1
|
Mani R, Babu SV, Murugesan N, Duraisamy R, Thayumanavan P. A Comparative Study of Quercetin/Rutin Loaded PEG Polymeric Nanoparticles: Controlled Drug Release and Its Biological Activity. J Biochem Mol Toxicol 2025; 39:e70269. [PMID: 40269608 DOI: 10.1002/jbt.70269] [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: 01/03/2025] [Revised: 03/17/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
Flavonoids are natural polyphenolic compounds that primarily possess antioxidant properties and play a significant role in opposing various diseases. Current chemotherapeutic approaches are largely ineffective, thus calling for the development of alternative strategies to combat this disease. In this regard, numerous studies have reported the anticancer effect of flavonoids in different types of cancer. To enhance its therapeutic value, polymeric nanoparticles (PEG NPs) represent an ideal delivery system. Further, surface modification of NPs with PEG holds tremendous potential for improving the bioavailability and circulation time of native drugs in the blood. The present study aimed to develop Quercetin/Rutin-loaded PEG polymeric NPs (Qu-PEG/Ru-PEG NPs) with enhanced encapsulation efficiency and sustained drug release. The synthesized Qu-PEG NPs & Ru-PEG NPs were characterized by UV-Vis Spectroscopy, FTIR spectrum, NMR, and XRD and SEM analysis. In-vitro drug release study exhibited a cumulative release of Quercetin & rutin for 24 h at pH 7.4. Further, the polymeric nano-formulations of Quercetin & Rutin showed enhanced antioxidant activity, leading to defense against oxidative stress. In-vitro cellular studies demonstrated that Qu-PEG NPs and Ru-PEG NPs significantly inhibit KB cell proliferation compared to free drugs alone. The current study also showed that Qu-PEG NPs & Ru-PEG NPs enhance intracellular ROS generation compared to the drug alone. Hence, our research findings revealed that successful encapsulation of Quercetin & Rutin in PEG NPs targets the tumor microenvironment and enhances the efficacy of drugs. Based on these preliminary results, flavonoid-loaded polymeric-based NPs might be potential therapeutic molecules against cancer in the future.
Collapse
Affiliation(s)
- Renuka Mani
- Department of Biochemistry, School of Bioscience, Periyar University, Salem, Tamil Nadu, India
| | - Swethaa Viswaresh Babu
- Department of Biochemistry, School of Bioscience, Periyar University, Salem, Tamil Nadu, India
| | - Nishanth Murugesan
- Department of Biochemistry, School of Bioscience, Periyar University, Salem, Tamil Nadu, India
| | - Ramachandhiran Duraisamy
- Department of Biotechnology and Biochemistry, Faculty of Science, Annamalai University, Chidambaram, Annamalai nagar, Tamilnadu, India
| | - Palvannan Thayumanavan
- Department of Biochemistry, School of Bioscience, Periyar University, Salem, Tamil Nadu, India
| |
Collapse
|
2
|
Reyes-Guzmán VL, Villarreal-Gómez LJ, Vázquez-Mora R, Méndez-Ramírez YI, Paz-González JA, Zizumbo-López A, Borbón H, Lizarraga-Medina EG, Cornejo-Bravo JM, Pérez-González GL, Ontiveros-Zepeda AS, Pérez-Sánchez A, Chavira-Martínez E, Huirache-Acuña R, Estévez-Martínez Y. Integrating an antimicrobial nanocomposite to bioactive electrospun fibers for improved wound dressing materials. Sci Rep 2024; 14:25118. [PMID: 39443526 PMCID: PMC11499993 DOI: 10.1038/s41598-024-75814-2] [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: 05/02/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
This study investigates the fabrication and characterization of electrospun poly (ε-caprolactone)/poly (vinyl pyrrolidone) (PCL/PVP) fibers integrated with a nanocomposite of chitosan, silver nanocrystals, and graphene oxide (ChAgG), aimed at developing advanced wound dressing materials. The ChAgG nanocomposite, recognized for its antimicrobial and biocompatible properties, was incorporated into PCL/PVP fibers through electrospinning techniques. We assessed the resultant fibers' morphological, physicochemical, and mechanical properties, which exhibited significant enhancements in mechanical strength and demonstrated effective antimicrobial activity against common bacterial pathogens. The findings suggest that the PCL/PVP-ChAgG fibers maintain biocompatibility and facilitate controlled therapeutic delivery, positioning them as a promising solution for managing chronic and burn-related wounds. This study underscores the potential of these advanced materials to improve healing outcomes cost-effectively, particularly in settings plagued by high incidences of burn injuries. Further clinical investigations are recommended to explore these innovative fibers' full potential and real-world applicability.
Collapse
Affiliation(s)
- Victoria Leonor Reyes-Guzmán
- Facultad de Ciencias de la Ingeniería y Tecnología, Universidad Autónoma de Baja California, Blvd. Universitario, #1000. Unidad Valle de las Palmas. Tijuana, Baja, Tijuana, CP. 21500, Baja California, México
| | - Luis Jesús Villarreal-Gómez
- Facultad de Ciencias de la Ingeniería y Tecnología, Universidad Autónoma de Baja California, Blvd. Universitario, #1000. Unidad Valle de las Palmas. Tijuana, Baja, Tijuana, CP. 21500, Baja California, México.
- Facultad de Ciencias Química e Ingeniería, Universidad Autónoma de Baja California, Universidad #14418, UABC, Parque Internacional Industrial Tijuana, Tijuana, 22424, Baja California, México.
| | - Rubi Vázquez-Mora
- Tecnológico Nacional de México, Unidad Tecnológica Acatlán, Campús Acatlán de Osorio, Carretera Acatlán - San Juan Ixcaquistla kilómetro 5.5, Del Maestro, Acatlán, 74949, Puebla, México
| | - Yesica Itzel Méndez-Ramírez
- Tecnológico Nacional de México, Unidad Tecnológica Acatlán, Campús Acatlán de Osorio, Carretera Acatlán - San Juan Ixcaquistla kilómetro 5.5, Del Maestro, Acatlán, 74949, Puebla, México
| | - Juan Antonio Paz-González
- Facultad de Ciencias de la Ingeniería y Tecnología, Universidad Autónoma de Baja California, Blvd. Universitario, #1000. Unidad Valle de las Palmas. Tijuana, Baja, Tijuana, CP. 21500, Baja California, México
| | - Arturo Zizumbo-López
- Tecnológico Nacional de México, Campus Tijuana, Blvd. Alberto Limón Padilla y Av. ITR Tijuana S/N, Colonia Mesa de Otay, Tijuana, C.P. 22500, Baja California, México
| | - Hugo Borbón
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Carr. Tijuana-Ensenada km107, C.I.C.E.S.E, Ensenada, 22860, Baja California, México
| | - Eder Germán Lizarraga-Medina
- Facultad de Ciencias de la Ingeniería y Tecnología, Universidad Autónoma de Baja California, Blvd. Universitario, #1000. Unidad Valle de las Palmas. Tijuana, Baja, Tijuana, CP. 21500, Baja California, México
| | - José Manuel Cornejo-Bravo
- Facultad de Ciencias Química e Ingeniería, Universidad Autónoma de Baja California, Universidad #14418, UABC, Parque Internacional Industrial Tijuana, Tijuana, 22424, Baja California, México
| | - Graciela Lizeth Pérez-González
- Facultad de Ciencias de la Ingeniería y Tecnología, Universidad Autónoma de Baja California, Blvd. Universitario, #1000. Unidad Valle de las Palmas. Tijuana, Baja, Tijuana, CP. 21500, Baja California, México
| | - Arturo Sinue Ontiveros-Zepeda
- Facultad de Ciencias de la Ingeniería, Administrativas y Sociales, Universidad Autónoma de Baja California, Blvrd Universidad 1, San Fernando, Tecate, 21460, Baja California, México
| | - Armando Pérez-Sánchez
- Facultad de Ciencias de la Ingeniería y Tecnología, Universidad Autónoma de Baja California, Blvd. Universitario, #1000. Unidad Valle de las Palmas. Tijuana, Baja, Tijuana, CP. 21500, Baja California, México
| | - Elizabeth Chavira-Martínez
- Instituto de Investigaciones en Materiales, Circuito Exterior S/N Circuito de la Investigación Científica, C.U, Ciudad de México, 04510, México.
| | - Rafael Huirache-Acuña
- Facultad de Ingeniería Química, Universidad Michoacana de San Nicolás de Hidalgo, Michoacán, 58060, Morelia, Mexico
| | - Yoxkin Estévez-Martínez
- Tecnológico Nacional de México, Unidad Tecnológica Acatlán, Campús Acatlán de Osorio, Carretera Acatlán - San Juan Ixcaquistla kilómetro 5.5, Del Maestro, Acatlán, 74949, Puebla, México.
| |
Collapse
|
3
|
Jenča A, Mills DK, Ghasemi H, Saberian E, Jenča A, Karimi Forood AM, Petrášová A, Jenčová J, Jabbari Velisdeh Z, Zare-Zardini H, Ebrahimifar M. Herbal Therapies for Cancer Treatment: A Review of Phytotherapeutic Efficacy. Biologics 2024; 18:229-255. [PMID: 39281032 PMCID: PMC11401522 DOI: 10.2147/btt.s484068] [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: 07/24/2024] [Accepted: 08/31/2024] [Indexed: 09/18/2024]
Abstract
Natural products have proven to be promising anti-cancer agents due to their diverse chemical structures and bioactivity. This review examines their central role in cancer treatment, focusing on their mechanisms of action and therapeutic benefits. Medicinal plants contain bioactive compounds, such as flavonoids, alkaloids, terpenoids and polyphenols, which exhibit various anticancer properties. These compounds induce apoptosis, inhibit cell proliferation and cell cycle progression, interfere with microtubule formation, act on topoisomerase targets, inhibit angiogenesis, modulate key signaling pathways, improve the tumor microenvironment, reverse drug resistance and activate immune cells. Herbal anti-cancer drugs offer therapeutic advantages, particularly selective toxicity against cancer cells, reducing the adverse side effects associated with conventional chemotherapy. Recent studies and clinical trials highlight the benefits of herbal medicines in alleviating side effects, improving tolerance to chemotherapy and the occurrence of synergistic effects with conventional treatments. For example, the herbal medicine SH003 was found to be safe and potentially effective in the treatment of solid cancers, while Fucoidan showed anti-inflammatory properties that are beneficial for patients with advanced cancer. The current research landscape on herbal anticancer agents is extensive. Numerous studies and clinical trials are investigating their efficacy, safety and mechanisms of action in various cancers such as lung, prostate, breast and hepatocellular carcinoma. Promising developments include the polypharmacological approach, combination therapies, immunomodulation and the improvement of quality of life. However, there are still challenges in the development and use of natural products as anti-cancer drugs, such as the need for further research into their mechanisms of action, possible drug interactions and optimal dosage. Standardizing herbal extracts, improving bioavailability and delivery, and overcoming regulatory and acceptance hurdles are critical issues that need to be addressed. Nonetheless, the promising anticancer effects and therapeutic benefits of natural products warrant further investigation and development. Multidisciplinary collaboration is essential to advance herbal cancer therapy and integrate these agents into mainstream cancer treatment.
Collapse
Affiliation(s)
- Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, UPJS LF, Kosice, Slovakia
| | - David K Mills
- Molecular Science and Nanotechnology, College of Engineering and Science, Louisiana Tech University, Ruston, LA, 71272, USA
| | - Hadis Ghasemi
- Department of Chemistry, College of Art and Science, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Elham Saberian
- Pavol Jozef Šafárik University, Klinika and Akadémia Košice Bacikova, Kosice, Slovakia
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, UPJS LF, Kosice, Slovakia
| | | | - Adriána Petrášová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, UPJS LF, Kosice, Slovakia
| | - Janka Jenčová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, UPJS LF, Kosice, Slovakia
| | - Zeinab Jabbari Velisdeh
- Molecular Science and Nanotechnology, College of Engineering and Science, Louisiana Tech University, Ruston, LA, 71272, USA
| | - Hadi Zare-Zardini
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | - Meysam Ebrahimifar
- Department of Toxicology, Faculty of Pharmacy, Islamic Azad University, Shahreza Branch, Shahreza
| |
Collapse
|
4
|
Li Z, Jiao Y, Wu Z, Liu H, Li Y, Cai Y, Wei W, Cao F. The role of quercetin in ameliorating bleomycin-induced pulmonary fibrosis: insights into autophagy and the SIRT1/AMPK signaling pathway. Mol Biol Rep 2024; 51:795. [PMID: 39001907 DOI: 10.1007/s11033-024-09752-7] [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: 04/30/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a disease of unknown etiology characterized by a constant incidence rate. Unfortunately, effective pharmacological treatments for this condition are lacking and the identification of novel therapeutic approaches and underlying pathological mechanisms are required. This study investigated the potential of quercetin in alleviating pulmonary fibrosis by promoting autophagy and activation of the SIRT1/AMPK pathway. METHODS Mouse models of IPF were divided into four treatment groups: control, bleomycin (BLM), quercetin (Q), and quercetin + EX-527 (Q + E) treatment. Pulmonary fibrosis was induced in the mouse models through intratracheal instillation of BLM. Various indexes were identified through histological staining, Western blotting analysis, enzyme-linked immunosorbent assay, immunohistochemistry, and transmission electron microscopy. RESULTS Quercetin treatment ameliorated the pathology of BLM-induced pulmonary fibrosis of mice by reducing α-smooth muscle actin (α-SMA), collagen I (Col I), and collagen III (Col III) levels, and also improved the level of E-cadherin in lung tissue. Furthermore, Quercetin significantly enhanced LC3II/LC3I levels, decreased P62 expression, and increased the number of autophagosomes in lung tissue. These effects were accompanied by the activation of the SIRT1/AMPK pathway. Treatment with EX-527, an inhibitor for SIRT1, reversed all effects induced by quercetin. CONCLUSION This study showed that quercetin could alleviate pulmonary fibrosis and improve epithelial-mesenchymal transition by acting on the SIRT1/AMPK signaling pathway, which may be achieved by regulating the level of autophagy.
Collapse
Affiliation(s)
- Zhipeng Li
- Graduate school, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Jiao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Zhisong Wu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Haoge Liu
- Zhejiang Provincial Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Chinese Medicine, Hangzhou, 310006, China
| | - Yang Li
- Graduate school, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yaodong Cai
- Graduate school, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wan Wei
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China.
| | - Fang Cao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China.
| |
Collapse
|
5
|
Chen F. Inhibiting Pink1/Parkin-mediated mitophagy enhances the anticancer effects of quercetin in hepatocellular carcinomaf. Biochem Biophys Res Commun 2024; 712-713:149899. [PMID: 38653003 DOI: 10.1016/j.bbrc.2024.149899] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024]
Abstract
Quercetin, a naturally occurring flavonoid, has been investigated for its potential anti-cancer effects in various types of cancer, including hepatocellular carcinoma (HCC). However, its suppressing effect on reactive oxygen species (ROS) production might limited its anti-cancer effects. In this study, we aimed to explore the interplay among quercetin, mitochondrial dynamics and mitophagy and whether mitophagy-inhibition synergistically enhances the anti-tumor effects of quercetin. Huh7 and Hep3B cells were utilized for in vitro and in vivo studies. Results showed that quercetin treatment significantly increased the expression of mitochondrial fusion genes (MFN1 and MFN2) and decreased the expression of fission genes (DRP1 and FIS1) in Huh7 and Hep3B cells, leading to a more fused and elongated mitochondrial network. Quercetin upregulated the expression of key mitophagy regulators, PINK1 and PARK2, and enhanced the colocalization of mitochondria with lysosomes, indicating increased mitophagy. Knockdown of PINK1, PARK2, or SIRT1 attenuated quercetin-induced mitophagy and reduction of intracellular ROS levels. Quercetin treatment upregulates SIRT1 expression, which subsequently enhances PINK1 and PARK2 expression in Huh7 and Hep3B cells. In vivo experiments using Hep3B xenograft models revealed that the combination of quercetin with the mitophagy inhibitor hydroxychloroquine or SIRT1 knockdown significantly enhanced the anticancer effects of quercetin, as evidenced by reduced tumor size and weight, increased necrosis and apoptosis, and decreased proliferation in tumor tissues. These findings suggest that quercetin-induced mitochondrial fusion and Pink1/Parkin-dependent mitophagy may negatively influence its anti-cancer effects in HCC. Targeting mitophagy may enhance the therapeutic potential of quercetin in HCC treatment.
Collapse
Affiliation(s)
- Fang Chen
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, 610072, China.
| |
Collapse
|
6
|
Fahmy HM, Shekewy S, Elhusseiny FA, Elmekawy A. Enhanced Biocompatibility by Evaluating the Cytotoxic and Genotoxic Effects of Magnetic Iron Oxide Nanoparticles and Chitosan on Hepatocellular Carcinoma Cells (HCC). Cell Biochem Biophys 2024; 82:1027-1042. [PMID: 38558242 PMCID: PMC11344728 DOI: 10.1007/s12013-024-01256-2] [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: 02/12/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024]
Abstract
Hepatocellular carcinoma (HCC), the fifth most prevalent cancer worldwide, is influenced by a myriad of clinic-pathological factors, including viral infections and genetic abnormalities. This study delineates the synthesis, characterization, and the biological efficacy of iron oxide nanoparticles (Fe3O4) and chitosan-coated iron oxide nanoparticles (Fe3O4-CS) against HCC. Analytical methods confirmed the successful synthesis of both nanoparticles, with Fe3O4-CS demonstrating a smaller, uniform spherical morphology and distinct surface and magnetic properties attributable to its chitosan coating. The prepared materials were analyzed using various techniques, and their potential cytotoxic effects on HepG2 cancer cells line for HCC were investigated. In biological evaluations against HepG2 cells, a notable distinction in cytotoxicity was observed. Fe3O4 showed modest anticancer activity with an IC50 of 383.71 ± 23.9 µg/mL, whereas Fe3O4 exhibited a significantly enhanced cytotoxic effect, with a much lower IC50 of 39.15 ± 39.2 µg/mL. The Comet assay further evidenced Fe3O4-CS potent DNA damaging effect, showcasing its superior ability to induce apoptosis through extensive DNA fragmentation. Biochemical analyses integrated into our results reveal that Fe3O4-CS not only induces significant DNA damage but also markedly alters oxidative stress markers. Compared to control and Fe3O4-treated cells, Fe3O4-CS exposure significantly elevated levels of oxidative stress markers: superoxide dismutase (SOD) increased to 192.07 U/ml, catalase (CAT) decreased to 0.03 U/L, glutathione peroxidase (GPx) rose dramatically to 18.76 U/gT, and malondialdehyde (MDA) levels heightened to 30.33 nmol/gT. These results underscore the potential of Fe3O4-CS nanoparticles not only in inducing significant DNA damage conducive to cancer cell apoptosis but also in altering enzymatic activities and oxidative stress markers, suggesting a dual mechanism of action that may underpin their therapeutic advantage in cancer treatment. Our findings advocate for the further exploration of Fe3O4-CS nanoparticles in the development of anticancer drugs, emphasizing their capability to trigger oxidative stress and enhance antioxidant defense mechanisms.
Collapse
Affiliation(s)
- Heba M Fahmy
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Samar Shekewy
- Physics Department, Faculty of Science, Tanta University, Tanta, Egypt
- Physics Department, Faculty of Science, Menofia University, Menofia, Egypt
| | | | - Ahmed Elmekawy
- Physics Department, Faculty of Science, Tanta University, Tanta, Egypt.
| |
Collapse
|
7
|
Wang Y, Li J, Xia L. Plant-derived natural products and combination therapy in liver cancer. Front Oncol 2023; 13:1116532. [PMID: 36865794 PMCID: PMC9971944 DOI: 10.3389/fonc.2023.1116532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Liver cancer is one of the malignant cancers globally and seriously endangers human health because of its high morbidity and mortality. Plant-derived natural products have been evaluated as potential anticancer drugs due to low side effects and high anti-tumor efficacy. However, plant-derived natural products also have defects of poor solubility and cumbersome extraction process. In recent years, a growing numbers of plant derived natural products have been used in combination therapy of liver cancer with conventional chemotherapeutic agents, which has improved clinical efficacy through multiple mechanisms, including inhibition of tumor growth, induction of apoptosis, suppression of angiogenesis, enhancement of immunity, reversal of multiple drug resistance and reduction of side effects. The therapeutic effects and mechanisms of plant-derived natural products and combination therapy on liver cancer are reviewed to provide references for developing anti-liver-cancer strategies with high efficacy and low side effects.
Collapse
Affiliation(s)
- Yuqin Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Jinyao Li
- *Correspondence: Jinyao Li, ; Lijie Xia,
| | - Lijie Xia
- *Correspondence: Jinyao Li, ; Lijie Xia,
| |
Collapse
|
8
|
Yüce H, Şahin Y, Türkmen NB, Özek DA, Ünüvar S, Çiftçi O. Apoptotic, Cytotoxic and Antimigratory Activities of Phenolic Compounds. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022060138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
9
|
González-García K, López-Martínez A, Velázquez-Enríquez JM, Zertuche-Martínez C, Carrasco-Torres G, Sánchez-Navarro LM, Villa-Treviño S, Baltiérrez-Hoyos R, Vásquez-Garzón VR. 3′5-Dimaleamylbenzoic Acid Attenuates Bleomycin-Induced Pulmonary Fibrosis in Mice. Int J Mol Sci 2022; 23:ijms23147943. [PMID: 35887292 PMCID: PMC9319702 DOI: 10.3390/ijms23147943] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 12/04/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease characterized by parenchymal scarring, leading progressively to alveolar architecture distortion, respiratory failure, and eventually death. Currently, there is no effective treatment for IPF. Previously, 3′5-dimaleamylbenzoic acid (3′5-DMBA), a maleimide, demonstrated pro-apoptotic, anti-inflammatory, and anti-cancer properties; however, its potential therapeutic effects on IPF have not been addressed. Bleomycin (BLM) 100 U/kg was administered to CD1 mice through an osmotic minipump. After fourteen days of BLM administration, 3′5-DMBA (6 mg/kg or 10 mg/kg) and its vehicle carboxymethylcellulose (CMC) were administered intragastrically every two days until day 26. On day 28, all mice were euthanized. The 3′5-DMBA effect was assessed by histological and immunohistochemical staining, as well as by RT-qPCR. The redox status on lung tissue was evaluated by determining the glutathione content and the GSH/GSSG ratio. 3′5-DMBA treatment re-established typical lung histological features and decreased the expression of BLM-induced fibrotic markers: collagen, α-SMA, and TGF-β1. Furthermore, 3′5-DMBA significantly reduced the expression of genes involved in fibrogenesis. In addition, it decreased reduced glutathione and increased oxidized glutathione content without promoting oxidative damage to lipids, as evidenced by the decrease in the lipid peroxidation marker 4-HNE. Therefore, 3′5-DMBA may be a promising candidate for IPF treatment.
Collapse
Affiliation(s)
- Karina González-García
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, Oaxaca C.P. 68020, Mexico; (K.G.-G.); (A.L.-M.); (J.M.V.-E.); (C.Z.-M.)
| | - Armando López-Martínez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, Oaxaca C.P. 68020, Mexico; (K.G.-G.); (A.L.-M.); (J.M.V.-E.); (C.Z.-M.)
| | - Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, Oaxaca C.P. 68020, Mexico; (K.G.-G.); (A.L.-M.); (J.M.V.-E.); (C.Z.-M.)
| | - Cecilia Zertuche-Martínez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, Oaxaca C.P. 68020, Mexico; (K.G.-G.); (A.L.-M.); (J.M.V.-E.); (C.Z.-M.)
| | - Gabriela Carrasco-Torres
- Departamento de Nanociencias y Nanotecnología, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN 2508, Ciudad de México C.P. 07360, Mexico;
| | - Luis Manuel Sánchez-Navarro
- Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, Oaxaca C.P. 68020, Mexico;
| | - Saúl Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico C.P. 07360, Mexico;
| | - Rafael Baltiérrez-Hoyos
- CONACYT-Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, Oaxaca C.P. 68020, Mexico
- Correspondence: (R.B.-H.); (V.R.V.-G.); Tel./Fax: +55-01-(951)-513-9784 (R.B.-H. & V.R.V.-G.)
| | - Verónica Rocío Vásquez-Garzón
- CONACYT-Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, Oaxaca C.P. 68020, Mexico
- Correspondence: (R.B.-H.); (V.R.V.-G.); Tel./Fax: +55-01-(951)-513-9784 (R.B.-H. & V.R.V.-G.)
| |
Collapse
|
10
|
Lee SH, Kim YJ, Kim YH, Kim HY, Bhang SH. Enhancing therapeutic efficacy of human adipose-derived stem cells by modulating photoreceptor expression for advanced wound healing. Stem Cell Res Ther 2022; 13:215. [PMID: 35619187 PMCID: PMC9137210 DOI: 10.1186/s13287-022-02892-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background Human adipose-derived stem cells (hADSCs) have been widely used for regenerative medicine because of their therapeutic efficacy and differentiation capacity. However, there are still limitations to use them intactly due to some difficulties such as poor cell engraftment and viability after cell transplantation. Therefore, techniques such as photobiomodulation (PBM) are required to overcome these limitations. This study probed improved preclinical efficacy of irradiated hADSCs and its underlying molecular mechanism.
Methods hADSCs were irradiated with green organic light-emitting diodes (OLEDs). Treated cells were analyzed for mechanism identification and tissue regeneration ability verification. Expression levels of genes and proteins associated with photoreceptor, cell proliferation, migration, adhesion, and wound healing were evaluated by performing multiple assays and immunostaining. Excision wound models were employed to test in vivo therapeutic effects. Results In vitro assessments showed that Opsin3 (OPN3) and OPN4 are both expressed in hADSCs. However, only OPN4 was stimulated by green OLED irradiation. Cell proliferation, migration, adhesion, and growth factor expression in treated hADSCs were enhanced compared to control group. Conditioned medium containing paracrine factors secreted from irradiated hADSCs increased proliferation of human dermal fibroblasts and normal human epidermal keratinocytes. Irradiated hADSCs exerted better wound healing efficacy in vivo than hADSCs without OLED irradiation. Conclusions Our study introduces an intracellular mechanism of PBM in hADSCs. Our results revealed that photoreceptor OPN4 known to activate Gq-protein and consequently lead to reactive oxygen species production responded to OLED irradiation with a wavelength peak of 532 nm. In conclusion, green OLED irradiation can promote wound healing capability of hADSCs, suggesting that green OLED has potential preclinical applications.
Collapse
Affiliation(s)
- Sang Ho Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yu-Jin Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Han Young Kim
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
11
|
Mirazimi SMA, Dashti F, Tobeiha M, Shahini A, Jafari R, Khoddami M, Sheida AH, EsnaAshari P, Aflatoonian AH, Elikaii F, Zakeri MS, Hamblin MR, Aghajani M, Bavarsadkarimi M, Mirzaei H. Application of Quercetin in the Treatment of Gastrointestinal Cancers. Front Pharmacol 2022; 13:860209. [PMID: 35462903 PMCID: PMC9019477 DOI: 10.3389/fphar.2022.860209] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/02/2022] [Indexed: 02/06/2023] Open
Abstract
Many cellular signaling pathways contribute to the regulation of cell proliferation, division, motility, and apoptosis. Deregulation of these pathways contributes to tumor cell initiation and tumor progression. Lately, significant attention has been focused on the use of natural products as a promising strategy in cancer treatment. Quercetin is a natural flavonol compound widely present in commonly consumed foods. Quercetin has shown significant inhibitory effects on tumor progression via various mechanisms of action. These include stimulating cell cycle arrest or/and apoptosis as well as its antioxidant properties. Herein, we summarize the therapeutic effects of quercetin in gastrointestinal cancers (pancreatic, gastric, colorectal, esophageal, hepatocellular, and oral).
Collapse
Affiliation(s)
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Tobeiha
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Shahini
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Raha Jafari
- Department of Medicine, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran
| | - Mehrad Khoddami
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Hossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Parastoo EsnaAshari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Hossein Aflatoonian
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fateme Elikaii
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Melika Sadat Zakeri
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Mohammad Aghajani
- Infectious Disease Research Center, School of Nursing and Midwifery, Kashan University of Medical Sciences, Kashan, Iran
| | - Minoodokht Bavarsadkarimi
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
12
|
Antiproliferative and cytotoxic activity of Geraniaceae plant extracts against five tumor cell lines. Future Sci OA 2022; 8:FSO775. [PMID: 35070357 PMCID: PMC8765112 DOI: 10.2144/fsoa-2021-0109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/18/2021] [Indexed: 11/23/2022] Open
Abstract
Aim: To determine the antiproliferative and cytotoxic activities of Geranium and Erodium species against human cancer and noncancer cell lines, respectively. Methods: Twenty-one species of Geranium and Erodium were extracted and screened against cancerous and noncancerous human cell lines. Results: In a dose-response manner, G. glaberrimum, G. asphodeloides, E. brandianum and E. leucanthum were able, with variable potency, to inhibit cellular proliferation. Except for E. brandianum, all extracts induced cellular autophagy in tumor cells with similar levels to that of rapamycin; but, only E. brandianum induced cellular apoptosis, likely through Bcl2 and BAX protein expressions. Discussion: This is the first study to report the potential antiproliferative effects of ethanol extracts of several Geraniaceae species.
Collapse
|
13
|
Mushtaq S, Shahzad K, Saeed T, Ul-Hamid A, Abbasi BH, Ahmad N, Khalid W, Atif M, Ali Z, Abbasi R. Biocompatibility and cytotoxicity in vitro of surface-functionalized drug-loaded spinel ferrite nanoparticles. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:1339-1364. [PMID: 34934608 PMCID: PMC8649206 DOI: 10.3762/bjnano.12.99] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/15/2021] [Indexed: 06/14/2023]
Abstract
In this study, poly(isobutylene-alt-maleic anhydride) (PMA)-coated spinel ferrite (MFe2O4, where M = Fe, Co, Ni, or Zn) nanoparticles (NPs) were developed as carriers of the anticancer drugs doxorubicin (DOX) and methotrexate (MTX). Physical characterizations confirmed the formation of pure cubic structures (14-22 nm) with magnetic properties. Drug-loaded NPs exhibited tumor specificity with significantly higher (p < 0.005) drug release in an acidic environment (pH 5.5). The nanoparticles were highly colloidal (zeta potential = -35 to -26 mV) in deionized water, phosphate buffer saline (PBS), and sodium borate buffer (SBB). They showed elevated and dose-dependent cytotoxicity in vitro compared to free drug controls. The IC50 values ranged from 0.81 to 3.97 μg/mL for HepG2 and HT144 cells, whereas IC50 values for normal lymphocytes were 10 to 35 times higher (18.35-43.04 µg/mL). Cobalt ferrite (CFO) and zinc ferrite (ZFO) NPs were highly genotoxic (p < 0.05) in cancer cell lines. The nanoparticles caused cytotoxicity via oxidative stress, causing DNA damage and activation of p53-mediated cell cycle arrest (significantly elevated expression, p < 0.005, majorly G1 and G2/M arrest) and apoptosis. Cytotoxicity testing in 3D spheroids showed significant (p < 0.05) reduction in spheroid diameter and up to 74 ± 8.9% of cell death after two weeks. In addition, they also inhibited multidrug resistance (MDR) pump activity in both cell lines suggesting effectivity in MDR cancers. Among the tested MFe2O4 NPs, CFO nanocarriers were the most favorable for targeted cancer therapy due to excellent magnetic, colloidal, cytotoxic, and biocompatible aspects. However, detailed mechanistic, in vivo cytotoxicity, and magnetic-field-assisted studies are required to fully exploit these nanocarriers in therapeutic applications.
Collapse
Affiliation(s)
- Sadaf Mushtaq
- Institute of Biomedical and Genetic Engineering, G-9/1, Islamabad, Pakistan
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Khuram Shahzad
- Department of Physics, Functional Materials Lab, Air University, Sector E-9, Islamabad, Pakistan
| | - Tariq Saeed
- Institute of Biomedical and Genetic Engineering, G-9/1, Islamabad, Pakistan
| | - Anwar Ul-Hamid
- Core Research Facilities, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
| | | | - Nafees Ahmad
- Institute of Biomedical and Genetic Engineering, G-9/1, Islamabad, Pakistan
| | - Waqas Khalid
- Department of Physics, Functional Materials Lab, Air University, Sector E-9, Islamabad, Pakistan
| | - Muhammad Atif
- Department of Physics, Functional Materials Lab, Air University, Sector E-9, Islamabad, Pakistan
| | - Zulqurnain Ali
- Department of Physics, Functional Materials Lab, Air University, Sector E-9, Islamabad, Pakistan
| | - Rashda Abbasi
- Institute of Biomedical and Genetic Engineering, G-9/1, Islamabad, Pakistan
| |
Collapse
|
14
|
Zou H, Ye H, Kamaraj R, Zhang T, Zhang J, Pavek P. A review on pharmacological activities and synergistic effect of quercetin with small molecule agents. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153736. [PMID: 34560520 DOI: 10.1016/j.phymed.2021.153736] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/25/2021] [Accepted: 09/05/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Quercetin is a natural flavonoid, which widely exists in nature, such as tea, coffee, apples, and onions. Numerous studies have showed that quercetin has multiple biological activities such as anti-oxidation, anti-inflammatory, and anti-aging. Hence, quercetin has a significant therapeutic effect on cancers, obesity, diabetes, and other diseases. In the past decades, a large number of studies have shown that quercetin combined with other agents can significantly improve the overall therapeutic effect, compared to single use. PURPOSE This work reviews the pharmacological activities of quercetin and its derivatives. In addition, this work also summarizes both in vivo and in vitro experimental evidence for the synergistic effect of quercetin against cancers and metabolic diseases. METHODS An extensive systematic search for pharmacological activities and synergistic effect of quercetin was performed considering all the relevant literatures published until August 2021 through the databases including NCBI PubMed, Scopus, Web of Science, and Google Scholar. The relevant literatures were extracted from the databases with following keyword combinations: "pharmacological activities" OR "biological activities" OR "synergistic effect" OR "combined" OR "combination" AND "quercetin" as well as free-text words. RESULTS Quercetin and its derivatives possess multiple pharmacological activities including anti-cancer, anti-oxidant, anti-inflammatory, anti-cardiovascular, anti-aging, and neuroprotective activities. In addition, the synergistic effect of quercetin with small molecule agents against cancers and metabolic diseases has also been confirmed. CONCLUSION Quercetin cooperates with agents to improve the therapeutic effect by regulating signal molecules and blocking cell cycle. Synergistic therapy can reduce the dose of agents and avoid the possible toxic and side effects in the treatment process. Although quercetin treatment has some potential side effects, it is safe under the expected use conditions. Hence, quercetin has application value and potential strength as a clinical drug. Furthermore, quercetin, as the main effective therapeutic ingredient in traditional Chinese medicine, may effectively treat and prevent coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Haoyang Zou
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Haiqing Ye
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Rajamanikkam Kamaraj
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, Hradec Kralove CZ500 05, Czech Republic
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Petr Pavek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Heyrovskeho 1203, Hradec Kralove CZ500 05, Czech Republic.
| |
Collapse
|
15
|
Thiopyrimidine derivatives induce cytotoxicity, cell cycle arrest and oxidative stress in breast cancer 3D-spheroids. CHEMICAL PAPERS 2021. [DOI: 10.1007/s11696-020-01383-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
16
|
More GK, Makola RT, Prinsloo G. In Vitro Evaluation of Anti-Rift Valley Fever Virus, Antioxidant and Anti-Inflammatory Activity of South African Medicinal Plant Extracts. Viruses 2021; 13:221. [PMID: 33572659 PMCID: PMC7912315 DOI: 10.3390/v13020221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/02/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Rift valley fever virus (RVFV) is a mosquito-borne virus endemic to sub-Saharan African countries, and the first sporadic outbreaks outside Africa were reported in the Asia-Pacific region. There are no approved therapeutic agents available for RVFV; however, finding an effective antiviral agent against RVFV is important. This study aimed to evaluate the antiviral, antioxidant and anti-inflammatory activity of medicinal plant extracts. Twenty medicinal plants were screened for their anti-RVFV activity using the cytopathic effect (CPE) reduction method. The cytotoxicity assessment of the extracts was done before antiviral screening using the MTT assay. Antioxidant and reactive oxygen/nitrogen species' (ROS/RNS) inhibitory activity by the extracts was investigated using non-cell-based and cell-based assays. Out of twenty plant extracts tested, eight showed significant potency against RVFV indicated by a decrease in tissue culture infectious dose (TCID50) < 105. The cytotoxicity of extracts showed inhibitory concentrations values (IC50) > 200 µg/mL for most of the extracts. The antioxidant activity and anti-inflammatory results revealed that extracts scavenged free radicals exhibiting an IC50 range of 4.12-20.41 µg/mL and suppressed the production of pro-inflammatory mediators by 60-80% in Vero cells. This study demonstrated the ability of the extracts to lower RVFV viral load and their potency to reduce free radicals.
Collapse
Affiliation(s)
- Garland K. More
- College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X6, Florida, Johannesburg 1710, South Africa;
| | - Raymond T. Makola
- Department of Biochemistry Microbiology and Biotechnology, School of Molecular and Life Science, University of Limpopo (Turfloop Campus) Sovenga, Polokwane 0727, South Africa;
- National institute of Communicable Diseases, Special Viral Pathogen/Arbovirus Unit, 1 Modderfontein Rd, Sandringham, Johannesburg 2192, South Africa
| | - Gerhard Prinsloo
- College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X6, Florida, Johannesburg 1710, South Africa;
| |
Collapse
|
17
|
Morawietz CM, Houhou H, Puckelwaldt O, Hehr L, Dreisbach D, Mokosch A, Roeb E, Roderfeld M, Spengler B, Haeberlein S. Targeting Kinases in Fasciola hepatica: Anthelminthic Effects and Tissue Distribution of Selected Kinase Inhibitors. Front Vet Sci 2020; 7:611270. [PMID: 33409299 PMCID: PMC7779637 DOI: 10.3389/fvets.2020.611270] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/13/2020] [Indexed: 11/13/2022] Open
Abstract
Protein kinases have been discussed as promising druggable targets in various parasitic helminths. New drugs are also needed for control of fascioliasis, a food-borne trematode infection and worldwide spread zoonosis, caused by the liver fluke Fasciola hepatica and related species. In this study, we intended to move protein kinases more into the spotlight of Fasciola drug research and characterized the fasciolicidal activity of two small-molecule inhibitors from human cancer research: the Abelson tyrosine kinase (ABL-TK) inhibitor imatinib and the polo-like 1 (PLK1) inhibitor BI2536. BI2536 reduced viability of 4-week-old immature flukes in vitro, while adult worms showed a blockade of egg production. Together with a significantly higher transcriptional expression of PLK1 in adult compared to immature worms, this argues for a role of PLK1 in fluke reproduction. Both fluke stages expressed ABL1-TK transcripts at similar high levels and were affected by imatinib. To study the uptake kinetic and tissue distribution of imatinib in F. hepatica, we applied matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) for the first time in this parasite. Drug imaging revealed the accumulation of imatinib in different fluke tissues from 20 min to 12 h of exposure. Furthermore, we show that imatinib is metabolized to N-desmethyl imatinib by F. hepatica, a bioactive metabolite also found in humans. Besides the vitellarium, gastrodermal tissue showed strong signal intensities. In situ hybridization demonstrated the gastrodermal presence of abl1 transcripts. Finally, we assessed transcriptional changes of physiologically important genes in imatinib-treated flukes. Moderately increased transcript levels of a gene encoding a multidrug resistance protein were detected, which may reflect an attempt to defend against imatinib. Increased expression levels of the cell cycle dependently expressed histone h2b and of two genes encoding superoxide dismutases (SODs) were also observed. In summary, our pilot study demonstrated cross-stage activity of imatinib but not BI2536 against immature and adult F. hepatica in vitro; a fast incorporation of imatinib within minutes, probably via the oral route; and imatinib-induced expression changes of physiologically relevant genes. We conclude that kinases are worth analyzing in more detail to evaluate the potential as therapeutic targets in F. hepatica.
Collapse
Affiliation(s)
- Carolin M Morawietz
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Hicham Houhou
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Oliver Puckelwaldt
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Laura Hehr
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Domenic Dreisbach
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Annika Mokosch
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Elke Roeb
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Martin Roderfeld
- Department of Gastroenterology, Justus Liebig University Giessen, Giessen, Germany
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Simone Haeberlein
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
18
|
Decoding Aging: Understanding the Complex Relationship among Aging, Free Radicals, and GSH. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3970860. [PMID: 33110472 PMCID: PMC7578726 DOI: 10.1155/2020/3970860] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/21/2020] [Accepted: 05/20/2020] [Indexed: 11/18/2022]
Abstract
N-aryl maleimides can undergo a 1,4-Michael-type addition reaction with reduced glutathione (GSH), leading to a decreased concentration of GSH and an increased concentration of free radicals (FRs) in cells. GSH is a critical scavenging molecule responsible for protecting cells from oxidation and for maintaining redox homeostasis. N-aryl maleimides disturb redox homeostasis in cells because they scavenge thiol-containing molecules, especially GSH. This study aimed at measuring the concentrations of GSH and FRs by electronic paramagnetic resonance (EPR), in the brain and liver tissue of male Wistar rats (ex vivo) at different ages and after treatment with 3,5-dimaleimylbenzoic acid (3,5-DMB). Our results showed a relationship between age and the concentrations of GSH and FRs in cells. In young rats, the concentration of GSH was higher than in old rats, while the concentration of FRs was higher in adult rats than in young rats, suggesting an inverse relationship between GSH and FRs. On the other hand, the reaction of 3,5-DMB (an electrophilic maleimide) with cellular GSH increased the FR content. The results of this study contribute to the awareness that the process of aging implies not only a loss of tissue function but also essential changes in the molecular contents of cells, especially the concentrations of FRs and GSH.
Collapse
|
19
|
Secondo LE, Avrutin V, Ozgur U, Topsakal E, Lewinski NA. Real-time monitoring of cellular oxidative stress during aerosol sampling: a proof of concept study. Drug Chem Toxicol 2020; 45:767-774. [PMID: 32529856 DOI: 10.1080/01480545.2020.1774774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The Portable In Vitro Exposure Cassette (PIVEC) was developed for on-site air quality testing using lung cells. Here, we describe the incorporation of a sensor within the PIVEC for real time monitoring of cellular oxidative stress during exposure to contaminated air. An electrochemical, enzymatic biosensor based on cytochrome c (cyt c) was selected to measure reactive oxygen species (ROS), including hydrogen peroxide and super oxides, due to the stability of signal over time. Human A549 lung cells were grown at the air-liquid interface and exposed within the PIVEC to dry 40 nm copper nanoparticle aerosols for 10 minutes. The generation of ROS compounds was measured during exposure and post-exposure for one hour using the biosensor and compared to intracellular ROS determined using the 2',7'-dichlorodihydrofluoroscein diacetate (DCFH-DA) assay. A similar increase in oxidative stress upon aerosol exposure was measured using both the cyt c biosensor and DCFH-DA assay. The incorporation of a biosensor within the PIVEC is a unique, first-of-its-kind system designed to monitor the real-time effect of aerosols.
Collapse
Affiliation(s)
- Lynn E Secondo
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, USA.,Department of Electrical and Computer Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Vitaliy Avrutin
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA
| | - Umit Ozgur
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA
| | - Erdem Topsakal
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA
| | - Nastassja A Lewinski
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
20
|
Quercetin Inhibits Lef1 and Resensitizes Docetaxel-Resistant Breast Cancer Cells. Molecules 2020; 25:molecules25112576. [PMID: 32492961 PMCID: PMC7321307 DOI: 10.3390/molecules25112576] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/29/2020] [Accepted: 05/30/2020] [Indexed: 02/07/2023] Open
Abstract
Drug resistance is a major problem for breast cancer patients. Docetaxel is an anti-mitotic agent that serves as first line of treatment in metastatic breast cancer, however it is susceptible to cellular drug resistance. Drug-resistant cells are able to spread during treatment, leading to treatment failure and eventually metastasis, which remains the main cause for cancer-associated death. In previous studies, we used single-cell technologies and identified a set of genes that exhibit increased expression in drug-resistant cells, and they are mainly regulated by Lef1. Furthermore, upregulating Lef1 in parental cells caused them to become drug resistant. Therefore, we hypothesized that inhibiting Lef1 could resensitize cells to docetaxel. Here, we confirmed that Lef1 inhibition, especially on treatment with the small molecule quercetin, decreased the expression of Lef1 and resensitized cells to docetaxel. Our results demonstrate that Lef1 inhibition also downregulated ABCG2, Vim, and Cav1 expression and equally decreased Smad-dependent TGF-β signaling pathway activation. Likewise, these two molecules worked in a synergetic manner, greatly reducing the viability of drug-resistant cells. Prior studies in phase I clinical trials have already shown that quercetin can be safely administered to patients. Therefore, the use of quercetin as an adjuvant treatment in addition to docetaxel for the treatment of breast cancer may be a promising therapeutic approach.
Collapse
|
21
|
Kong N, Ji X, Wang J, Sun X, Chen G, Fan T, Liang W, Zhang H, Xie A, Farokhzad OC, Tao W. ROS-Mediated Selective Killing Effect of Black Phosphorus: Mechanistic Understanding and Its Guidance for Safe Biomedical Applications. NANO LETTERS 2020; 20:3943-3955. [PMID: 32243175 DOI: 10.1021/acs.nanolett.0c01098] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Black phosphorus (BP)-based nanomaterials have distinguished advantages and potential applications in various biomedical fields. However, their biological effects in physiological systems remain largely unexplored. Here, we systematically revealed a reactive oxygen species (ROS)-mediated mechanism for the selective killing of cancer cells by BP-based nanosheets. The treatment with BP-based materials can induce higher levels of ROS in cancer cells than in normal cells, leading to significant changes in the cytoskeleton, cell cycle arrest, DNA damage, and apoptosis in tumor cell lines. We revealed that the decreased superoxide dismutase activity by lipid peroxides could be an essential mechanism of the selectively higher ROS generation induced by BP-based nanosheets in cancer cells. In addition, the selective killing effect only occurred within a certain dosage range (named "SK range" in this study). Once exceeding the SK range, BP-based materials could also induce a high ROS production in normal tissues, leading to detectable DNA damage and pathological characteristics in normal organs and raising safety concerns. These findings not only shed light on a new mechanism for the selective killing of cancer cells by BP-based materials but also provide deep insights into the safe use of BP-based therapies.
Collapse
Affiliation(s)
- Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Xiaoyuan Ji
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Junqing Wang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Xiuna Sun
- Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Guoqiao Chen
- Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Taojian Fan
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Weiyuan Liang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Han Zhang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Anyong Xie
- Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
22
|
Markowicz-Piasecka M, Komeil I, Huttunen J, Sikora J, Huttunen KM. Effective Cellular Transport of Ortho-Halogenated Sulfonamide Derivatives of Metformin Is Related to Improved Antiproliferative Activity and Apoptosis Induction in MCF-7 Cells. Int J Mol Sci 2020; 21:E2389. [PMID: 32235654 PMCID: PMC7177633 DOI: 10.3390/ijms21072389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 01/08/2023] Open
Abstract
Metformin is a substrate for plasma membrane monoamine transporters (PMAT) and organic cation transporters (OCTs); therefore, the expression of these transporters and interactions between them may affect the uptake of metformin into tumor cells and its anticancer efficacy. The aim of this study was to evaluate how chemical modification of metformin scaffold into benzene sulfonamides with halogen substituents (compounds 1-9) may affect affinity towards OCTs, cellular uptake in two breast cancer cell lines (MCF-7 and MDA-MB-231) and antiproliferative efficacy of metformin. The uptake of most sulfonamides was more efficient in MCF-7 cells than in MDA-MB-231 cells. The presence of a chlorine atom in the aromatic ring contributed to the highest uptake in MCF-7 cells. For instance, the uptake of compound 1 with o-chloro substituent in MCF-7 cells was 1.79 ± 0.79 nmol/min/mg protein, while in MDA-MB-231 cells, the uptake was considerably lower (0.005 ± 0.0005 nmol/min/mg protein). The elevated uptake of tested compounds in MCF-7 was accompanied by high antiproliferative activity, with compound 1 being the most active (IC50 = 12.6 ± 1.2 µmol/L). Further studies showed that inhibition of MCF-7 growth is associated with the induction of early and late apoptosis and cell cycle arrest at the G0/G1 phase. In summary, the chemical modification of the biguanide backbone into halogenated sulfonamides leads to improved transporter-mediated cellular uptake in MCF-7 and contributes to the greater antiproliferative potency of studied compounds through apoptosis induction and cell cycle arrest.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland;
| | - Ibrahim Komeil
- Department of Pharmaceutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University, Alexandria 21311, Egypt;
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland; (J.H.); (K.M.H.)
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland;
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland; (J.H.); (K.M.H.)
| |
Collapse
|
23
|
Hafezi K, Hemmati AA, Abbaszadeh H, Valizadeh A, Makvandi M. Anticancer activity and molecular mechanisms of α-conidendrin, a polyphenolic compound present in Taxus yunnanensis, on human breast cancer cell lines. Phytother Res 2020; 34:1397-1408. [PMID: 31971313 DOI: 10.1002/ptr.6613] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/18/2019] [Accepted: 12/31/2019] [Indexed: 12/14/2022]
Abstract
α-Conidendrin is a polyphenolic compound found mainly in Taxus yunnanensis, as the source of chemotherapy drug paclitaxel, which has been used in traditional medicine for treatment of cancer. This study aimed to investigate the anticancer activity and molecular mechanisms of α-conidendrin on breast cancer cell lines. The results of the present study show that α-conidendrin possesses potent antiproliferative effects on breast cancer cell lines MCF-7 and MDA-MB-231. α-Conidendrin significantly induced apoptosis in breast cancer cells via reactive oxygen species generation, upregulation of p53 and Bax, downregulation of Bcl-2, depolarization of mitochondrial membrane potential (MMP), release of cytochrome c from mitochondria, and activation of caspases-3 and -9. α-Conidendrin remarkably inhibited the proliferation of breast cancer cells through induction of cell cycle arrest by upregulating p53 and p21 and downregulating cyclin D1 and CDK4. Unlike breast cancer cells, the antiproliferative effect of α-conidendrin on human foreskin fibroblast cells (normal cells) was very small. In normal cells, reactive oxygen species levels, loss of MMP, release of cytochrome c, mRNA expression of p53, p21, cyclin D1, CDK4, Bax, and Bcl-2 as well as mRNA expression and activity of caspases-3 and -9 were significantly less affected by α-conidendrin compared with cancer cells. These results suggest that α-conidendrin can be a promising agent for treatment of breast cancer with little or no toxicity against normal cells.
Collapse
Affiliation(s)
- Katayoon Hafezi
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Asghar Hemmati
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hassan Abbaszadeh
- Department of Pharmacology, School of Pharmacy, Medicinal Plants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Armita Valizadeh
- Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Manoochehr Makvandi
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
24
|
Alehaideb Z, AlGhamdi S, Yahya WB, Al-Eidi H, Alharbi M, Alaujan M, Albaz A, Tukruni M, Nehdi A, Abdulla MH, Matou-Nasri S. Anti-Proliferative and Pro-Apoptotic Effects of Calligonum comosum (L'Her.) Methanolic Extract in Human Triple-Negative MDA-MB-231 Breast Cancer Cells. J Evid Based Integr Med 2020; 25:2515690X20978391. [PMID: 33302699 PMCID: PMC7734547 DOI: 10.1177/2515690x20978391] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 10/29/2020] [Accepted: 11/10/2020] [Indexed: 12/21/2022] Open
Abstract
Triple-negative breast cancer (TNBC), the most aggressive subtype, does not respond to targeted therapy due to the lack of hormone receptors. There is an urgent need for alternative therapies, including natural product-based anti-cancer drugs, at lower cost. We investigated the impact of a Calligonum comosum L'Hér. methanolic extract (CcME) on the TNBC MDA-MB-231 cell line proliferation and related cell death mechanisms performing cell viability and cytotoxicity assays, flow cytometry to detect apoptosis and cell cycle analysis. The apoptosis-related protein array and cellular reactive oxygen species (ROS) assay were also carried out. We showed that the CcME inhibited the TNBC cell viability, in a dose-dependent manner, with low cytotoxic effects. The CcME-treated TNBC cells underwent apoptosis, associated with a concomitant increase of apoptosis-related protein expression, including cytochrome c, cleaved caspase-3, cyclin-dependent kinase inhibitor p21, and the anti-oxidant enzyme catalase, compared with the untreated cells. The CcME also enhanced the mitochondrial transition pore opening activity and induced G0/G1 cell growth arrest, which confirmed the cytochrome c release and the increase of the p21 expression detected in the CcME-treated TNBC cells. The CcME-treated TNBC cells resulted in intracellular ROS production, which, when blocked with a ROS scavenger, did not reduce the CcME-induced apoptosis. In conclusion, CcME exerts anti-proliferative effects against TNBC cells through the induction of apoptosis and cell growth arrest. In vivo studies are justified to verify the CcME anti-proliferative activities and to investigate any potential anti-metastatic activities of CcME against TNBC development and progression.
Collapse
Affiliation(s)
- Zeyad Alehaideb
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Saleh AlGhamdi
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Clinical Research Department, Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Wesam Bin Yahya
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Hamad Al-Eidi
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Mashael Alharbi
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Monira Alaujan
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abeer Albaz
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Muruj Tukruni
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Atef Nehdi
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Maha-Hamadien Abdulla
- Department of Surgery, King Khalid University Hospital and College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sabine Matou-Nasri
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
25
|
Hu S, Li SW, Yan Q, Hu XP, Li LY, Zhou H, Pan LX, Li J, Shen CP, Xu T. Natural products, extracts and formulations comprehensive therapy for the improvement of motor function in alcoholic liver disease. Pharmacol Res 2019; 150:104501. [PMID: 31689520 DOI: 10.1016/j.phrs.2019.104501] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/08/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023]
|
26
|
Fernández-Palanca P, Fondevila F, Méndez-Blanco C, Tuñón MJ, González-Gallego J, Mauriz JL. Antitumor Effects of Quercetin in Hepatocarcinoma In Vitro and In Vivo Models: A Systematic Review. Nutrients 2019; 11:nu11122875. [PMID: 31775362 PMCID: PMC6950472 DOI: 10.3390/nu11122875] [Citation(s) in RCA: 48] [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: 10/21/2019] [Revised: 11/14/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022] Open
Abstract
Quercetin is a flavonoid present in fruits, vegetables and plants with antioxidant, anti-inflammatory and anticancer properties. Its beneficial activities have been demonstrated in different human pathologies, including hepatoprotective effects against liver disorders. High mortality and late diagnosis of the primary liver tumor hepatocarcinoma (HCC) makes this cancer an interesting target for the study of quercetin effects. Our aim was to systematically review antitumor activities of quercetin in HCC preclinical studies employing single, encapsulated, combined or derived quercetin forms. Literature search was conducted in PubMed, Scopus and Web of Science (WOS), and 39 studies were finally included. We found that 17 articles evaluated quercetin effects alone, six used encapsulated strategy, 10 combined this flavonoid, two decided to co-encapsulate it and only four studied effects of quercetin derivatives, highlighting that only nine included in vivo models. Results evidence the quercetin antiproliferative and proapoptotic properties against HCC either alone and with the mentioned strategies; nevertheless, few investigations assessed specific activities on different processes related with cancer progression. Overall, further studies including animal models are needed to deeper investigate the precise mechanisms of action of quercetin as antitumor agent, as well as the potential of novel strategies aimed to improve quercetin effects in HCC.
Collapse
Affiliation(s)
- Paula Fernández-Palanca
- Institute of Biomedicine, University of León, 24071 León, Spain; (P.F.-P.); (F.F.); (C.M.-B.); (M.J.T.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Flavia Fondevila
- Institute of Biomedicine, University of León, 24071 León, Spain; (P.F.-P.); (F.F.); (C.M.-B.); (M.J.T.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Carolina Méndez-Blanco
- Institute of Biomedicine, University of León, 24071 León, Spain; (P.F.-P.); (F.F.); (C.M.-B.); (M.J.T.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - María J. Tuñón
- Institute of Biomedicine, University of León, 24071 León, Spain; (P.F.-P.); (F.F.); (C.M.-B.); (M.J.T.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Javier González-Gallego
- Institute of Biomedicine, University of León, 24071 León, Spain; (P.F.-P.); (F.F.); (C.M.-B.); (M.J.T.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - José L. Mauriz
- Institute of Biomedicine, University of León, 24071 León, Spain; (P.F.-P.); (F.F.); (C.M.-B.); (M.J.T.); (J.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
27
|
Wu L, Li J, Liu T, Li S, Feng J, Yu Q, Zhang J, Chen J, Zhou Y, Ji J, Chen K, Mao Y, Wang F, Dai W, Fan X, Wu J, Guo C. Quercetin shows anti-tumor effect in hepatocellular carcinoma LM3 cells by abrogating JAK2/STAT3 signaling pathway. Cancer Med 2019; 8:4806-4820. [PMID: 31273958 PMCID: PMC6712453 DOI: 10.1002/cam4.2388] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 05/31/2019] [Accepted: 06/17/2019] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Hepatocellular carcinima is one of the most common tumors in clinic and also one of the leading causes of death from cancer worldwide. Quercetin shows significant effects on blocking the development of various cancers. METHODS We used the human hepatocellular carcinoma LM3 and nude mice tumor model to assess the effects of quercetin in hepatocellular carcinoma and clarify its mechanism of action. We collected LM3 cell line treated with different doses of quercetin at different time periods and determined the vital indexes. The liver tissues of mice were collected and used for western boltting (WB), Hematoxylin and Eosin (H&E) and TUNEL staining. RESULTS Results indicated that quercetin suppressed the Hepatocellular carcinoma (HCC) growth both in vivo and in vitro. Quercetin could disturb LM3 cells proliferation and cell cycle distribution, thus inducing apoptosis. At the same time, quercetin inhibited LM3 cells migration and invasion and promoted HCC autophagy. These effects at least partly depended on the down-regulation of the activation of JAK2 and STAT3 by quercetin. CONCLUSION Quercetin inhibited hepatocellular carcinoma progression by modulating cell apoptosis, migration, invasion, and autophagy; and its effects were at least partly related with the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Liwei Wu
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
| | - Jingjing Li
- Department of GastroenterologyPutuo People's Hospital, Tongji University School of MedicineShanghaiChina
| | - Tong Liu
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
| | - Sainan Li
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
| | - Jiao Feng
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
| | - Qiang Yu
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
- Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Jie Zhang
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
- Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Jiaojiao Chen
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
- Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Yuting Zhou
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
- Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Jie Ji
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
| | - Kan Chen
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
| | - Yuqing Mao
- Department of GerontologyShanghai General Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Fan Wang
- Department of OncologyShanghai General Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Weiqi Dai
- Department of GastroenterologyZhongshan Hospital of Fudan UniversityShanghaiChina
- Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan UniversityShanghaiChina
| | - Xiaoming Fan
- Department of GastroenterologyJinshan Hospital of Fudan University, JinshanShanghaiChina
| | - Jianye Wu
- Department of GastroenterologyPutuo People's Hospital, Tongji University School of MedicineShanghaiChina
| | - Chuanyong Guo
- Department of GastroenterologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghaiChina
| |
Collapse
|
28
|
Reyes-Farias M, Carrasco-Pozo C. The Anti-Cancer Effect of Quercetin: Molecular Implications in Cancer Metabolism. Int J Mol Sci 2019; 20:E3177. [PMID: 31261749 PMCID: PMC6651418 DOI: 10.3390/ijms20133177] [Citation(s) in RCA: 402] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/19/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022] Open
Abstract
Cancer is a problem with worldwide importance and is the second leading cause of death globally. Cancer cells reprogram their metabolism to support their uncontrolled expansion by increasing biomass (anabolic metabolism-glycolysis) at the expense of their energy (bioenergetics- mitochondrial function) requirements. In this aspect, metabolic reprogramming stands out as a key biological process in understanding the conversion of a normal cell into a neoplastic precursor. Quercetin is the major representative of the flavonoid subclass of flavonols. Quercetin is ubiquitously present in fruits and vegetables, being one of the most common dietary flavonols in the western diet. The anti-cancer effects of quercetin include its ability to promote the loss of cell viability, apoptosis and autophagy through the modulation of PI3K/Akt/mTOR, Wnt/-catenin, and MAPK/ERK1/2 pathways. In this review, we discuss the role of quercetin in cancer metabolism, addressing specifically its ability to target molecular pathways involved in glucose metabolism and mitochondrial function.
Collapse
Affiliation(s)
- Marjorie Reyes-Farias
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, 08916 Barcelona, Spain
| | - Catalina Carrasco-Pozo
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| |
Collapse
|
29
|
Carrasco-Torres G, Valdés-Madrigal MA, Vásquez-Garzón VR, Baltiérrez-Hoyos R, De la Cruz-Burelo E, Román-Doval R, Valencia-Lazcano AA. Effect of Silk Fibroin on Cell Viability in Electrospun Scaffolds of Polyethylene Oxide. Polymers (Basel) 2019; 11:E451. [PMID: 30960435 PMCID: PMC6473723 DOI: 10.3390/polym11030451] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/12/2019] [Accepted: 02/28/2019] [Indexed: 01/06/2023] Open
Abstract
In this study, a coating from electrospun silk fibroin was performed with the aim to modify the surface of breast implants. We evaluated the effect of fibroin on polymeric matrices of poly (ethylene oxide) (PEO) to enhance cell viability, adhesion, and proliferation of HaCaT human keratinocytes to enhance the healing process on breast prosthesis implantation. We electrospun six blends of fibroin and PEO at different concentrations. These scaffolds were characterized by scanning electron microscopy, contact angle measurements, ATR-FTIR spectroscopy, and X-ray diffraction. We obtained diverse network conformations at different combinations to examine the regulation of cell adhesion and proliferation by modifying the microstructure of the matrix to be applied as a potential scaffold for coating breast implants. The key contribution of this work is the solution it provides to enhance the healing process on prosthesis implantation considering that the use of these PEO⁻fibroin scaffolds reduced (p < 0.05) the amount of pyknotic nuclei. Therefore, viability of HaCaT human keratinocytes on PEO⁻fibroin matrices was significantly improved (p < 0.001). These findings provide a rational strategy to coat breast implants improving biocompatibility.
Collapse
Affiliation(s)
- Gabriela Carrasco-Torres
- Departamento de Nanociencias y Nanotecnología. Centro de Investigación y de Estudios Avanzados del IPN. Av. IPN 2508, la laguna Ticomán, Ciudad de México 07360, Mexico.
| | - Manuel A Valdés-Madrigal
- Departamento de Nanociencias y Nanotecnología. Centro de Investigación y de Estudios Avanzados del IPN. Av. IPN 2508, la laguna Ticomán, Ciudad de México 07360, Mexico.
- Instituto Tecnológico Superior de Ciudad Hidalgo. Av. Ing. Carlos Rojas Gutiérrez 2120, fracc. Valle de la herradura, Michoacán 61100, Mexico.
| | - Verónica R Vásquez-Garzón
- CONACYT-Facultad de Medicina y Cirugía Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Carretera a San Felipe del Agua S/N, Oaxaca 68020, Mexico.
| | - Rafael Baltiérrez-Hoyos
- CONACYT-Facultad de Medicina y Cirugía Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Carretera a San Felipe del Agua S/N, Oaxaca 68020, Mexico.
| | - Eduard De la Cruz-Burelo
- Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN 2508, la laguna Ticomán, Ciudad de México 07360, Mexico.
| | - Ramón Román-Doval
- Departamento de investigación y posgrado en alimentos, Facultad de Química, Universidad Autónoma de Querétaro, Centro universitario, Santiago de Querétaro, Querétaro 76010, Mexico.
| | - Anaí A Valencia-Lazcano
- Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN 2508, la laguna Ticomán, Ciudad de México 07360, Mexico.
| |
Collapse
|
30
|
Erukainure OL, Narainpersad N, Singh M, Olakunle S, Islam MS. Clerodendrum volubile inhibits key enzymes linked to type 2 diabetes but induces cytotoxicity in human embryonic kidney (HEK293) cells via exacerbated oxidative stress and proinflammation. Biomed Pharmacother 2018; 106:1144-1152. [PMID: 30119181 DOI: 10.1016/j.biopha.2018.07.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/29/2018] [Accepted: 07/01/2018] [Indexed: 12/12/2022] Open
Abstract
The toxicity and safety associated with the use of medicinal plants remains a major concern. In this study, the antidiabetic properties of the dichloromethane (DCM) fraction of C. volubile leaves were investigated in vitro. Its cytotoxic effect and mechanism of toxicity were also investigated in Human Embryonic Kidney (HEK293) cells. The fraction was subjected to in vitro antioxidant assays using the 2,2'-diphenyl-1-picrylhydrazyl (DPPH) scavenging and Ferric reducing antioxidant power (FRAP) protocols. Its enzyme-inhibitory properties were investigated on α-glucosidase and α-amylase activities. Gas Chromatography Mass Spectroscopy (GCMS) and Fourier Transform Infrared (FTIR) spectroscopic analysis were used to identify its phytoconstituents. Cytotoxicity was determined via MTT assay. The treated cells were assayed for reduced glutathione (GSH), non-protein thiol, nitric oxide and malondialdehyde (MDA) levels, as well as Superoxide Dismutase (SOD), catalase, myeloperoxidase and ATPase activities. Cell apoptosis and/or morphological changes were determined using the acridine orange and ethidium bromide (AO/EB) dual staining method. The fraction showed significant (p < 0.05) antioxidant and enzyme-inhibitory activity. It showed significant (p < 0.05) cytotoxic effect against HEK293 cells with concomitant depletion of antioxidative and elevation of proinflammatory biomarkers. Morphological changes were examined in the cells with an apoptotic index of 0.84. 1,1-Dodecanediol, diacetate was identified as the most predominant compound, while aromatics and amines as the most functional groups present in the fraction. These results suggest the antidiabetic and cytotoxic effects of C. volubile leaves. The toxicity can be attributed to induced oxidative stress and proinflammation with concomitant depletion of ATP leading to apoptosis of the cells.
Collapse
Affiliation(s)
- Ochuko L Erukainure
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa; Nutrition and Toxicology Division, Federal Institute of Industrial Research, Lagos, Nigeria
| | - Nicholisha Narainpersad
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Moganavelli Singh
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Sanni Olakunle
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa.
| |
Collapse
|
31
|
Garcia AA, Rayevski A, Andrade-Jorge E, Trujillo-Ferrara JG. Structural and biological overview of Boron-containing amino acids in the medicinal chemistry field. Curr Med Chem 2018; 26:5077-5089. [PMID: 30259808 DOI: 10.2174/0929867325666180926150403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 09/06/2018] [Accepted: 09/06/2018] [Indexed: 11/22/2022]
Abstract
Amino acids are the basic structural units of proteins as well as the precursors of many compounds with biological activity. The addition of boron reportedly induces changes in the chemical-biological profile of amino acids. METHODS We compiled information on the biological effect of some compounds and discuss the structure-activity relationship of the addition of boron. The specific focus presently is on borinic derivatives of α-amino acids, the specific changes in biological activity caused by the addition of a boron-containing moiety, and the identification of some attractive compounds for testing as potential new drugs. RESULTS Borinic derivatives of α-amino acids have been widely synthesized and tested as potential new therapeutic tools. The B-N (1.65 A°) or B-C (1.61 A°) or B-O (1.50 A°) bond is often key for the stability at different pHs and temperatures and activity of these compounds. The chemical features of synthesized derivatives, such as the specific moieties and the logP, polarizability and position of the boron atom are clearly linked to their pharmacodynamic and pharmacokinetic profiles. Some mechanisms of action have been suggested or demonstrated, while those responsible for other effects remain unknown. CONCLUSION The increasing number of synthetic borinic derivatives of α-amino acids as well as the recently reported crystal structures are providing new insights into the stability of these compounds at different pHs and temperatures, their interactions on drug targets, and the ring formation of five-membered heterocycles. Further research is required to clarify the ways to achieve specific synthesis, the mechanisms involved in the observed biological effect, and the toxicological profile of this type of boron-containing compounds (BCCs).
Collapse
Affiliation(s)
- Antonio Abad Garcia
- Departamento de Bioquimica y Seccion de Estudios de Posgrado e Investigación. Escuela Superior de Medicina. Plan de San Luis y Diaz Miron s/n, 11340, Mexico City. Mexico
| | - Alexey Rayevski
- Chuiko Institute of Surface Chemistry, National Academy of Science of Ukranie. 17 Generala Naumova St., 03164, Kyiv. Ukraine
| | - Erik Andrade-Jorge
- Departamento de Bioquimica y Seccion de Estudios de Posgrado e Investigacion. Escuela Superior de Medicina. Plan de San Luis y Diaz Miron s/n, 11340, Mexico City. Mexico
| | - Jose G Trujillo-Ferrara
- Departamento de Bioquímica y Sección de Estudios de Posgrado e Investigación. Escuela Superior de Medicina. Plan de San Luis y Diaz Mirón s/n, 11340, Mexico City. Mexico
| |
Collapse
|
32
|
Erukainure OL, Ashraf N, Naqvi AS, Zaruwa MZ, Muhammad A, Odusote AD, Elemo GN. Fatty Acids Rich Extract From Clerodendrum volubile Suppresses Cell Migration; Abates Oxidative Stress; and Regulates Cell Cycle Progression in Glioblastoma Multiforme (U87 MG) Cells. Front Pharmacol 2018; 9:251. [PMID: 29615913 PMCID: PMC5870396 DOI: 10.3389/fphar.2018.00251] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 03/06/2018] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a malignant primary type of brain cancer with high proliferation and metastasis rates due to involvement of the microglial cell. It is resistant against available chemotherapy. Many strategic protocols have been developed but prognosis and patient life has not improved substantially. In this study, the anti-metastatic and antioxidant effect of fatty acids from Clerodendrum volubile leaves were investigated in U87-MG (Human Glioblastoma Multiforme) cell lines. The extracted fatty acids were incubated with U87-MG cells for 48 h. The anti-proliferative effect was determined by MTT assay, while apoptosis and cell cycle were analyzed with BD FACSCalibur. The transwell assay protocol was utilized in the analysis of cell migration and invasion. The treated cell lines were also assessed for reduced glutathione (GSH) level, catalase, superoxide dismutase (SOD) and lipid peroxidation. The fatty acid extract showed significant inhibitory activity on cell proliferation and cell cycle progression, mitigated oxidative stress, and suppressed migration and invasion in U-87 MG cell lines. These results give credence to the therapeutic potential of this plant against cancer, especially GBM.
Collapse
Affiliation(s)
- Ochuko L Erukainure
- Nutrition and Toxicology Division, Federal Institute of Industrial Research Oshodi, Lagos, Nigeria
| | - Nadia Ashraf
- Faculty of Pharmacy, Barrett Hodgson University, Karachi, Pakistan
| | - Asma S Naqvi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Moses Z Zaruwa
- Department of Biochemistry, Adamawa State University, Mubi, Nigeria
| | - Aliyu Muhammad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan.,Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Adenike D Odusote
- Analytical Division, Federal Institute of Industrial Research Oshodi, Lagos, Nigeria
| | - Gloria N Elemo
- Nutrition and Toxicology Division, Federal Institute of Industrial Research Oshodi, Lagos, Nigeria
| |
Collapse
|