Kaiser VF, Haupt J, Gacche RN. Repurposing of neprilysin inhibitor 'sacubitrilat' as an anti-cancer drug by modulating epigenetic and apoptotic regulators. Sci Rep 2023; 13:9952. [PMID: 37336927 DOI: 10.1038/s41598-023-36872-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/12/2023] [Indexed: 06/21/2023] Open
Abstract
Modifications in the epigenetic landscape have been considered a hallmark of cancer. Histone deacetylation is one of the crucial epigenetic modulations associated with the aggressive progression of various cancer subtypes. Herein, we have repurposed the neprilysin inhibitor sacubitrilat as a potent anticancer agent using in-silico protein-ligand interaction profiler (PLIP) analysis, molecular docking, and in vitro studies. The screening of PLIP profiles between vorinostat/panobinostat and HDACs/LTA4H followed by molecular docking resulted in five (Sacubitrilat, B65, BDS, BIR, and NPV) FDA-approved, experimental and investigational drugs. Sacubitrilat has demonstrated promising anticancer activity against colorectal cancer (SW-480) and triple-negative breast cancer (MDA-MB-231) cells, with IC50 values of 14.07 μg/mL and 23.02 μg/mL, respectively. FACS analysis revealed that sacubitrilat arrests the cell cycle at the G0/G1 phase and induces apoptotic-mediated cell death in SW-480 cells. In addition, sacubitrilat inhibited HDAC isoforms at the transcriptomic level by 0.7-0.9 fold and at the proteomic level by 0.5-0.6 fold as compared to the control. Sacubitrilat increased the protein expression of tumor-suppressor (p53) and pro-apoptotic makers (Bax and Bid) by 0.2-2.5 fold while decreasing the expression of anti-apoptotic Bcl2 and Nrf2 proteins by 0.2-0.5 fold with respect to control. The observed cleaved PARP product indicates that sacubitrilat induces apoptotic-mediated cell death. This study may pave the way to identify the anticancer potential of sacubitrilat and can be explored in human clinical trials.
Collapse
Affiliation(s)
- Navanath Kumbhar
- Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra (MS), 411007, India
| | - Snehal Nimal
- Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra (MS), 411007, India
| | - Deeksha Patil
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra (MS), 411007, India
| | | | | | - Rajesh N Gacche
- Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra (MS), 411007, India.
| |
Collapse
|
36
|
Hatshan MR, Saquib Q, Siddiqui MA, Faisal M, Ahmad J, Al-Khedhairy AA, Shaik MR, Khan M, Wahab R, Matteis VD, Adil SF. Effectiveness of Nonfunctionalized Graphene Oxide Nanolayers as Nanomedicine against Colon, Cervical, and Breast Cancer Cells. Int J Mol Sci 2023; 24:9141. [PMID: 37298090 PMCID: PMC10252622 DOI: 10.3390/ijms24119141] [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: 03/05/2023] [Revised: 04/26/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Recent studies in nanomedicine have intensively explored the prospective applications of surface-tailored graphene oxide (GO) as anticancer entity. However, the efficacy of nonfunctionalized graphene oxide nanolayers (GRO-NLs) as an anticancer agent is less explored. In this study, we report the synthesis of GRO-NLs and their in vitro anticancer potential in breast (MCF-7), colon (HT-29), and cervical (HeLa) cancer cells. GRO-NLs-treated HT-29, HeLa, and MCF-7 cells showed cytotoxicity in the MTT and NRU assays via defects in mitochondrial functions and lysosomal activity. HT-29, HeLa, and MCF-7 cells treated with GRO-NLs exhibited substantial elevations in ROS, disturbances of the mitochondrial membrane potential, an influx of Ca2+, and apoptosis. The qPCR quantification showed the upregulation of caspase 3, caspase 9, bax, and SOD1 genes in GRO-NLs-treated cells. Western blotting showed the depletion of P21, P53, and CDC25C proteins in the above cancer cell lines after GRO-NLs treatment, indicating its function as a mutagen to induce mutation in the P53 gene, thereby affecting P53 protein and downstream effectors P21 and CDC25C. In addition, there may be a mechanism other than P53 mutation that controls P53 dysfunction. We conclude that nonfunctionalized GRO-NLs exhibit prospective biomedical application as a putative anticancer entity against colon, cervical, and breast cancers.
Collapse
Affiliation(s)
- Mohammad Rafe Hatshan
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| | - Quaiser Saquib
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Maqsood A. Siddiqui
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Mohammad Faisal
- Botany and Microbiology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | - Javed Ahmad
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Abdulaziz A. Al-Khedhairy
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Mohammed Rafi Shaik
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| | - Mujeeb Khan
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| | - Rizwan Wahab
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Valeria De Matteis
- Department of Mathematics and Physics “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy;
| | - Syed Farooq Adil
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| |
Collapse
|
37
|
Abolhassani Y, Mirzaei S, Nejabat M, Talebian S, Gholamhosseinian H, Iranshahi M, Rassouli FB, Jamialahmadi K. 7-Geranyloxcycoumarin enhances radio sensitivity in human prostate cancer cells. Mol Biol Rep 2023:10.1007/s11033-023-08439-9. [PMID: 37217617 DOI: 10.1007/s11033-023-08439-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 04/06/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Prostate cancer is the second most prevalent and the fifth deadliest cancer among men worldwide. To improve radiotherapy outcome, we investigated the effects of 7-geranyloxycoumarin, also known as auraptene (AUR), on radiation response of prostate cancer cells. METHODS AND RESULTS PC3 cells were pretreated with 20 and 40 µM AUR for 24, 48 and 72 h, followed by X-ray exposure (2, 4 and 6 Gy). After 72 h recovery, cell viability was determined by alamar Blue assay. Flow cytometric analysis was performed to assess apoptosis induction, clonogenic assay was carried out to investigate clonogenic survival, and the expression of P53, BAX, BCL2, CCND1 and GATA6 was analyzed by quantitative polymerase chain reaction (qPCR). Cell viability assay indicated that toxic effects of radiation was enhanced by AUR, which was also confirmed by increased numbers of apoptotic cells and reduced amount of survival fraction. The qPCR results demonstrated significant induction of P53 and BAX, while the expression of BCL2, GATA6, and CCND1 was significantly downregulated. CONCLUSION The findings of the present study indicated, for the first time, that AUR improved radio sensitivity in prostate cancer cells, and thus, has the potential to be used in future clinical trials.
Collapse
Affiliation(s)
- Yasaman Abolhassani
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Mirzaei
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Masoud Nejabat
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyedehsaba Talebian
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh B Rassouli
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Khadijeh Jamialahmadi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
38
|
Zhang Y, Zeng M, Li B, Zhang B, Cao B, Wu Y, Ye S, Xu R, Zheng X, Feng W. Ephedra Herb extract ameliorates adriamycin-induced nephrotic syndrome in rats via the CAMKK2/AMPK/mTOR signaling pathway. Chin J Nat Med 2023; 21:371-382. [PMID: 37245875 DOI: 10.1016/s1875-5364(23)60454-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Indexed: 05/30/2023]
Abstract
This study aimed to investigate the effect and mechanisms of Ephedra Herb (EH) extract on adriamycin-induced nephrotic syndrome (NS), providing an experimental basis for the clinical treatment of NS. Hematoxylin and eosin staining, creatinine, urea nitrogen, and kidn injury molecule-1 were used to evaluate the activities of EH extract on renal function. The levels of inflammatory factors and oxidative stress were detected by kits. The levels of reactive oxygen species, immune cells, and apoptosis were measured by flow cytometry. A network pharmacological approach was used to predict the potential targets and mechanisms of EH extract in the treatment of NS. The protein levels of apoptosis-related proteins and CAMKK2, p-CAMKK2, AMPK, p-AMPK, mTOR and p-mTOR in the kidneys were detected by Western blot. The effective material basis of EH extract was screened by MTT assay. The AMPK pathway inhibitor (compound C, CC) was added to investigate the effect of the potent material basis on adriamycin-induced cell injury. EH extract significantly improved renal injury and relieve inflammation, oxidative stress, and apoptosis in rats. Network pharmacology and Western blot results showed that the effect of EH extract on NS may be associated with the CAMKK2/AMPK/mTOR signaling pathway. Moreover, methylephedrine significantly ameliorated adriamycin-induced NRK-52e cell injury. Methylephedrine also significantly improved the phosphorylation of AMPK and mTOR, which were blocked by CC. In sum, EH extract may ameliorate renal injury via the CAMKK2/AMPK/mTOR signaling pathway. Moreover, methylephedrine may be one of the material bases of EH extract.
Collapse
Affiliation(s)
- Yuhan Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Mengnan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., Zhengzhou 450000, China
| | - Benke Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Beibei Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Bing Cao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Yuanyuan Wu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Shan Ye
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Ruiqi Xu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., Zhengzhou 450000, China.
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450000, China; The Engineering and Technology Center for Chinese Medicine Development of Henan province, Zhengzhou 450000, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R., Zhengzhou 450000, China.
| |
Collapse
|
39
|
Yao Q, Duan S, Yang Q, Ma X, Li Z, Wu K, Chang P, Cao M, Chen X, Wang Z, Zhong X, Zhou Q, Zhao H. Mep50 is essential for embryonic development in medaka fish. Gene 2023; 868:147387. [PMID: 36963734 DOI: 10.1016/j.gene.2023.147387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/07/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023]
Abstract
Mep50 as a partner promotes the activity and substrate affinity of Prmt5. Prmt5 and Mep50 function together in multiple bioprocesses of the cells. Both Prmt5 and Mep50 are necessary for maintenance of the stem cells and are indispensable in the embryogenesis in the mammals. However, the role of Mep50 is rarely studied in fish. This study was to investigate the role of Mep50 in embryonic development of medaka. Medaka mep50 was mutated by genomic editing with CRISPR-Cas9 technology. Two mutants with a deletion of 22 and 46 bp separately in mep50 caused premature stopping of translation. The homozygotes of these mutant fish were obtained by self-crossing of the heterozygotes. These homozygotic mutants could reproduce embryos but the offspring were not viable. The apoptotic cells were significantly more in the mutant embryos than that in the wild type indicated by TUNEL assay. Quantitative RT-PCR showed that the expression of oct4 and sox2 were significantly decreased, but p53 was increased in the mutant embryos. These results suggest that disruption of mep50 severely interferes with embryogenesis and mep50 is necessary for embryonic development by maintaining stem cells and repression of apoptosis in medaka.
Collapse
Affiliation(s)
- Qiting Yao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Shi Duan
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Qing Yang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Xiaoqin Ma
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Zhenyu Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Kongyue Wu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Pei Chang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Mengxi Cao
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Xinhua Chen
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Zequn Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Xueping Zhong
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Qingchun Zhou
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Haobin Zhao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China.
| |
Collapse
|
40
|
Elzahhar PA, Nematalla HA, Al-Koussa H, Abrahamian C, El-Yazbi AF, Bodgi L, Bou-Gharios J, Azzi J, Al Choboq J, Labib HF, Kheir WA, Abu-Serie MM, Elrewiny MA, El-Yazbi AF, Belal ASF. Inclusion of Nitrofurantoin into the Realm of Cancer Chemotherapy via Biology-Oriented Synthesis and Drug Repurposing. J Med Chem 2023; 66:4565-4587. [PMID: 36921275 DOI: 10.1021/acs.jmedchem.2c01408] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Structural modifications of the antibacterial drug nitrofurantoin were envisioned, employing drug repurposing and biology-oriented drug synthesis, to serve as possible anticancer agents. Eleven compounds showed superior safety in non-cancerous human cells. Their antitumor efficacy was assessed on colorectal, breast, cervical, and liver cancer cells. Three compounds induced oxidative DNA damage in cancer cells with subsequent cellular apoptosis. They also upregulated the expression of Bax while downregulated that of Bcl-2 along with activating caspase 3/7. The DNA damage induced by these compounds, demonstrated by pATM nuclear shuttling, was comparable in both MCF7 and MDA-MB-231 (p53 mutant) cell lines. Mechanistic studies confirmed the dependence of these compounds on p53-mediated pathways as they suppressed the p53-MDM2 interaction. Indeed, exposure of radiosensitive prostatic cancer cells to low non-cytotoxic concentrations of compound 1 enhanced the cytotoxic response to radiation indicating a possible synergistic effect. In vivo antitumor activity was verified in an MCF7-xenograft animal model.
Collapse
Affiliation(s)
- Perihan A Elzahhar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Hisham A Nematalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22516, Egypt
| | - Houssam Al-Koussa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut 11072020, Lebanon
| | - Carla Abrahamian
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Amira F El-Yazbi
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Larry Bodgi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 11072020, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 11072020, Lebanon
| | - Jolie Bou-Gharios
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 11072020, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 11072020, Lebanon
| | - Joyce Azzi
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 11072020, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 11072020, Lebanon
| | - Joelle Al Choboq
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 11072020, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 11072020, Lebanon
| | - Hala F Labib
- Department of Pharmaceutical Chemistry, College of Pharmacy, Arab Academy of Science Technology and Maritime Transport, Alexandria 21913, Egypt
| | - Wassim Abou Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 11072020, Lebanon
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Alexandria 21934, Egypt
| | - Mohamed A Elrewiny
- Faculty of Pharmacy and the Research and Innovation Hub, Alamein International University, Alamein 5060335, Egypt
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut 11072020, Lebanon.,Faculty of Pharmacy and the Research and Innovation Hub, Alamein International University, Alamein 5060335, Egypt.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed S F Belal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
41
|
Sanesi L, Storlino G, Dicarlo M, Oranger A, Zerlotin R, Pignataro P, Suriano C, Guida G, Grano M, Colaianni G, Colucci SC. Time-dependent unloading effects on muscle and bone and involvement of FNDC5/irisin axis. NPJ Microgravity 2023; 9:4. [PMID: 36658231 PMCID: PMC9852594 DOI: 10.1038/s41526-023-00251-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
The identification of biomarkers and countermeasures to prevent the adverse effects on the musculoskeletal system caused by the absence of mechanical loading is the main goal of space biomedical research studies. In this study, we analyzed over 4 weeks of unloading, the modulation in the expression of key proteins in Vastus lateralis, Gastrocnemius and cortical bone in parallel with the modulation of irisin serum levels and its precursor FNDC5 in skeletal muscle of hind limb unloaded (HU) mice. Here we report that Atrogin-1 was up-regulated as early as 1- and 2-week of unloading, whereas Murf-1 at 2- and 3-weeks, along with a marked modulation in the expression of myosin heavy chain isoforms during unloading. Since HU mice showed reduced irisin serum levels at 4-weeks, as well as FNDC5 decrease at 3- and 4-weeks, we treated HU mice with recombinant irisin for 4 weeks, showing that unloading-dependent decline of myosin heavy chain isoforms, MyHCIIα and MyHCIIx, and the anti-apoptotic factor Bcl2, were prevented. In parallel, irisin treatment inhibited the increase of the senescence marker p53, and the pro-apoptotic factor Bax. Overall, these results suggest that the myokine irisin could be a possible therapy to counteract the musculoskeletal impairment caused by unloading.
Collapse
Affiliation(s)
- Lorenzo Sanesi
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Giuseppina Storlino
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Manuela Dicarlo
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Angela Oranger
- grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Roberta Zerlotin
- grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Patrizia Pignataro
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy ,grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Clelia Suriano
- grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Gabriella Guida
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Maria Grano
- grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Graziana Colaianni
- grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Silvia Concetta Colucci
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| |
Collapse
|
42
|
In vitro study of ochratoxin A in the expression of genes associated with neuron survival and viability. Toxicology 2023; 483:153376. [PMID: 36400265 DOI: 10.1016/j.tox.2022.153376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/27/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Ochratoxin A (OTA) is a common mycotoxin and known contaminant of crops, foods and drinks. As OTA crosses the blood-brain barrier, this study investigated the role of OTA, as an environmental hazard, on neuronal survival and viability. The impact of a range of OTA concentrations on the expression of MAPT, BAX, P53, BDNF and TPPP genes was investigated using human neuroblastoma (SH-SY5Y) cells. The absence of altered gene expression determined using reverse transcription quantitative PCR demonstrated that exposure to a typical daily dose of OTA delivered to the brain (2 fM), may not trigger neuronal dysfunction. However, a dose of OTA (2 pM) decreased BDNF expression. BDNF and TPPP expression were significantly reduced after 1 day and significantly increased after 2 days of exposure to 1 µM OTA. The expression of P53, MAPT, and BAX was reduced at both days. Thus, despite OTA cytotoxicity, SH-SY5Y cells entered a survival state following a strong toxic insult. A typical daily environmental OTA exposure does not appear to carry an increased risk of neurodegenerative disease. However, BDNF dysfunction may occur through prolonged exposure to a dose one thousand times higher than the typical daily consumed OTA dose potentially causing adverse effects on neuronal health.
Collapse
|
43
|
Wang F, Yan X, Hua Y, Song J, Liu D, Yang C, Peng F, Kang F, Hui Y. PI3K/AKT/mTOR pathway and its related molecules participate in PROK1 silence-induced anti-tumor effects on pancreatic cancer. Open Life Sci 2023; 18:20220538. [PMID: 37070074 PMCID: PMC10105552 DOI: 10.1515/biol-2022-0538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/28/2022] [Accepted: 11/21/2022] [Indexed: 04/19/2023] Open
Abstract
The PI3K/AKT/mTOR (phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin) pathway can be initiated by PROK1 (prokineticin 1), but its effect and mechanism of action in pancreatic carcinoma (PC) are not fully understood. In this study, we elucidated the roles of PROK1 and its related molecules in PC in vivo. PANC-1 cells with PROK1 knockdown were injected into BALB/c nude mice. The growth and weight of the tumor were monitored and measured, which was followed by TUNEL (terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling), immunohistochemical staining, and hematoxylin and eosin staining. The key proteins related to proliferation, apoptosis, and the PI3K/AKT/mTOR pathway were determined by Western blotting. We also used public databases to identify the molecules related to PROK1. The reduction of PROK1 inhibited angiopoiesis and promoted apoptosis in vivo. PCNA-1, cyclin D1, and Bcl-2 decreased considerably, while Bax and cleaved caspase-3 increased significantly after PROK1 inhibition. The PI3K/AKT/mTOR signal inhibition was also closely associated with PROK1 knockdown. The possible related molecules of PROK1, such as von Willebrand factor, were screened and considered to be involved in the aberrant activation of PI3K/AKT. In conclusion, PROK1 knockdown significantly prevented tumor growth and promoted apoptosis of human PC cells in vivo, where the PI3K/AKT/mTOR pathway was probably inhibited. Therefore, PROK1, along with its related molecules, might be important targets for PC therapy.
Collapse
Affiliation(s)
- Feng Wang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Xiaogang Yan
- Department of Surgical Oncology, The First People’s Hospital of Yinchuan, Yinchuan750001, China
| | - Yongqiang Hua
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, Shanghai200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Jianjun Song
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Di Liu
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Chun Yang
- Department of Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan750001, China
| | - Fei Peng
- Department of Hepatobiliary Pancreatic Surgery, Edong Healthcare Huangshi Central Hospital, Huangshi435002, Hubei, China
| | - Fuping Kang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Yongfeng Hui
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| |
Collapse
|
44
|
Seyedi SMR, Asoodeh A, Darroudi M. The human immune cell simulated anti-breast cancer nanorobot: the efficient, traceable, and dirigible anticancer bio-bot. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00150-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abstract
Background
Various types of cancer therapy strategies have been investigated and successfully applied so far. There are a few modern strategies for improving drug selectivity and biocompatibility, such as nanoparticle-based drug delivery systems. Herein, we designed the traceable enzyme-conjugated magnetic nanoparticles to target human breast cancer cells by simulating the innate immune cell’s respiratory explosion response.
Methods
The human immune cell simulated anti-breast cancer-nanorobot (hisABC-NB) was produced by conjugating the mouse-derived iNOS and human-originated MPO enzymes on the folate-linked chitosan-coated Fe3O4 nanoparticles. The synthesized nanoparticles were functionalized with folic acid as the breast cancer cell detector. Then, the hisABC-NB’s stability and structural properties were characterized by studying Zeta-potential, XRD, FTIR, VSM, FESEM, and DLS analysis. Next, the selectivity and anti-tumor activity of the hisABC-NB were comparatively analyzed on both normal (MCF-10) and cancerous (MCF-7) human breast cells by analyzing the cells’ survival, apoptotic gene expression profile (P53, BAX, BCL2), and flow cytometry data. Finally, the hisABC-NB’s traceability was detected by T2-weighted MRI imaging on the balb-c breast tumor models.
Results
The hisABC-NB significantly reduced the MCF-7 human breast cancer cells by inducing apoptosis response and arresting the cell cycle at the G2/M phase compared with the normal cell type (MCF-10). Moreover, the hisABC-NB exhibited a proper MRI contrast at the tumor region of treated mice compared with the non-treated type, which approved their appropriate MRI-mediated traceability.
Conclusion
The hisABC-NB’s traceability, dirigibility, and selective cytotoxicity were approved, which are the three main required factors for an efficient anticancer compound. Therefore, it has the potential to be used as an intelligent safe anticancer agent for human breast cancer treatment. However, several in vitro and in vivo studies are required to clarify its selectivity, stability, and safety.
Collapse
|
45
|
Kalın ŞN, Altay A, Budak H. Inhibition of thioredoxin reductase 1 by vulpinic acid suppresses the proliferation and migration of human breast carcinoma. Life Sci 2022; 310:121093. [DOI: 10.1016/j.lfs.2022.121093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/05/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022]
|
46
|
Saleh NAM, El-bary RBEDA, Mpingirika EZ, Essa HL, El-Sayed MMH, Sherbetjian MS, Elfandi HF, Wahed MAA, Arafeh R, Amleh A. Evaluating the Potential Anticancer Properties of Salvia triloba in Human-Osteosarcoma U2OS Cell Line and Ovarian Adenocarcinoma SKOV3 Cell Line. APPLIED SCIENCES 2022; 12:11545. [DOI: 10.3390/app122211545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Salvia triloba (S. triloba) is an herb inherently linked to traditional medicine systems in the Eastern Mediterranean region. There is minimal experimental evidence however, regarding the anticancer effects of S. triloba in both osteosarcoma and ovarian cancer. In this study, we investigated the effects of crude (macerated) S. triloba ethanol and acetone leaf extracts on viability, migratory ability, and the expression of genes regulating these activities in U2OS and SKOV3 cells using MTT assay, scratch-wound healing/trans-well migration assay, and RT-qPCR respectively. MTT assay results indicated that the acetone extract significantly reduced both U2OS and SKOV3 cell viability with half-maximal inhibitory concentrations (IC50) of 54.51 ± 1.10 µg/mL and 75.96 ± 1.0237 µg/mL respectively; these concentrations further displayed negligible hemolytic activity. The combination of acetone extract (19 µg/mL) and paclitaxel (0.787 µg/mL) displayed synergy and reduced SKOV3 cell viability by over 90%. Additionally, the trans-well migration assay illustrated that the acetone extract (IC50) inhibited both U2OS and SKOV3 cell migration by more than 50%. Moreover, S. triloba acetone extract significantly downregulated the steady-state mRNA expression of key genes involved in driving select cancer hallmarks. Four fractions were generated from the acetone extract by thin layer chromatography (TLC), and the obtained retention factors (Rf) (ranging from 0.2 to 0.8) suggested a mixture of high and moderately polar compounds whose bioactivities require further investigation. In addition, FTIR measurements of the extract revealed peaks corresponding to OH, aliphatic CH, and ester groups suggesting the presence of phenolic compounds, terpenes, and polysaccharides. Altogether, these results suggest that S. triloba possesses potential therapeutic compounds that inhibit cell proliferation and migration, and modulate several genes involved in osteosarcoma and ovarian carcinoma progression.
Collapse
|
47
|
Abu-Elfotuh K, Abdel-Sattar SA, Abbas AN, Mahran YF, Alshanwani AR, Hamdan AME, Atwa AM, Reda E, Ahmed YM, Zaghlool SS, El-Din MN. The protective effect of thymoquinone or/and thymol against monosodium glutamate-induced attention-deficit/hyperactivity disorder (ADHD)-like behavior in rats: Modulation of Nrf2/HO-1, TLR4/NF-κB/NLRP3/caspase-1 and Wnt/β-Catenin signaling pathways in rat model. Biomed Pharmacother 2022; 155:113799. [PMID: 36271575 DOI: 10.1016/j.biopha.2022.113799] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 11/26/2022] Open
Abstract
Both thymoquinone (TQ) and thymol (T) have been proved to possess a positive impact on human health. In this research, we aimed to investigate the effect of these compounds separately and together on the Attention-deficit/hyperactivity disorder (ADHD)-like behavior induced by monosodium glutamate (MSG) in rats. Forty male, Spargue Dawley rat pups (postnatal day 21), were randomly allocated into five groups: Normal saline (NS), MSG, MSG+TQ, MSG+T, and MSG+TQ+T. MSG (0.4 mg/kg/day), TQ (10 mg/kg/day) and T (30 mg/kg/day) were orally administered for 8 weeks. The behavioral tests proved that rats treated with TQ and/or T showed improved locomotor, attention and cognitive functions compared to the MSG group with more pronounced effect displayed with their combination. All treated groups showed improvement in MSG-induced aberrations in brain levels of GSH, IL-1β, TNF-α, GFAP, glutamate, calcium, dopamine, norepinephrine, Wnt3a, β-Catenin and BDNF. TQ and/or T treatment also enhanced the mRNA expression of Nrf2, HO-1 and Bcl2 while reducing the protein expression of TLR4, NFκB, NLRP3, caspase 1, Bax, AIF and GSK3β as compared to the MSG group. However, the combined therapy showed more significant effects in all measured parameters. All of these findings were further confirmed by the histopathological examinations. Current results concluded that the combined therapy of TQ and T had higher protective effects than their individual supplementations against MSG-induced ADHD-like behavior in rats.
Collapse
|
48
|
Manturthi S, Bhattacharya D, Sakhare KR, Narayan KP, Patri SV. Cimetidine-Based Cationic Amphiphiles for In Vitro Gene Delivery Targetable to Colon Cancer. ACS OMEGA 2022; 7:31388-31402. [PMID: 36092589 PMCID: PMC9453813 DOI: 10.1021/acsomega.2c03777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/10/2022] [Indexed: 06/15/2023]
Abstract
Cimetidine, a histamine-2 (H2) receptor antagonist, has been found to have anticancer properties against a number of cancer-type cells. In this report, we have demonstrated that cimetidine can acts as a hydrophilic domain in cationic lipids and targetable to the gastric system by carrying reporter genes and therapeutic genes through in vitro transfection. Two lipids, namely, Toc-Cim and Chol-Cim consisting cimetidine as the main head group and hydrophobic moieties as alpha-tocopherol or cholesterol, respectively, were designed and synthesized. 1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) is a well-known co-lipid employed to produce liposomes as uniform vesicles. The liposomes and lipoplexes were structurally and functionally evaluated for global surface charges and hydrodynamic diameters, and results found that both liposome and lipoplex size and surface charges are optimal to screen the transfection potentials. DNA-binding studies were analyzed as complete binding at all formulated N/P ratios. The liposomes and lipoplexes of both the lipids Toc-Cim and Chol-Cim show minimal cytotoxicity even though at higher concentrations. The results of the transfection experiments revealed that tocopherol-based cationic lipids (Toc-Cim) show finer transfection efficacy with optimized N/P ratios (2:1 and 4:1) in the colon cancer cell line. Toc-Cim lipoplexes show higher cellular uptake compare to Chol-Cim in the colon cancer cell line at 2:1 and 4:1 N/P ratios. Toc-Cim and Chol-Cim lipids showed highly compatible serum, examined up to 50% of the serum concentration. To evaluate the apoptotic cell death in CT-26 cells, exposed to Toc-Cim:p53 and Chol-Cim:p53 lipoplexes at 2:1 N/P ratios, superior results showed with Toc-Cim:p53. An effect of TP53 protein expression in CT-26 cell lines assayed by western blot, transfected with Toc-Cim:p53 and Chol-Cim:p53 lipoplexes, demonstrated the superior efficacy of Toc-Cim. All of the findings suggest that Toc-Cim lipid is relatively secure and is an effective transfection agent to colon cancer gene delivery.
Collapse
Affiliation(s)
- Shireesha Manturthi
- Department
of Chemistry, National Institute of Technology
Warangal, Hanamkonda, Telangana 506004, India
| | - Dwaipayan Bhattacharya
- Department
of Biological Science, Bits Pilani-Hyderabad, Hyderabad, Telangana 500078, India
| | - Kalyani Rajesh Sakhare
- Department
of Biological Science, Bits Pilani-Hyderabad, Hyderabad, Telangana 500078, India
| | - Kumar Pranav Narayan
- Department
of Biological Science, Bits Pilani-Hyderabad, Hyderabad, Telangana 500078, India
| | - Srilakshmi V. Patri
- Department
of Chemistry, National Institute of Technology
Warangal, Hanamkonda, Telangana 506004, India
| |
Collapse
|
49
|
Taghizabet N, Bahmanpour S, Zarei-fard N, Mohseni G, Aliakbari F, Dehghani F. Effect of endometrial cell-conditioned medium and platelet-rich plasma on the developmental competence of mouse preantral follicles: An in vitro study. Clin Exp Reprod Med 2022; 49:175-184. [PMID: 36097733 PMCID: PMC9468696 DOI: 10.5653/cerm.2022.05260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
Objective The aim of this study was to evaluate the impacts of platelet-rich plasma (PRP) and conditioned medium (CM) derived from endometrial stromal cells on mouse preantral follicle culture in a two-dimensional system to produce competent mature oocytes for fertilization. Methods In total, 240 preantral follicles were isolated from female mouse ovarian tissue and divided into four groups. The preantral follicles were isolated three times for each group and then cultured, respectively, in the presence of alpha minimum essential medium (control), PRP, CM, and PRP+CM. The in vitro growth, in vitro maturation, and cleavage percentage of the preantral follicles were investigated. Immunocytochemistry (IHC) was also conducted to monitor the meiotic progression of the oocytes. Additionally, the mRNA expression levels of the two folliculogenesis-related genes (Gdf9 and Bmp15) and two apoptosis-related genes (Bcl2 and Bax) were investigated using real-time polymerase chain reaction. Results In the PRP, CM, and PRP+CM groups, the preantral follicle maturation (evaluated by identifying polar bodies) were greater than the control group. The cleavage rate in the CM, and PRP+CM groups were also greater than the control group. IHC analysis demonstrated that in each treatment group, meiotic spindle was normal. In the PRP+CM group, the gene expression levels of Bmp15, Gdf9, and Bcl2 were greater than in the other groups. The Bax gene was more strongly expressed in the PRP and control groups than in the other groups. Conclusion Overall, the present study suggests that the combination of CM and PRP can effectively increase the growth and cleavage rate of mouse preantral follicles in vitro.
Collapse
Affiliation(s)
- Neda Taghizabet
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soghra Bahmanpour
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nehleh Zarei-fard
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Mohseni
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Aliakbari
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzaneh Dehghani
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Corresponding Author: Farzaneh Dehghani Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences and Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Emam Hossein Ave, Zand St, Shiraz 7134853185, Iran Tel: +91-7717-0758, E-mail:
| |
Collapse
|
50
|
Dong LY, Cao TY, Guo YH, Chen R, Zhao YS, Zhao Y, Kong H, Qu HH. Aristolochic Acid Nephropathy: A Novel Suppression Strategy of Carbon Dots Derived from Astragali Radix Carbonisata. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Despite strict restrictions on the use of aristolochic acids (AAs)-containing merchandise or drugs in many countries, a substantial amounts of occurrences aristolochic acid nephropathy (AAN) had been accounted worldwide. Clinically, there is no effective incurable therapy regimen to
reverse the progression of AAN. Although carbon dots have shown surprising bioactivity, research on the acute kidney injury caused by AAs is lacking. Here, a novel biomass-carbon dots from Astragali Radix (AR) as precursors was synthesized through one-step pyrolysis treatment. The ARC-carbon
dots (ARC-CDs) was demonstrated in detail for its inhibitory effect on aristolochic acid nephropathy in a mice model. The indexes of inflammatory cytokines as well as oxidative stress were significantly reduced by the ARC-CDs in kidney tissue cells. Additionally, the ARC-CDs administration
resulted in a large decrease in positive apoptotic cells according to TUNEL labeling and western blotting, which may be connected to the ARC-CDs’ modulation of the protein in the Akt/Mdm2/p53 signaling pathway. These findings show that ARC-CDs have remarkable anti-inflammatory, antioxidant,
and anti-apoptotic capabilities against acute kidney injury spurred by aristolochic acids via the AKT/Mdm2/p53 signaling pathway.
Collapse
Affiliation(s)
- Li-Yang Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Tian-You Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Ying-Hui Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Rui Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Yu-Sheng Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Yan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Hui Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Hui-Hua Qu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| |
Collapse
|