1
|
Wang Y, Li C, Li Z, Moalin M, den Hartog GJM, Zhang M. Computational Chemistry Strategies to Investigate the Antioxidant Activity of Flavonoids-An Overview. Molecules 2024; 29:2627. [PMID: 38893503 PMCID: PMC11173571 DOI: 10.3390/molecules29112627] [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: 04/28/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Despite several decades of research, the beneficial effect of flavonoids on health is still enigmatic. Here, we focus on the antioxidant effect of flavonoids, which is elementary to their biological activity. A relatively new strategy for obtaining a more accurate understanding of this effect is to leverage computational chemistry. This review systematically presents various computational chemistry indicators employed over the past five years to investigate the antioxidant activity of flavonoids. We categorize these strategies into five aspects: electronic structure analysis, thermodynamic analysis, kinetic analysis, interaction analysis, and bioavailability analysis. The principles, characteristics, and limitations of these methods are discussed, along with current trends.
Collapse
Affiliation(s)
- Yue Wang
- Department of Pharmacology and Personalized Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (Y.W.); (C.L.); (G.J.M.d.H.)
| | - Chujie Li
- Department of Pharmacology and Personalized Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (Y.W.); (C.L.); (G.J.M.d.H.)
| | - Zhengwen Li
- School of Pharmacy, Chengdu University, 2025 Chengluo Avenue, Chengdu 610106, China;
| | - Mohamed Moalin
- Research Centre Material Sciences, Zuyd University of Applied Science, 6400 AN Heerlen, The Netherlands;
| | - Gertjan J. M. den Hartog
- Department of Pharmacology and Personalized Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (Y.W.); (C.L.); (G.J.M.d.H.)
| | - Ming Zhang
- Hainan University-HSF/LWL Collaborative Innovation Laboratory, College of Food Sciences & Engineering, Hainan University, 58 People Road, Haikou 570228, China
| |
Collapse
|
2
|
Zhang Z, Huang C, Guan S, Wang L, Yin H, Yin J, Liu J, Wu J. Hybrid gelatin-ascorbyl phosphate scaffolds accelerate diabetic wound healing via ROS scavenging, angiogenesis and collagen remodeling. BIOMATERIALS ADVANCES 2024; 158:213779. [PMID: 38277902 DOI: 10.1016/j.bioadv.2024.213779] [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: 10/05/2023] [Revised: 12/26/2023] [Accepted: 01/17/2024] [Indexed: 01/28/2024]
Abstract
Skin wound healing, particularly diabetic wound healing, is challenging in clinical management. Impaired wound healing is associated with persistent oxidative stress, altered inflammatory responses, unsatisfactory angiogenesis and epithelialization. Magnesium ascorbyl phosphate (MAP), which is an ascorbic acid derivative and active ingredient in cosmetics, has been reported to scavenge reactive oxygen species (ROS), and is considered a potential therapeutic agent for diabetic wounds. Herein, we report a hybrid gelatin-MAP scaffolds that can reduces oxidative stress damage, enhances angiogenesis and collagen remodeling to accelerate diabetic wound repair. Preliminary insights based on network pharmacology indicate that MAP may accelerate wound repair through multiple biological pathways, including extracellular matrix remodeling and anti-apoptosis. In vitro studies showed that the hybrid hydrogel scaffold had suitable mechanical properties, excellent biocompatibility and bioactivity. Further animal experiments demonstrated that the hydrogel accelerated full-thickness wound repair in diabetic mice (repair rate MAP vs Control=91.791±3.306 % vs 62.962±6.758 %) through antioxidant, neuroangiogenesis, collagen remodeling, and up-regulated the expression of the related factors COL-1, CD31, VEGF, and CGRP. Overall, we developed a bioactive hybrid hydrogel encapsulating MAP that synergistically promotes diabetic wound repair through multiple biological effects. This potentially integrated therapeutic scaffold may enrich future surgical approaches for treating diabetic wounds.
Collapse
Affiliation(s)
- Zhen Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Chunlin Huang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Shiyao Guan
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
| | - Liying Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Hanxiao Yin
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Junqiang Yin
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China.
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Jun Wu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China; Division of Life Science, The Hong Kong University of Science and Technology, 999077, Hong Kong.
| |
Collapse
|
3
|
Kanner J. Food Polyphenols as Preventive Medicine. Antioxidants (Basel) 2023; 12:2103. [PMID: 38136222 PMCID: PMC10740609 DOI: 10.3390/antiox12122103] [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: 10/29/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Reactive oxygen species (ROS) are the initiators in foods and in the stomach of oxidized dietary lipids, proteins, and lipid-oxidation end-products (ALEs), inducing in humans the development of several chronic diseases and cancer. Epidemiological, human clinical and animal studies supported the role of dietary polyphenols and derivatives in prevention of development of such chronic diseases. There is much evidence that polyphenols/derivatives at the right timing and concentration, which is critical, acts mostly in the aerobic stomach and generally in the gastrointestinal tract as reducing agents, scavengers of free radicals, trappers of reactive carbonyls, modulators of enzyme activity, generators of beneficial gut microbiota and effectors of cellular signaling. In the blood system, at low concentration, they act as generators of electrophiles and low concentration of H2O2, acting mostly as cellular signaling, activating the PI3K/Akt-mediated Nrf2/eNOS pathways and inhibiting the inflammatory transcription factor NF-κB, inducing the cells, organs and organism for eustress, adaptation and surviving.
Collapse
Affiliation(s)
- Joseph Kanner
- Department of Food Science, ARO, Volcani Center, Bet-Dagan 7505101, Israel; or
- Institute of Biochemistry, Food Science and Nutrtion, Faculty of Agriculture Food and Environment, The Hebrew University of Jerusalem, Rehovot 9190501, Israel
| |
Collapse
|
4
|
Wang Q, Lin D, Liu XF, Dai F, Jin XJ, Zhou B. Engineering piperlongumine-inspired analogs as Nrf2-dependent neuroprotectors against oxidative damage by an electrophilicity-based strategy. Free Radic Biol Med 2023; 194:298-307. [PMID: 36528122 DOI: 10.1016/j.freeradbiomed.2022.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Oxidative stress contributes significantly to the development of neurodegenerative diseases, thus developing nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent neuroprotectors is highly required for either prevention or treatment of these diseases. This work highlights an electrophilicity-based strategy that allows finding more active Nrf2-dependent neuroprotectors than natural piperlongumine (PL). Electrophilic modification was applied on both the exocylic and endocyclic Michael acceptors of PL, which includes placement of an electron-withdrawing trifluoromethyl group on its aromatic ring in the ortho, meta, or para position to the exocyclic olefin, and further introduction of an electron-withdrawing α-chlorine on its lactam ring. From a panel of PL analogs, we identified PLCl-4CF3, characterized by the presence of p-trifluoromethyl group and α-chlorine, to be significantly superior to the parent PL in protecting PC12 cells from oxidative damage induced by 6-hydroxydopamine hydrochloride. Mechanistic studies reveal that the increased electrophilicity of PLCl-4CF3 in its two Michael acceptors allows its ability to covalently modify Cys-151 at Keap1, facilitating inhibition against Nrf2 ubiquitination, translocation of Nrf2 into the nucleus, induction of phase 2 enzymes and final protection of PC12 cells from oxidative damage.
Collapse
Affiliation(s)
- Qi Wang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Dong Lin
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Xue-Feng Liu
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu, 730000, China; Gansu University Key Laboratory for Molecular Medicine and Chinese Medicine Prevention and Treatment of Major Diseases, College of Pharmacy, Gansu University of Chinese Medicine, 35 Dingxi East Road, Gansu, 730000, China
| | - Fang Dai
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu, 730000, China.
| | - Xiao-Jie Jin
- Gansu University Key Laboratory for Molecular Medicine and Chinese Medicine Prevention and Treatment of Major Diseases, College of Pharmacy, Gansu University of Chinese Medicine, 35 Dingxi East Road, Gansu, 730000, China.
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
5
|
Molecular and Cellular Interactions in Pathogenesis of Sporadic Parkinson Disease. Int J Mol Sci 2022; 23:ijms232113043. [PMID: 36361826 PMCID: PMC9657547 DOI: 10.3390/ijms232113043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/16/2022] [Accepted: 10/25/2022] [Indexed: 11/23/2022] Open
Abstract
An increasing number of the population all around the world suffer from age-associated neurodegenerative diseases including Parkinson’s disease (PD). This disorder presents different signs of genetic, epigenetic and environmental origin, and molecular, cellular and intracellular dysfunction. At the molecular level, α-synuclein (αSyn) was identified as the principal molecule constituting the Lewy bodies (LB). The gut microbiota participates in the pathogenesis of PD and may contribute to the loss of dopaminergic neurons through mitochondrial dysfunction. The most important pathogenetic link is an imbalance of Ca2+ ions, which is associated with redox imbalance in the cells and increased generation of reactive oxygen species (ROS). In this review, genetic, epigenetic and environmental factors that cause these disorders and their cause-and-effect relationships are considered. As a constituent of environmental factors, the example of organophosphates (OPs) is also reviewed. The role of endothelial damage in the pathogenesis of PD is discussed, and a ‘triple hit hypothesis’ is proposed as a modification of Braak’s dual hit one. In the absence of effective therapies for neurodegenerative diseases, more and more evidence is emerging about the positive impact of nutritional structure and healthy lifestyle on the state of blood vessels and the risk of developing these diseases.
Collapse
|
6
|
Famurewa AC, Mukherjee AG, Wanjari UR, Sukumar A, Murali R, Renu K, Vellingiri B, Dey A, Valsala Gopalakrishnan A. Repurposing FDA-approved drugs against the toxicity of platinum-based anticancer drugs. Life Sci 2022; 305:120789. [PMID: 35817170 DOI: 10.1016/j.lfs.2022.120789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022]
Abstract
Platinum-based anticancer drugs (PADs), mainly cisplatin, carboplatin, and oxaliplatin, are widely used efficacious long-standing anticancer agents for treating several cancer types. However, clinicians worry about PAD chemotherapy and its induction of severe non-targeted organ toxicity. Compelling evidence has shown that toxicity of PAD on delicate body organs is associated with free radical generation, DNA impairment, endocrine and mitochondrial dysfunctions, oxidative inflammation, apoptosis, endoplasmic reticulum stress, and activation of regulator signaling proteins, cell cycle arrest, apoptosis, and pathways. The emerging trend is the repurposing of FDA-approved non-anticancer drugs (FNDs) for combating the side effects toxicity of PADs. Thus, this review chronicled the mechanistic preventive and therapeutic effects of FNDs against PAD organ toxicity in preclinical studies. FNDs are potential clinical drugs for the modulation of toxicity complications associated with PAD chemotherapy. Therefore, FNDs may be suggested as non-natural agent inhibitors of unpalatable side effects of PADs.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike lkwo, Nigeria.
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Aarthi Sukumar
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal 700073, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
7
|
Braveboy-Wagner J, Sharoni Y, Lelkes PI. Nutraceuticals Synergistically Promote Osteogenesis in Cultured 7F2 Osteoblasts and Mitigate Inhibition of Differentiation and Maturation in Simulated Microgravity. Int J Mol Sci 2021; 23:136. [PMID: 35008559 PMCID: PMC8745420 DOI: 10.3390/ijms23010136] [Citation(s) in RCA: 4] [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: 12/02/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 02/08/2023] Open
Abstract
Microgravity is known to impact bone health, similar to mechanical unloading on Earth. In the absence of countermeasures, bone formation and mineral deposition are strongly inhibited in Space. There is an unmet need to identify nutritional countermeasures. Curcumin and carnosic acid are phytonutrients with anticancer, anti-inflammatory, and antioxidative effects and may exhibit osteogenic properties. Zinc is a trace element essential for bone formation. We hypothesized that these nutraceuticals could counteract the microgravity-induced inhibition of osteogenic differentiation and function. To test this hypothesis, we cultured 7F2 murine osteoblasts in simulated microgravity (SMG) in a Random Positioning Machine in the presence and absence of curcumin, carnosic acid, and zinc and evaluated cell proliferation, function, and differentiation. SMG enhanced cell proliferation in osteogenic medium. The nutraceuticals partially reversed the inhibitory effects of SMG on alkaline phosphatase (ALP) activity and did not alter the SMG-induced reduction in the expression of osteogenic marker genes in osteogenic medium, while they promoted osteoblast proliferation and ALP activity in the absence of traditional osteogenic media. We further observed a synergistic effect of the intermix of the phytonutrients on ALP activity. Intermixes of phytonutrients may serve as convenient and effective nutritional countermeasures against bone loss in space.
Collapse
Affiliation(s)
- Justin Braveboy-Wagner
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA;
| | - Yoav Sharoni
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel;
| | - Peter I. Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA;
| |
Collapse
|
8
|
Yadav AM, Bagade MM, Ghumnani S, Raman S, Saha B, Kubatzky KF, Ashma R. The phytochemical plumbagin reciprocally modulates osteoblasts and osteoclasts. Biol Chem 2021; 403:211-229. [PMID: 34882360 DOI: 10.1515/hsz-2021-0290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/08/2021] [Indexed: 12/28/2022]
Abstract
Bone metabolism is essential for maintaining bone mineral density and bone strength through a balance between bone formation and bone resorption. Bone formation is associated with osteoblast activity whereas bone resorption is linked to osteoclast differentiation. Osteoblast progenitors give rise to the formation of mature osteoblasts whereas monocytes are the precursors for multi-nucleated osteoclasts. Chronic inflammation, auto-inflammation, hormonal changes or adiposity have the potential to disturb the balance between bone formation and bone loss. Several plant-derived components are described to modulate bone metabolism and alleviate osteoporosis by enhancing bone formation and inhibiting bone resorption. The plant-derived naphthoquinone plumbagin is a bioactive compound that can be isolated from the roots of the Plumbago genus. It has been used as traditional medicine for treating infectious diseases, rheumatoid arthritis and dermatological diseases. Reportedly, plumbagin exerts its biological activities primarily through induction of reactive oxygen species and triggers osteoblast-mediated bone formation. It is plausible that plumbagin's reciprocal actions - inhibiting or inducing death in osteoclasts but promoting survival or growth of osteoblasts - are a function of the synergy with bone-metabolizing hormones calcitonin, Parathormone and vitamin D. Herein, we develop a framework for plausible molecular modus operandi of plumbagin in bone metabolism.
Collapse
Affiliation(s)
- Avinash M Yadav
- Department of Zoology, Savitribai Phule Pune University, Pune 411007, Maharashtra, India
| | - Manali M Bagade
- Department of Zoology, Savitribai Phule Pune University, Pune 411007, Maharashtra, India
| | - Soni Ghumnani
- Department of Zoology, Savitribai Phule Pune University, Pune 411007, Maharashtra, India
| | - Sujatha Raman
- Center for Complementary and Integrative Health (CCIH), Interdisciplinary School of Health Sciences (ISHS), Savitribai Phule Pune University, Pune 411007, Maharashtra, India
| | - Bhaskar Saha
- National Center for Cell Science, Pune-411007, Maharashtra, India
| | - Katharina F Kubatzky
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, D-69120 Heidelberg, Germany
| | - Richa Ashma
- Department of Zoology, Savitribai Phule Pune University, Pune 411007, Maharashtra, India
| |
Collapse
|
9
|
Lu Q, Zhang Y, Zhao C, Zhang H, Pu Y, Yin L. Copper induces oxidative stress and apoptosis of hippocampal neuron via pCREB/BDNF/ and Nrf2/HO-1/NQO1 pathway. J Appl Toxicol 2021; 42:694-705. [PMID: 34676557 DOI: 10.1002/jat.4252] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 01/17/2023]
Abstract
Disordered copper metabolism has been suggested to occur to several neurological conditions, including Alzheimer's disease and Parkinson's disease. However, the underlying mechanism was still unclear. This might link to copper-induced hippocampal neuronal apoptosis and decrease in neurons viability. Our vitro experiment showed copper exposure induced oxidative stress and promoted apoptosis of HT22 murine hippocampal neuronal cell. Mechanistically, we found copper, on the one hand, prevented phosphorylation of cAMP response element binding protein (CREB) to decrease expression its downstream target protein Brain-derived neurotrophic factor (BDNF), and to decrease mitochondrial membrane potential and Bcl-2/Bax ratio; on the other hand, copper-induced reactive oxygen species (ROS), promoted lipid peroxidation, reduced antioxidant enzyme activity of GSH-Px. Copper-induced oxidative damage further decreased the phosphorylation of CREB, decreased expression of Bcl-2, enhanced expression of Bax, and accelerated the dissociation of keap1-Nrf2 complex, promoted the nuclear translocation of Nrf2, stimulate the expression of antioxidant molecules HO-1 and NQO1. In conclusion, we found copper inhibited pCREB/BDNF signaling pathway by prevent CREB from phosphorylation, further found that oxidative damage not only inhibited neuroprotective signaling pathways and induced apoptosis, but activated antioxidant protection signals Nrf2/HO-1/NQO1 signaling pathway.
Collapse
Affiliation(s)
- Qiang Lu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Ying Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Chao Zhao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Hu Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
10
|
Sadovnikova IS, Gureev AP, Ignatyeva DA, Gryaznova MV, Chernyshova EV, Krutskikh EP, Novikova AG, Popov VN. Nrf2/ARE Activators Improve Memory in Aged Mice via Maintaining of Mitochondrial Quality Control of Brain and the Modulation of Gut Microbiome. Pharmaceuticals (Basel) 2021; 14:607. [PMID: 34201885 PMCID: PMC8308546 DOI: 10.3390/ph14070607] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 01/01/2023] Open
Abstract
Aging is one of the most serious factors for central nervous dysfunctions, which lead to cognitive impairment. New highly effective drugs are required to slow the development of cognitive dysfunction. This research studied the effect of dimethyl fumarate (DMF), methylene blue (MB), and resveratrol (RSV) on the cognitive functions of 15-month-old mice and their relationship to the maintenance of mitochondrial quality control in the brain and the bacterial composition of the gut microbiome. We have shown that studied compounds enhance mitochondrial biogenesis, mitophagy, and antioxidant defense in the hippocampus of 15-month-old mice via Nrf2/ARE pathway activation, which reduces the degree of oxidative damage to mtDNA. It is manifested in the improvement of short-term and long-term memory. We have also shown that memory improvement correlates with levels of Roseburia, Oscillibacter, ChristensenellaceaeR-7, Negativibacillus, and Faecalibaculum genera of bacteria. At the same time, long-term treatment by MB induced a decrease in gut microbiome diversity, but the other markers of dysbiosis were not observed. Thus, Nrf2/ARE activators have an impact on mitochondrial quality control and are associated with a positive change in the composition of the gut microbiome, which together lead to an improvement in memory in aged mice.
Collapse
Affiliation(s)
- Irina S. Sadovnikova
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (I.S.S.); (D.A.I.); (M.V.G.); (E.V.C.); (E.P.K.); (A.G.N.); (V.N.P.)
| | - Artem P. Gureev
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (I.S.S.); (D.A.I.); (M.V.G.); (E.V.C.); (E.P.K.); (A.G.N.); (V.N.P.)
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technology, 394036 Voronezh, Russia
| | - Daria A. Ignatyeva
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (I.S.S.); (D.A.I.); (M.V.G.); (E.V.C.); (E.P.K.); (A.G.N.); (V.N.P.)
| | - Maria V. Gryaznova
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (I.S.S.); (D.A.I.); (M.V.G.); (E.V.C.); (E.P.K.); (A.G.N.); (V.N.P.)
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technology, 394036 Voronezh, Russia
| | - Ekaterina V. Chernyshova
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (I.S.S.); (D.A.I.); (M.V.G.); (E.V.C.); (E.P.K.); (A.G.N.); (V.N.P.)
| | - Ekaterina P. Krutskikh
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (I.S.S.); (D.A.I.); (M.V.G.); (E.V.C.); (E.P.K.); (A.G.N.); (V.N.P.)
| | - Anastasia G. Novikova
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (I.S.S.); (D.A.I.); (M.V.G.); (E.V.C.); (E.P.K.); (A.G.N.); (V.N.P.)
| | - Vasily N. Popov
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia; (I.S.S.); (D.A.I.); (M.V.G.); (E.V.C.); (E.P.K.); (A.G.N.); (V.N.P.)
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technology, 394036 Voronezh, Russia
| |
Collapse
|
11
|
Aliahmadi M, Amiri F, Bahrami LS, Hosseini AF, Abiri B, Vafa M. Effects of raw red beetroot consumption on metabolic markers and cognitive function in type 2 diabetes patients. J Diabetes Metab Disord 2021; 20:673-682. [PMID: 34222085 PMCID: PMC8212206 DOI: 10.1007/s40200-021-00798-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/08/2021] [Indexed: 12/30/2022]
Abstract
OBJECTIVES This study aimed to investigate the effects of raw red beetroot consumption on metabolic markers and cognitive function in type 2 diabetes patients. METHODS In a quasi-experimental study, 44 type 2 diabetes patients (57 ± 4.5 years) consumed raw red beetroot (100 g, daily), for 8 weeks. Metabolic markers including body weight, glucose and lipid profile parameters, inflammatory and oxidative stress markers, paraoxonase-1 activity, hepatic enzymes, blood pressure and cognitive function were measured at the beginning and end of 8 weeks. RESULTS Raw red beetroot consumption resulted in a significant decrease in fasting blood sugar (FBS) levels (-13.53 mg/dL), glycosylated hemoglobin (HbA1c)(-0.34%), apolipoproteinB100 (ApoB100) (-8.25 mg/dl), aspartate aminotransferase (AST) (-1.75 U/L), alanine aminotransferase (ALT) (-3.7 U/L), homocysteine (-7.88 μmol/l), systolic (-0.73 mmHg) and diastolic blood pressure (-0.34 mmHg), anda significant increase in total antioxidant capacity (TAC) (105 μmol/L) and cognitive function tests (all P values <0.05). Other variables did not change significantly after the intervention. CONCLUSIONS Raw red beetroot consumption for 8 weeks in T2DM patients has beneficial impacts on cognitive function, glucose metabolism and other metabolic markers.
Collapse
Affiliation(s)
- Mitra Aliahmadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemehsadat Amiri
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Sadat Bahrami
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Agha Fatemeh Hosseini
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Behnaz Abiri
- Department of Nutrition, Faculty of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammadreza Vafa
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Sharma S, Advani D, Das A, Malhotra N, Khosla A, Arora V, Jha A, Yadav M, Ambasta RK, Kumar P. Pharmacological intervention in oxidative stress as a therapeutic target in neurological disorders. J Pharm Pharmacol 2021; 74:461-484. [PMID: 34050648 DOI: 10.1093/jpp/rgab064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/01/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Oxidative stress is a major cellular burden that triggers reactive oxygen species (ROS) and antioxidants that modulate signalling mechanisms. Byproducts generated from this process govern the brain pathology and functions in various neurological diseases. As oxidative stress remains the key therapeutic target in neurological disease, it is necessary to explore the multiple routes that can significantly repair the damage caused due to ROS and consequently, neurodegenerative disorders (NDDs). Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase is the critical player of oxidative stress that can also be used as a therapeutic target to combat NDDs. KEY FINDINGS Several antioxidants signalling pathways are found to be associated with oxidative stress and show a protective effect against stressors by increasing the release of various cytoprotective enzymes and also exert anti-inflammatory response against this oxidative damage. These pathways along with antioxidants and reactive species can be the defined targets to eliminate or reduce the harmful effects of neurological diseases. SUMMARY Herein, we discussed the underlying mechanism and crucial role of antioxidants in therapeutics together with natural compounds as a pharmacological tool to combat the cellular deformities cascades caused due to oxidative stress.
Collapse
Affiliation(s)
- Sudhanshu Sharma
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Ankita Das
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Nishtha Malhotra
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Akanksha Khosla
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Vanshika Arora
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Ankita Jha
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Megha Yadav
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| |
Collapse
|
13
|
Feng W, Yang X, Feng M, Pan H, Liu J, Hu Y, Wang S, Zhang D, Ma F, Mao Y. Alginate Oligosaccharide Prevents against D-galactose-mediated Cataract in C57BL/6J Mice via Regulating Oxidative Stress and Antioxidant System. Curr Eye Res 2020; 46:802-810. [PMID: 33153341 DOI: 10.1080/02713683.2020.1842456] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Alginate oligosaccharides (AOS), obtained from depolymerizing alginate, has multiple pharmacological benefits. Cataract is a common disease caused by turbidity of the lens protein due to lens metabolism disorders. This study aimed to test the effects and the underlying mechanisms of AOS on D-galactose (D-gal)-mediated cataract. MATERIALS AND METHODS A total of 45 8-week-old C57BL/6 J male mice were randomly divided into 5 groups. After eight weeks' intervention, the score of cataract was calculated depending on the turbidity of the lens. Hematoxylin and eosin (HE) and transmission electron microscope (TEM) images were observed. Superoxide dismutase (SOD) activity and malondialdehyde (MDA) level were measured by corresponding detection kits, respectively. SOD1, SOD2, catalase (CAT) and p53 protein expressions were examined by Western blot. Nuclear factor erythroid-2 related factor (Nrf2) and heme oxygenase-1 (HO-1) mRNA expressions were examined by Quantitative Real Time-PCR (RT-qPCR). RESULTS The score of the turbidity of the lens showed that AOS significantly delayed the cataractogenesis. HE staining and TEM imaging showed that AOS decreased the damage and senescence of lenses in D-gal-induced C57BL/6 J mice. We further detected aging marker p53 expression in crystalline lenses, and our result showed that AOS significantly inhibited p53 protein expression in D-gal-induced mice. In addition, SOD activity and MDA level detection results showed that AOS significantly increased the activity of SOD, and decreased the level of MDA in crystalline lenses homogenates of D-gal-induced aging mice. Western blot results showed that AOS attenuated the damage of D-gal in the protein expressions of antioxidative enzymes SOD1, SOD2 and CAT. RT-qPCR results showed that AOS suppressed the down-regulation of Nrf2 and HO-1 mRNA expressions induced by D-gal. CONCLUSIONS AOS prevents against D-gal-mediated cataract in C57BL/6 J mice via inhibiting oxidative stress and up-regulating antioxidant system. Consequently, our results suggest that AOS may be an effective therapeutic strategy against cataract.
Collapse
Affiliation(s)
- Wenjing Feng
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, Qingdao, Shandong, China
| | - Xuejiao Yang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Meiping Feng
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Pan
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianya Liu
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yi Hu
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shan Wang
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, Qingdao, Shandong, China
| | - Fenghua Ma
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yongjun Mao
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
14
|
Polyphenols by Generating H 2O 2, Affect Cell Redox Signaling, Inhibit PTPs and Activate Nrf2 Axis for Adaptation and Cell Surviving: In Vitro, In Vivo and Human Health. Antioxidants (Basel) 2020; 9:antiox9090797. [PMID: 32867057 PMCID: PMC7555200 DOI: 10.3390/antiox9090797] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/29/2022] Open
Abstract
Human health benefits from different polyphenols molecules consumption in the diet, derived mainly by their common activities in the gastrointestinal tract and at the level of blood micro-capillary. In the stomach, intestine and colon, polyphenols act as reducing agents preventing lipid peroxidation, generation and absorption of AGEs/ALEs (advanced glycation end products/advanced lipid oxidation end products) and postprandial oxidative stress. The low absorption of polyphenols in blood does not support their activity as antioxidants and their mechanism of activity is not fully understood. The results are from in vitro, animal and human studies, detected by relevant oxidative stress markers. The review carries evidences that polyphenols, by generating H2O2 at nM concentration, exogenous to cells and organs, act as activators of signaling factors increasing cell Eustress. When polyphenols attain high concentration in the blood system, they generate H2O2 at µM concentration, acting as cytotoxic agents and Distress. Pre-treatment of cells or organisms with polyphenols, by generating H2O2 at low levels, inhibits cellular PTPs (protein tyrosine phosphatases), inducing cell signaling through transcription of the Nrf2 (nuclear factor erythroid 2-related factor 2) axis of adaptation and protection to oxidation stress. Polyphenols ingestion at the right amount and time during the meal acts synergistically at the level of the gastrointestinal tract (GIT) and blood system, for keeping the redox homeostasis in our organism and better balancing human health.
Collapse
|
15
|
Computational repositioning of dimethyl fumarate for treating alcoholic liver disease. Cell Death Dis 2020; 11:641. [PMID: 32811823 PMCID: PMC7434920 DOI: 10.1038/s41419-020-02890-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022]
Abstract
Alcoholic liver disease (ALD) is a chronic alcohol-induced disorder of the liver for which there are few effective therapies for severe forms of ALD and for those who do not achieve alcohol abstinence. In this study, we used a systematic drug-repositioning bioinformatics approach querying a large compendium of gene-expression profiles to identify candidate U.S. Food and Drug Administration (FDA)–approved drugs to treat ALD. One of the top compounds predicted to be therapeutic for ALD by our approach was dimethyl fumarate (DMF), an nuclear factor erythroid 2-related factor 2 (NRF2) inducer. We experimentally validated DMF in liver cells and in vivo. Our work demonstrates that DMF is able to significantly upregulate the NRF2 protein level, increase NRF2 phosphorylation, and promote NRF2 nuclear localization in liver cells. DMF also reduced the reactive oxygen species (ROS) level, lipid peroxidation, and ferroptosis. Furthermore, DMF treatment could prevent ethanol-induced liver injury in ALD mice. Our results provide evidence that DMF might serve as a therapeutic option for ALD in humans, and support the use of computational repositioning to discover therapeutic options for ALD.
Collapse
|
16
|
Škandík M, Mrvová N, Bezek Š, Račková L. Semisynthetic quercetin-quinone mitigates BV-2 microglia activation through modulation of Nrf2 pathway. Free Radic Biol Med 2020; 152:18-32. [PMID: 32142880 DOI: 10.1016/j.freeradbiomed.2020.02.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/12/2022]
Abstract
During brain ageing, microglia, the resident immune cells of the CNS, are immunologically activated and contribute to neuroinflammation, a vicious cycle that supports development of neurological disorders. Therapeutic approaches focus mainly on downregulation of their pro-inflammatory activated state that is associated with health benefits. Electrophilic compounds, such as natural quinones and their reduced pro-electrophilic precursors, flavonoids, represent a wide group of diverse substances with important biological effects. They can cause considerable cytotoxicity when used at higher dosages, but on the other hand, they have versatile health benefits at lower dosages. In this study, we investigated the cytotoxicity and prooxidant profile of synthetic conjugate of two electrophilic compounds, quercetin and 1,4-naphthoquinone, 4'-O-(2-chloro-1,4-naphthoquinone-3-yloxy) quercetin (CHNQ), and its attenuation of inflammatory responses and modulation of Nrf2 pathway in BV-2 microglial cells. CHNQ showed higher cytotoxicity than its precursors, accompanied by promotion of production of reactive oxygen species along with G2/M cell cycle arrest at higher concentrations tested. Nevertheless, at a lower non-toxic concentration, CHNQ, more significantly than did its precursors, downregulated LPS-stimulated microglia cells as documented by decreased iNOS, COX-2 and TNFα protein levels. Moreover, CHNQ most effectively upregulated expression of phase II antioxidant enzyme HO-1 and β5 subunit of constitutive proteasome. The enhanced anti-inflammatory effect of CHNQ was accompanied by prominent increase in cytosolic expression of Nrf2 and c-Jun, however, induction effect on nuclear Nrf2 translocation was comparable to QUER. Moreover, a conditioned medium from activated BV-2 cells co-treated with quercetin and CHNQ maintained viability of neuron-like PC12 cells. The compounds tested did not show any disturbance of phagocytosis of live or dead PC12 cells. The present experimental data predict a preventive and therapeutic potential of semisynthetic derivative CHNQ in ageing and related pathologies, mediated by activation of proteins of the antioxidant response.
Collapse
Affiliation(s)
- Martin Škandík
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravská cesta 9, 841 04, Bratislava, Slovak Republic
| | - Nataša Mrvová
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravská cesta 9, 841 04, Bratislava, Slovak Republic
| | - Štefan Bezek
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravská cesta 9, 841 04, Bratislava, Slovak Republic
| | - Lucia Račková
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravská cesta 9, 841 04, Bratislava, Slovak Republic.
| |
Collapse
|
17
|
Vargas-Mendoza N, Morales-González Á, Morales-Martínez M, Soriano-Ursúa MA, Delgado-Olivares L, Sandoval-Gallegos EM, Madrigal-Bujaidar E, Álvarez-González I, Madrigal-Santillán E, Morales-Gonzalez JA. Flavolignans from Silymarin as Nrf2 Bioactivators and Their Therapeutic Applications. Biomedicines 2020; 8:122. [PMID: 32423098 PMCID: PMC7277158 DOI: 10.3390/biomedicines8050122] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
Silymarin (SM) is a mixture of flavolignans extracted from the seeds of species derived from Silybum marianum, commonly known as milk thistle or St. Mary'sthistle. These species have been widely used in the treatment of liver disorders in traditional medicine since ancient times. Several properties had been attributed to the major SM flavolignans components, identified as silybin, isosilybin, silychristin, isosilychristin, and silydianin. Previous research reported antioxidant and protective activities, which are probably related to the activation of the nuclear factor erythroid 2 (NFE2)-related factor 2 (Nrf2), known as a master regulator of the cytoprotector response. Nrf2 is a redox-sensitive nuclear transcription factor able to induce the downstream-associated genes. The disruption of Nrf2 signaling has been associated with different pathological conditions. Some identified phytochemicals from SM had shown to participate in the Nrf2 signaling pathway; in particular, they have been suggested as activators that disrupt interactions in the Keap1-Nrf2 system, but also as antioxidants or with additional actions regarding Nrf2 regulation. Thus, the study of these molecules makes them appear attractive as novel targets for the treatment or prevention of several diseases.
Collapse
Affiliation(s)
- Nancy Vargas-Mendoza
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México Escuela Superior de Medicina, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico;
| | - Ángel Morales-González
- Escuela Superior de Cómputo, Instituto Politécnico Nacional, Av. Juan de Dios Bátiz s/n esquina Miguel Othón de Mendizabal, Unidad Profesional Adolfo López Mateos, Mexico City CP 07738, Mexico;
| | - Mauricio Morales-Martínez
- Licenciatura en Nutrición, Universidad Intercontinental, Insurgentes Sur 4303, Santa Úrsula Xitla, Alcaldía Tlalpan, Mexico City CP 14420, Mexico;
| | - Marvin A. Soriano-Ursúa
- Academia de Fisiología Humana, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Del. Miguel Hidalgo, Mexico City 11340, Mexico;
| | - Luis Delgado-Olivares
- Centro de Investigación Interdisciplinario, Área Académica de Nutrición, Instituto de Ciencias de la Salud. Universidad Autónoma del Estado de Hidalgo. Circuito Actopan-Tilcuauttla, s/n, Ex hacienda La Concepción, San Agustín Tlaxiaca, Hidalgo CP 42160, Mexico; (L.D.-O.); (E.M.S.-G.)
| | - Eli Mireya Sandoval-Gallegos
- Centro de Investigación Interdisciplinario, Área Académica de Nutrición, Instituto de Ciencias de la Salud. Universidad Autónoma del Estado de Hidalgo. Circuito Actopan-Tilcuauttla, s/n, Ex hacienda La Concepción, San Agustín Tlaxiaca, Hidalgo CP 42160, Mexico; (L.D.-O.); (E.M.S.-G.)
| | - Eduardo Madrigal-Bujaidar
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, “Unidad Profesional A. López Mateos”. Av. Wilfrido Massieu. Col., Zacatenco, Mexico City 07738, Mexico; (E.M.-B.); (I.Á.-G.)
| | - Isela Álvarez-González
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, “Unidad Profesional A. López Mateos”. Av. Wilfrido Massieu. Col., Zacatenco, Mexico City 07738, Mexico; (E.M.-B.); (I.Á.-G.)
| | - Eduardo Madrigal-Santillán
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México Escuela Superior de Medicina, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico;
| | - José A. Morales-Gonzalez
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México Escuela Superior de Medicina, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico;
| |
Collapse
|
18
|
AlKahtane AA, Ghanem E, Bungau SG, Alarifi S, Ali D, AlBasher G, Alkahtani S, Aleya L, Abdel-Daim MM. Carnosic acid alleviates chlorpyrifos-induced oxidative stress and inflammation in mice cerebral and ocular tissues. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:11663-11670. [PMID: 31965510 DOI: 10.1007/s11356-020-07736-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 01/14/2020] [Indexed: 06/10/2023]
Abstract
Chlorpyrifos is an organophosphate pesticide whose exposure leads to inhibition of acetylcholinesterase (AChE) enzyme and induces oxidative stress, inflammation, and neurotoxicity. The current study was designed to evaluate the efficacy of carnosic acid (CA) in ameliorating CPF-induced cytotoxicity in mice brain and eye tissues. We allocated 40 male Swiss albino mice to receive DMSO 1% solution, oral CA 60 mg/kg/day bw, CPF 12 mg/kg/day bw via gastric gavage, or CPF plus CA at 30 and 60 mg/kg/day bw. Carnosic acid was administered once/day for 14 days, while CPF was administered in the last 7 days of the experiment. Biochemical analysis showed that CPF administration was associated with significant increases in the serum concentrations of interleukin-1β, IL-6, and tumor necrosis factor-α, while it was associated with significant reductions in serum AChE levels in mice. Moreover, CPF-intoxicated mice exhibited significantly higher levels of malondialdehyde and nitric oxide in the brain and eye tissues. However, they had significantly lower levels of reduced glutathione, glutathione peroxidase, superoxide dismutase, and catalase in comparison with normal controls. Pretreatment with CA at 30 and 60 mg/kg/day bw for 14 days significantly alleviated all the aforementioned CPF-induced alterations in a dose-dependent manner; more frequent restorations of the normal control ranges were observed in the higher dose group. In conclusion, CA offers a neuroprotective effect against CPF-induced oxidative stress and inflammation and should be further studied in upcoming experimental and clinical research.
Collapse
Affiliation(s)
- Abdullah A AlKahtane
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Esraa Ghanem
- Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Simona G Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Saud Alarifi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Daoud Ali
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Gadah AlBasher
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, 25030, Besançon Cedex, France
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia.
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
19
|
Red Beetroot Extract Abrogates Chlorpyrifos-Induced Cortical Damage in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2963020. [PMID: 32215171 PMCID: PMC7085382 DOI: 10.1155/2020/2963020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 12/14/2019] [Accepted: 01/18/2020] [Indexed: 02/07/2023]
Abstract
Organophosphorus insecticides including chlorpyrifos (CPF) are mainly used for agriculture, household, and military purposes; their application is associated with various adverse reactions in animals and humans. This study was conducted to evaluate the potential neuroprotective effect of red beetroot methanolic extract (RBR) against CPF-induced cortical damage. Twenty-eight adult male Wistar albino rats were divided into 4 groups (n = 7 in each group): the control group was administered physiological saline (0.9% NaCl), the CPF group was administered CPF (10 mg/kg), the RBR group was administered RBR (300 mg/kg), and the RBR+CPF group was treated with RBR (300 mg/kg) 1 hr before CPF (10 mg/kg) supplementation. All groups were treated for 28 days. Rats exposed to CPF exhibited a significant decrease in cortical acetylcholinesterase activity and brain-derived neurotrophic factor and a decrease in glial fibrillary acidic protein. CPF intoxication increased lipid peroxidation, inducible nitric oxide synthase expression, and nitric oxide production. This was accompanied by a decrease in glutathione content and in the activities of glutathione peroxidase, glutathione reductase, superoxide dismutase, and catalase in the cortical tissue. Additionally, CPF enhanced inflammatory response, indicated by increased levels and expression of interleukin-1β and tumor necrosis factor-α. CPF triggered neuronal apoptosis by upregulating Bax and caspase-3 and downregulating Bcl-2. However, RBR reversed the induced neuronal alterations following CPF intoxication. Our findings suggest that RBR can minimize and prevent CPF neurotoxicity through its antioxidant, anti-inflammatory, and antiapoptotic activities.
Collapse
|
20
|
Teil M, Arotcarena ML, Faggiani E, Laferriere F, Bezard E, Dehay B. Targeting α-synuclein for PD Therapeutics: A Pursuit on All Fronts. Biomolecules 2020; 10:biom10030391. [PMID: 32138193 PMCID: PMC7175302 DOI: 10.3390/biom10030391] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/26/2020] [Accepted: 02/29/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's Disease (PD) is characterized both by the loss of dopaminergic neurons in the substantia nigra and the presence of cytoplasmic inclusions called Lewy Bodies. These Lewy Bodies contain the aggregated α-synuclein (α-syn) protein, which has been shown to be able to propagate from cell to cell and throughout different regions in the brain. Due to its central role in the pathology and the lack of a curative treatment for PD, an increasing number of studies have aimed at targeting this protein for therapeutics. Here, we reviewed and discussed the many different approaches that have been studied to inhibit α-syn accumulation via direct and indirect targeting. These analyses have led to the generation of multiple clinical trials that are either completed or currently active. These clinical trials and the current preclinical studies must still face obstacles ahead, but give hope of finding a therapy for PD with time.
Collapse
Affiliation(s)
- Margaux Teil
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Marie-Laure Arotcarena
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Emilie Faggiani
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Florent Laferriere
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Erwan Bezard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Benjamin Dehay
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; (M.T.); (M.-L.A.); (E.F.); (F.L.); (E.B.)
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
21
|
Zhou R, Li X, Li L, Zhang H. Theaflavins alleviate sevoflurane-induced neurocytotoxicity via Nrf2 signaling pathway. Int J Neurosci 2019; 130:1-8. [PMID: 31518514 DOI: 10.1080/00207454.2019.1667788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aim: Sevoflurane could induce apoptosis of rat hippocampal neurons, while theaflavins (TFs) have antioxidant and anti-inflammatory properties. This study aims to explore whether TFs could alleviate sevoflurane-induced neuronal cell injury.Materials and methods: Cells were treated by concentration gradient of sevoflurane and TFs. Cell viability, level of reactive oxygen species (ROS) and apoptosis rate were determined by cell counting kit-8 (CCK-8) and flow cytometry, respectively. Quantitative PCR (qPCR) and western blot were performed to determine mRNA and protein expressions.Results: TFs promoted viability of cells under the treatment of sevoflurane, while it suppressed apoptosis and down-regulated ROS level in a concentration-dependent manner. TFs could also down-regulate expression levels of caspase-3 and caspase-9 and cytosol and intranuclear nuclear factor E2-related factor 2 (Nrf2) in rat hippocampal nerve cells, while it up-regulated those of heme oxygenase 1 (HO-1), NADPH quinine oxidoreductase 1 (NQO1), glutamate cysteine ligase (GCL) and peroxiredoxin 1 (Prx1).Conclusions: Our study suggests that TFs exert protective effects on sevoflurane-induced neurocytotoxicity and therefore could be used as a potential drug for treatment of neuronal injury.
Collapse
Affiliation(s)
- Rongsheng Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaogang Li
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lu Li
- Department of the Second Anesthesia, The Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Han Zhang
- Department of the Second Anesthesia, The Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
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
|
23
|
Abstract
Targeted covalent modification is assuming consolidated importance in drug discovery. In this context, the electrophilic tuning of redox-dependent cell signaling is attracting major interest, as it opens prospect for treating numerous pathologic conditions. Herein, we discuss the rationale and the issues of electrophile-based approaches, focusing on the transcriptional Nrf2-Keap1 pathway as a test case. We also highlight relevant medicinal chemistry strategies researchers have devised to meet the ambitious goal, dwelling on the investigational and therapeutic potential of modulating redox-signaling networks through regulatory cysteine switches.
Collapse
|
24
|
Zhang Y, Ahmad KA, Khan FU, Yan S, Ihsan AU, Ding Q. Chitosan oligosaccharides prevent doxorubicin-induced oxidative stress and cardiac apoptosis through activating p38 and JNK MAPK mediated Nrf2/ARE pathway. Chem Biol Interact 2019; 305:54-65. [PMID: 30928397 DOI: 10.1016/j.cbi.2019.03.027] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/14/2019] [Accepted: 03/26/2019] [Indexed: 11/19/2022]
Abstract
Doxorubicin (DOX) is one of the most effective chemotherapeutic drugs; however, the incidence of cardiotoxicity compromises its therapeutic index. Oxidative stress and apoptosis are believed to be involved in DOX-induced cardiotoxicity. Chitosan oligosaccharides (COS), the enzymatic hydrolysates of chitosan, have been reported to possess diverse biological activities including antioxidant and anti-apoptotic properties. The objective of the present study was to investigate the potential role of COS against DOX-induced cardiotoxicity, and the effects of COS on apoptosis and oxidative stress in rats and H9C2 cells. Furthermore, we also shed light on the involved pathways during the whole process. For this purpose, first, we demonstrated that COS exhibited a significant protective effect on cardiac tissue by not only inducing a decrease in body and heart growth but also ameliorated oxidative damage and ECG alterations in DOX-treated rats. Second, we found that COS reversed the decrease of cell viability induced by DOX, reduced the intracellular reactive oxygen species (ROS), increased the mitochondrial membrane potential (MMP) and Bcl-2/Bax ratio. COS treatment also results in reduced caspase-3 and caspase-9 expressions, and an increase in the phosphorylation of MAPKs (mitogen-activated protein kinases) in DOX-exposed H9C2 cells. Additionally, cellular homeostasis was re-established via stabilization of MAPK mediated nuclear factor erythroid 2-related factor 2/antioxidant-response element (Nrf2/ARE) signaling and transcription of downstream cytoprotective genes. In summary, these findings suggest that COS could be a potential candidate for the prevention and treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Yongtian Zhang
- Experimental and Teaching Center of Medical Basis for Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Khalil Ali Ahmad
- Experimental and Teaching Center of Medical Basis for Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China; Shanghai Jiao Tong University, School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - Farhan Ullah Khan
- Shanghai Jiao Tong University, School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China; Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, China
| | - Simin Yan
- Experimental and Teaching Center of Medical Basis for Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Awais Ullah Ihsan
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, China
| | - Qilong Ding
- Experimental and Teaching Center of Medical Basis for Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
25
|
Pinto A, El Ali Z, Moniot S, Tamborini L, Steegborn C, Foresti R, De Micheli C. Effects of 3-Bromo-4,5-dihydroisoxazole Derivatives on Nrf2 Activation and Heme Oxygenase-1 Expression. ChemistryOpen 2018; 7:858-864. [PMID: 30397576 PMCID: PMC6207109 DOI: 10.1002/open.201800185] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Indexed: 12/31/2022] Open
Abstract
Natural and synthetic electrophilic compounds have been shown to activate the antioxidant protective Nrf2 (nuclear factor erythroid 2-related factor 2)/heme oxygenase-1 (HO-1) axis in cells and tissues. Here, we tested the ability of different isoxazoline-based electrophiles to up-regulate Nrf2/HO-1. The potency of activation is dependent on the leaving group at the 3-position of the isoxazoline nucleus, and an additional ring on the molecule limits the Nrf2/HO-1 activating properties. Among the synthetized compounds, we identified 3-bromo-5-phenyl-4,5-dihydroisoxazole 1 as the derivative with best activating properties in THP-1 human monocytic cells. We have confirmed that the target of our compounds is the Cys151 of the BTB domain of Keap1 by using mass spectrometry analyses and X-ray crystallography. Our findings demonstrate that these compounds affect the Nrf2/HO-1 axis and highlight a positive activity that can be of relevance from a therapeutic perspective in inflammation and infection.
Collapse
Affiliation(s)
- Andrea Pinto
- Department of Food, Environmental and Nutritional Sciences (DeFENS)University of MilanviaCeloria 220133MilanItaly
| | - Zeina El Ali
- Inserm U955, Equipe 12Créteil94000France
- Université Paris EstFaculté de MédecineCréteil94000France
| | - Sébastien Moniot
- Department of BiochemistryUniversity of BayreuthUniversitaetsstr. 3095447BayreuthGermany
| | - Lucia Tamborini
- Department of Pharmaceutical SciencesUniversity of MilanviaMangiagalli 2520133MilanItaly
| | - Clemens Steegborn
- Department of BiochemistryUniversity of BayreuthUniversitaetsstr. 3095447BayreuthGermany
| | - Roberta Foresti
- Inserm U955, Equipe 12Créteil94000France
- Université Paris EstFaculté de MédecineCréteil94000France
| | - Carlo De Micheli
- Department of Pharmaceutical SciencesUniversity of MilanviaMangiagalli 2520133MilanItaly
| |
Collapse
|
26
|
Du YT, Zheng YL, Ji Y, Dai F, Hu YJ, Zhou B. Applying an Electrophilicity-Based Strategy to Develop a Novel Nrf2 Activator Inspired from Dietary [6]-Shogaol. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:7983-7994. [PMID: 29987924 DOI: 10.1021/acs.jafc.8b02442] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Activation of nuclear factor erythroid-2-related factor 2 (Nrf2) is a crucial cellular defense mechanisms against oxidative stress and also an effective means to decrease the risk of oxidative stress-related diseases including cancer. Thus, identifying novel Nrf2 activators is highly anticipated. Inspired from [6]-shogaol (6S), an active component of ginger, herein we developed a novel potent Nrf2 activator, (1E,4E)-1-(4-hydroxy-3-methoxyphenyl)-7-methylocta-1,4,6-trien-3-one (SA) by an electrophilicity-based strategy. Compared with the parent 6S, SA bearing a short but entirely conjugated unsaturated ketone chain manifested the improved electrophilicity and cytoprotection (cell viability for the 10 μM 6S- and SA-treated group being 48.9 ± 5.3% and 76.1 ± 3.2%, respectively) against tert-butylhydroperoxide ( t-BHP)-induced cell death (cell viability for the t-BHP-stimulated group being 42.4 ± 0.4%) of HepG2. Mechanistic study uncovers that SA works as a potent Nrf2 activator by inducing Keap1 modification, inhibiting Nrf2 ubiquitylation and phosphorylating ERK in a Michael acceptor-dependent fashion. Taking 6S as an example, this works illustrates the feasibility and importance of applying an electrophilicity-based strategy to develop Nrf2 activators with dietary molecules as an inspiration due to their low toxicity and extraordinarily diverse chemical scaffolds.
Collapse
Affiliation(s)
- Yu-Ting Du
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , China
| | - Ya-Long Zheng
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , China
| | - Yuan Ji
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , China
| | - Fang Dai
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , China
| | - Yong-Jing Hu
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , China
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry , Lanzhou University , 222 Tianshui Street South , Lanzhou , Gansu 730000 , China
| |
Collapse
|
27
|
Protective Effect of Boswellic Acids against Doxorubicin-Induced Hepatotoxicity: Impact on Nrf2/HO-1 Defense Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018. [PMID: 29541348 PMCID: PMC5818967 DOI: 10.1155/2018/8296451] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The current study aimed to investigate the potential protective role of boswellic acids (BAs) against doxorubicin- (DOX-) induced hepatotoxicity. Also, the possible mechanisms underlying this protection; antioxidant, as well as the modulatory effect on the Nrf2 transcription factor/hem oxygenase-1 (Nrf2/HO-1) pathway in liver tissues, was investigated. Animals were allocated to five groups: group 1: the saline control, group 2: the DOX group, animals received DOX (6 mg/kg, i.p.) weekly for a period of three weeks, and groups 3–5: animals received DOX (6 mg/kg, i.p.) weekly and received protective doses of BAs (125, 250, and 500 mg/kg/day). Treatment with BAs significantly improved the altered liver enzyme activities and oxidative stress markers. This was coupled with significant improvement in liver histopathological features. BAs increased the Nrf2 and HO-1 expression, which provided protection against DOX-induced oxidative insult. The present results demonstrated that BAs appear to scavenge ROS and inhibit lipid peroxidation and DNA damage of DOX-induced hepatotoxicity. The antioxidant efficacy of BAs might arise from its modulation of the Nrf2/HO-1 pathway and thereby protected liver from DOX-induced oxidative injury.
Collapse
|
28
|
Goncharov NV, Nadeev AD, Jenkins RO, Avdonin PV. Markers and Biomarkers of Endothelium: When Something Is Rotten in the State. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9759735. [PMID: 29333215 PMCID: PMC5733214 DOI: 10.1155/2017/9759735] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 09/05/2017] [Indexed: 12/14/2022]
Abstract
Endothelium is a community of endothelial cells (ECs), which line the blood and lymphatic vessels, thus forming an interface between the tissues and the blood or lympha. This strategic position of endothelium infers its indispensable functional role in controlling vasoregulation, haemostasis, and inflammation. The state of endothelium is simultaneously the cause and effect of many diseases, and this is coupled with modifications of endothelial phenotype represented by markers and with biochemical profile of blood represented by biomarkers. In this paper, we briefly review data on the functional role of endothelium, give definitions of endothelial markers and biomarkers, touch on the methodological approaches for revealing biomarkers, present an implicit role of endothelium in some toxicological mechanistic studies, and survey the role of reactive oxygen species (ROS) in modulation of endothelial status.
Collapse
Affiliation(s)
- Nikolay V. Goncharov
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Saint Petersburg, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint Petersburg, Russia
| | - Alexander D. Nadeev
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint Petersburg, Russia
- Institute of Cell Biophysics RAS, Pushchino, Russia
| | - Richard O. Jenkins
- School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | | |
Collapse
|
29
|
Development of Novel Nrf2/ARE Inducers Bearing Pyrazino[2,1-a]isoquinolin Scaffold with Potent In Vitro Efficacy and Enhanced Physicochemical Properties. Molecules 2017; 22:molecules22091541. [PMID: 28902165 PMCID: PMC6151508 DOI: 10.3390/molecules22091541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/09/2017] [Indexed: 11/16/2022] Open
Abstract
Pyrazino[2,1-a]isoquinolin analogues were reported as potent activators of Nrf2/ARE signaling both in vitro and in vivo by our group. In this study, we simplified the ring system to investigate the functions of various parts of the pyrazino[2,1-a]isoquinolin scaffold. We proved that the tetrahydroisoquinoline was not essential for activity and the pyrido[1,2-a]pyrazin analogues 3b and 3g retained the cellular Nrf2/ARE activation activity. Besides, this simplification significantly enhanced water solubility and membrane permeability, indicating that these compounds are more favourable for the further development of therapeutic agents around Nrf2 activation.
Collapse
|
30
|
Simoni E, Serafini MM, Caporaso R, Marchetti C, Racchi M, Minarini A, Bartolini M, Lanni C, Rosini M. Targeting the Nrf2/Amyloid-Beta Liaison in Alzheimer's Disease: A Rational Approach. ACS Chem Neurosci 2017; 8:1618-1627. [PMID: 28421738 DOI: 10.1021/acschemneuro.7b00100] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyloid is a prominent feature of Alzheimer's disease (AD). Yet, a linear linkage between amyloid-β peptide (Aβ) and the disease onset and progression has recently been questioned. In this context, the crucial partnership between Aβ and Nrf2 pathways is acquiring paramount importance, offering prospects for deciphering the Aβ-centered disease network. Here, we report on a new class of antiaggregating agents rationally designed to simultaneously activate transcription-based antioxidant responses, whose lead 1 showed interesting properties in a preliminary investigation. Relying on the requirements of Aβ recognition, we identified the catechol derivative 12. In SH-SY5Y neuroblastoma cells, 12 combined remarkable free radical scavenger properties to the ability to trigger the Nrf2 pathway and induce the Nrf2-dependent defensive gene NQO1 by means of electrophilic activation of the transcriptional response. Moreover, 12 prevented the formation of cytotoxic stable oligomeric intermediates, being significantly more effective, and per se less toxic, than prototype 1. More importantly, as different chemical features were exploited to regulate Nrf2 and Aβ activities, the two pathways could be tuned independently. These findings point to compound 12 and its derivatives as promising tools for investigating the therapeutic potential of the Nrf2/Aβ cellular network, laying foundation for generating new drug leads to confront AD.
Collapse
Affiliation(s)
- Elena Simoni
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Melania M. Serafini
- Department of Drug Sciences (Pharmacology
Section), University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy
- Scuola Universitaria Superiore IUSS Pavia, P.zza Vittoria, 15, 27100 Pavia, Italy
| | - Roberta Caporaso
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Chiara Marchetti
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Marco Racchi
- Department of Drug Sciences (Pharmacology
Section), University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy
| | - Anna Minarini
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Manuela Bartolini
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Cristina Lanni
- Department of Drug Sciences (Pharmacology
Section), University of Pavia, V.le Taramelli 14, 27100 Pavia, Italy
| | - Michela Rosini
- Department of Pharmacy
and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| |
Collapse
|
31
|
Carnosic acid attenuates RANKL-induced oxidative stress and osteoclastogenesis via induction of Nrf2 and suppression of NF-κB and MAPK signalling. J Mol Med (Berl) 2017; 95:1065-1076. [PMID: 28674855 DOI: 10.1007/s00109-017-1553-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 05/03/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022]
Abstract
Nuclear factor-erythroid 2-related factor 2 (Nrf2) is a redox-sensitive transcription factor, which plays an important role in the cellular defense against oxidative stress by induction of anti-oxidant and cytoprotective enzymes. In the current study, we sought to investigate the osteoprotective effect of carnosic acid (CA), a phenolic (catecholic) diterpene. It is widely identified for its electrophilic nature under oxidative stress conditions and thus anticipated to counter osteoporosis by facilitation of Nrf2 signalling. Osteoclast differentiation was induced by incubation of RAW 264.7 (mouse macrophage) cells and mouse bone marrow macrophages (BMMs) in the presence of receptor activator of NF-κB ligand (RANKL) (100 ng/ml). After treatment, osteoclastogenesis was assessed using tartrate-resistant acid phosphatase (TRAP) assay. We observed that 6 h pretreatment with CA (1.25, 2.5, 5 μM) decreased RANKL-induced osteoclast formation and abolished RANKL-induced ROS generation by activating Nrf2 and its transcriptional targets. Further, CA also inhibited RANKL-induced activation of NF-κB and MAPK signalling. RANKL-induced mRNA expression of osteoclast related genes and transcription factors was also diminished by CA. In vivo osteolysis was developed in C57BL/6 male mice using lipopolysaccharide (LPS). Consistent with in vitro results, in vivo μ-CT analysis of femurs showed that bone mineral density (BMD), bone mineral content (BMC), and bone architecture parameters such as trabecular thickness (Tb.Th) and trabecular space (Tb.Sp) were positively modulated by CA in LPS-injected mice. The results obtained in this study support that CA inhibits RANKL-induced osteoclastogenesis by maintaining redox homeostasis through modulation of Nrf2 and NF-κB pathways. KEY MESSAGES Carnosic acid (CA) inhibits RANKL-induced osteoclastogenesis. CA inhibits RANKL-induced oxidative stress by upregulating Nrf2 transcriptional targets. CA attenuates RANKL-induced NF-κB and MAPK signalling activation. CA decreases NFATc1 and c-Fos expression. In vivo μ-CT analysis reveals that CA prevents bone loss in LPS-injected mice.
Collapse
|
32
|
Cipak Gasparovic A, Zarkovic N, Zarkovic K, Semen K, Kaminskyy D, Yelisyeyeva O, Bottari SP. Biomarkers of oxidative and nitro-oxidative stress: conventional and novel approaches. Br J Pharmacol 2017; 174:1771-1783. [PMID: 27864827 PMCID: PMC5446576 DOI: 10.1111/bph.13673] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 10/04/2016] [Accepted: 10/11/2016] [Indexed: 12/14/2022] Open
Abstract
The concept of oxidative stress (OS) that connects altered redox biology with various diseases was introduced 30 years ago and has generated intensive research over the past two decades. Whereas it is now commonly accepted that macromolecule oxidation in response to ROS is associated with a variety of pathologies, the emergence of NO as a key regulator of redox signalling has led to the discovery of the pathophysiological significance of reactive nitrogen species (RNS). RNS can elicit various modifications of macromolecules and lead to nitrative or nitro-OS. In order to investigate oxidative and nitro-OS in human and in live animal models, circulating biomarker assays have been developed. This article provides an overview of key biomarkers used to assess lipid peroxidation and NO/NO2 signalling, thereby stressing the necessity to analyse several OS biomarkers in relation to the overall (aerobic) metabolism and health condition of patients. In addition, the potential interest of heart rate variability as the non-invasive integrative biomarker of OS is discussed. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
| | | | - Kamelija Zarkovic
- Division of Pathology, Clinical Hospital Centre, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Khrystyna Semen
- Department of Propedeutics of Internal Medicine #2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Danylo Kaminskyy
- Department of Pharmaceutical, Organic, and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Olha Yelisyeyeva
- Department of Histology, Cytology and Embryology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Serge P Bottari
- Institute for Advanced Biosciences, INSERM U1029, CNRS UMR 5309, Grenoble-Alps University Medical School, Grenoble, France
- Radioanalysis Laboratory, CHU Grenoble-Alpes, Grenoble, France
| |
Collapse
|
33
|
Nogueira L, Mello DF, Trevisan R, Garcia D, da Silva Acosta D, Dafre AL, de Almeida EA. Hypoxia effects on oxidative stress and immunocompetence biomarkers in the mussel Perna perna (Mytilidae, Bivalvia). MARINE ENVIRONMENTAL RESEARCH 2017; 126:109-115. [PMID: 28260615 DOI: 10.1016/j.marenvres.2017.02.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/21/2017] [Accepted: 02/22/2017] [Indexed: 06/06/2023]
Abstract
This study investigated the effects of hypoxia on oxidative stress response and immune function in mussels Perna perna exposed to air for 6, 12, 24 and 48 h. In air-exposed mussels, the antioxidant enzymes superoxide dismutase (SOD), catalase, and glutathione reductase (GR) activities were lower in gill tissues (24-48 h) and digestive gland (12 h), while the glutathione peroxidase and GR activities were increased in the digestive gland (48 h). In both tissues, aerial exposure promoted a rapid (6 h) and persistent (up to 48 h) increase of glutathione levels. Decreased hemocyte count and viability, as well as increased phagocytic activity and cellular adhesion capacity were detected after prolonged aerial exposure (>12 h). In summary, induction of thiol pools, altered antioxidant enzyme activities, and activation of immune responses were detected in hypoxia exposed brown mussels, indicating hypoxia induced tissue-specific responses in both antioxidant and immune systems.
Collapse
Affiliation(s)
- Lílian Nogueira
- Laboratório de Biomarcadores de Contaminação Aquática, Departamento de Química e Ciências Ambientais, Universidade Estadual Paulista (IBILCE/UNESP), 15054-000, São José do Rio Preto, SP, Brazil
| | - Danielle Ferraz Mello
- Laboratório de Defesas Celulares, Departamento de Bioquímica, Universidade Federal de Santa Catarina, 88040-900, Florianópolis, SC, Brazil
| | - Rafael Trevisan
- Laboratório de Defesas Celulares, Departamento de Bioquímica, Universidade Federal de Santa Catarina, 88040-900, Florianópolis, SC, Brazil
| | - Danielly Garcia
- Laboratório de Biomarcadores de Contaminação Aquática, Departamento de Química e Ciências Ambientais, Universidade Estadual Paulista (IBILCE/UNESP), 15054-000, São José do Rio Preto, SP, Brazil
| | - Daiane da Silva Acosta
- Laboratório de Defesas Celulares, Departamento de Bioquímica, Universidade Federal de Santa Catarina, 88040-900, Florianópolis, SC, Brazil
| | - Alcir Luiz Dafre
- Laboratório de Defesas Celulares, Departamento de Bioquímica, Universidade Federal de Santa Catarina, 88040-900, Florianópolis, SC, Brazil
| | - Eduardo Alves de Almeida
- Laboratório de Biomarcadores de Contaminação Aquática, Departamento de Química e Ciências Ambientais, Universidade Estadual Paulista (IBILCE/UNESP), 15054-000, São José do Rio Preto, SP, Brazil; Departamento de Ciências Naturais, Fundação Universidade Regional de Blumenau (FURB), 89030-903, Blumenau, SC, Brazil
| |
Collapse
|
34
|
Kasarełło K, Cudnoch-Jędrzejewska A, Członkowski A, Mirowska-Guzel D. Mechanism of action of three newly registered drugs for multiple sclerosis treatment. Pharmacol Rep 2017; 69:702-708. [PMID: 28550802 DOI: 10.1016/j.pharep.2017.02.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/16/2017] [Accepted: 02/16/2017] [Indexed: 11/16/2022]
Abstract
Multiple sclerosis (MS) is a disease of suspected autoimmune origin leading to neurodegeneration. The disease pathomechanism is considered to be primarily based on neuroinflammation directed against myelin antigens caused by autoreactive T cells. MS etiology remains still unknown, which makes it difficult to create an efficient therapy, therefore, MS treatment targets mechanisms involved in disease pathology. In this review, we present the mechanism of action of three newly registered drugs for MS. Dimethyl fumarate (DMF) is an agent presenting a broad spectrum of action. Its main activity is based on activating the nuclear factor E2 dependent pathway leading to antioxidant enzyme synthesis. DMF in general suppresses the pro-inflammatory immune activity and exerts a neuroprotective action. Teriflunomide is a more focused drug, acting as an inhibitor of pyrimidines synthesis, important for rapidly dividing cells such as activated lymphocytes. Similarly, alemtuzumab, an anti-CD52 antibody, causes depletion of mainly lymphocytes. Since in MS pathology, T and B cells are involved, this mode of action is promising.
Collapse
Affiliation(s)
- Kaja Kasarełło
- Department of Experimental and Clinical Physiology, Medical University of Warsaw, Warszawa, Poland
| | | | - Andrzej Członkowski
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warszawa, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warszawa, Poland; Second Department of Neurology, Institute of Psychiatry and Neurology, Warszawa, Poland.
| |
Collapse
|
35
|
Qin J, He Y, Duan M, Luo M. Effects of Nuclear Factor-E2-related factor 2/Heme Oxygenase 1 on splanchnic hemodynamics in experimental cirrhosis with portal hypertension. Microvasc Res 2016; 111:12-19. [PMID: 28025064 DOI: 10.1016/j.mvr.2016.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 01/07/2023]
Abstract
OBJECTIVE We explored the effects of Nuclear Factor-E2-related factor 2 (Nrf2) and Heme Oxygenase 1 (HO-1) on splanchnic hemodynamics in portal hypertensive rats. METHODS Experimental cirrhosis with portal hypertension was induced by intraperitoneal injection of carbon tetrachloride. The expression of proteins was examined by immunoblotting. Hemodynamic studies were performed by radioactive microspheres. The vascular perfusion system was used to measure the contractile response of mesentery arterioles in rats. RESULTS Nrf2 expression in the nucleus and HO-1 expression in cytoplasm was significantly enhanced in portal hypertensive rats. Portal pressure, as well as regional blood flow, increased significantly in portal hypertension and can be blocked by tin protoporphyrin IX. The expression of endogenous nitric oxide synthase and vascular endothelial growth factors increased significantly compared to normal rats, while HO-1 inhibition decreased the expression of these proteins significantly. The contractile response of mesenteric arteries decreased in portal hypertension, but can be partially recovered through tin protoporphyrin IX treatment. CONCLUSIONS The expression of Nrf2/HO-1 increased in mesenteric arteries of portal hypertensive rats, which was related to oxidative stress. HO-1was involved in increased portal pressure and anomaly splanchnic hemodynamics in portal hypertensive rats.
Collapse
Affiliation(s)
- Jun Qin
- Department of General Surgery, Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yue He
- Department of General Surgery, Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ming Duan
- Department of General Surgery, Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Meng Luo
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
36
|
Sun Z, Park SY, Hwang E, Park B, Seo SA, Cho JG, Zhang M, Yi TH. Dietary Foeniculum vulgare Mill extract attenuated UVB irradiation-induced skin photoaging by activating of Nrf2 and inhibiting MAPK pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1273-1284. [PMID: 27765346 DOI: 10.1016/j.phymed.2016.06.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 06/08/2016] [Accepted: 06/11/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Foeniculum vulgare Mill (FV) has long been prescribed in traditional medicine due to its antioxidant anti-inflammatory properties. However, little research has been done on the use of FV to alleviate changes in UVB-induced photoaging PURPOSE: This study was to investigate the photoprotective effects and mechanism of FV in vitro and in vivo. METHODS The anti-photoaging effect of FV was assessed in normal human dermal fibroblasts (NHDFs) in vitro. The secretion of reactive oxygen species (ROS), lactate dehydrogenase (LDH), GSH, matrix metalloproteinases (MMPs), procollagen type I, IL-6 and transforming growth factor-β1 (TGF-β1) were measured by kits. Additionally, the level of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), p-ERK and p38 were evaluated by western blotting. In vivo, H&E and Masson's trichrome staining were employed. The expression of MMP-1, procollagen type I, TGF-β1 and elastin were measured by western blot. RESULTS FV significantly increased the production of collagen, elastin and TGF-β1 levels, while blocked matrix metalloproteinases (MMPs) production in UVB irradiation induced hairless mice, which were consistent with the result in NHDFs. Furthermore, FV dose-dependently decreased the production of ROS and LDH by promoting the nuclear amount of Nrf2 and enhancing the expression of cytoprotective antioxidants such as GSH. FV also significantly quenched UVB-induced phosphorylation of ERK and p38 in NHDFs. CONCLUSION Our results indicate that FV is a potential botanical agent for the treatment of skin damage induced by UV irradiation.
Collapse
Affiliation(s)
- Zhengwang Sun
- Department of Oriental Medicinal Material and Processing, College of Life Science, Kyung Hee University Global Campus, 1732 Deokyoungdaero, Giheung-gu, Yongin-si, Gyeonggi-do, 446-701, Republic of Korea
| | - Sang Yong Park
- Department of Oriental Medicinal Material and Processing, College of Life Science, Kyung Hee University Global Campus, 1732 Deokyoungdaero, Giheung-gu, Yongin-si, Gyeonggi-do, 446-701, Republic of Korea
| | - Eunson Hwang
- Department of Oriental Medicinal Material and Processing, College of Life Science, Kyung Hee University Global Campus, 1732 Deokyoungdaero, Giheung-gu, Yongin-si, Gyeonggi-do, 446-701, Republic of Korea
| | - Bom Park
- Department of Oriental Medicinal Material and Processing, College of Life Science, Kyung Hee University Global Campus, 1732 Deokyoungdaero, Giheung-gu, Yongin-si, Gyeonggi-do, 446-701, Republic of Korea
| | - Seul A Seo
- Department of Oriental Medicinal Material and Processing, College of Life Science, Kyung Hee University Global Campus, 1732 Deokyoungdaero, Giheung-gu, Yongin-si, Gyeonggi-do, 446-701, Republic of Korea
| | - Jin-Gyeong Cho
- Department of Oriental Medicinal Material and Processing, College of Life Science, Kyung Hee University Global Campus, 1732 Deokyoungdaero, Giheung-gu, Yongin-si, Gyeonggi-do, 446-701, Republic of Korea
| | - Mengyang Zhang
- Department of Oriental Medicinal Material and Processing, College of Life Science, Kyung Hee University Global Campus, 1732 Deokyoungdaero, Giheung-gu, Yongin-si, Gyeonggi-do, 446-701, Republic of Korea
| | - Tae-Hoo Yi
- Department of Oriental Medicinal Material and Processing, College of Life Science, Kyung Hee University Global Campus, 1732 Deokyoungdaero, Giheung-gu, Yongin-si, Gyeonggi-do, 446-701, Republic of Korea..
| |
Collapse
|
37
|
Dong Q, Hou H, Wu J, Chen Y. The Nrf2-ARE pathway is associated with Schisandrin b attenuating benzo(a)pyrene-Induced HTR cells damages in vitro. ENVIRONMENTAL TOXICOLOGY 2016; 31:1439-1449. [PMID: 25946486 DOI: 10.1002/tox.22149] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/11/2015] [Indexed: 06/04/2023]
Abstract
As is ubiquitous in the environmental sources, benzo(a)pyrene (BaP) has been reported to induce reprotoxicity in previous studies. Toxicity to trophoblast cells may be one key factor, but evidences were absent. We speculated that BaP can induce cytotoxicity in human trophoblast HTR-8/SVneo (HTR) cells, and Schisandrin B (Sch B) as a potential protector can inhibit the cytotoxicity. MTS assay identified that BaP induced HTR cells death while Sch B played a cytoprotective role. And after Nrf2 interference, the ability of Sch B-induced cytoprotection was declined. Furthermore, PCR, western blot, ELISA, and SOD assays were found that Sch B significantly increased the mRNA and protein expression of Nrf2, HO1, NQO1, and SOD in the Nrf2-ARE pathway, and the extents of increase were declined after Nrf2 interference. These results demonstrated that the Nrf2-ARE pathway plays an important role in Sch B attenuating BaP-induced HTR cells damages in vitro. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1439-1449, 2016.
Collapse
Affiliation(s)
- Qulong Dong
- Department of Obstetrics and Gynecology, Affiliated Hospital of Logistics University of the Chinese People's Armed Forces, Tianjin, 300162, China
| | - Haiyan Hou
- Department of Obstetrics and Gynecology, Affiliated Hospital of Logistics University of the Chinese People's Armed Forces, Tianjin, 300162, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Jun Wu
- Program in Public Health and Department of Epidemiology, University of California, Irvine, 92697, USA
| | - Yaqiong Chen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Logistics University of the Chinese People's Armed Forces, Tianjin, 300162, China.
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, 300162, China.
| |
Collapse
|
38
|
Jiang T, Sun Q, Chen S. Oxidative stress: A major pathogenesis and potential therapeutic target of antioxidative agents in Parkinson's disease and Alzheimer's disease. Prog Neurobiol 2016; 147:1-19. [PMID: 27769868 DOI: 10.1016/j.pneurobio.2016.07.005] [Citation(s) in RCA: 446] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/19/2016] [Accepted: 07/11/2016] [Indexed: 12/14/2022]
Abstract
Oxidative stress reflects an imbalance between the overproduction and incorporation of free radicals and the dynamic ability of a biosystem to detoxify reactive intermediates. Free radicals produced by oxidative stress are one of the common features in several experimental models of diseases. Free radicals affect both the structure and function of neural cells, and contribute to a wide range of neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. Although the precise mechanisms that result in the degeneration of neurons and the relevant pathological changes remain unclear, the crucial role of oxidative stress in the pathogenesis of neurodegenerative diseases is associated with several proteins (such as α-synuclein, DJ-1, Amyloid β and tau protein) and some signaling pathways (such as extracellular regulated protein kinases, phosphoinositide 3-kinase/Protein Kinase B pathway and extracellular signal-regulated kinases 1/2) that are tightly associated with the neural damage. In this review, we present evidence, gathered over the last decade, concerning a variety of pathogenic proteins, their important signaling pathways and pathogenic mechanisms associated with oxidative stress in Parkinson's disease and Alzheimer's disease. Proper control and regulation of these proteins' functions and the related signaling pathways may be a promising therapeutic approach to the patients. We also emphasizes antioxidative options, including some new neuroprotective agents that eliminate excess reactive oxygen species efficiently and have a certain therapeutic effect; however, controversy surrounds some of them in terms of the dose and length of therapy. These agents require further investigation by clinical application in patients suffering Parkinson's disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Tianfang Jiang
- Department of Neurology, Institute of Neurology and the Collaborative Innovation Center for Brain Science, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian Sun
- Department of Neurology, Institute of Neurology and the Collaborative Innovation Center for Brain Science, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shengdi Chen
- Department of Neurology, Institute of Neurology and the Collaborative Innovation Center for Brain Science, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Laboratory of Neurodegenerative Diseases, Institute of Health Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Science & Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
39
|
Rojo de la Vega M, Dodson M, Chapman E, Zhang DD. NRF2-targeted therapeutics: New targets and modes of NRF2 regulation. CURRENT OPINION IN TOXICOLOGY 2016; 1:62-70. [PMID: 29082352 DOI: 10.1016/j.cotox.2016.10.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pharmacological activation of the transcription factor nuclear factor-erythroid derived 2-like 2 (NRF2), the key regulator of the cellular antioxidant response, has been recognized as a feasible strategy to reduce oxidative/electrophilic stress and prevent carcinogenesis or other chronic illnesses, such as diabetes and chronic kidney disease. In contrast, due to the discovery of the "dark side" of NRF2, where prolonged activation of NRF2 causes tissue damage, cancer progression, or chemoresistance, efforts have been devoted to identify inhibitors. Currently, only one NRF2 activator has been approved for use in the clinic, while no specific NRF2 inhibitors have been discovered. Future development of NRF2-targeted therapeutics should be based on our current understanding of the regulatory mechanisms of this protein. In addition to the KEAP1-dependent mechanisms, the recent discovery of other pathways involved in the degradation of NRF2 have opened up new possibilities for the development of safe and specific therapeutics. Here, we review available and putative NRF2-targeted therapeutics and discuss their modes of action as well as their potential for disease prevention and treatment.
Collapse
Affiliation(s)
- Montserrat Rojo de la Vega
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721.,Arizona Cancer Center, University of Arizona, Tucson, AZ, USA, 85724
| |
Collapse
|
40
|
Deny LJ, Traboulsi H, Cantin AM, Marsault É, Richter MV, Bélanger G. Bis-Michael Acceptors as Novel Probes to Study the Keap1/Nrf2/ARE Pathway. J Med Chem 2016; 59:9431-9442. [PMID: 27682717 DOI: 10.1021/acs.jmedchem.6b01132] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulator that promotes the transcription of cytoprotective genes in response to oxidative/electrophilic stress. Various Michael-type compounds were designed and synthesized, and their potency to activate the Keap1/Nrf2/ARE pathway was evaluated. Compounds bearing two Michael-type acceptors proved to be the most active. Tether length and rigidity between the acceptors was crucial. This study will help to understand how this feature disrupts the interaction between Keap1 and Nrf2.
Collapse
Affiliation(s)
- Ludovic J Deny
- Département de Chimie, Université de Sherbrooke , 2500 Boulevard Université, Sherbrooke, Québec J1K 2R1, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke , Sherbrooke, Québec J1H 5N4, Canada
| | - Hussein Traboulsi
- Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke , Sherbrooke, Québec J1H 5N4, Canada.,Département de Médecine, Service de Pneumologie et Centre de Recherche Clinique, Centre Hospitalier de l'Université de Sherbrooke , Sherbrooke, Québec J1H 5N4, Canada
| | - André M Cantin
- Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke , Sherbrooke, Québec J1H 5N4, Canada.,Département de Médecine, Service de Pneumologie et Centre de Recherche Clinique, Centre Hospitalier de l'Université de Sherbrooke , Sherbrooke, Québec J1H 5N4, Canada
| | - Éric Marsault
- Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke , Sherbrooke, Québec J1H 5N4, Canada.,Département de Pharmacologie, Université de Sherbrooke , Sherbrooke, Québec J1K 2R1, Canada
| | - Martin V Richter
- Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke , Sherbrooke, Québec J1H 5N4, Canada.,Département de Médecine, Service de Pneumologie et Centre de Recherche Clinique, Centre Hospitalier de l'Université de Sherbrooke , Sherbrooke, Québec J1H 5N4, Canada
| | - Guillaume Bélanger
- Département de Chimie, Université de Sherbrooke , 2500 Boulevard Université, Sherbrooke, Québec J1K 2R1, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke , Sherbrooke, Québec J1H 5N4, Canada
| |
Collapse
|
41
|
VCE-003.2, a novel cannabigerol derivative, enhances neuronal progenitor cell survival and alleviates symptomatology in murine models of Huntington's disease. Sci Rep 2016; 6:29789. [PMID: 27430371 PMCID: PMC4949444 DOI: 10.1038/srep29789] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/24/2016] [Indexed: 01/01/2023] Open
Abstract
Cannabinoids have shown to exert neuroprotective actions in animal models by acting at different targets including canonical cannabinoid receptors and PPARγ. We previously showed that VCE-003, a cannabigerol (CBG) quinone derivative, is a novel neuroprotective and anti-inflammatory cannabinoid acting through PPARγ. We have now generated a non-thiophilic VCE-003 derivative named VCE-003.2 that preserves the ability to activate PPARγ and analyzed its neuroprotective activity. This compound exerted a prosurvival action in progenitor cells during neuronal differentiation, which was prevented by a PPARγ antagonist, without affecting neural progenitor cell proliferation. In addition, VCE-003.2 attenuated quinolinic acid (QA)-induced cell death and caspase-3 activation and also reduced mutant huntingtin aggregates in striatal cells. The neuroprotective profile of VCE-003.2 was analyzed using in vivo models of striatal neurodegeneration induced by QA and 3-nitropropionic acid (3NP) administration. VCE-003.2 prevented medium spiny DARPP32+ neuronal loss in these Huntington’s-like disease mice models improving motor deficits, reactive astrogliosis and microglial activation. In the 3NP model VCE-003.2 inhibited the upregulation of proinflammatory markers and improved antioxidant defenses in the brain. These data lead us to consider VCE-003.2 to have high potential for the treatment of Huntington’s disease (HD) and other neurodegenerative diseases with neuroinflammatory traits.
Collapse
|
42
|
Wang X, Bai Y, Cheng G, Ihsan A, Zhu F, Wang Y, Tao Y, Chen D, Dai M, Liu Z, Yuan Z. Genomic and proteomic analysis of the inhibition of synthesis and secretion of aldosterone hormone induced by quinocetone in NCI-H295R cells. Toxicology 2016; 350-352:1-14. [PMID: 27046791 DOI: 10.1016/j.tox.2016.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 11/26/2022]
Abstract
Quinoxaline 1,4-dioxides (QdNOs) are widely used as a kind of antibacterial growth promoter in animal husbandry. The adrenal cortex was found to be one of the main toxic targets of QdNOs, accompanied by a decreased aldosterone level. However, the way in which QdNOs decrease production of the hormone aldosterone is far from clear. To illustrate the mechanism by which QdNOs damage the adrenal cortex and decrease aldosterone hormone levels, the QdNOs were screened to choose the drug with most toxic effects on aldosterone production, and then to reveal the mechanism between the gene and protein profiles in human adrenocortical cells (NCI-H295R cells). The results found that quinocetone (QCT) showed the highest adrenal toxic effect among QdNOs. After exposing H295R cells to 10 and 20μM QCT for 24h, compared with blank cells, the gene and protein expression profiles obtained were analyzed by microarray and MALDI TOF/TOF mass spectrometry, respectively. The results of microarray analysis suggested that ABCG1 and SREBF1, which were involved in the cholesterol biosynthetic and metabolic processes, and CYP17A1, NR4A2 and G6PD, which were related to aldosterone biosynthesis, were important molecular targets. It has been speculated that PKC and ERK pathways might be involved in the reduction of aldosterone production caused by QCT, through enhanced mRNA expression of CYP17A1. Additionally, JNK and p38MAPK signal transduction pathways might participate in apoptosis induced by QCT. Twenty-nine and 32 protein spots were successfully identified when cells were treated with 10 and 20μM QCT, respectively. These identified proteins mainly included material synthesis and energy metabolism-related proteins, transcription/translation processing-related proteins, signal transduction proteins, cytoskeletal proteins, molecular chaperones, proteins related to response to stress, and transport proteins. Further investigations suggested that oxidative stress caused by QCT was exacerbated through disruption of the Keap1/Nrf2/ARE anti-oxidative stress pathway. Taken together, the data demonstrated for the first time that the Keap1/Nrf2/ARE pathway plays a crucial role in adrenal toxicity, and that CYP17A1 was the key switch to reduce the aldosterone production induced by QCT. Furthermore, large numbers of genes and proteins and entry points for research in the inhibition of aldosterone synthesis induced by QCT were offered, which will provide new insight into the adrenal toxicity of QdNOs and help to provide a theoretical foundation for the formulation of safety controls for products obtained from animals and to design new QdNOs with less harmful effects.
Collapse
Affiliation(s)
- Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yijie Bai
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Guyue Cheng
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Awais Ihsan
- Department of Biosciences, COMSATS Institute of Information Technology, Sahiwal, Pakistan
| | - Feng Zhu
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yulian Wang
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yanfei Tao
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China
| | - Dongmei Chen
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China
| | - Menghong Dai
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China
| | - Zhengli Liu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China
| | - Zonghui Yuan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China.
| |
Collapse
|
43
|
Islam MT, da Silva CB, de Alencar MVOB, Paz MFCJ, Almeida FRDC, Melo-Cavalcante AADC. Diterpenes: Advances in Neurobiological Drug Research. Phytother Res 2016; 30:915-28. [DOI: 10.1002/ptr.5609] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 02/10/2016] [Accepted: 02/20/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Md. Torequl Islam
- Northest Biotechnology Network (RENORBIO), Post-graduation Program in Biotechnology; Federal University of Piauí; 64.049-550 Teresina Brazil
- Department of Biochemistry and Pharmacology, Post-graduation Program in Pharmaceutical Science; Federal University of Piauí; 64.049-550 Teresina Brazil
- Department of Pharmacy; Southern University Bangladesh; 22-Shahid Mirza Lane (E), Academic building-II, 1st floor, 739/A, Mehedibag Road, Mehedibag-4000 Chittagong Bangladesh
| | - Claucenira Bandeira da Silva
- Northest Biotechnology Network (RENORBIO), Post-graduation Program in Biotechnology; Federal University of Piauí; 64.049-550 Teresina Brazil
| | - Marcus Vinícius Oliveira Barros de Alencar
- Northest Biotechnology Network (RENORBIO), Post-graduation Program in Biotechnology; Federal University of Piauí; 64.049-550 Teresina Brazil
- Department of Biochemistry and Pharmacology, Post-graduation Program in Pharmaceutical Science; Federal University of Piauí; 64.049-550 Teresina Brazil
| | - Márcia Fernanda Correia Jardim Paz
- Northest Biotechnology Network (RENORBIO), Post-graduation Program in Biotechnology; Federal University of Piauí; 64.049-550 Teresina Brazil
- Department of Biochemistry and Pharmacology, Post-graduation Program in Pharmaceutical Science; Federal University of Piauí; 64.049-550 Teresina Brazil
| | - Fernanda Regina de Castro Almeida
- Northest Biotechnology Network (RENORBIO), Post-graduation Program in Biotechnology; Federal University of Piauí; 64.049-550 Teresina Brazil
| | - Ana Amélia de Carvalho Melo-Cavalcante
- Northest Biotechnology Network (RENORBIO), Post-graduation Program in Biotechnology; Federal University of Piauí; 64.049-550 Teresina Brazil
- Department of Biochemistry and Pharmacology, Post-graduation Program in Pharmaceutical Science; Federal University of Piauí; 64.049-550 Teresina Brazil
| |
Collapse
|
44
|
Vnukov VV, Gutsenko OI, Milutina NP, Ananyan AA, Danilenko AO, Panina SB, Kornienko IV. Influence of SkQ1 on Expression of Nrf2 Transcription Factor Gene, ARE-Controlled Genes of Antioxidant Enzymes and Their Activity in Rat Blood Leukocytes. BIOCHEMISTRY (MOSCOW) 2016; 80:586-91. [PMID: 26071777 DOI: 10.1134/s0006297915050107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
This study demonstrated that pretreatment of rats with mitochondria-targeted antioxidant SkQ1 (50 nmol/kg during 5 days) significantly increased the mRNA levels of Nrf2 transcription factor and Nrf2-induced genes encoding antioxidant enzymes SOD1, SOD2, CAT, and GPx4 in rat peripheral blood leukocytes. The increase in expression of these genes with SkQ1 addition was accompanied by increased activities of catalase, glutathione peroxidase, and glutathione-S-transferase in leukocytes. These results indicate that antioxidant properties of SkQ1 might be realized via induction of expression of the genes regulating activity of antioxidant system elements.
Collapse
Affiliation(s)
- V V Vnukov
- Southern Federal University, Academy of Biology and Biotechnology, Department of Biochemistry and Microbiology, Rostov-on-Don, 344090, Russia.
| | | | | | | | | | | | | |
Collapse
|
45
|
Figueiredo-Pereira ME, Corwin C, Babich J. Prostaglandin J2: a potential target for halting inflammation-induced neurodegeneration. Ann N Y Acad Sci 2016; 1363:125-37. [PMID: 26748744 DOI: 10.1111/nyas.12987] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Prostaglandins (PGs) are produced via cyclooxygenases, which are enzymes that play a major role in neuroinflammation. Epidemiological studies show that chronic treatment with low levels of cyclooxygenase inhibitors (nonsteroidal anti-inflammatory drugs (NSAIDs)) lowers the risk for Alzheimer's disease (AD) and Parkinson's disease (PD) by as much as 50%. Unfortunately, inhibiting cyclooxygenases with NSAIDs blocks the synthesis of downstream neuroprotective and neurotoxic PGs, thus producing adverse side effects. We focus on prostaglandin J2 (PGJ2) because it is highly neurotoxic compared to PGA1, D2, and E2. Unlike other PGs, PGJ2 and its metabolites have a cyclopentenone ring with reactive α,β-unsaturated carbonyl groups that form covalent Michael adducts with key cysteines in proteins and GSH. Cysteine-binding electrophiles such as PGJ2 are considered to play an important role in determining whether neurons will live or die. We discuss in vitro and in vivo studies showing that PGJ2 induces pathological processes relevant to neurodegenerative disorders such as AD and PD. Further, we discuss our work showing that increasing intracellular cAMP with the lipophilic peptide PACAP27 counteracts some of the PGJ2-induced detrimental effects. New therapeutic strategies that neutralize the effects of specific neurotoxic PGs downstream from cyclooxygenases could have a significant impact on the treatment of chronic neurodegenerative disorders with fewer adverse side effects.
Collapse
Affiliation(s)
| | - Chuhyon Corwin
- Department of Biological Sciences, Hunter College and the Graduate Center, CUNY, New York, New York
| | - John Babich
- Department of Radiology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
46
|
Park JH, Choi JW, Ju EJ, Pae AN, Park KD. Antioxidant and Anti-Inflammatory Activities of a Natural Compound, Shizukahenriol, through Nrf2 Activation. Molecules 2015; 20:15989-6003. [PMID: 26364630 PMCID: PMC6332350 DOI: 10.3390/molecules200915989] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/18/2015] [Accepted: 08/28/2015] [Indexed: 11/21/2022] Open
Abstract
Imbalance in the antioxidant defense system leads to detrimental consequences, such as neurological disorders. The Nrf2 signaling is known as a main pathway involved in cellular defense system. Nrf2 is a transcription factor that regulates oxidative stress response by inducing expression of various antioxidant enzyme genes. In this study, we screened several pure natural compounds for Nrf2 activator. Among them, shizukahenriol (SZH), isolated from Chloranthus henryi, activated Nrf2, and induced expression of the Nrf2-dependent antioxidant enzymes HO-1, GCLC, and GCLM in BV-2 microglial cells. This natural compound was also effective in suppressing production of inflammatory molecules NO, TNF-α, and inhibition of NF-κB p65 translocation to the nucleus in a dose-dependent manner. We also examined whether SZH rescued the microglial cells from oxidative stress-induced cell death. Pretreatment with SZH dose-dependently attenuated H2O2-induced cytotoxicity in BV-2 microglial cells. These results suggested SZH as a potential neuroprotective agent for neurological disorders.
Collapse
Affiliation(s)
- Jong-Hyun Park
- Center for Neuro-Medicine, Korea Institute of Science and Technology, Seoul 136-791, Korea.
| | - Ji Won Choi
- Center for Neuro-Medicine, Korea Institute of Science and Technology, Seoul 136-791, Korea.
- Department of Biotechnology, Yonsei University, Seoul 120-749, Korea.
| | - Eun Ji Ju
- Center for Neuro-Medicine, Korea Institute of Science and Technology, Seoul 136-791, Korea.
- Department of Biotechnology, Yonsei University, Seoul 120-749, Korea.
| | - Ae Nim Pae
- Center for Neuro-Medicine, Korea Institute of Science and Technology, Seoul 136-791, Korea.
- Department of Biological Chemistry, University of Science and Technology, Daejeon 305-350, Korea.
| | - Ki Duk Park
- Center for Neuro-Medicine, Korea Institute of Science and Technology, Seoul 136-791, Korea.
- Department of Biological Chemistry, University of Science and Technology, Daejeon 305-350, Korea.
| |
Collapse
|
47
|
Wang Y, Mei X, Yuan J, Lu W, Li B, Xu D. Taurine zinc solid dispersions attenuate doxorubicin-induced hepatotoxicity and cardiotoxicity in rats. Toxicol Appl Pharmacol 2015; 289:1-11. [PMID: 26335259 DOI: 10.1016/j.taap.2015.08.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 07/29/2015] [Accepted: 08/24/2015] [Indexed: 12/17/2022]
Abstract
The clinical efficacy of anthracycline anti-neoplastic agents is limited by cardiac and hepatic toxicities. The aim of this study was to assess the hepatoprotective and cardioprotective effects of taurine zinc solid dispersions, which is a newly-synthesized taurine zinc compound, against doxorubicin-induced toxicity in Sprague-Dawley rats intraperitoneally injected with doxorubicin hydrochloride (3mg/kg) three times a week (seven injections) over 28 days. Hemodynamic parameters, levels of liver toxicity markers and oxidative stress were assessed. Taurine zinc significantly attenuated the reductions in blood pressure, left ventricular pressure and ± dp/dtmax, increases in serum alanine aminotransferase and aspartate aminotransferase activities, and reductions in serum Zn(2+) and albumin levels (P<0.05 or 0.01) induced by doxorubicin. In rats treated with doxorubicin, taurine zinc dose-dependently increased liver superoxide dismutase activity and glutathione concentration, and decreased malondialdehyde level (P<0.01). qBase(+) was used to evaluate the stability of eight candidate reference genes for real-time quantitative reverse-transcription PCR. Taurine zinc dose-dependently increased liver heme oxygenase-1 and UDP-glucuronyl transferase mRNA and protein expression (P<0.01). Western blotting demonstrated that taurine zinc inhibited c-Jun N-terminal kinase phosphorylation by upregulating dual-specificity phosphoprotein phosphatase-1. Additionally, taurine zinc inhibited cardiomyocyte apoptosis as there was decreased TUNEL/DAPI positivity and protein expression of caspase-3. These results indicate that taurine zinc solid dispersions prevent the side-effects of anthracycline-based anticancer therapy. The mechanisms might be associated with the enhancement of antioxidant defense system partly through activating transcription to synthesize endogenous phase II medicine enzymes and anti-apoptosis through inhibiting JNK phosphorylation.
Collapse
Affiliation(s)
- Yu Wang
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Xueting Mei
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Jingquan Yuan
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Wenping Lu
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Binglong Li
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Donghui Xu
- Lab of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China.
| |
Collapse
|
48
|
Activation of the Nrf2-regulated antioxidant cell response inhibits HEMA-induced oxidative stress and supports cell viability. Biomaterials 2015; 56:114-28. [PMID: 25934285 DOI: 10.1016/j.biomaterials.2015.03.047] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 03/20/2015] [Accepted: 03/27/2015] [Indexed: 11/22/2022]
Abstract
Oxidative stress due to increased formation of reactive oxygen species (ROS) in target cells of dental resin monomers like 2-hydroxyethyl methacrylate (HEMA) is a major mechanism underlying the disturbance of vital cell functions including mineralization and differentiation, responses of the innate immune system, and the induction of cell death via apoptosis. Although a shift in the equilibrium between cell viability and apoptosis is related to the non-enzymatic antioxidant glutathione (GSH) in HEMA-exposed cells, the major mechanisms of adaptive antioxidant cell responses to maintain cellular redox homeostasis are still unknown. The present study provides insight into the induction of a communicating network of pathways under the control of the redox-sensitive transcription factor Nrf2, a major transcriptional activator of genes coding for enzymatic antioxidants. Here, oxidative stress was indicated by DCF fluorescence in cells after a short exposure (1 h) to HEMA, while DHR123 fluorescence significantly increased about 1.8-fold after a long exposure period (24 h) showing the formation of hydrogen peroxide (H2O2). The corresponding expression of Nrf2 was activated immediately after HEMA exposure (1 h) and remained constant up to 24 h. Nrf2-regulated expression of enzymes of the glutathione metabolism (glutathione peroxidase 1/2, glutathione reductase) decreased in HEMA-exposed cells as a result of GSH depletion, and superoxide dismutase expression was downregulated after H2O2 overproduction. However, the expression of Nrf2-controlled enzymatic antioxidants (catalase, peroxiredoxin, thioredoxin 1, thioredoxin reductase, heme oxygenase-1) and the NADPH-regenerating system (glucose 6-phosphate dehydrogenase, transaldolase) was increased. Phenolic tert-butylhydroquinone (tBHQ), a classic inducer of the Nrf2 pathway, reduced oxidative stress and protected cells from HEMA-induced cell death through a shift in the number of cells in necrosis to apoptosis. The expression of Nrf2 and related enzymatic antioxidants downstream was enhanced by tBHQ in parallel. In conclusion, this investigation expanded the detailed understanding of the underlying mechanisms of HEMA-induced oxidative stress, and highlighted the cross-talk and interdependence between various Nrf2-regulated antioxidant pathways as a major adaptive cell response. The current results demonstrate that modulation of the Nrf2-mediated cellular defense response is an effective means for manipulating the sensitivity of cells to dental resin monomers.
Collapse
|
49
|
The potential benefits of red beetroot supplementation in health and disease. Nutrients 2015; 7:2801-22. [PMID: 25875121 PMCID: PMC4425174 DOI: 10.3390/nu7042801] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 03/17/2015] [Accepted: 04/09/2015] [Indexed: 12/15/2022] Open
Abstract
In recent years there has been a growing interest in the biological activity of red beetroot (Beta vulgaris rubra) and its potential utility as a health promoting and disease preventing functional food. As a source of nitrate, beetroot ingestion provides a natural means of increasing in vivo nitric oxide (NO) availability and has emerged as a potential strategy to prevent and manage pathologies associated with diminished NO bioavailability, notably hypertension and endothelial function. Beetroot is also being considered as a promising therapeutic treatment in a range of clinical pathologies associated with oxidative stress and inflammation. Its constituents, most notably the betalain pigments, display potent antioxidant, anti-inflammatory and chemo-preventive activity in vitro and in vivo. The purpose of this review is to discuss beetroot’s biological activity and to evaluate evidence from studies that specifically investigated the effect of beetroot supplementation on inflammation, oxidative stress, cognition and endothelial function.
Collapse
|
50
|
Mazzetti AP, Fiorile MC, Primavera A, Lo Bello M. Glutathione transferases and neurodegenerative diseases. Neurochem Int 2015; 82:10-8. [PMID: 25661512 DOI: 10.1016/j.neuint.2015.01.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 01/23/2015] [Accepted: 01/27/2015] [Indexed: 02/08/2023]
Abstract
There is substantial agreement that the unbalance between oxidant and antioxidant species may affect the onset and/or the course of a number of common diseases including Parkinson's and Alzheimer's diseases. Many studies suggest a crucial role for oxidative stress in the first phase of aging, or in the pathogenesis of various diseases including neurological ones. Particularly, the role exerted by glutathione and glutathione-related enzymes (Glutathione Transferases) in the nervous system appears more relevant, this latter tissue being much more vulnerable to toxins and oxidative stress than other tissues such as liver, kidney or muscle. The present review addresses the question by focusing on the results obtained by specimens from patients or by in vitro studies using cells or animal models related to Parkinson's and Alzheimer's diseases. In general, there is an association between glutathione depletion and Parkinson's or Alzheimer's disease. In addition, a significant decrease of glutathione transferase activity in selected areas of brain and in ventricular cerebrospinal fluid was found. For some glutathione transferase genes there is also a correlation between polymorphisms and onset/outcome of neurodegenerative diseases. Thus, there is a general agreement about the protective effect exerted by glutathione and glutathione transferases but no clear answer about the mechanisms underlying this crucial role in the insurgence of neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Mario Lo Bello
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|