1
|
Faruk EM, Ibrahim F, Hassan MM, Kamal KM, Hassan DAA, Awwad AAE, Taha NM, Hablas MGA, Zaazaa AM, Ibrahim MH. Protective effects of quercetin against tongue injury and oxidative stress triggered by irinotecan: a histopathological, biochemical and molecular study. Toxicol Res (Camb) 2024; 13:tfae214. [PMID: 39703341 PMCID: PMC11652611 DOI: 10.1093/toxres/tfae214] [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: 08/05/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction About 80% of patients receiving chemotherapeutics suffer from side effects related to the gastrointestinal tract. Irinotecan (CPT-11) is a chemotherapeutic agent usually used in treating solid tumors. Quercetin (QRT), a bioflavonoid, is an antioxidant and scavenger reactive oxygen species scavenger. Objective The current study explored the possible protective effects of QRT against mucosal tongue injury caused by CPT-11. Methods The study included four equal groups: group 1/control, group 2/QRT, group 3/CPT-11, and group 4/CPT-11 + QRT. Results CPT-11-induced tongue injury in the form of non-healed ulcers, absent lingual papillae, mononuclear cells infiltration, marked deposition of collagen fibers, and overexpression of CD86 and tumor necrosis factor- α (TNF-α). The increased malondialdehyde levels, decreased superoxide dismutase and total antioxidant capacity revealed that there was an oxidative stress. Also, there was a decreased countenance of Ki-67 and Bcl-2 and an increased countenance of NF-κB. The QRT-treated group showed complete ulcer healing, with histological features almost like the control group, along with minimal collagen fiber deposition, decreased reactivity to CD86 and TNF-α and improvement of oxidative stress status and the molecular study results as well. Conclusion QRT possess protective properties against CPT-11-triggered tongue injury.
Collapse
Affiliation(s)
- Eman Mohamed Faruk
- Anatomy Department, College of Medicine, Umm Al-Qura University, Al Abidiyah, Makkah, Saudi Arabia
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Al Nadi Al Ryadi, Qism Benha 13518, Benha, Al-Qalyubia Governorate, Egypt
| | - Fatma Ibrahim
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig Rd inside Zagazig University, Shaibet an Nakareyah, Zagazig 44519, Al-Sharqia Governorate,Egypt
| | - Mahmoud M Hassan
- Department of Physiology, Faculty of Medicine, Benha University, Al Nadi Al Ryadi, Qism Benha, Al-Qalyubia Governorate, Benha 13518, Benha, Egypt
| | - Kamal M Kamal
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Al Nadi Al Ryadi, Qism Benha, Al-Qalyubia Governorate, Benha 13518, Benha, Egypt
| | | | - Ayat Abu-elnasr Awwad
- Department of Otorhinolaryngology, Faculty of Medicine, Al-Azhar University, Cairo 11751, Cairo, Egypt
| | - Neama Mahmoud Taha
- Department of Physiology, Umm Al-Qura University, Al Abidiyah, Makkah, Saudi Arabia
| | | | - Ahmed Mohammed Zaazaa
- Student at Faculty of Medicine, Benha National University, Benha Colleges in Cairo, Main Axis of El-Obour City, Egypt
| | - Mai Hassan Ibrahim
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Al Nadi Al Ryadi, Qism Benha, Al-Qalyubia Governorate, Benha 13518, Benha, Egypt
| |
Collapse
|
2
|
Alsirhani AM, Abu-Almakarem AS, Alwaili MA, Aljohani S, Alali I, AlRashidi AA, Abuzinadah NY, Alkhodair SA, Mobasher MA, Alothaim T, Eid TM, El-Said KS. Syzygium aromaticum Extract Mitigates Doxorubicin-Induced Hepatotoxicity in Male Rats. Int J Mol Sci 2024; 25:12541. [PMID: 39684253 DOI: 10.3390/ijms252312541] [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: 10/07/2024] [Revised: 11/09/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Doxorubicin (DOX), an anticancer drug, is used to treat several types of tumors, but it has detrimental side effects that restrict its therapeutic efficacy. One is the iron-dependent form of ferroptosis, which is characterized by elevated ROS production and iron overload. Syzygium aromaticum has a diverse range of biological and pharmaceutical actions due to their antioxidant properties. This study investigated the effect of S. aromaticum extract (SAE) on hepatotoxicity caused by DOX in rats. Phytochemical analysis was performed to assess compounds in SAE. The ADMETlab 2.0 web server was used to predict the pharmacokinetic properties of the most active components of SAE when DOX was injected into rats. Molecular docking studies were performed using AutoDock Vina. Forty male Sprague Dawley rats were divided into four groups of ten rats each (G1 was a negative control group, G2 was given 1/10 of SAE LD50 by oral gavage (340 mg/kg), G3 was given 4 mg/kg of DOX intraperitoneally (i.p.) once a week for a month, and G4 was administered DOX as in G3 and SAE as in G2). After a month, biochemical and histopathological investigations were performed. Rats given SAE had promising levels of phytochemicals, which could significantly ameliorate DOX-induced hepatotoxicity by restoring biochemical alterations, mitigating ferroptosis, and upregulating the NRF-2-SLC7A-11-GPX-4 signaling pathway. These findings suggest that SAE could potentially alleviate DOX-induced hepatotoxicity in rats.
Collapse
Affiliation(s)
- Alaa Muqbil Alsirhani
- Department of Chemistry, College of Science, Jouf University, Sakaka 72341, Saudi Arabia
| | - Amal S Abu-Almakarem
- Department of Basic Medical Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al Baha 65431, Saudi Arabia
| | - Maha Abdullah Alwaili
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Salwa Aljohani
- Chemistry Department, Faculty of Science, Taibah University, Yanbu Branch, Yanbu 46423, Saudi Arabia
| | - Ibtisam Alali
- Department of Chemistry, College of Science, Jouf University, Sakaka 72341, Saudi Arabia
| | | | - Najlaa Yousef Abuzinadah
- Department of Biological Science, College of Science, University of Jeddah, Jeddah 23714, Saudi Arabia
| | | | - Maysa A Mobasher
- Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
| | - Tahiyat Alothaim
- Department of Biology, College of Science, Qassim University, Buraydah 51452, Saudi Arabia
| | - Thamir M Eid
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Karim Samy El-Said
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
3
|
Nguyen MH, Nguyen TYN, Le THN, Le TNT, Chau NTN, Le TMH, Huy Nguyen BQ. Medicinal plants as a potential resource for the discovery of novel structures towards cancer drug resistance treatment. Heliyon 2024; 10:e39229. [PMID: 39492898 PMCID: PMC11530815 DOI: 10.1016/j.heliyon.2024.e39229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/23/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
Despite extensive research in chemotherapy, global cancer concerns persist, exacerbated by the challenge of drug resistance, which imposes economic and medical burdens. Natural compounds, particularly secondary metabolites from medicinal plants, present promising avenues for overcoming cancer drug resistance due to their diverse structures and essential pharmacological effects. This review provides a comprehensive exploration of cancer cell resistance mechanisms and target actions for reversing resistance and highlights the in vitro and in vivo efficacy of noteworthy alkaloids, flavonoids, and other compounds, emphasizing their potential as therapeutic agents. The molecular properties supporting ligand interactions are thoroughly examined, providing a robust theoretical foundation. The review concludes by discussing methods including quantitative structure-activity relationships and molecular docking, offering insights into screening potential candidates. Current trends in clinical treatment, contributing to a holistic understanding of the multifaceted approaches to address cancer drug resistance are also outlined.
Collapse
Affiliation(s)
- Minh Hien Nguyen
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
- Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc City, Ho Chi Minh city, Viet Nam
| | - Thi Yen Nhi Nguyen
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
- Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc City, Ho Chi Minh city, Viet Nam
- Faculty of Applied Science, Ho Chi Minh City University of Technology, Vietnam National University Ho Chi Minh City, 268 Ly Thuong Kiet Street Ward 14, District 10, Ho Chi Minh City, Viet Nam
| | - Thien Han Nguyen Le
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
| | - Thi Ngoc Tam Le
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
| | - Ngoc Trong Nghia Chau
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
| | - Tu Manh Huy Le
- University of Health Sciences, Vietnam National University Ho Chi Minh City, YA1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Di An City, Binh Duong Province, Viet Nam
| | - Bui Quoc Huy Nguyen
- The University of Danang - VN-UK Institute for Research and Executive Education, 41 Le Duan Street, Hai Chau 1 Ward, Hai Chau District, Danang City, Viet Nam
| |
Collapse
|
4
|
Pourmasoumi P, Abdouss M, Farhadi M, Jameie SB, Khonakdar HA. Co-delivery of temozolomide and quercetin with folic acid-conjugated exosomes in glioblastoma treatment. Nanomedicine (Lond) 2024; 19:2271-2287. [PMID: 39360642 PMCID: PMC11487946 DOI: 10.1080/17435889.2024.2395234] [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: 07/04/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024] Open
Abstract
Aim: The study aims to improve glioblastoma multiforme (GBM) treatment by combining temozolomide (TMZ) and quercetin (Qct), using folic acid (FA)-conjugated exosomes to overcome TMZ resistance and enhance blood-brain barrier (BBB) penetration.Methods: Exosomes were isolated and after characterizing and modifying their surfaces with FA, drug loading of TMZ and Qct into exosomes was done. In vitro assays, including cell viability tests, RT-PCR, Western-blotting and flow-cytometry, were performed using U87MG and U251MG GBM cell lines. In vivo analysis included administering exosome-drug formulations to glioblastoma-bearing Wistar rats, monitored through optical imaging and PET scans, followed by post-mortem immunohistochemistry and histological examination.Results: The results showed successful exosome isolation and FA conjugation, with drug release studies indicating accelerated release of TMZ and Qct in acidic conditions, enhancing cytotoxicity. Immunofluorescence indicated greater exosome uptake in GBM cells due to FA conjugation. Cell viability assays demonstrated increased toxicity of the combination therapy, correlating with elevated apoptosis. In vivo studies revealed significant tumor size reduction, alongside increased apoptosis and reduced angiogenesis, particularly in the TMZ-Qct-Exo-FA group.Conclusion: FA-conjugated exosomes loaded with TMZ and Qct represent a promising strategy to enhance GBM treatment efficacy by improving drug delivery, apoptosis induction and inhibiting the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Parvin Pourmasoumi
- Department of Biomedical Engineering, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Majid Abdouss
- Department of Chemistry, Amirkabir University of Technology, P.O. Box 15875-4413, Tehran, Iran
| | - Mona Farhadi
- Department of Microbiology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | | | - Hossein Ali Khonakdar
- Department of Polymer Processing, Iran Polymer & Petrochemical Institute, P.O. Box 14965-115, Tehran, Iran
- Max Bergman Center for Biomaterials, Institute of Materials Science, Technische Universität Dresden, 01069, Dresden, Germany
| |
Collapse
|
5
|
Dikeocha IJ, Wardill HR, Coller JK, Bowen JM. Dietary interventions and tumor response to chemotherapy in breast cancer: A comprehensive review of preclinical and clinical data. Clin Nutr ESPEN 2024; 63:462-475. [PMID: 39018241 DOI: 10.1016/j.clnesp.2024.06.048] [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: 05/21/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND & AIMS Optimizing treatment efficacy is still a critical part in advancing the treatment of breast cancer. Dietary interventions have drawn significant attention for their potential to increase tumor sensitivity, with a plethora of strategies evaluated both preclinically and clinically. The aim of this paper is to explore these strategies, ranging from entire dietary programs to specific supplements, for their potential to directly enhance tumor sensitivity and chemotherapy adherence. METHODS PubMed, Scopus, Embase and Web of Science databases were searched up to September 2023. In this comprehensive review, preclinical and clinical research on dietary interventions used in conjunction with chemotherapy for breast cancer was examined and synthesized, to identify potential causal mechanisms. RESULTS 42 studies in total were identified and synthesized, 32 pre-clinical and 8 clinical studies. CONCLUSION Although a topic of intense interest, the heterogeneity in approaches has resulted in a large but minimally impactful evidence base, further complicated by a limited understanding of the mechanisms at play. This review highlights the areas for further research to increase opportunities for nutritional-based interventions as adjuvant to chemotherapy for breast cancer.
Collapse
Affiliation(s)
- Ifeoma J Dikeocha
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA 5000, Australia.
| | - Hannah R Wardill
- Supportive Oncology Research Group, Precision Cancer Medicine, The South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Janet K Coller
- Discipline of Pharmacology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA 5000, Australia
| | - Joanne M Bowen
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA 5000, Australia
| |
Collapse
|
6
|
Malla R, Jyosthsna K, Rani G, Purnachandra Nagaraju G. CD44/PD-L1-mediated networks in drug resistance and immune evasion of breast cancer stem cells: Promising targets of natural compounds. Int Immunopharmacol 2024; 138:112613. [PMID: 38959542 DOI: 10.1016/j.intimp.2024.112613] [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: 01/29/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Cancer stem cells (CSCs) significantly interfere with immunotherapy, leading to challenges such as low response rates and acquired resistance. PD-L1 expression is associated with the CSC population's overexpression of CD44. Mounting evidence suggests that the breast cancer stem cell (BCSC) marker CD44 and the immune checkpoint PD-L1 contribute to treatment failure through their networks. Natural compounds can overcome therapy resistance in breast cancer by targeting mechanisms underlying resistance in BCSCs. This review provides an updated insight into the CD44 and PD-L1 networks of BCSCs in mediating metastasis and immune evasion. The review critically examines existing literature, providing a comprehensive understanding of the topic and emphasizing the impact of natural flavones on the signaling pathways of BCSCs. Additionally, the review discusses the potential of natural compounds in targeting CD44 and PD-L1 in breast cancer (BC). Natural compounds consistently show potential in targeting regulatory mechanisms of BCSCs, inducing loss of stemness, and promoting differentiation. They offer a promising approach for developing alternative therapeutic strategies to manage breast cancer.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India; Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India.
| | - Kattula Jyosthsna
- Department of Biotechnology, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - G Rani
- Department of Biotechnology, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA
| |
Collapse
|
7
|
Rostamkhani N, Salimi M, Adibifar A, Karami Z, Agh-Atabay AH, Rostamizadeh K, Abdi Z. Enhanced anti-tumor and anti-metastatic activity of quercetin using pH-sensitive Alginate@ZIF-8 nanocomposites: in vitroand in vivostudy. NANOTECHNOLOGY 2024; 35:475102. [PMID: 39163872 DOI: 10.1088/1361-6528/ad713f] [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: 05/12/2024] [Accepted: 08/20/2024] [Indexed: 08/22/2024]
Abstract
Quercetin (Qc) possesses anti-cancer properties, such as cell signaling, growth suppression, pro-apoptotic, anti-proliferative, and antioxidant effects. In this study, we developed an alginate-modified ZIF-8 (Alg@ZIF-8) to enhance the anti-tumor efficacy of Qc. The developed alginate-modified quercetin-loaded ZIF-8 (Alg@Qc@ZIF-8) was characterized using scanning electron microscope (SEM), dynamic light scattering (DLS), fourier transform infrared spectroscopy Thermogravimetric analysis, Brunauer-Emmett-Teller, and x-ray diffraction. The drug release pattern was evaluated at pH 5.4 and 7.4. The cytotoxicity of nanoparticles was assessed on the 4T1 cell line. Finally, the anti-tumor activity of Alg@Qc@ZIF-8 was evaluated in 4T1 tumor-bearing mice. SEM showed that the nanoparticles were spherical with a diameter of mainly below 50 nm. The DLS showed that the developed nanoparticles' hydrodynamic diameter, zeta potential, and polydispersity index were 154.9 ± 7.25 nm, -23.8 ± 5.33 mV, and 0.381 ± 0.09, respectively. The drug loading capacity was 10.40 ± 0.02%. Alg@Qc@ZIF-8 exhibited pH sensitivity, releasing more Qc at pH 5.4 (about 3.62 times) than at pH 7.4 after 24 h. Furthermore, the IC50value of Alg@Qc@ZIF-8 on the 4T1 cell line was 2.16 times lower than net Qc. Importantly, in tumor-bearing mice, Alg@Qc@ZIF-8 demonstrated enhanced inhibitory effects on tumor growth and lung metastasis compared to net Qc. Considering thein vitroandin vivooutcomes, Alg@Qc@ZIF-8 might hold great potential for effective breast cancer management.
Collapse
Affiliation(s)
- Neda Rostamkhani
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Maryam Salimi
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Arghavan Adibifar
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Zahra Karami
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Kobra Rostamizadeh
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Psychiatry and Behavioral Sciences, Department of Pharmacology, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Zahra Abdi
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
8
|
Wang S, Wang K, Li C, Chen J, Kong X. Role of flavonoids in inhibiting triple-negative breast cancer. Front Pharmacol 2024; 15:1411059. [PMID: 39257397 PMCID: PMC11384598 DOI: 10.3389/fphar.2024.1411059] [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: 04/02/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
Increasing incidences of metastasis or recurrence (or both) in triple-negative breast cancer (TNBC) are a growing concern worldwide, as these events are intricately linked to higher mortality rates in patients with advanced breast cancer. Flavonoids possess several pharmaceutical advantages with multi-level, multi-target, and coordinated intervention abilities for treating TNBC, making them viable for preventing tumor growth and TNBC metastasis. This review focused on the primary mechanisms by which flavonoids from traditional Chinese medicine extracts inhibit TNBC, including apoptosis, blocking of cell cycle and movement, regulation of extracellular matrix degradation, promotion of anti-angiogenesis, inhibition of aerobic glycolysis, and improvement in tumor microenvironment. This review aims to improve the knowledge of flavonoids as a promising pharmacological intervention for patients with TNBC.
Collapse
Affiliation(s)
- Shuai Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Kuanyu Wang
- The Second Department of Surgery, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Cheng Li
- The Second Department of Surgery, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Jing Chen
- The Second Department of Surgery, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xiangding Kong
- The Second Department of Surgery, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
9
|
Fang T, Wang J, Sun S, Deng X, Xue M, Han F, Sun B, Chen L. JinLiDa granules alleviates cardiac hypertrophy and inflammation in diabetic cardiomyopathy by regulating TP53. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155659. [PMID: 38759318 DOI: 10.1016/j.phymed.2024.155659] [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: 11/26/2023] [Revised: 03/14/2024] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND JinLiDa granules (JLD) is a traditional Chinese medicine (TCM) used to treat type 2 diabetes mellitus with Qi and Yin deficiency. Clinical evidence has shown that JLD can alleviate diabetic cardiomyopathy, but the exact mechanism is not yet clear. PURPOSE The purpose of this study was to examine the potential role and mechanism of JLD in the treatment of diabetic cardiomyopathy through network pharmacological analysis and basic experiments. METHODS The targets of JLD associated with diabetic cardiomyopathy were examined by network pharmacology. Protein interaction analysis was performed on the targets, and the associated pathways were searched by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Diabetic mice were treated with low or high doses of JLD by gavage, and AC16 and H9C2 cardiomyocytes exposed to high-glucose conditions were treated with JLD. The analysis results were verified by various experimental techniques to examine molecular mechanisms. RESULTS Network pharmacological analysis revealed that JLD acted on the tumor suppressor p53 (TP53) during inflammation and fibrosis associated with diabetic cardiomyopathy. The results of basic experiments showed that after JLD treatment, ventricular wall thickening in diabetic mouse hearts was attenuated, cardiac hypertrophy and myocardial inflammation were alleviated, and the expression of cardiac hypertrophy- and inflammation-related factors in cardiomyocytes exposed to a high-glucose environment was decreased. Cardiomyocyte morphology also improved after JLD treatment. TP53 expression and the tumor necrosis factor (TNF) and transforming growth factor beta-1 (TGFβ1) signaling pathways were significantly altered, and inhibiting TP53 expression effectively alleviated the activation of the TNF and TGFβ1 signaling pathways under high glucose conditions. Overexpression of TP53 activated these signaling pathways. CONCLUSIONS JLD acted on TP53 to regulate the TNF and TGFβ1 signaling pathways, effectively alleviating cardiomyocyte hypertrophy and inflammation in high glucose and diabetic conditions. Our study provides a solid foundation for the future treatment of diabetic cardiomyopathy with JLD.
Collapse
Affiliation(s)
- Ting Fang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Jingyi Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Shengnan Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Xiaoqing Deng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Mei Xue
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
| | - Fei Han
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China.
| | - Liming Chen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China.
| |
Collapse
|
10
|
El Oirdi M. Harnessing the Power of Polyphenols: A New Frontier in Disease Prevention and Therapy. Pharmaceuticals (Basel) 2024; 17:692. [PMID: 38931359 PMCID: PMC11206774 DOI: 10.3390/ph17060692] [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: 04/28/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
There are a wide variety of phytochemicals collectively known as polyphenols. Their structural diversity results in a broad range of characteristics and biological effects. Polyphenols can be found in a variety of foods and drinks, including fruits, cereals, tea, and coffee. Studies both in vitro and in vivo, as well as clinical trials, have shown that they possess potent antioxidant activities, numerous therapeutic effects, and health advantages. Dietary polyphenols have demonstrated the potential to prevent many health problems, including obesity, atherosclerosis, high blood sugar, diabetes, hypertension, cancer, and neurological diseases. In this paper, the protective effects of polyphenols and the mechanisms behind them are investigated in detail, citing the most recent available literature. This review aims to provide a comprehensive overview of the current knowledge on the role of polyphenols in preventing and managing chronic diseases. The cited publications are derived from in vitro, in vivo, and human-based studies and clinical trials. A more complete understanding of these naturally occurring metabolites will pave the way for the development of novel polyphenol-rich diet and drug development programs. This, in turn, provides further evidence of their health benefits.
Collapse
Affiliation(s)
- Mohamed El Oirdi
- Department of Life Sciences, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia;
- Department of Basic Sciences, Preparatory Year, King Faisal University, Al Ahsa 31982, Saudi Arabia
| |
Collapse
|
11
|
Sakao K, Hamamoto S, Urakawa D, He Z, Hou DX. Anticancer Activity and Molecular Mechanisms of Acetylated and Methylated Quercetin in Human Breast Cancer Cells. Molecules 2024; 29:2408. [PMID: 38792269 PMCID: PMC11124128 DOI: 10.3390/molecules29102408] [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: 04/16/2024] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Quercetin, a flavonoid polyphenol found in many plants, has garnered significant attention due to its potential cancer chemoprevention. Our previous studies have shown that acetyl modification of the hydroxyl group of quercetin altered its antitumor effects in HepG2 cells. However, the antitumor effect in other cancer cells with different gene mutants remains unknown. In this study, we investigated the antitumor effect of quercetin and its methylated derivative 3,3',4',7-O-tetramethylquercetin (4Me-Q) and acetylated derivative 3,3',4',7-O-tetraacetylquercetin (4Ac-Q) on two human breast cancer cells, MCF-7 (wt-p53, caspase-3-ve) and MDA-MB-231 (mt-p53, caspase-3+ve). The results demonstrated that 4Ac-Q exhibited significant cell proliferation inhibition and apoptosis induction in both MCF-7 and MDA-MB-231 cells. Conversely, methylation of quercetin was found to lose the activity. The human apoptosis antibody array revealed that 4Ac-Q might induce apoptosis in MCF-7 cells via a p53-dependent pathway, while in MDA-MB-231 cells, it was induced via a caspase-3-dependent pathway. Furthermore, an evaluation using a superoxide inhibitor, MnTBAP, revealed 4Ac-Q-induced apoptosis in MCF-7 cells in a superoxide-independent manner. These findings provide valuable insights into the potential of acetylated quercetin as a new approach in cancer chemoprevention and offer new avenues for health product development.
Collapse
Affiliation(s)
- Kozue Sakao
- The United Graduate School of Agriculture Sciences, Kagoshima University, Kagoshima 890-0065, Japan; (D.U.); (Z.H.); (D.-X.H.)
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| | - Shihomi Hamamoto
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| | - Daigo Urakawa
- The United Graduate School of Agriculture Sciences, Kagoshima University, Kagoshima 890-0065, Japan; (D.U.); (Z.H.); (D.-X.H.)
| | - Ziyu He
- The United Graduate School of Agriculture Sciences, Kagoshima University, Kagoshima 890-0065, Japan; (D.U.); (Z.H.); (D.-X.H.)
| | - De-Xing Hou
- The United Graduate School of Agriculture Sciences, Kagoshima University, Kagoshima 890-0065, Japan; (D.U.); (Z.H.); (D.-X.H.)
- Graduate School of Agriculture, Forestry and Fisheries, Kagoshima University, Kagoshima 890-0065, Japan
| |
Collapse
|
12
|
Wang L, He S, Liu R, Xue Y, Quan Y, Shi R, Yang X, Lin Q, Sun X, Zhang Z, Zhang L. A pH/ROS dual-responsive system for effective chemoimmunotherapy against melanoma via remodeling tumor immune microenvironment. Acta Pharm Sin B 2024; 14:2263-2280. [PMID: 38799639 PMCID: PMC11119573 DOI: 10.1016/j.apsb.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 05/29/2024] Open
Abstract
Chemotherapeutics can induce immunogenic cell death (ICD) in tumor cells, offering new possibilities for cancer therapy. However, the efficiency of the immune response generated is insufficient due to the inhibitory nature of the tumor microenvironment (TME). Here, we developed a pH/reactive oxygen species (ROS) dual-response system to enhance chemoimmunotherapy for melanoma. The system productively accumulated in tumors by specific binding of phenylboronic acid (PBA) to sialic acids (SA). The nanoparticles (NPs) rapidly swelled and released quercetin (QUE) and doxorubicin (DOX) upon the stimulation of tumor microenvironment (TME). The in vitro and in vivo results consistently demonstrated that the NPs improved anti-tumor efficacy and prolonged survival of mice, significantly enhancing the effects of the combination. Our study revealed DOX was an ICD inducer, stimulating immune responses and promoting maturation of dendritic cells (DCs). Additionally, QUE served as a TME regulator by inhibiting the cyclooxygenase-2 (COX2)-prostaglandin E2 (PGE2) axis, which influenced various immune cells, including increasing cytotoxic T cells (CLTs) infiltration, promoting M1 macrophage polarization, and reducing regulatory T cells (Tregs) infiltration. The combination synergistically facilitated chemoimmunotherapy efficacy by remodeling the immunosuppressive microenvironment. This work presents a promising strategy to increase anti-tumor efficiency of chemotherapeutic agents.
Collapse
Affiliation(s)
- Leilei Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shanshan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rong Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuan Xue
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuan Quan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rongying Shi
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xueying Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Qing Lin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ling Zhang
- Med-X Center for Materials, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
13
|
Almohammad Aljabr B, Zihlif M, Abu-Dahab R, Zalloum H. Effect of quercetin on doxorubicin cytotoxicity in sensitive and resistant human MCF7 breast cancer cell lines. Biomed Rep 2024; 20:58. [PMID: 38414625 PMCID: PMC10895388 DOI: 10.3892/br.2024.1745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 09/26/2023] [Indexed: 02/29/2024] Open
Abstract
Chemoresistance is the major cause of cancer recurrence, relapse and eventual death. Doxorubicin resistance is one such challenge in breast cancer. The use of quercetin, an antioxidant, in combination with doxorubicin has been investigated for offering protection to normal cells from the toxic side effects of doxorubicin in addition to modulation of its resistance. The present study aimed to investigate the effects of quercetin in prevention of a doxorubicin-chemoresistant phenotype in both doxorubicin-sensitive and -resistant human MCF-7 breast cancer cell lines. A doxorubicin-resistant MCF-7 cell line was established. The development of resistant cells was closely monitored for changes in morphological features. Sensitivity to doxorubicin and the doxorubicin/quercetin combination was assessed using the tetrazolium assay. To determine the mechanism by which quercetin sensitizes the doxorubicin MCF-7-resistant cell line to doxorubicin, gene expression alterations in breast cancer-related genes were examined using the reverse transcription-quantitative PCR (RT-qPCR) array technology. Resistant MCF cells were successfully developed and the inhibitory concentration (IC50) value of doxorubicin increased from 0.133 to 4 µM (wild-type to resistant). The effects of the quercetin/doxorubicin combination exhibited different effects on wild-type vs. resistant cells. The IC50 of doxorubicin was reduced in wild cells, whereas resistant cells showed an increase in cell viability at lower concentrations and a potentiation of the effects of doxorubicin only at higher concentrations. Annexin V/propidium iodide staining demonstrated that quercetin drives cells into late apoptosis and necrosis, but in resistant cells, necrosis predominates. RT-qPCR results revealed that quercetin led to a reversal in doxorubicin effects via up- and downregulation of important genes such as SNAI2, PLAU and CSF1 genes. Downregulation of cell migration genes, SNAI2 (-31.23-fold) and plasminogen activator, urokinase (PLAU; -30.62-fold), and the apoptotic pathway gene, colony stimulating factor 1 (CSF1; -17.25-fold) were the most important querticin-associated events. Other gene alterations were also observed involving cell cycle arrest and DNA repair pathways. The results of the present study indicated that quercetin could lead to a reversal of doxorubicin resistance in breast cancer cells via downregulation of the expression of important genes, such as SNAI2, PLAU and CSF1. Such findings may represent a potential strategy for reversing breast cancer cell-related chemoresistance.
Collapse
Affiliation(s)
- Bayan Almohammad Aljabr
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Malek Zihlif
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Rana Abu-Dahab
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Hiba Zalloum
- Hamdi Mango Research Center for Scientific Research, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
14
|
Jadhav PV, Prasath NJ, Gajbhiye SG, Rane UA, Agnihotri TG, Gomte SS, Jain A. Empowering the Battle: Bioenhancers as Allies Against Cancer Drug Resistance. Curr Pharm Biotechnol 2024; 25:1552-1563. [PMID: 37957922 DOI: 10.2174/0113892010192038231107051715] [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: 03/29/2023] [Revised: 08/14/2023] [Accepted: 08/25/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Drug resistance has been a great hindrance in the path of counteracting diseases like cancer and is driven by drugs misuse and overuse. In terms of cancer, resistance has been developed due to cellular changes, altered growth activation pathways, increased expression of efflux proteins, and changes in the local physiology of cancer (blood supply, tissue hydrodynamics, increased mutation rate/epigenetics, tumor cell heterogeneity). One of the approaches to address these challenges is the use of bioenhancers, which can overcome drug resistance, thereby improving bioavailability (BA). CONCLUSION Bioenhancers when combined with drugs can elicit pharmacological activity. They are generally combined with therapeutic agents at low doses, which increase the BA or therapeutic activity of active pharmaceutical ingredient (API). This review sheds light on the synthesis and classification of bio-enhancers. It also discusses different applications of bio-enhancers like piperine, ginger, quercetin, curcumin, etc. in the treatment of cancer. The review also presents some of the recent advancements in terms of nanocarriers for delivering API combined with bioenhancers.
Collapse
Affiliation(s)
- Pratiksha Vasant Jadhav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Naga Jothi Prasath
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Saurabh Ghannil Gajbhiye
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Utkarsha Arun Rane
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| |
Collapse
|
15
|
Yang Y, Yan J, Huang J, Wu X, Yuan Y, Yuan Y, Zhang S, Mo F. Exploring the mechanism by which quercetin re-sensitizes breast cancer to paclitaxel: network pharmacology, molecular docking, and experimental verification. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3045-3059. [PMID: 37148401 DOI: 10.1007/s00210-023-02510-9] [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: 04/01/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
This study is aimed to explore the potential molecular mechanism of quercetin reversing paclitaxel (PTX) resistance in breast cancer (BC) by network pharmacology, molecular docking, and experimental verification. Pharmacological platform databases are used to predict quercetin targets and BC PTX-resistance genes and constructed the expression profile of quercetin chemosensitization. The overlapping targets were input into the STRING database and used Cytoscape v3.9.0 to construct the protein-protein interaction (PPI) network. Subsequently, these targets were performed with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses and molecular docking. Finally, we further detected the potential role of quercetin in improving PTX sensitivity in BC in vitro experiments. Compounds and targets screening hinted that 220 quercetin predicted targets, 244 BC PTX resistance-related genes, and 66 potential sensitive target genes (PSTGs). Network pharmacology screening revealed the top-15 crucial targets in PPI network of quercetin reversing the sensitivity of BC to PTX. KEGG analysis revealed that they were mainly enriched in the EGFR/ERK signaling pathway. Molecular docking showed that both quercetin and PTX could stably bind to the key targets in the EGFR/ERK signaling pathway. In vitro experiments further confirmed that quercetin inhibited the key targets in the EGFR/ERK axis to the suppression of cell proliferation and promotion of apoptosis in PTX-resistance BC cells, and restoring the activity of the resistant cells to PTX. Our results suggested that quercetin increased the sensitivity of BC to PTX through inhibiting EGFR/ERK axis, and it is an effective treatment for reversing PTX resistance.
Collapse
Affiliation(s)
- Ye Yang
- Department of Basic Clinical Laboratory Medicine, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, China
| | - Jiaoyan Yan
- Department of Basic Clinical Laboratory Medicine, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, China
| | - Jian Huang
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Xiangyi Wu
- Department of Basic Clinical Laboratory Medicine, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, China
| | - Yan Yuan
- Department of Basic Clinical Laboratory Medicine, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, China
| | - Yan Yuan
- Department of Clinical Laboratory, The First People's Hospital of Guiyang, Guiyang, 550002, China
| | - Shu Zhang
- Department of Basic Clinical Laboratory Medicine, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, China.
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| | - Fei Mo
- Department of Basic Clinical Laboratory Medicine, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, China.
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
16
|
El-Said KS, Haidyrah AS, Mobasher MA, Khayyat AIA, Shakoori A, Al-Sowayan NS, Barnawi IO, Mariah RA. Artemisia annua Extract Attenuate Doxorubicin-Induced Hepatic Injury via PI-3K/Akt/Nrf-2-Mediated Signaling Pathway in Rats. Int J Mol Sci 2023; 24:15525. [PMID: 37958509 PMCID: PMC10647718 DOI: 10.3390/ijms242115525] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Doxorubicin (DOX), which is used to treat cancer, has harmful effects that limit its therapeutic application. Finding preventative agents to thwart DOX-caused injuries is thus imperative. Artemisia annua has numerous biomedical uses. This study aims to investigate the attenuative effect of Artemisia annua leaf extract (AALE) treatment on DOX-induced hepatic toxicity in male rats. A phytochemical screening of AALE was evaluated. Forty male rats were used; G1 was a negative control group, G2 was injected with AALE (150 mg/kg) intraperitoneally (i.p) daily for a month, 4 mg/kg of DOX was given i.p to G3 once a week for a month, and G4 was injected with DOX as G3 and with AALE as G2. Body weight changes and biochemical, molecular, and histopathological investigations were assessed. The results showed that AALE contains promising phytochemical constituents that contribute to several potential biomedical applications. AALE mitigated the hepatotoxicity induced by DOX in rats as evidenced by restoring the alterations in the biochemical parameters, antioxidant gene expression, and hepatic histopathological alterations in rats. Importantly, the impact of AALE against the hepatic deterioration resulting from DOX treatment is through activation of the PI-3K/Akt/Nrf-2 signaling, which in turn induces the antioxidant agents.
Collapse
Affiliation(s)
- Karim Samy El-Said
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt;
| | - Ahmed S. Haidyrah
- Digital & Smart Laboratories (DSL), King Abdulaziz City for Science & Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Maysa A. Mobasher
- Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Arwa Ishaq A. Khayyat
- Biochemistry Department, Science College, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Afnan Shakoori
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | | | - Ibrahim Omar Barnawi
- Department of Biological Sciences, Faculty of Science, Taibah University, Al-Madinah Al-Munawwarah 41321, Saudi Arabia;
| | - Reham A. Mariah
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
17
|
Attar ES, Chaudhari VH, Deokar CG, Dyawanapelly S, Devarajan PV. Nano Drug Delivery Strategies for an Oral Bioenhanced Quercetin Formulation. Eur J Drug Metab Pharmacokinet 2023; 48:495-514. [PMID: 37523008 DOI: 10.1007/s13318-023-00843-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 08/01/2023]
Abstract
Quercetin, a naturally occurring flavonoid, has been credited with a wide spectrum of therapeutic properties. However, the oral use of quercetin is limited due to its poor water solubility, low bioavailability, rapid metabolism, and rapid plasma clearance. Quercetin has been studied extensively when used with various nanodelivery systems for enhancing quercetin bioavailability. To enhance its oral bioavailability and efficacy, various quercetin-loaded nanosystems such as nanosuspensions, polymer nanoparticles, metal nanoparticles, emulsions, liposomes or phytosomes, micelles, solid lipid nanoparticles, and other lipid-based nanoparticles have been investigated in in-vitro cells, in-vivo animal models, and humans. Among the aforementioned nanosystems, quercetin phytosomes are attracting more interest and are available on the market. The present review covers insights into the possibilities of harnessing quercetin for several therapeutic applications and a special focus on anticancer applications and the clinical benefits of nanoquercetin formulations.
Collapse
Affiliation(s)
- Esha S Attar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, N.P. Marg, Matunga, Mumbai, Maharashtra, 400019, India
| | - Vanashree H Chaudhari
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, N.P. Marg, Matunga, Mumbai, Maharashtra, 400019, India
| | - Chaitanya G Deokar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, N.P. Marg, Matunga, Mumbai, Maharashtra, 400019, India
| | - Sathish Dyawanapelly
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, N.P. Marg, Matunga, Mumbai, Maharashtra, 400019, India
| | - Padma V Devarajan
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, N.P. Marg, Matunga, Mumbai, Maharashtra, 400019, India.
| |
Collapse
|
18
|
Monteiro-Alfredo T, dos Santos JM, Antunes KÁ, Cunha J, da Silva Baldivia D, Pires AS, Marques I, Abrantes AM, Botelho MF, Monteiro L, Gonçalves AC, Botelho WH, Paula de Araújo Boleti A, Cabral C, Oliveira PJ, Lucas dos Santos E, Matafome P, de Picoli Souza K. Acrocomia aculeata associated with doxorubicin: cardioprotection and anticancer activity. Front Pharmacol 2023; 14:1223933. [PMID: 37654604 PMCID: PMC10466431 DOI: 10.3389/fphar.2023.1223933] [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: 05/25/2023] [Accepted: 08/01/2023] [Indexed: 09/02/2023] Open
Abstract
Doxorubicin (Dox) is a chemotherapeutic agent widely used in the clinic, whose side effects include cardiotoxicity, associated with decreased antioxidant defenses and increased oxidative stress. The association of Dox with natural antioxidants can extend its use if not interfering with its pharmacological potential. In this study, we aimed to understand the effects and mechanisms of the aqueous extract of Acrocomia aculeata leaves (EA-Aa) in cancer cells and the co-treatment with Dox, in in vitro and in vivo models. It was found that EA-Aa showed a relevant decrease in the viability of cancer cells (K562 and MCF-7) and increased apoptosis and death. The Dox cytotoxic effect in co-treatment with EA-Aa was increased in cancer cells. The therapeutic association also promoted a change in cell death, leading to a higher rate of apoptosis compared to the Dox group, which induced necrosis. In addition, in non-cancer cells, EA-Aa enhanced red blood cell (RBC) redox state with lower hemolysis and malondialdehyde (MDA) content and had no in vitro nor in vivo toxicity. Furthermore, EA-Aa showed antioxidant protection against Dox-induced cytotoxicity in H9c2 cells (cardiomyoblast), partially mediated by the NRF2 pathway. In vivo, EA-Aa treatment showed a relevant decrease in MDA levels in the heart, kidney, and brain, evaluated in C57Bl/6 mice induced to cardiotoxicity by Dox. Together, our results proved the effectiveness of EA-Aa in potentiating Dox anticancer effects, with antioxidant and cardioprotective activity, suggesting EA-Aa as a potential Dox pharmacological adjuvant.
Collapse
Affiliation(s)
- Tamaeh Monteiro-Alfredo
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
- Faculty of Medicine, Institute of Physiology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| | - Jéssica Maurino dos Santos
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Kátia Ávila Antunes
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Janielle Cunha
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Debora da Silva Baldivia
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Ana Salomé Pires
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University Coimbra, Coimbra, Portugal
| | - Inês Marques
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University Coimbra, Coimbra, Portugal
| | - Ana Margarida Abrantes
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University Coimbra, Coimbra, Portugal
| | - Maria Filomena Botelho
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University Coimbra, Coimbra, Portugal
| | - Lúcia Monteiro
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University Coimbra, Coimbra, Portugal
| | - Ana Cristina Gonçalves
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University Coimbra, Coimbra, Portugal
| | - Wellington Henrique Botelho
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Ana Paula de Araújo Boleti
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Célia Cabral
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Paulo J. Oliveira
- CNC—Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Edson Lucas dos Santos
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| | - Paulo Matafome
- Faculty of Medicine, Institute of Physiology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
- Department of Complementary Sciences, Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - Kely de Picoli Souza
- Research Group on Biotechnology and Bioprospection Applied to Metabolism and Cancer (GEBBAM), Federal University of Grande Dourados, Dourados, Brazil
| |
Collapse
|
19
|
Macrì R, Bava I, Scarano F, Mollace R, Musolino V, Gliozzi M, Greco M, Foti D, Tucci L, Maiuolo J, Carresi C, Tavernese A, Palma E, Muscoli C, Mollace V. In Vitro Evaluation of Ferutinin Rich- Ferula communis L., ssp. glauca, Root Extract on Doxorubicin-Induced Cardiotoxicity: Antioxidant Properties and Cell Cycle Modulation. Int J Mol Sci 2023; 24:12735. [PMID: 37628916 PMCID: PMC10454821 DOI: 10.3390/ijms241612735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
The clinical use of anthracycline Doxorubicin as an antineoplastic drug in cancer therapy is limited by cardiotoxic effects that can lead to congestive heart failure. Recent studies have shown several promising activities of different species of the genus Ferula belonging to the Apiaceae Family. Ferula communis is the main source of Ferutinin-a bioactive compound isolated from many species of Ferula-studied both in vitro and in vivo because of their different effects, such as estrogenic, antioxidant, anti-inflammatory, and also antiproliferative and cytotoxic activity, performed in a dose-dependent and cell-dependent way. However, the potential protective role of Ferutinin in myocardium impairment, caused by chemotherapeutic drugs, still represents an unexplored field. The aim of this study was to test the effects of Ferutinin rich-Ferula communis L. root extract (FcFE) at different concentrations on H9C2 cells. Moreover, we evaluated its antioxidant properties in cardiomyocytes in order to explore new potential therapeutic activities never examined before in other experimental works. FcFE, at a concentration of 0.25 µM, in the H9C2 line, significantly reduced the ROS production induced by H2O2 (50 µM and 250 µM) and traced the cell mortality of the H9C2 co-treated with Ferutinin 0.25 µM and Doxorubicin (0.5 µM and 1 µM) to control levels. These results showed that FcFE could protect against Doxorubicin-induced cardiotoxicity. Further molecular characterization of this natural compound may open the way for testing FcFE at low concentrations in vivo and in clinical studies as an adjuvant in cancer therapy in association with anthracyclines to prevent side effects on heart cells.
Collapse
Affiliation(s)
- Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Irene Bava
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Federica Scarano
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy
- Division of Cardiology, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| | - Vincenzo Musolino
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (V.M.); (J.M.)
| | - Micaela Gliozzi
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Marta Greco
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (D.F.)
| | - Daniela Foti
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (D.F.)
| | - Luigi Tucci
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Jessica Maiuolo
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (V.M.); (J.M.)
| | - Cristina Carresi
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (C.C.); (E.P.)
| | - Annamaria Tavernese
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Ernesto Palma
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (C.C.); (E.P.)
| | - Carolina Muscoli
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Lamezia Terme, 88046 Catanzaro, Italy
| |
Collapse
|
20
|
Hosseinzadeh A, Poursoleiman F, Biregani AN, Esmailzadeh A. Flavonoids target different molecules of autophagic and metastatic pathways in cancer cells. Cancer Cell Int 2023; 23:114. [PMID: 37308913 DOI: 10.1186/s12935-023-02960-4] [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: 03/12/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023] Open
Abstract
Despite the success of cancer therapy, it has encountered a major obstacle due to the complicated nature of cancer, namely resistance. The recurrence and metastasis of cancer occur when anti-cancer therapeutic agents fail to eradicate all cancer cells. Cancer therapy aims to find the best agent that targets all cancer cells, including those sensitive or resistant to treatment. Flavonoids, natural products from our diet, show anti-cancer effects in different studies. They can inhibit metastasis and the recurrence of cancers. This review discusses metastasis, autophagy, anoikis in cancer cells, and their dynamic relationship. We present evidence that flavonoids can block metastasis and induce cell death in cancer cells. Our research suggests that flavonoids can serve as potential therapeutic agents in cancer therapy.
Collapse
Affiliation(s)
- Aysooda Hosseinzadeh
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Faezeh Poursoleiman
- Department of Cellular and Molecular Nutrition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Naghdipour Biregani
- Department of Nutrition, School of Health, Shahid Sadoughi University of Medical Scinences, Yazd, Iran
| | - Ahmad Esmailzadeh
- Students' Scientific Center, Tehran University of Medical Sciences, Tehran, Iran.
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
- Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
21
|
Linders AN, Dias IB, Ovchinnikova ES, Vermeer MC, Hoes MF, Markousis Mavrogenis G, Deiman FE, Arevalo Gomez KF, Bliley JM, Nehme J, Vink A, Gietema J, de Boer RA, Westenbrink D, Sillje HH, Hilfiker-Kleiner D, van Laake LW, Feinberg AW, Demaria M, Bomer N, van der Meer P. Evaluation of Senescence and Its Prevention in Doxorubicin-Induced Cardiotoxicity Using Dynamic Engineered Heart Tissues. JACC CardioOncol 2023; 5:298-315. [PMID: 37397084 PMCID: PMC10308053 DOI: 10.1016/j.jaccao.2023.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 07/04/2023] Open
Abstract
Background Doxorubicin is an essential cancer treatment, but its usefulness is hampered by the occurrence of cardiotoxicity. Nevertheless, the pathophysiology underlying doxorubicin-induced cardiotoxicity and the respective molecular mechanisms are poorly understood. Recent studies have suggested involvement of cellular senescence. Objectives The aims of this study were to establish whether senescence is present in patients with doxorubicin-induced cardiotoxicity and to investigate if this could be used as a potential treatment target. Methods Biopsies from the left ventricles of patients with severe doxorubicin-induced cardiotoxicity were compared with control samples. Additionally, senescence-associated mechanisms were characterized in 3-dimensional dynamic engineered heart tissues (dyn-EHTs) and human pluripotent stem cell-derived cardiomyocytes. These were exposed to multiple, clinically relevant doses of doxorubicin to recapitulate patient treatment regimens. To prevent senescence, dyn-EHTs were cotreated with the senomorphic drugs 5-aminoimidazole-4-carboxamide ribonucleotide and resveratrol. Results Senescence-related markers were significantly up-regulated in the left ventricles of patients with doxorubicin-induced cardiotoxicity. Treatment of dyn-EHTs resulted in up-regulation of similar senescence markers as seen in the patients, accompanied by tissue dilatation, decreased force generation, and increased troponin release. Treatment with senomorphic drugs led to decreased expression of senescence-associated markers, but this was not accompanied by improved function. Conclusions Senescence was observed in the hearts of patients with severe doxorubicin-induced cardiotoxicity, and this phenotype can be modeled in vitro by exposing dyn-EHTs to repeated clinically relevant doses of doxorubicin. The senomorphic drugs 5-aminoimidazole-4-carboxamide ribonucleotide and resveratrol prevent senescence but do not result in functional improvements. These findings suggest that preventing senescence by using a senomorphic during doxorubicin administration might not prevent cardiotoxicity.
Collapse
Affiliation(s)
- Annet N. Linders
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Itamar B. Dias
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ekaterina S. Ovchinnikova
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mathilde C.S.C. Vermeer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martijn F. Hoes
- Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
- Department of Cardiology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - George Markousis Mavrogenis
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Frederik E. Deiman
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Karla F. Arevalo Gomez
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jacqueline M. Bliley
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Jamil Nehme
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jourik Gietema
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Daan Westenbrink
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Herman H.W. Sillje
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Denise Hilfiker-Kleiner
- Institute of Cardiovascular Complications in Pregnancy and in Oncologic Therapies, Philipps-Universität Marburg, Marburg, Germany
| | - Linda W. van Laake
- Division of Heart and Lungs and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Adam W. Feinberg
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Marco Demaria
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
22
|
Michlewska S, Garaiova Z, Šubjakova V, Hołota M, Kubczak M, Grodzicka M, Okła E, Naziris N, Balcerzak Ł, Ortega P, de la Mata FJ, Hianik T, Waczulikova I, Bryszewska M, Ionov M. Lipid-coated ruthenium dendrimer conjugated with doxorubicin in anti-cancer drug delivery: Introducing protocols. Colloids Surf B Biointerfaces 2023; 227:113371. [PMID: 37244201 DOI: 10.1016/j.colsurfb.2023.113371] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/12/2023] [Accepted: 05/23/2023] [Indexed: 05/29/2023]
Abstract
One of the major limitations for the treatment of many diseases is an inability of drugs to cross the cell membrane barrier. Different kinds of carriers are being investigated to improve drug bioavailability. Among them, lipid or polymer-based systems are of special interest due to their biocompatibility. In our study, we combined dendritic and liposomal carriers and analysed the biochemical and biophysical properties of these formulations. Two preparation methods of Liposomal Locked-in Dendrimers (LLDs) systems have been established and compared. Carbosilane ruthenium metallodendrimer was complexed with an anti-cancer drug (doxorubicin) and locked in a liposomal structure, using both techniques. The LLDs systems formed by hydrophilic locking had more efficient transfection profiles and interacted with the erythrocyte membrane better than systems using the hydrophobic method. The results indicate these systems have improved transfection properties when compared to non-complexed components. The coating of dendrimers with lipids significantly reduced their hemotoxicity and cytotoxicity. The nanometric size, low polydispersity index and reduced positive zeta potential of such complexes made them attractive for future application in drug delivery. The formulations prepared by the hydrophobic locking protocol were not effective and will not be considered furthermore as prospective drug delivery systems. In contrast, the formulations formed by the hydrophilic loading method have shown promising results where the cytotoxicity of LLD systems with doxorubicin was more effective against cancer than normal cells.
Collapse
Affiliation(s)
- Sylwia Michlewska
- Laboratory of Microscopic Imaging & Specialized Biological Techniques. Faculty of Biology & Environmental Protection. University of Lodz, Banacha 12/16, Lodz 90-237, Poland.
| | - Zuzana Garaiova
- Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Comenius University, 842 48 Bratislava, Slovakia
| | - Veronika Šubjakova
- Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Comenius University, 842 48 Bratislava, Slovakia
| | - Marcin Hołota
- Department of General Biophysics. Faculty of Biology & Environmental Protection. University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | - Małgorzata Kubczak
- Department of General Biophysics. Faculty of Biology & Environmental Protection. University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | - Marika Grodzicka
- Department of General Biophysics. Faculty of Biology & Environmental Protection. University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | - Elżbieta Okła
- Department of General Biophysics. Faculty of Biology & Environmental Protection. University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | - Nikolaos Naziris
- Department of General Biophysics. Faculty of Biology & Environmental Protection. University of Lodz, Pomorska 141/143, Lodz 90-236, Poland; Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, Athens 15771, Greece
| | - Łucja Balcerzak
- Laboratory of Microscopic Imaging & Specialized Biological Techniques. Faculty of Biology & Environmental Protection. University of Lodz, Banacha 12/16, Lodz 90-237, Poland
| | - Paula Ortega
- Networking Research Center on Bioengineering. Biomaterials &Nanomedicine (CIBER-BBN), Monforte de Lemos 3-5, Pabell on 11, Planta 028029, Madrid, Spain; Universidad de Alcalá. Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), Spain and Instituto Ramon y Cajal de Investigacion Sanitaria, IRYCIS, Colmenar Viejo Road, Km 9, 100, 28034 Madrid, Spain
| | - Francisco Javier de la Mata
- Networking Research Center on Bioengineering. Biomaterials &Nanomedicine (CIBER-BBN), Monforte de Lemos 3-5, Pabell on 11, Planta 028029, Madrid, Spain; Universidad de Alcalá. Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), Spain and Instituto Ramon y Cajal de Investigacion Sanitaria, IRYCIS, Colmenar Viejo Road, Km 9, 100, 28034 Madrid, Spain
| | - Tibor Hianik
- Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Comenius University, 842 48 Bratislava, Slovakia
| | - Iveta Waczulikova
- Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Comenius University, 842 48 Bratislava, Slovakia
| | - Maria Bryszewska
- Department of General Biophysics. Faculty of Biology & Environmental Protection. University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | - Maksim Ionov
- Department of General Biophysics. Faculty of Biology & Environmental Protection. University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| |
Collapse
|
23
|
Sheikhi-Mohammareh S, Oroojalian F, Beyzaei H, Moghaddam-Manesh M, Salimi A, Azizollahi F, Shiri A. Domino protocol for the synthesis of diversely functionalized derivatives of a novel fused pentacyclic antioxidant/anticancer fluorescent scaffold: Pyrazolo[5'',1'':2',3']pyrimido[4',5':5,6][1,4]thiazino[2,3-b]quinoxaline. Talanta 2023; 262:124723. [PMID: 37245433 DOI: 10.1016/j.talanta.2023.124723] [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: 02/09/2023] [Revised: 05/04/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Rising to the challenge of formidable multi-step reaction needed for the synthesis of polycyclic compounds, an efficient one-pot two-step procedure for the synthesis of densely functionalized novel pyrazolo[5″,1'':2',3']pyrimido[4',5':5,6] [1,4]thiazino[2,3-b]quinoxalines from synthetically accessible starting materials 6-bromo-7-chloro-3-cyano-2-(ethylthio)-5-methylpyrazolo[1,5-a]pyrimidine, 3-aminoquinoxaline-2-thiol and some readily accessible alkyl halides was established. The domino reaction pathway involves cyclocondensation/N-alkylation sequence in K2CO3/N,N-dimethyl formamide under heating condition. DPPH free radical scavenging activity of all synthesized pyrazolo[5″,1'':2',3']pyrimido[4',5':5,6][1,4]thiazino[2,3-b]quinoxalines was evaluated to determine their antioxidant potentials. IC50 values were recorded in the range of 29-71 μM. N-benzyl substituted derivative represented the most effective antioxidant activity as well as antiproliferative activity against MCF-7 cells. Moreover, fluorescence in solution for these compounds exhibited strong red emission in the visible region (λflu. = 536-558 nm) with good to excellent quantum yields (61-95%). Due to their interesting fluorescence properties, these novel pentacyclic fluorophores can be used as fluorescent markers and probes for studies in biochemistry and pharmacology.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hamid Beyzaei
- Department of Chemistry, Faculty of Science, University of Zabol, Zabol, Iran
| | | | - Alireza Salimi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fatemeh Azizollahi
- Department of Advanced Sciences and Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Ali Shiri
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
24
|
Lobo CL, Shetty A, M M, Dubey A, El-Zahaby SA. Non-systemic Approaches for Ductal Carcinoma In Situ: Exploring the Potential of Ultra-flexible Combisomes as a Novel Drug Delivery Strategy-a Review. AAPS PharmSciTech 2023; 24:119. [PMID: 37173545 DOI: 10.1208/s12249-023-02574-z] [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: 02/16/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Ductal carcinoma in situ (DCIS) is currently treated through breast-conserving surgery (lumpectomy), radiation therapy, breast-removing surgery (mastectomy), and hormone therapy to prevent further progression into invasive breast cancer and recurrence. Discrepancies concerning the prognosis of DCIS have sparked controversy about adequate treatment. Considering the severe medical and psychological consequences of mastectomy, developing a treatment approach that arrests the progression of DCIS to the invasive stage without affecting the non-cancerous cells is of utmost importance. In the current review, the problems associated with the diagnosis and management of DCIS have been thoroughly discussed. A summary of the route of administration and drug delivery systems to manage DCIS was also provoked. Innovative ultra-flexible combisomes were also proposed for the effective management of DCIS. Prevention is essential in managing the risk of DCIS and reducing the risk of progression to invasive breast cancer. While prevention is vital, it is not always possible to prevent DCIS, and in some cases, treatment may be necessary. Hence, this review recommends that ultra-flexible combisomes administered as a topical gel provide a non-systemic approach for managing DCIS and thus significantly minimize the side effects and costs associated with existing therapies.
Collapse
Affiliation(s)
- Cynthia Lizzie Lobo
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India
| | - Amitha Shetty
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India
| | - Manohar M
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India.
| | - Sally A El-Zahaby
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, King Salman International University, South Sinai, Egypt
| |
Collapse
|
25
|
Jakobušić Brala C, Karković Marković A, Kugić A, Torić J, Barbarić M. Combination Chemotherapy with Selected Polyphenols in Preclinical and Clinical Studies-An Update Overview. Molecules 2023; 28:molecules28093746. [PMID: 37175156 PMCID: PMC10180288 DOI: 10.3390/molecules28093746] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
This review article describes studies published over the past five years on the combination of polyphenols, which are the most studied in the field of anticancer effects (curcumin, quercetin, resveratrol, epigallocatechin gallate, and apigenin) and chemotherapeutics such as cisplatin, 5-fluorouracil, oxaliplatin, paclitaxel, etc. According to WHO data, research has been limited to five cancers with the highest morbidity rate (lung, colorectal, liver, gastric, and breast cancer). A systematic review of articles published in the past five years (from January 2018 to January 2023) was carried out with the help of all Web of Science databases and the available base of clinical studies. Based on the preclinical studies presented in this review, polyphenols can enhance drug efficacy and reduce chemoresistance through different molecular mechanisms. Considering the large number of studies, curcumin could be a molecule in future chemotherapy cocktails. One of the main problems in clinical research is related to the limited bioavailability of most polyphenols. The design of a new co-delivery system for drugs and polyphenols is essential for future clinical research. Some polyphenols work in synergy with chemotherapeutic drugs, but some polyphenols can act antagonistically, so caution is always required.
Collapse
Affiliation(s)
- Cvijeta Jakobušić Brala
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Ana Karković Marković
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Azra Kugić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Jelena Torić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Monika Barbarić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| |
Collapse
|
26
|
Karim S, Madani B, Burzangi AS, Alsieni M, Bazuhair MA, Jamal M, Daghistani H, Barasheed MO, Alkreathy H, Khan MA, Khan LM. Urolithin A's Antioxidative, Anti-Inflammatory, and Antiapoptotic Activities Mitigate Doxorubicin-Induced Liver Injury in Wistar Rats. Biomedicines 2023; 11:biomedicines11041125. [PMID: 37189743 DOI: 10.3390/biomedicines11041125] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 03/29/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Human colon microbiota produce a metabolite called urolithin A (URO A) from ellagic acid and linked compounds, and this metabolite has been demonstrated to have antioxidant, anti-inflammatory, and antiapoptotic activities. The current work examines the various mechanisms through which URO A protects against doxorubicin (DOX)-induced liver injury in Wistar rats. In this experiment, Wistar rats were administered DOX intraperitoneally (20 mg kg-1) on day 7 while given URO A intraperitoneally (2.5 or 5 mg kg-1 d-1) for 14 days. The serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and gamma glutamyl transferase (GGT) were measured. Hematoxylin and eosin (HE) staining was used to evaluate histopathological characteristics, and then antioxidant and anti-inflammatory properties were evaluated in tissue and serum, respectively. We also looked at how active caspase 3 and cytochrome c oxidase were in the liver. The findings demonstrated that supplementary URO A therapy clearly mitigated DOX-induced liver damage. The antioxidant enzymes SOD and CAT were elevated in the liver, and the levels of inflammatory cytokines, such as TNF-α, NF-kB, and IL-6, in the tissue were significantly attenuated, all of which complemented the beneficial effects of URO A in DOX-induced liver injury. In addition, URO A was able to alter the expression of caspase 3 and cytochrome c oxidase in the livers of rats that were subjected to DOX stress. These results showed that URO A reduced DOX-induced liver injury by reducing oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Shahid Karim
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Batoul Madani
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdulhadi S Burzangi
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammed Alsieni
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammed A Bazuhair
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Maha Jamal
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hussam Daghistani
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Regenerative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammed O Barasheed
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Huda Alkreathy
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Lateef M Khan
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
27
|
Homayoonfal M, Gilasi H, Asemi Z, Mahabady MK, Asemi R, Yousefi B. Quercetin modulates signal transductions and targets non-coding RNAs against cancer development. Cell Signal 2023; 107:110667. [PMID: 37023996 DOI: 10.1016/j.cellsig.2023.110667] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/22/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
In recent decades, various investigations have indicated that natural compounds have great potential in the prevention and treatment of different chronic disorders including different types of cancer. As a bioactive flavonoid, Quercetin (Qu) is a dietary ingredient enjoying high pharmacological values and health-promoting effects due to its antioxidant and anti-inflammatory characterization. Conclusive in vitro and in vivo evidence has revealed that Qu has great potential in cancer prevention and development. Qu exerts its anticancer influences by altering various cellular processes such as apoptosis, autophagy, angiogenesis, metastasis, cell cycle, and proliferation. In this way, Qu by targeting numerous signaling pathways as well as non-coding RNAs regulates several cellular mechanisms to suppress cancer occurrence and promotion. This review aimed to summarize the impact of Qu on the molecular pathways and non-coding RNAs in modulating various cancer-associated cellular mechanisms.
Collapse
Affiliation(s)
- Mina Homayoonfal
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamidreza Gilasi
- Department of Biostatistics and Epidemiology, Kashan University of Medical Sciences, Kashan, Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Reza Asemi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Islam R, Yan MP, Yen KP, Rasol NE, Meng CK, Wai LK. Synthesis and biological evaluation of chromone derivatives against triple-negative breast cancer cells. Med Chem Res 2023. [DOI: 10.1007/s00044-023-03048-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
|
29
|
Abstract
Flavonoids are polyphenolic phytochemicals, which occur naturally in plants and possess both anti-oxidant and pro-oxidant properties. Flavonoids are gaining increasing popularity in the pharmaceutical industry as healthy and cost-effective compounds. Flavonoids show beneficial pharmacological activities in the treatment and prevention of various types of diseases. They are natural and less toxic agents for cancer chemotherapy and radiotherapy via regulation of multiple cell signaling pathways and pro-oxidant effects. In this review, we have summarized the mechanisms of action of selected flavonoids, and their pharmacological implications and potential therapeutic applications in cancer therapy.
Collapse
Affiliation(s)
- Prabha Tiwari
- Riken Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Kaushala Prasad Mishra
- Ex Bhabha Atomic Research Center, Foundation for Education and Research, Mumbai, Maharashtra, India
| |
Collapse
|
30
|
Metabolomics Analysis Reveals Novel Targets of Chemosensitizing Polyphenols and Omega-3 Polyunsaturated Fatty Acids in Triple Negative Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24054406. [PMID: 36901842 PMCID: PMC10002396 DOI: 10.3390/ijms24054406] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer with typically poorer outcomes due to its aggressive clinical behavior and lack of targeted treatment options. Currently, treatment is limited to the administration of high-dose chemotherapeutics, which results in significant toxicities and drug resistance. As such, there is a need to de-escalate chemotherapeutic doses in TNBC while also retaining/improving treatment efficacy. Dietary polyphenols and omega-3 polyunsaturated fatty acids (PUFAs) have been demonstrated to have unique properties in experimental models of TNBC, improving the efficacy of doxorubicin and reversing multi-drug resistance. However, the pleiotropic nature of these compounds has caused their mechanisms to remain elusive, preventing the development of more potent mimetics to take advantage of their properties. Using untargeted metabolomics, we identify a diverse set of metabolites/metabolic pathways that are targeted by these compounds following treatment in MDA-MB-231 cells. Furthermore, we demonstrate that these chemosensitizers do not all target the same metabolic processes, but rather organize into distinct clusters based on similarities among metabolic targets. Common themes in metabolic targets included amino acid metabolism (particularly one-carbon and glutamine metabolism) and alterations in fatty acid oxidation. Moreover, doxorubicin treatment alone generally targeted different metabolites/pathways than chemosensitizers. This information provides novel insights into chemosensitization mechanisms in TNBC.
Collapse
|
31
|
Costa AR, Duarte AC, Costa-Brito AR, Gonçalves I, Santos CRA. Bitter taste signaling in cancer. Life Sci 2023; 315:121363. [PMID: 36610638 DOI: 10.1016/j.lfs.2022.121363] [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: 11/08/2022] [Revised: 12/21/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023]
Abstract
Pharmacoresistance of cancer cells to many drugs used in chemotherapy remains a major challenge for the treatment of cancer. Multidrug resistance transporters, especially ATP-binding cassette (ABC) transporters, are a major cause of cancer drug resistance since they translocate a broad range of drug compounds across the cell membrane, extruding them out of the cells. The regulation of ABC transporters by bitter taste receptors (TAS2Rs), which might be activated by specific bitter tasting compounds, was described in several types of cells/organs, becoming a potential target for cancer therapy. TAS2Rs expression has been reported in many organs and several types of cancer, like breast, ovarian, prostate, and colorectal cancers, where their activation was shown to be involved in various biological actions (cell survival, apoptosis, molecular transport, among others). Moreover, many TAS2Rs' ligands, such as flavonoids and alkaloids, with well-recognized beneficial properties, including several anticancer effects, have been reported as potential adjuvants in cancer therapies. In this review, we discuss the potential therapeutic role of TAS2Rs and bitter tasting compounds in different types of cancer as a possible way to circumvent chemoresistance.
Collapse
Affiliation(s)
- Ana R Costa
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - Ana C Duarte
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal; CPIRN-IPG - Centro de Potencial e Inovação de Recursos Naturais, Instituto Politécnico da Guarda, Guarda, Portugal
| | - Ana R Costa-Brito
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal; Research Unit for Inland Development (UDI), Polytechnic of Guarda, Guarda, Portugal
| | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - Cecília R A Santos
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal.
| |
Collapse
|
32
|
Espinoza MJC, Lin KS, Weng MT, Kunene SC, Lin YS, Lin YT. Synthesis and characterization of silica nanoparticles from rice ashes coated with chitosan/cancer cell membrane for hepatocellular cancer treatment. Int J Biol Macromol 2023; 228:487-497. [PMID: 36581030 DOI: 10.1016/j.ijbiomac.2022.12.235] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022]
Abstract
Dual pH-sensitive smart nanocarriers based on silica nanoparticles (SNPs) extracted from rice husk ashes (RHAs) to effectively inhibit liver cancer cell proliferation were investigated. The SNPs were coated with chitosan (CH) and loaded with doxorubicin (DOX), then functionalized with cell membrane (CM) for homologous targeting ability. The FTIR spectra showed an absorption wave number at 1083 cm-1 which confirmed the existence of the SiOSi group, ratifying that the nanocarriers belong to silica species. The Korsmeyer-Peppas kinetic model reported R2 values of 0.996 and 0.931 for pH = 5.4 and pH = 7.4, respectively, demonstrating pH-responsive behavior of the nanocarriers. The cytotoxicity test confirmed that the HepG2 cell line treated with different SNP-CH-CM concentrations had no detectable significant cell toxicity, however, SNP-CH-DOX-CM induced greater cell death. In vivo tests revealed that SNP-CH-DOX-CM suppressed liver cancer growth in nude mice, demonstrating high pharmaceutical capability. Histological examination of vital organs showed that the targeted drug delivery system (DDS) had minor in vivo toxicity. In the light of its high treatment efficacy and minimal side effects, the investigated DDS is promising for the therapy of liver cancer.
Collapse
Affiliation(s)
- Maria Janina Carrera Espinoza
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li District, Taoyuan City 32003, Taiwan
| | - Kuen-Song Lin
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li District, Taoyuan City 32003, Taiwan.
| | - Meng-Tzu Weng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100233, Taiwan; Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302, Taiwan.
| | - Sikhumbuzo Charles Kunene
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li District, Taoyuan City 32003, Taiwan
| | - You-Sheng Lin
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li District, Taoyuan City 32003, Taiwan
| | - Yi-Ting Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100233, Taiwan
| |
Collapse
|
33
|
Kundrapu DB, Malla RR. Advances in Quercetin for Drug-Resistant Cancer Therapy: Mechanisms, Applications, and Delivery Systems. Crit Rev Oncog 2023; 28:15-26. [PMID: 38050978 DOI: 10.1615/critrevoncog.2023049513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Quercetin (QUE), a natural flavone abundantly discovered in fruits, has gained attention for its potential health benefits due to its unique structure. In addition, epidemiological and clinical studies have shown promising antioxidant activity of QUE aiming to treat various diseases, including cancer. This article's purpose is to provide an overview of recent advances in the use of QUE for drug-resistant cancer therapies, focusing on its mechanisms, applications, and delivery systems. The review discusses the structure-function relationship of QUE and its role in mitigating various disorders. Furthermore, it highlights the impact of QUE on cancer and cancer stem cells, elucidating the signaling pathways at the cellular and molecular levels involved. Additionally, the review explores the mechanistic role of QUE in reversing drug resistance in different types of drug-resistant cancers. Moreover, it presents a comprehensive analysis of drug diverse delivery strategies employed for effective cancer treatment using QUE. Clinical studies investigating the safety and bioavailability of QUE are also discussed. Finally, the review concludes with future directions, emphasizing the use of cost-effective and efficient protein and peptide-based self-assembling hydrogels for targeted delivery of QUE.
Collapse
Affiliation(s)
- Durga Bhavani Kundrapu
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Rama Rao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, Gandhi Institute of Technology and Management (GITAM) (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India; Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| |
Collapse
|
34
|
Song F, Lu CL, Wang CG, Hu CW, Zhang Y, Wang TL, Han L, Chen Z. Uncovering the mechanism of Kang-ai injection for treating intrahepatic cholangiocarcinoma based on network pharmacology, molecular docking, and in vitro validation. Front Pharmacol 2023; 14:1129709. [PMID: 36937833 PMCID: PMC10017963 DOI: 10.3389/fphar.2023.1129709] [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: 12/22/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Objective: Kang-ai injection (KAI) has been a popular adjuvant treatment for solid tumors, but its anti-tumor mechanism in intrahepatic cholangiocarcinoma (ICC) remains poorly understood. This study applied a network pharmacology-based approach to unveil KAI's anti-tumor activity, key targets, and potential pharmacological mechanism in ICC by integrating molecular docking and in vitro validation. Methods: The KAI-compound-target-ICC network was constructed to depict the connections between active KAI compounds and ICC-related targets based on the available data sources. The crucial ingredients, potential targets, and signaling pathways were screened using GO, KEGG enrichment analysis, and the PPI network. Molecular docking was performed to visualize the interactions between hub targets and components. In vitro experiments were carried out to validate the findings. Results: Among the 87 active components of KAI and 80 KAI-ICC-related targets, bioinformatics analysis identified quercetin as a possible candidate. GO and KEGG enrichment analysis indicated that the PI3K-AKT signaling pathway might be essential in ICC pharmacotherapy. The PPI network and its sub-networks screened 10 core target genes, including AKT1 and IL1β. Molecular docking results showed stable binding between AKT1 and IL1β with KAI active ingredients. The in vitro experiments confirmed that KAI might suppress the proliferation of ICC cell lines by inhibiting the PI3K/AKT signaling pathway, consistent with the network pharmacology approach and molecular docking predictions. Conclusion: The study sheds light on KAI's biological activity, potential targets, and molecular mechanisms in treating ICC and provides a promising strategy for understanding the scientific basis and therapeutic mechanisms of herbal treatments for ICC. This research has important implications for developing new, targeted therapies for ICC and highlights the importance of network pharmacology-based approaches in investigating complex herbal formulations.
Collapse
Affiliation(s)
- Fei Song
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Chang-Liang Lu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Cheng-Gui Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Chen-Wei Hu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yu Zhang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Tian-Lun Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Lu Han
- Jiangsu Vocational College of Medicine, Yancheng, China
| | - Zhong Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- *Correspondence: Zhong Chen,
| |
Collapse
|
35
|
Xing H, Zhu X, Liao J, Kong Y, Lu Y, Zhao D, Li N, Chen X, Qin Z. Pharmacokinetic study of Strongylocentrotus nudus egg polysaccharide in rats and beagles using a 3H-labeling method. Front Pharmacol 2023; 14:1109084. [PMID: 36937847 PMCID: PMC10017969 DOI: 10.3389/fphar.2023.1109084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Strongylocentrotus nudus egg polysaccharide (SEP) extracted from sea urchins has potential anticancer activity. However, little is known about its pharmacokinetic properties. To investigate the pharmacokinetics of SEP, it was radiolabeled with tritium. Furthermore, a sensitive, selective, and rapid liquid scintillation counter (LSC) method for quantifying 3H-SEP in biological matrix was validated. The lower quantification limit of the method was 4 Bq. The relative standard deviations (RSDs) of the intra- and inter-day precision were <3.0% and <3.9%, respectively. 3H-SEP was successfully applied to investigate the pharmacokinetics of SEP after intravenous administration of 20, 40, and 80 mg/kg (40 μCi/kg) in rats and 5, 10, and 20 mg/kg (6 μCi/kg) in beagles. The AUC(0-t) of SEP at three different doses was 487.81 ± 39.99 mg/L*h, 1,003.10 ± 95.94 mg/L*h, and 2,188.84 ± 137.73 mg/L*h in rats and 144.12 ± 3.78 mg/L*h, 322.62 ± 28.03 mg/L*h, and 754.17 ± 37.79 mg/L*h in beagles. The terminal elimination half-life (t1/2) of SEP was longer in beagles (204.29 ± 139.34 h) than in rats (35.48 ± 6.04 h). The concentration of SEP in plasma declined rapidly in both rats and beagles. All the study results provide detailed pharmacokinetic profiles of SEP in two kinds of animals, which will be helpful for further development.
Collapse
Affiliation(s)
- Han Xing
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Application and Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Xiaojie Zhu
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Jianmin Liao
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Ying Kong
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Yayuan Lu
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Di Zhao
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Ning Li
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, Jiangsu Province, China
- *Correspondence: Zhiying Qin, ; Xijing Chen,
| | - Zhiying Qin
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Application and Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhiying Qin, ; Xijing Chen,
| |
Collapse
|
36
|
Cell Dome as an Evaluation Platform for Organized HepG2 Cells. Cells 2022; 12:cells12010069. [PMID: 36611862 PMCID: PMC9818560 DOI: 10.3390/cells12010069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/06/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Human-hepatoblastoma-derived cell line, HepG2, has been widely used in liver and liver cancer studies. HepG2 spheroids produced in a three-dimensional (3D) culture system provide a better biological model than cells cultured in a two-dimensional (2D) culture system. Since cells at the center of spheroids exhibit specific behaviors attributed to hypoxic conditions, a 3D cell culture system that allows the observation of such cells using conventional optical or fluorescence microscopes would be useful. In this study, HepG2 cells were cultured in "Cell Dome", a micro-dome in which cells are enclosed in a cavity consisting of a hemispherical hydrogel shell. HepG2 cells formed hemispherical cell aggregates which filled the cavity of Cell Domes on 18 days of culture and the cells could continue to be cultured for 29 days. The cells at the center of hemispherical cell aggregates were observed using a fluorescence microscope. The cells grew in Cell Domes for 18 days exhibited higher Pi-class Glutathione S-Transferase enzymatic activity, hypoxia inducible factor-1α gene expression, and higher tolerance to mitomycin C than those cultured in 2D on tissue culture dishes (* p < 0.05). These results indicate that the center of the glass adhesive surface of hemispherical cell aggregates which is expected to have the similar environment as the center of the spheroids can be directly observed through glass plates. In conclusion, Cell Dome would be useful as an evaluation platform for organized HepG2 cells.
Collapse
|
37
|
Chuang CH, Lin YC, Yang J, Chan ST, Yeh SL. Quercetin supplementation attenuates cisplatin induced myelosuppression in mice through regulation of hematopoietic growth factors and hematopoietic inhibitory factors. J Nutr Biochem 2022; 110:109149. [PMID: 36049669 DOI: 10.1016/j.jnutbio.2022.109149] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 04/27/2022] [Accepted: 08/09/2022] [Indexed: 01/13/2023]
Abstract
The present study investigated the effects of quercetin on cisplatin (CDDP)-induced common side effect, myelosuppression, and the possible mechanisms in Balb/c mice. The mice were randomly treated with CDDP alone or in combination with quercetin for 14 days. Quercetin was given by intraperitoneal injection (10 mg/kg, 3 times a week; IQ) or by a diet containing 0.1% or 1% quercetin (LQ and HQ, respectively). We found that quercetin supplementation especially HQ and IQ, significantly restored the decrease in number of bone marrow cells, total white blood cells, red blood cells and platelets, and the body weight in mice exposed to CDDP (P≤.05). Similar trends were observed in the number of neutrophils, lymphocytes and monocytes in the plasma. HQ and IQ also increased the levels of hematopoietic growth factors (HGFs), especially in granulocyte-macrophage-colony stimulating factor and IL-9 (P<.05), but decreased the levels of hematopoietic inhibitory factors (HIFs) and oxidative stress in the plasma and the bone marrow in CDDP-exposed mice. Furthermore, both quercetin and quercetin-3-O-glucuronide (Q3G) significantly increase cell viability and inhibited apoptosis at 48 or 72 h (P≤.05), accompanied by increasing HGF levels and decreasing HIF levels in the cultured medium in 32D cells exposed to CDDP. IL-9 siRNA transfection suppressed the effects of quercetin and Q3G on cell viability (P≤.05) in32D cells. In conclusion, our results indicate that quercetin attenuates CDDP-induced myelosuppression through the mechanisms associated with regulation of HGFs and HIFs.
Collapse
Affiliation(s)
- Cheng-Hung Chuang
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Taichung, Taiwan, Republic of China
| | - Yi-Chin Lin
- Department of Nutritional Science, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Jung Yang
- Department of Nutritional Science, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Shu-Ting Chan
- Department of Nutritional Science, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Shu-Lan Yeh
- Department of Nutritional Science, Chung Shan Medical University, Taichung, Taiwan, Republic of China; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China.
| |
Collapse
|
38
|
Morishita H, Perera LMB, Zhang X, Mizoi K, Ito MA, Yano K, Ogihara T. P-Glycoprotein-Mediated Pharmacokinetic Interactions Increase Pimozide hERG Channel Inhibition. J Pharm Sci 2022; 111:3411-3416. [PMID: 36181876 DOI: 10.1016/j.xphs.2022.09.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/23/2022] [Accepted: 09/23/2022] [Indexed: 01/05/2023]
Abstract
Pimozide, an antipsychotic drug, is a potent inhibitor of the hERG channel. A case of death due to cardiac arrest has been reported in a boy who received pimozide together with sertraline and aripiprazole. In this study, we focused on drug-drug interactions and investigated the relationships between transporter-mediated intracellular accumulation and the hERG inhibitory effect of pimozide. The accumulation of pimozide in cardiomyocyte-derived AC16 cells was significantly increased by sertraline and aripiprazole, which are thought to have a P-glycoprotein (P-gp) inhibitory effect, and under P-gp siRNA conditions. These results suggest P-gp inhibition increases pimozide accumulation in AC16 cells. We introduced the hERG plasmid into AC16 cells and investigated the concentration-dependent hERG inhibitory effect of pimozide from within AC16 cells. Addition of 10 nM or more pimozide significantly inhibited the hERG current with concentration dependence. These results indicate P-gp-mediated pharmacokinetic interaction increases pimozide accumulation in AC16 cells, and the subsequent elevated pimozide levels within the cells may result in an increased risk of hERG channel inhibition. Our present study calls attention to the risks associated with the combined use of cardiotoxic P-gp substrate(s) and P-gp inhibitory medicines.
Collapse
Affiliation(s)
- Hiroki Morishita
- Department of Pharmacy, Saiseikai Maebashi Hospital, 564-1 Kamishinden-machi, Maebashi-shi, Gunma 371-0821, Japan; Laboratory of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma 370-0033, Japan.
| | - Liyanage Manosika Buddhini Perera
- Laboratory of Biopharmaceutics, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma 370-0033, Japan
| | - Xieyi Zhang
- Laboratory of Biopharmaceutics, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma 370-0033, Japan
| | - Kenta Mizoi
- Laboratory of Biopharmaceutics, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma 370-0033, Japan
| | - Masa-Aki Ito
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma 370-0033, Japan
| | - Kentaro Yano
- Laboratory of Drug Metabolism and Pharmacokinetics, Yokohama University of Pharmacy, 601 Matano-cho, Totsuka-ku, Yokohama, Kanagawa 245-0066, Japan
| | - Takuo Ogihara
- Laboratory of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma 370-0033, Japan; Laboratory of Biopharmaceutics, Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki-shi, Gunma 370-0033, Japan
| |
Collapse
|
39
|
Wen L, Wang Y, Zhang J. Nano‐gold micelles loaded Dox and Elacridar for reversing drug resistance of breast cancer. IET Nanobiotechnol 2022; 17:49-60. [PMID: 36341719 PMCID: PMC10116014 DOI: 10.1049/nbt2.12102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/05/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
The aim of this study was to provide a new effective carrier for rescuing the sensitivity of drug-resistant in breast cancer cells. Nano-gold micelles loaded with Dox and Elacridar (FP-ssD@A-E) were chemically synthesised. With the increase in the amount of Dox and Elacridar, the encapsulation rate of FP-ssD@A-E gradually increased, and the drug loading rate gradually decreased. FP-ss@A-E had a sustained-release effect. Dox, Elacridar, FP-ss@AuNPs, and FP-ssD@A-E significantly improved cell apoptosis, in which, FP-ssD@A-E was the most significant. FP-ssD@A-E significantly decreased the cell viability and improved the Dox uptake. The levels of VEGFR-1, P-gp, IL-6, and i-NOS were significantly decreased after Dox, Dox + Elacridar, FP-ss@AuNPs, and FP-ssD@A-E treatment. It was worth noting that FP-ssD@A-E had the most significant effects. The prepared FP-ssD@A-E micelles, which were spherical in shape, uniform in particle size distribution, and had good drug loading performance and encapsulation efficiency.
Collapse
Affiliation(s)
- Liu‐Jing Wen
- Department of Pharmacy Tianjin Medical University Cancer Institute and Hospital National Clinical Research Center for Cancer Key Laboratory of Cancer Prevention and Therapy Tianjin's Clinical Research Center for Cancer Tianjin China
| | - Yue‐Sheng Wang
- Department of Dentistry Second Hospital Affiliated to Tianjin Medical University Tianjin China
| | - Jie Zhang
- Department of Pharmacy Tianjin Medical University Cancer Institute and Hospital National Clinical Research Center for Cancer Key Laboratory of Cancer Prevention and Therapy Tianjin's Clinical Research Center for Cancer Tianjin China
| |
Collapse
|
40
|
Telomerase inhibitor MST-312 and quercetin synergistically inhibit cancer cell proliferation by promoting DNA damage. Transl Oncol 2022; 27:101569. [PMID: 36274541 PMCID: PMC9596868 DOI: 10.1016/j.tranon.2022.101569] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 09/29/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Quercetin is a natural flavonoid with well-established anti-proliferative activities against a variety of cancers. Telomerase inhibitor MST-312 also exhibits anti-proliferative effect on various cancer cells independent of its effect on telomere shortening. However, due to their low absorption and toxicity at higher doses, their clinical development is limited. In the present study, we examine the synergistic potential of their combination in cancer cells, which may result in a decrease in the therapeutic dosage of these compounds. We report that MST-312 and quercetin exhibit strong synergism in ovarian cancer cells with combination index range from 0.2 to 0.7. Co-treatment with MST-312 and quercetin upregulates the DNA damage and augments apoptosis when compared to treatment with either compound alone or a vehicle. We also examined the effect of these compounds on the proliferation of normal ovarian surface epithelial cells (OSEs). MST-312 has a cytoprotective impact in OSEs at lower dosages, but is inhibitory at higher doses. Quercetin did not affect the OSEs proliferation at low concentrations while at higher concentrations it is inhibitory. Notably, combination of MST-312 and quercetin had no discernible impact on OSEs. These observations have significant implications for future efforts towards maximizing efficacy in cancer therapeutics as this co-treatment specifically affects cancer cells and reduces the effective dosage of both the compounds.
Collapse
|
41
|
The Utilization of Physiologically Active Molecular Components of Grape Seeds and Grape Marc. Int J Mol Sci 2022; 23:ijms231911165. [PMID: 36232467 PMCID: PMC9570270 DOI: 10.3390/ijms231911165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/01/2022] [Accepted: 09/16/2022] [Indexed: 11/18/2022] Open
Abstract
Nutritional interventions may highly contribute to the maintenance or restoration of human health. Grapes (Vitis vinifera) are one of the oldest known beneficial nutritional components of the human diet. Their high polyphenol content has been proven to enhance human health beyond doubt in statistics-based public health studies, especially in the prevention of cardiovascular disease and cancer. The current review concentrates on presenting and classifying polyphenol bioactive molecules (resveratrol, quercetin, catechin/epicatechin, etc.) available in high quantities in Vitis vinifera grapes or their byproducts. The molecular pathways and cellular signaling cascades involved in the effects of these polyphenol molecules are also presented in this review, which summarizes currently available in vitro and in vivo experimental literature data on their biological activities mostly in easily accessible tabular form. New molecules for different therapeutic purposes can also be synthesized based on existing polyphenol compound classes available in high quantities in grape, wine, and grape marc. Therefore an overview of these molecular structures is provided. Novel possibilities as dendrimer nanobioconjugates are reviewed, too. Currently available in vitro and in vivo experimental literature data on polyphenol biological activities are presented in easily accessible tabular form. The scope of the review details the antidiabetic, anticarcinogenic, antiviral, vasoprotective, and neuroprotective roles of grape-origin flavonoids. The novelty of the study lies in the description of the processing of agricultural by-products (grape seeds and skins) of industrial relevance, and the detailed description of the molecular mechanisms of action. In addition, the review of the clinical therapeutic applications of polyphenols is unique as no summary study has yet been done.
Collapse
|
42
|
The effect of ciprofloxacin on doxorubicin cytotoxic activity in the acquired resistance to doxorubicin in DU145 prostate carcinoma cells. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:194. [PMID: 36071289 DOI: 10.1007/s12032-022-01787-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/28/2022] [Indexed: 10/14/2022]
Abstract
The present study aimed to assess the influence of ciprofloxacin (CIP) against the doxorubicin (DOX)-resistant androgen-independent prostate cancer DU145 cells. The DOX-resistant DU145 (DU145/DOX20) cells were established by exposing DU145 cells to the increasing concentrations of DOX. The antiproliferative effect of CIP was examined through employing MTT, colony formation, and 3D culture assays. DU145/DOX20 cells exhibited a twofold higher IC50 value for DOX, an increased ABCB1 transporter activity, and some morphological changes accompanied by a decrease in spheroid size, adhesive and migration potential compared to DU145 cells. CIP (5 and 25 µg mL-1) resulted in a higher reduction in the viability of DU145/DOX20 cells than in DU145 cells. DU145/DOX20 cells were more resistant to CIP in 3D culture compared to the 2D one. No spheroid formation was observed for DU145/DOX20 cells treated with DOX and CIP combination. CIP and DOX, alone or in combination, significantly reduced the growth of DU145 spheroids. CIP in combination with 20 nM DOX prevented the colony formation of DU145 cells. The clonogenicity of DU145/DOX20 cells could not be estimated due to their low adhesive potential. CIP alone caused a significant reduction in the migration of DU145 cells and resulted in a more severe decrease in the wound closure ability of DOX-exposed ones. We identified that CIP enhanced DOX sensitivity in DU145 and DU145/DOX20 cells. This study suggested the co-delivery of low concentrations of CIP and DOX may be a promising strategy in treating the DOX-resistant and -sensitive hormone-refractory prostate cancer.
Collapse
|
43
|
Talib WH, Awajan D, Hamed RA, Azzam AO, Mahmod AI, AL-Yasari IH. Combination Anticancer Therapies Using Selected Phytochemicals. Molecules 2022; 27:5452. [PMID: 36080219 PMCID: PMC9458090 DOI: 10.3390/molecules27175452] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is still one of the most widespread diseases globally, it is considered a vital health challenge worldwide and one of the main barriers to long life expectancy. Due to the potential toxicity and lack of selectivity of conventional chemotherapeutic agents, discovering alternative treatments is a top priority. Plant-derived natural products have high potential in cancer treatment due to their multiple mechanisms of action, diversity in structure, availability in nature, and relatively low toxicity. In this review, the anticancer mechanisms of the most common phytochemicals were analyzed. Furthermore, a detailed discussion of the anticancer effect of combinations consisting of natural product or natural products with chemotherapeutic drugs was provided. This review should provide a strong platform for researchers and clinicians to improve basic and clinical research in the development of alternative anticancer medicines.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Dima Awajan
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Reem Ali Hamed
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Aya O. Azzam
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan
| | - Intisar Hadi AL-Yasari
- Department of Genetic Engineering, College of Biotechnology, Al-Qasim Green University, Babylon 964, Iraq
| |
Collapse
|
44
|
Morsi AA, Faruk EM, Medhat E, Taha NM, Ebrahim UFA. Modulatory effects of concomitant quercetin/sitagliptin administration on the ovarian histological and biochemical alterations provoked by doxorubicin in a streptozotocin-induced diabetic rat model. J Histotechnol 2022; 46:65-79. [DOI: 10.1080/01478885.2022.2105481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Ahmed A. Morsi
- Department of Histology and Cell Biology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Eman Mohamed Faruk
- Department Anatomy, College of Medicine, Umm Al-Qura University, Mecca, KSA
- Department of Histology and Cytology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Engy Medhat
- Department Medical Biochemistry and Molecular Biology, Faculty of Medicine Cairo University, Cairo, Egypt
| | - Neama M. Taha
- Physiology Department, Umm Al-Qura University, Mecca, KSA
| | | |
Collapse
|
45
|
Saleh DO, Mahmoud SS, Hassan A, Sanad EF. Doxorubicin-induced hepatic toxicity in rats: Mechanistic protective role of Omega-3 fatty acids through Nrf2/HO-1 activation and PI3K/Akt/GSK-3β axis modulation. Saudi J Biol Sci 2022; 29:103308. [PMID: 35677895 PMCID: PMC9167977 DOI: 10.1016/j.sjbs.2022.103308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/15/2022] [Accepted: 05/15/2022] [Indexed: 11/14/2022] Open
Abstract
Doxorubicin (DOX), a common antibiotic used to treat a variety of tumors, has several substantial adverse effects that limit its clinical use. As a result, finding effective protective agents to combat DOX-induced organ damage is a necessity. The current study was set to delineate the hepatoprotective role of omega‐3 fatty acids (ω-3FA) against DOX-mediated acute liver damage in rats and the underlined mechanism of GSK-3β inhibition. Five groups of rats were orally received either saline (groups 1 & 2) or ω-3FA (25, 50 and 100 mg/kg/day; groups 3, 4 & 5, respectively) for 28 consecutive days. Single DOX intraperitoneal injection (20 mg/kg) was used to induce hepatic toxicity in all groups except group 1 (negative control). Blood samples and liver tissues were collected 48-hr after injection. Our results revealed that pre-administration of ω-3FA (25, 50 and 100 mg/kg) to DOX-induced hepatic injured rats showed a significant reduction in serum hepatic injury biomarkers (ALT, AST, total and direct bilirubin) as well as hepatic contents of MDA, GSH, Nrf2 and HO-1. Additionally, hepatic PI3K, pAkt and GSK-3β have been restored significantly in a dose-dependent manner. Furthermore, all the hepatic histopathological features have been retained upon ω-3FA treatment together with the immunostaining intensity of tumor necrosis factor-α and caspase-3. These results suggest that ω-3FA have shown a marked activation of the Nrf2/HO-1 signaling pathway and modulation of the PI3K/pAkt/GSK-3β axis against DOX-induced hepatotoxicity.
Collapse
|
46
|
Michala AS, Pritsa A. Quercetin: A Molecule of Great Biochemical and Clinical Value and Its Beneficial Effect on Diabetes and Cancer. Diseases 2022; 10:37. [PMID: 35892731 PMCID: PMC9326669 DOI: 10.3390/diseases10030037] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/01/2023] Open
Abstract
Quercetin belongs to the broader category of polyphenols. It is found, in particular, among the flavonols, and along with kaempferol, myricetin and isorhamnetin, it is recognized as a foreign substance after ingestion in contrast to vitamins. Quercetin occurs mainly linked to sugars with the most common compounds being quercetin-3-O-glucoside or as an aglycone, especially in the plant population. The aim of this review is to present a recent bibliography on the mechanisms of quercetin absorption and metabolism, bioavailability, and antioxidant and the clinical effects in diabetes and cancer. The literature reports a positive effect of quercetin on oxidative stress, cancer, and the regulation of blood sugar levels. Moreover, research-administered drug dosages of up to 2000 mg per day showed mild to no symptoms of overdose. It should be noted that quercetin is no longer considered a carcinogenic substance. The daily intake of quercetin in the diet ranges 10 mg-500 mg, depending on the type of products consumed. This review highlights that quercetin is a valuable dietary antioxidant, although a specific daily recommended intake for this substance has not yet been determined and further studies are required to decide a beneficial concentration threshold.
Collapse
Affiliation(s)
| | - Agathi Pritsa
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University (IHU), P.O. 141 Sindos, 57400 Thessaloniki, Greece;
| |
Collapse
|
47
|
Targeting Breast Cancer-Derived Stem Cells by Dietary Phytochemicals: A Strategy for Cancer Prevention and Treatment. Cancers (Basel) 2022; 14:cancers14122864. [PMID: 35740529 PMCID: PMC9221436 DOI: 10.3390/cancers14122864] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is heterogeneous disease with variable prognosis and therapeutic response. Approximately, 70% of diagnosed breast cancer represents the luminal A subtype. This subpopulation has a fair prognosis with a lower rate of relapse than the other clinical subtypes. Acquisition of stemness in luminal A subtype modifies the phenotype plasticity to accomplish increased aggressiveness and therapeutic resistance. Therefore, targeting luminal A-derived breast cancer stem cells (BCSCs) could be a promising strategy for its prevention and treatment. Extensive studies reveal that dietary phytochemicals have the potential to target BCSCs by modulating the molecular and signal transduction pathways. Dietary phytochemicals alone or in combination with standard therapeutic modalities exert higher efficacy in targeting BCSCs through changes in stemness, self-renewal properties and hypoxia-related factors. These combinations offer achieving higher radio- and chemo- sensitization through alteration in the key signaling pathways such as AMPK, STAT3, NF-ĸB, Hedgehog, PI3K/Akt/mTOR, Notch, GSK3β, and Wnt related to cancer stemness and drug resistance. In this review, we highlight the concept of targeting luminal A-derived BCSCs with dietary phytochemicals by summarizing the pathways and underlying mechanism(s) involved during therapeutic resistance.
Collapse
|
48
|
LC-MS Profiled Chemical Constituents, Molecular Modeling, and In vitro Bioactivity Evaluations of Suaeda vermiculata Extracts as Anti-Hepatocellular Carcinoma Preparation: Assessment of the Constituents’ Role, and Receptor Docking Feasibility Based Activity Projections. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
49
|
Jehan S, Huang J, Farooq U, Basheer I, Zhou W. Combinatorial effect of thymoquinone with chemo agents for tumor therapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153936. [PMID: 35114449 DOI: 10.1016/j.phymed.2022.153936] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 12/04/2021] [Accepted: 01/12/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Most chemotherapeutics used in cancer therapies exhibit considerable side effects to the patients. Thus, developing new chemo agents to treat cancer patients with minimal toxic and side effects is urgently needed. Recently, the combination of different chemotherapeutics has become a promising strategy to treat malignancies. Thymoquinone (TQ) is a primary bioactive compound derived from the folk medicinal plant Nigella sativa, which has been found an antitumor, chemopreventive and chemopotentiating agent against human neoplastic diseases. PURPOSE We briefly summarize the current research of the biomolecular mechanisms of TQ and evaluate the existing literature on TQ adjuvant therapies against various cancers. METHOD The data in this review were gathered by several search engines including, Google Scholar, PubMed and ScienceDirect. We highlighted and classified the outcomes of both in vitro and in vivo experiments of TQ adjuvant therapies against human cancers and their chemopreventive activities on vital organs. RESULTS Several studies have shown that TQ synergistically potentiated the antitumor activity of numerous chemo agents against human neoplastic disease, including lung, breast, liver, colorectal, skin, prostate, stomach, bone and blood cancers. TQ also acted as a chemopreventive agent and reduced the toxicity of many chemo agents to vital organs, such as the heart, liver, kidneys and lungs. CONCLUSION In summary, we highly recommend an advanced evaluation of TQ adjuvant therapies at the level of preclinical and clinical trials, which could lead to a novel combinatorial therapy for cancer treatment with low or tolerable adverse effects on patients.
Collapse
Affiliation(s)
- Shah Jehan
- Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo Road 172, Changsha, Hunan 410013, China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Jiaxin Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo Road 172, Changsha, Hunan 410013, China
| | - Umar Farooq
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Irum Basheer
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo Road 172, Changsha, Hunan 410013, China.
| |
Collapse
|
50
|
Huang M, Zeng L, Zhu R, Chen G, Wu H, Fan B, Liu C, Guo B, Zhong H. Hyaluronic Acid Stabilized Doxorubicin Nano-Precipitations for Osteosarcoma Treatment. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Doxorubicin (Dox) is a wide-spectrum drug to treat different kinds of cancers. However, in clinical practice, Dox usually showed untargeted distributions to the other organs, which can cause serious side effects, such as cardiotoxity. Herein, the formulation of Dox into nanoparticles
is critical to enhance its distribution to tumors. Herein, we used polysaccharide, hyaluronic acid, to stabilize the Dox to form nano-precipitations (PD NPs) for the therapy of osteosarcoma. The PD NPs showed enhanced drug accumulation to tumor cells and realized better anticancer effects
than free drugs.
Collapse
Affiliation(s)
- Mouzhang Huang
- Department of Orthopedic Surgery, Ganzhou People’s Hospital (The Affitiated Ganzhou Hospital of Nanchang University), Ganzhou, Jiangxi 341000, China
| | - Limei Zeng
- Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Rongping Zhu
- Department of Orthopedic Surgery, Ganzhou People’s Hospital (The Affitiated Ganzhou Hospital of Nanchang University), Ganzhou, Jiangxi 341000, China
| | - Gongqun Chen
- Department of Orthopedic Surgery, Ganzhou People’s Hospital (The Affitiated Ganzhou Hospital of Nanchang University), Ganzhou, Jiangxi 341000, China
| | - Haijian Wu
- Department of Orthopedic Surgery, Ganzhou People’s Hospital (The Affitiated Ganzhou Hospital of Nanchang University), Ganzhou, Jiangxi 341000, China
| | - Bin Fan
- Department of Orthopedic Surgery, Ganzhou People’s Hospital (The Affitiated Ganzhou Hospital of Nanchang University), Ganzhou, Jiangxi 341000, China
| | - Changtie Liu
- Department of Orthopedic Surgery, Ganzhou People’s Hospital (The Affitiated Ganzhou Hospital of Nanchang University), Ganzhou, Jiangxi 341000, China
| | - Bowen Guo
- Department of Orthopedic Surgery, Ganzhou People’s Hospital (The Affitiated Ganzhou Hospital of Nanchang University), Ganzhou, Jiangxi 341000, China
| | - Hongfa Zhong
- Department of Orthopedic Surgery, Ganzhou People’s Hospital (The Affitiated Ganzhou Hospital of Nanchang University), Ganzhou, Jiangxi 341000, China
| |
Collapse
|