1
|
Cascajosa-Lira A, Prieto AI, Pichardo S, Jos A, Cameán AM. Protective effects of sulforaphane against toxic substances and contaminants: A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155731. [PMID: 38824824 DOI: 10.1016/j.phymed.2024.155731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Sulforaphane (SFN) is a dietary isothiocyanate, derived from glucoraphanin, present in cruciferous vegetables belonging to the Brassica genus. It is a biologically active phytochemical that acts as a nuclear factor erythroid 2-related factor 2 (Nrf2) inducer. Thus, it has been reported to have multiple protective functions including anticancer responses and protection against a toxic agent's action. PURPOSE The present work systematically reviewed and synthesised the protective properties of sulforaphane against a toxic agent. This review reveals the mechanism of the action of SFN in each organ or system. METHODS The PRISMA guideline was followed in this sequence: researched literature, organised retrieved documents, abstracted relevant information, assessed study quality and bias, synthesised data, and prepared a comprehensive report. Searches were conducted on Science Direct and PubMed using the keywords "Sulforaphane" AND ("protective effects" OR "protection against"). RESULTS Reports showed that liver and the nervous system are the target organs on which attention was focused, and this might be due to the key role of oxidative stress in liver and neurodegenerative diseases. However, protective activities have also been demonstrated in the lungs, heart, immune system, kidneys, and endocrine system. SFN exerts its protective effects by activating the Nrf2 pathway, which enhances antioxidant defenses and reduces oxidative stress. It also suppresses inflammation by decreasing interleukin production. Moreover, SFN inhibits apoptosis by preventing caspase 3 cleavage and increasing Bcl2 levels. Overall, SFN demonstrates multifaceted mechanisms to counteract the adverse effects of toxic agents. CONCLUSION SFN has potential clinical applications as a chemoprotective agent. Nevertheless, more studies are necessary to set the safe doses of SFN in humans.
Collapse
Affiliation(s)
- Antonio Cascajosa-Lira
- Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, España.
| | - Ana I Prieto
- Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, España.
| | - Silvia Pichardo
- Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, España.
| | - Angeles Jos
- Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, España.
| | - Ana M Cameán
- Área de Toxicología, Facultad de Farmacia, Universidad de Sevilla, Profesor García González n 2, 41012 Seville, España.
| |
Collapse
|
2
|
Haritwal T, Kalra N, Agrawala PK. Mitigation of radiation injury to reproductive system of male mice by Trichostatin A. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 881:503522. [PMID: 36031339 DOI: 10.1016/j.mrgentox.2022.503522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 06/14/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
Trichostatin A (TSA), derived from the bacteria Streptomyces hygroscopicus, is a hydroxamic acid having various biological properties such as histone deacetylase inhibition, anticancer and radiomitigative action. However the mitigative activity of TSA against radiation-induced damages in the mouse reproductive system has not yet been elucidated. The present study unraveled the effects of 2 Gy whole body irradiation (60Co γ- radiation) on C57BL/6 mice male reproductive system including structural damages to testes, increase in apoptosis and reduction in germ cell viability, reduced fertility as well as increased genomic instability in the next generation. Moreover, hematological study and micronuclei assay were used to record chances of radiation-induced hematologic cancer and disruption of genomic integrity in F1 generation. Interestingly, TSA administration 1 and 24 h post-irradiation attenuated radiation-induced morphological damage and cellular apoptosis in testes. In male mice, TSA restored hematological parameters and micronuclei frequency to normal levels, restored sperm viability, and helped them overcome radiation-induced temporary sterility 5 weeks after the irradiation. Thus our results showed that TSA reduced the probability of radiation-induced hematologic cancers as well as genotoxicity and restored genomic integrity in the progenies of paternally exposed mice by reducing radiation-induced apoptosis in spermatogenic cells and restoring cell proliferation. This study suggested that TSA could be used as potential radiomitigator for male reproductive system.
Collapse
Affiliation(s)
- Teena Haritwal
- Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi 110054, India
| | - Namita Kalra
- Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi 110054, India
| | - Paban K Agrawala
- Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi 110054, India.
| |
Collapse
|
3
|
Xu Y, Zeng F, Jiang J, Huo J, Zhao C, Yan Z, Li L. The Hematopoietic Function of Medicinal Wine Maoji Jiu Revealed in Blood Deficiency Model Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:1025504. [PMID: 35911170 PMCID: PMC9325634 DOI: 10.1155/2022/1025504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022]
Abstract
Maoji Jiu (MJ), a medicinal wine, has been used commonly by the Chinese to enrich and nourish the blood. In this study, the aim is to examine the hematopoietic function of MJ and investigate its hematopoietic regulation mechanism. Thirty-six female Sprague-Dawley rats (200 ± 20 g) were randomly divided into six groups with six rats in each group. The blood deficiency model was induced by injecting hypodermically with N-acetylphenylhydrazine (APH) and injecting intraperitoneally with cyclophosphamide (CTX), and treatment drugs were given by oral gavage twice a day for continuous 10 days from the start of the experiments. The administration of MJ improved the levels of white blood cells (WBCs), red blood cells (RBCs), hemoglobin (HGB), and hematocrit (HCT) in the blood deficiency model rats. Hematopoietic effect involves regulating the antioxidant activity in the liver and the levels of Bcl-2, Bax, erythropoietin (EPO), transforming growth factor-beta-1 (TGF-β1), and macrophage colony-stimulating factor (M-CSF) mRNA in spleen tissues to enhance extramedullary hematopoiesis. This study suggests that MJ has a beneficial effect on blood deficiency model rats.
Collapse
Affiliation(s)
- Yongli Xu
- Guangxi Botanical Garden of Medicinal Plants, Nanning 530023, Guangxi, China
| | - Fanqiang Zeng
- Department of Pharmacy, Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang 537100, Guangxi, China
| | - Jianping Jiang
- Guangxi Botanical Garden of Medicinal Plants, Nanning 530023, Guangxi, China
| | - Juan Huo
- Guangxi Botanical Garden of Medicinal Plants, Nanning 530023, Guangxi, China
| | - Chengjian Zhao
- Guangxi Botanical Garden of Medicinal Plants, Nanning 530023, Guangxi, China
| | - Zhigang Yan
- Guangxi Botanical Garden of Medicinal Plants, Nanning 530023, Guangxi, China
| | - Li Li
- Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning 530022, Guangxi, China
| |
Collapse
|
4
|
Farag MR, Moselhy AAA, El-Mleeh A, Aljuaydi SH, Ismail TA, Di Cerbo A, Crescenzo G, Abou-Zeid SM. Quercetin Alleviates the Immunotoxic Impact Mediated by Oxidative Stress and Inflammation Induced by Doxorubicin Exposure in Rats. Antioxidants (Basel) 2021; 10:antiox10121906. [PMID: 34943009 PMCID: PMC8750303 DOI: 10.3390/antiox10121906] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent against hematogenous and solid tumors with undesirable side effects including immunosuppression. Quercetin (QUR), a natural flavonoid abundant in fruits and vegetables, has a potent antioxidant activity. The aim of the current study was to assess the impact of QUR on DOX-induced hematological and immunological dysfunctions in a rodent model. Randomly grouped rats were treated as follows: control, QUR alone (50 mg/kg for 15 days per os), DOX alone (2.5 mg/kg I/P, three times a week, for two weeks), and co-treated rats with QUR for 15 days prior to and concomitantly with DOX (for two weeks), at the doses intended for groups two and three. DOX alone significantly disrupted the erythrogram and leukogram variables. Serum immunoglobulin (IgG, IgM, and IgE) levels and the activities of catalase (CAT) and superoxide dismutase (SOD) in spleen were declined. The DNA damage traits in spleen were elevated with an upregulation of the expression of the apoptotic markers (p53 and Caspase-3 genes) and the proinflammatory cytokines (IL-6 and TNF-α genes), while the expression of CAT gene was downregulated. These biochemical changes were accompanied by morphological changes in the spleen of DOX-treated rats. Co-treatment with QUR abated most of the DOX-mediated alterations in hematological variables, serum immunoglobulins, and spleen antioxidant status, pro-inflammatory and apoptotic responses, and histopathological alterations. In essence, these data suggest that QUR alleviated DOX-induced toxicities on the bone marrow, spleen, and antibody-producing cells. Supplementation of chemotherapy patients with QUR could circumvent the DOX-induced inflammation and immunotoxicity, and thus prevent chemotherapy failure.
Collapse
Affiliation(s)
- Mayada R. Farag
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
- Correspondence: (M.R.F.); (A.D.C.)
| | - Attia A. A. Moselhy
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Amany El-Mleeh
- Department of Pharmacology, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkoum 32511, Egypt;
| | - Samira H. Aljuaydi
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Tamer Ahmed Ismail
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
- Correspondence: (M.R.F.); (A.D.C.)
| | - Giuseppe Crescenzo
- Department of Veterinary Medicine, University of Bari ‘Aldo Moro’, 70121 Bari, Italy;
| | - Shimaa M. Abou-Zeid
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 6012201, Egypt;
| |
Collapse
|
5
|
Wei J, Zhao Q, Zhang Y, Shi W, Wang H, Zheng Z, Meng L, Xin Y, Jiang X. Sulforaphane-Mediated Nrf2 Activation Prevents Radiation-Induced Skin Injury through Inhibiting the Oxidative-Stress-Activated DNA Damage and NLRP3 Inflammasome. Antioxidants (Basel) 2021; 10:antiox10111850. [PMID: 34829721 PMCID: PMC8614868 DOI: 10.3390/antiox10111850] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/16/2022] Open
Abstract
This article mainly observed the protective effect of sulforaphane (SFN) on radiation-induced skin injury (RISI). In addition, we will discuss the mechanism of SFN's protection on RISI. The RISI model was established by the irradiation of the left thigh under intravenous anesthesia. Thirty-two C57/BL6 mice were randomly divided into control group (CON), SFN group, irradiation (IR) group, and IR plus SFN (IR/SFN) group. At eight weeks after irradiation, the morphological changes of mouse skin tissues were detected by H&E staining. Then, the oxidative stress and inflammatory response indexes in mouse skin tissues, as well as the expression of Nrf2 and its downstream antioxidant genes, were evaluated by ELISA, real-time PCR, and Western blotting. The H&E staining showed the hyperplasia of fibrous tissue in the mouse dermis and hypodermis of the IR group. Western blotting and ELISA results showed that the inflammasome of NLRP3, caspase-1, and IL-1β, as well as oxidative stress damage indicators ROS, 4-HNE, and 3-NT, in the skin tissues of mice in the IR group were significantly higher than those in the control group (p < 0.05). However, the above pathological changes declined sharply after SFN treatment (p < 0.05). In addition, the expressions of Nrf2 and its regulated antioxidant enzymes, including CAT and HO-1, were higher in the skin tissues of SFN and IR/SFN groups, but lower in the control and IR groups (p < 0.05). SFN may be able to suppress the oxidative stress by upregulating the expression and function of Nrf2, and subsequently inhibiting the activation of NLRP3 inflammasome and DNA damage, so as to prevent and alleviate the RISI.
Collapse
Affiliation(s)
- Jinlong Wei
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Qin Zhao
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Yuyu Zhang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Weiyan Shi
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Huanhuan Wang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Zhuangzhuang Zheng
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
- Correspondence: (Y.X.); ; (X.J.); Tel.: +86-13504310452 (Y.X.); +86-15804302750 (X.J.)
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
- Correspondence: (Y.X.); ; (X.J.); Tel.: +86-13504310452 (Y.X.); +86-15804302750 (X.J.)
| |
Collapse
|
6
|
Haritwal T, Goyal N, Gupta N, Parvez S, Agrawala PK. Trichostatin A mitigates radiation induced teratogenesis in C57 Bl/6 mice. Mutagenesis 2021; 36:303-309. [PMID: 34086940 DOI: 10.1093/mutage/geab018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 06/03/2021] [Indexed: 11/13/2022] Open
Abstract
Radiation exposure in utero is known to lead to serious concerns to both the mother and children, including developmental anomalies in the children. In the recent past trichostatn A, an HDAC inhibitor and epigenetic modifier, has been shown to mitigate radiation induced anomalies in the male reproductive system of C57BL/6 mice. Therefore, the current study was undertaken to evaluate the mitigating effects of trichostatin A against radiation induced developmental anomalies in mice. Fetuses of in utero whole body gamma irradiated mice during active organogenesis period were examined for developmental anomalies on 8.5 and 18.5 day of gestation. In utero radiation exposure caused developmental anomalies like microcephaly, microphthalamia, gastroschisis and kinky tail besides prenatal mortality. Trichostatin A administration post irradiation was observed to reduce 50% of prenatal mortality at E18.5 by reducing congenital and developmental anomalies. Observation of such results could be corroborated with the HDAC inhibitory potential of trichostatin A knowing that developmental anomalies may have epigenetic origin. Trichostatin A therefore can be considered as a potential radiomitigator.
Collapse
Affiliation(s)
- Teena Haritwal
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig SK Mazumdar Road, Timarpur, Delhi 110054, INDIA
| | - Nikita Goyal
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig SK Mazumdar Road, Timarpur, Delhi 110054, INDIA
| | - Noopur Gupta
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig SK Mazumdar Road, Timarpur, Delhi 110054, INDIA
| | - Suhel Parvez
- Department of Medical Entomology and Toxicology, School of Life Sciences, Jamia Hamdard University, Hamdard Nagar, New Delhi 110062, INDIA
| | - Paban K Agrawala
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig SK Mazumdar Road, Timarpur, Delhi 110054, INDIA
| |
Collapse
|
7
|
Katoch O, Tiwari M, Kalra N, Agrawala PK. Mechanism of Action of Diallyl Sulphide in Ameliorating the Hematopoietic Radiation Injury. JOURNAL OF HEALTH AND ALLIED SCIENCES NU 2021. [DOI: 10.1055/s-0041-1730094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractDiallyl sulphide (DAS), the pungent component of garlic, is known to have several medicinal properties and has recently been shown to have radiomitigative properties. The present study was performed to better understand its mode of action in rendering radiomitigation. Evaluation of the colonogenic ability of hematopoietic progenitor cells (HPCs) on methocult media, proliferation and differentiation of hematopoietic stem cells (HSCs), and transplantation of stem cells were performed. The supporting tissue of HSCs was also evaluated by examining the histology of bone marrow and in vitro colony-forming unit–fibroblast (CFU-F) count. Alterations in the levels of IL-5, IL-6 and COX-2 were studied as a function of radiation or DAS treatment. It was observed that an increase in proliferation and differentiation of hematopoietic stem and progenitor cells occurred by postirradiation DAS administration. It also resulted in increased circulating and bone marrow homing of transplanted stem cells. Enhancement in bone marrow cellularity, CFU-F count, and cytokine IL-5 level were also evident. All those actions of DAS that could possibly add to its radiomitigative potential and can be attributed to its HDAC inhibitory properties, as was observed by the reversal radiation induced increase in histone acetylation.
Collapse
Affiliation(s)
- Omika Katoch
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| | - Mrinalini Tiwari
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| | - Namita Kalra
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| | - Paban K. Agrawala
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Timarpur, New Delhi, India
| |
Collapse
|
8
|
Proshkina E, Shaposhnikov M, Moskalev A. Genome-Protecting Compounds as Potential Geroprotectors. Int J Mol Sci 2020; 21:E4484. [PMID: 32599754 PMCID: PMC7350017 DOI: 10.3390/ijms21124484] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
Throughout life, organisms are exposed to various exogenous and endogenous factors that cause DNA damages and somatic mutations provoking genomic instability. At a young age, compensatory mechanisms of genome protection are activated to prevent phenotypic and functional changes. However, the increasing stress and age-related deterioration in the functioning of these mechanisms result in damage accumulation, overcoming the functional threshold. This leads to aging and the development of age-related diseases. There are several ways to counteract these changes: 1) prevention of DNA damage through stimulation of antioxidant and detoxification systems, as well as transition metal chelation; 2) regulation of DNA methylation, chromatin structure, non-coding RNA activity and prevention of nuclear architecture alterations; 3) improving DNA damage response and repair; 4) selective removal of damaged non-functional and senescent cells. In the article, we have reviewed data about the effects of various trace elements, vitamins, polyphenols, terpenes, and other phytochemicals, as well as a number of synthetic pharmacological substances in these ways. Most of the compounds demonstrate the geroprotective potential and increase the lifespan in model organisms. However, their genome-protecting effects are non-selective and often are conditioned by hormesis. Consequently, the development of selective drugs targeting genome protection is an advanced direction.
Collapse
Affiliation(s)
- Ekaterina Proshkina
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (M.S.)
| | - Mikhail Shaposhnikov
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (M.S.)
| | - Alexey Moskalev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (M.S.)
- Pitirim Sorokin Syktyvkar State University, 55 Oktyabrsky prosp., 167001 Syktyvkar, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
9
|
Venkateswaran K, Shrivastava A, Prasad AK, Parmar VS, Dwarakanath BS. Developing polyphenolic acetates as radiation countermeasure agents: current status and future perspectives. Drug Discov Today 2020; 25:781-786. [PMID: 32062010 DOI: 10.1016/j.drudis.2020.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/22/2020] [Accepted: 02/07/2020] [Indexed: 10/25/2022]
Abstract
Total-body exposure to ionizing radiation (TBI) results in life-threatening acute radiation syndrome (ARS), which encompasses hematopoietic and gastrointestinal (GI) injuries and results in dose-dependent morbidity and mortality. Management of ARS warrants the deployment of effective medical countermeasure agents (MCM) that protect against and/or mitigate lethal radiation injury. The polyphenolic acetate (PA) 7,8-diacetoxy-4-methylthiocoumarin (DAMTC) has been identified as a potential MCM against ARS by virtue of it mitigating the lethal effects of TBI in C57BL/6 mice. Herein, we describe current evidence, including mechanistic aspects, for the use of PAs as MCMs against ARS and provide perspectives for their further development as approved drugs for the mitigation of ARS.
Collapse
Affiliation(s)
| | | | - Ashok K Prasad
- Bioorganic Laboratory, Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Virinder S Parmar
- Bioorganic Laboratory, Department of Chemistry, University of Delhi, Delhi 110007, India; Department of Chemistry and Environmental Science, Medgar Evers College, The City University of New York, New York NY, USA
| | | |
Collapse
|
10
|
Negrette-Guzmán M. Combinations of the antioxidants sulforaphane or curcumin and the conventional antineoplastics cisplatin or doxorubicin as prospects for anticancer chemotherapy. Eur J Pharmacol 2019; 859:172513. [PMID: 31260654 DOI: 10.1016/j.ejphar.2019.172513] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/17/2022]
Abstract
Drugs used in clinical oncology have narrow therapeutic indices with adverse toxicity often involving oxidative damage. Chemoresistance to these conventional antineoplastics is usually mediated by oxidative stress-upregulated pathways such as those of nuclear factor-kappa B (NF-κB) and hypoxia-inducible factor-1 alpha (HIF-1α). Accordingly, the use of antioxidants in combinational approaches has begun to be considered for fighting cancer because of both the protective role against adverse effects and the ability to sensitize chemoresistant cancer cells. Nuclear factor erythroid 2-related factor 2 (Nrf2) has been identified as a mediator of the cytoprotection but it is not regularly associated with tumor chemosensitization. However, some Nrf2 inducers could be exerting cytoprotective and chemosensitizing roles through a simple integrated mechanism in which the cellular level of reactive oxygen species is controlled, thus inhibiting the oxidative damage in non-target tissues and the tumor chemoresistance mediated by NF-κB or HIF-1α. As examples to show the general idea of this antioxidant combination chemotherapy, this review explores the preclinical information available for four combinations, each composed by a paradigmatic oncological drug (cisplatin or doxorubicin) and a recognized antioxidant (sulforaphane or curcumin). The issues for translating these outcomes to clinical trials are briefly discussed.
Collapse
Affiliation(s)
- Mario Negrette-Guzmán
- Centro de Investigaciones en Enfermedades Tropicales (CINTROP), Departamento de Ciencias Básicas, Escuela de Medicina, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, 68002, Colombia.
| |
Collapse
|
11
|
Mielczarek L, Krug P, Mazur M, Milczarek M, Chilmonczyk Z, Wiktorska K. In the triple-negative breast cancer MDA-MB-231 cell line, sulforaphane enhances the intracellular accumulation and anticancer action of doxorubicin encapsulated in liposomes. Int J Pharm 2019; 558:311-318. [PMID: 30641176 DOI: 10.1016/j.ijpharm.2019.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/03/2018] [Accepted: 01/02/2019] [Indexed: 01/06/2023]
Abstract
A new combination of sulforaphane (a natural compound obtained from Brassicaceae vegetables) and the cytostatic drug doxorubicin was entrapped in nanometer-sized liposomes. In vitro experiments were performed to investigate the cytotoxicity of these structures on the human breast cancer cell line MDA-MB-231. Confocal microscopy studies revealed enhanced cellular endocytotic internalization, followed by the release of the examined combination from the lysosomes. The in vitro interaction analysis using the Chou-Talalay approach showed high synergistic activity of the examined combination. This synergistic activity enables a considerable reduction in cytostatic dosage and an increase in cancer treatment efficiency.
Collapse
Affiliation(s)
- Lidia Mielczarek
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland; Department of Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Pamela Krug
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland; Department of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Maciej Mazur
- Department of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Małgorzata Milczarek
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland
| | - Zdzisław Chilmonczyk
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland
| | - Katarzyna Wiktorska
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland.
| |
Collapse
|
12
|
Haritwal T, Maan K, Rana P, Parvez S, Singh AK, Khushu S, Agrawala PK. Trichostatin A, an epigenetic modifier, mitigates radiation-induced androphysiological anomalies and metabolite changes in mice as evident from NMR-based metabolomics. Int J Radiat Biol 2018; 95:443-451. [PMID: 30307353 DOI: 10.1080/09553002.2018.1524989] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
PURPOSE Ionizing radiation is known to damage male reproductive system. Current study aims to study the mitigative effects of trichostatin A on male reproductive system and accompanying metabolite changes in testicular tissue of mice. MATERIALS AND METHODS Eight-week-old male C57 Bl/6J mice were exposed to 2 Gy γ-radiation with or without trichostatin A administration. The animals were sacrificed at various time intervals for organ body weight index, sperm head abnormality assay, sperm mobility assay, and study of various metabolites in testicular tissue using NMR spectroscopy. RESULTS Ionizing radiation induced no significant change in organ body weight index at any time points studied, however a significant increase in sperm head abnormality and significant decrease in sperm mobility was evident on fifth postirradiation week. trichostatin A administration, 1 and 24 h postirradiation, could efficiently mitigate radiation-induced changes studied. NMR metabolome profile also showed prominent changes associated with energy metabolism, osmolytes and membrane metabolism at 24 h postirradiation and some of these changes (choline, glycerolphosphoethanol amine, and glycine) were persistent till fifth postirradiation week. Trichostatin A administration resulted in reverting metabolic profile of the irradiated animals to normal level suggesting its mitigative role. CONCLUSION Results obtained suggest that trichostatin A could restore normal metabolic profile of testicular tissue of irradiated male mice and also restored certain morphological and functional properties of sperms. Trichostatin A thus could further be exploited for its radio-mitigative properties.
Collapse
Affiliation(s)
- Teena Haritwal
- a Department of Radiation Genetics and Epigenetics , Institute for Nuclear Medicine and Allied Sciences , Delhi , India
| | - Kiran Maan
- b NMR Research Centre, Institute for Nuclear Medicine and Allied Sciences , Delhi , India
| | - Poonam Rana
- b NMR Research Centre, Institute for Nuclear Medicine and Allied Sciences , Delhi , India
| | - Suhel Parvez
- c Department of Toxicology , Jamia Hamdard University , New Delhi , India
| | - Ajay K Singh
- a Department of Radiation Genetics and Epigenetics , Institute for Nuclear Medicine and Allied Sciences , Delhi , India
| | - Subash Khushu
- b NMR Research Centre, Institute for Nuclear Medicine and Allied Sciences , Delhi , India
| | - Paban K Agrawala
- a Department of Radiation Genetics and Epigenetics , Institute for Nuclear Medicine and Allied Sciences , Delhi , India
| |
Collapse
|
13
|
Alkharashi NAO, Periasamy VS, Athinarayanan J, Alshatwi AA. Assessment of sulforaphane-induced protective mechanisms against cadmium toxicity in human mesenchymal stem cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:10080-10089. [PMID: 29383641 DOI: 10.1007/s11356-018-1228-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/04/2018] [Indexed: 06/07/2023]
Abstract
Cd is a hazardous substance and carcinogen that is present in the environment; it is known to cause toxic effects in living organisms. Sulforaphane is a naturally available phytochemical with antioxidant, anti-inflammatory, and anticarcinogenic properties. However, the effects of sulforaphane on Cd toxicity in human mesenchymal stem cells (hMSCs) are unknown. In the present study, we investigated the molecular mechanisms of the effects of sulforaphane on Cd toxicity in hMSCs by using MTT assays, acridine orange/ethidium bromide staining, Hoechst staining, LysoRed staining, assessment of mitochondrial membrane potential, and gene expression analysis. Cd decreased hMSC viability in a dose-dependent manner with an IC50 value of 56.5 μM. However, sulforaphane did not induce any significant reduction in cell viability. Nuclear morphological analysis revealed that Cd induced necrotic cell death. Additionally, Cd caused mitochondrial membrane potential loss in hMSCs. The treatment of Cd-exposed cells with sulforaphane (Cd-sulforaphane co-treatment) resulted in a significant recovery of the cell viability and nuclear morphological changes compared with that of cells treated with Cd only. The gene expression pattern of cells co-treated with Cd-sulforaphane was markedly different from that of Cd-treated cells, owing to the reduction in Cd toxicity. Our results clearly indicated that sulforaphane reduced Cd-induced toxic effects in hMSCs. Overall, the results of our study suggested that sulforaphane-rich vegetables and fruits can help to improve human health through amelioration of the molecular effects of Cd poisoning.
Collapse
Affiliation(s)
- Nouf Abdulkareem Omer Alkharashi
- Nanobiotechnology and Molecular Biology Research Laboratory, Department of Food Science and Nutrition, College of Food and Agriculture Science, King Saud University, P.O. Box 2460, Riyadh, 11451, Kingdom of Saudi Arabia
- Department of Home Economics, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Vaiyapuri Subbarayan Periasamy
- Nanobiotechnology and Molecular Biology Research Laboratory, Department of Food Science and Nutrition, College of Food and Agriculture Science, King Saud University, P.O. Box 2460, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Jegan Athinarayanan
- Nanobiotechnology and Molecular Biology Research Laboratory, Department of Food Science and Nutrition, College of Food and Agriculture Science, King Saud University, P.O. Box 2460, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Ali A Alshatwi
- Nanobiotechnology and Molecular Biology Research Laboratory, Department of Food Science and Nutrition, College of Food and Agriculture Science, King Saud University, P.O. Box 2460, Riyadh, 11451, Kingdom of Saudi Arabia.
| |
Collapse
|
14
|
Kainthola A, Haritwal T, Tiwari M, Gupta N, Parvez S, Tiwari M, Prakash H, Agrawala PK. Immunological Aspect of Radiation-Induced Pneumonitis, Current Treatment Strategies, and Future Prospects. Front Immunol 2017; 8:506. [PMID: 28512460 PMCID: PMC5411429 DOI: 10.3389/fimmu.2017.00506] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/12/2017] [Indexed: 12/21/2022] Open
Abstract
Delivery of high doses of radiation to thoracic region, particularly with non-small cell lung cancer patients, becomes difficult due to subsequent complications arising in the lungs of the patient. Radiation-induced pneumonitis is an early event evident in most radiation exposed patients observed within 2-4 months of treatment and leading to fibrosis later. Several cytokines and inflammatory molecules interplay in the vicinity of the tissue developing radiation injury leading to pneumonitis and fibrosis. While certain cytokines may be exploited as biomarkers, they also appear to be a potent target of intervention at transcriptional level. Initiation and progression of pneumonitis and fibrosis thus are dynamic processes arising after few months to year after irradiation of the lung tissue. Currently, available treatment strategies are challenged by the major dose limiting complications that curtails success of the treatment as well as well being of the patient's future life. Several approaches have been in practice while many other are still being explored to overcome such complications. The current review gives a brief account of the immunological aspects, existing management practices, and suggests possible futuristic approaches.
Collapse
Affiliation(s)
- Anup Kainthola
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Teena Haritwal
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Mrinialini Tiwari
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Noopur Gupta
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard University, New Delhi, India
| | - Manisha Tiwari
- Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Hrideysh Prakash
- School of Life Sciences, Science complex, University of Hyderabad, Hyderabad, India
| | - Paban K. Agrawala
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| |
Collapse
|
15
|
EGCG, a tea polyphenol, as a potential mitigator of hematopoietic radiation injury in mice. Biomed Pharmacother 2017; 88:203-209. [DOI: 10.1016/j.biopha.2016.12.129] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 12/26/2016] [Accepted: 12/31/2016] [Indexed: 11/20/2022] Open
|
16
|
Tiwari M, Parvez S, Agrawala PK. Role of some epigenetic factors in DNA damage response pathway. AIMS GENETICS 2017; 4:69-83. [PMID: 31435504 PMCID: PMC6690236 DOI: 10.3934/genet.2017.1.69] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/17/2017] [Indexed: 01/01/2023]
Abstract
The current review gives a brief account of the DNA damage response pathway and involvement of various epigenetic mechanisms in DNA damage response pathway. The main focus is on histone modifications leading to structural alterations in chromatin since the compact chromatin structure poses a major limitation in the DNA repair process. Based on this hypothesis, our laboratory has also evaluated certain histone deacetylase inhibitors as potential radiomitigators and the same has been discussed in brief at the end of the review.
Collapse
Affiliation(s)
- Mrinalini Tiwari
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Timarpur, Delhi 110054 India
| | - Suhel Parvez
- Department of Toxicology, Jamia Hamdard University, Hamdard Nagar, Delhi 110062 India
| | - Paban K Agrawala
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Timarpur, Delhi 110054 India
| |
Collapse
|
17
|
Kumar A, Selvan TG, Tripathi AM, Choudhary S, Khan S, Adhikari JS, Chaudhury NK. Sesamol attenuates genotoxicity in bone marrow cells of whole-body γ-irradiated mice. Mutagenesis 2015; 30:651-61. [PMID: 25863274 DOI: 10.1093/mutage/gev026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
18
|
Shelke S, Das B. Dose response and adaptive response of non-homologous end joining repair genes and proteins in resting human peripheral blood mononuclear cells exposed to γ radiation. Mutagenesis 2014; 30:365-79. [PMID: 25473122 DOI: 10.1093/mutage/geu081] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ionising radiation induces single-strand breaks, double-strand breaks (DSB) and base damages in human cell. DSBs are the most deleterious and if not repaired may lead to genomic instability and cell death. DSB can be repaired through non-homologous end joining (NHEJ) pathway in resting lymphocytes. In this study, NHEJ genes and proteins were studied in irradiated human peripheral blood mononuclear cells (PBMC) at resting stage. Dose-response, time point kinetics and adaptive-response studies were conducted in irradiated PBMC at various end points such as DNA damage quantitation, transcription and protein expression profile. Venous blood samples were collected from 20 random, normal and healthy donors with written informed consent. PBMC was separated and irradiated with various doses between 0.1 and 2.0 Gy ((60)CO-γ source) for dose-response study. Repair kinetics of DNA damage and time point changes in expression of genes and proteins were studied in post-irradiated PBMC at 2.0 Gy at various time points up to 240 min. Adaptive-response study was conducted with a priming dose of 0.1 Gy followed by a challenging dose of 2.0 Gy after 4-h incubation. Our results revealed that Ku70, Ku80, XLF and Ligase IV were significantly upregulated (P < 0.05) at 4-h post-irradiation at transcript and protein level. Adaptive-response study showed significantly increased expression of the proteins involved in NHEJ, suggesting their role in adaptive response in human PBMC at G0/G1, which has important implications to human health.
Collapse
Affiliation(s)
- Shridevi Shelke
- Low Level Radiation Research Section, Radiation Biology and Health Sciences Division, Bio-Sciences Group, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| | - Birajalaxmi Das
- Low Level Radiation Research Section, Radiation Biology and Health Sciences Division, Bio-Sciences Group, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| |
Collapse
|
19
|
Lin H, Wei B, Li G, Zheng J, Sun J, Chu J, Zeng R, Niu Y. Sulforaphane reverses glucocorticoid-induced apoptosis in osteoblastic cells through regulation of the Nrf2 pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:973-82. [PMID: 25071366 PMCID: PMC4111650 DOI: 10.2147/dddt.s65410] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Apoptosis of osteoblasts triggered by high-dose glucocorticoids (GCs) has been identified as a major cause of osteoporosis. However, the underlying molecular mechanisms accounting for this action remain elusive, which has impeded the prevention and cure of this side effect. Sulforaphane (SFP) is a naturally occurring isothiocyanate that has huge health benefits for humans. In this study, by using osteoblastic MC3T3-E1 cells as a model, we demonstrate the protective effects of SFP against dexamethasone (Dex)-induced apoptosis and elucidate the underlying molecular mechanisms. The results show that SFP could effectively inhibit the Dex-induced growth inhibition and release of lactate dehydrogenase in MC3T3-E1 cells. Treatment with Dex induced caspase-dependent apoptosis in MC3T3-E1 cells, as evidenced by an increase in the Sub-G1 phase, chromatin condensation, and deoxyribonucleic acid fragmentation, which were significantly suppressed by coincubation with SFP. Mitochondria-mediated apoptosis pathway contributed importantly to Dex-induced apoptosis, as revealed by the activation of caspase-3/-9 and subsequent cleavage of poly adenosine diphosphate ribose polymerase, which was also effectively blocked by SFP. Moreover, treatments of Dex strongly induced overproduction of reactive oxygen species and inhibited the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and the downstream effectors HO1 and NQO1. However, cotreatment with SFP effectively reversed this action of Dex. Furthermore, silencing of Nrf2 by small interfering ribonucleic acid significantly blocked the cytoprotective effects of SFP against Dex-induced apoptosis, which suggest the important role of Nrf2 signaling pathway and cell apoptosis induced by Dex. Taken together, this study provides a novel strategy for molecular intervention against Dex-induced osteoporosis using phytochemicals.
Collapse
Affiliation(s)
- Hao Lin
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, People's Republic of China
| | - Bo Wei
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, People's Republic of China
| | - Guangsheng Li
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, People's Republic of China
| | - Jinchang Zheng
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, People's Republic of China
| | - Jiecong Sun
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, People's Republic of China
| | - Jiaqi Chu
- Laboratory Institute of Minimally Invasive Orthopedic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, People's Republic of China
| | - Rong Zeng
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, People's Republic of China
| | - Yanru Niu
- Laboratory Institute of Minimally Invasive Orthopedic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, People's Republic of China
| |
Collapse
|