101
|
Bayo Jimenez MT, Frenis K, Hahad O, Steven S, Cohen G, Cuadrado A, Münzel T, Daiber A. Protective actions of nuclear factor erythroid 2-related factor 2 (NRF2) and downstream pathways against environmental stressors. Free Radic Biol Med 2022; 187:72-91. [PMID: 35613665 DOI: 10.1016/j.freeradbiomed.2022.05.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/23/2022] [Accepted: 05/19/2022] [Indexed: 12/14/2022]
Abstract
Environmental risk factors, including noise, air pollution, chemical agents, ultraviolet radiation (UVR) and mental stress have a considerable impact on human health. Oxidative stress and inflammation are key players in molecular pathomechanisms of environmental pollution and risk factors. In this review, we delineate the impact of environmental risk factors and the protective actions of the nuclear factor erythroid 2-related factor 2 (NRF2) in connection to oxidative stress and inflammation. We focus on well-established studies that demonstrate the protective actions of NRF2 and its downstream pathways against different environmental stressors. State-of-the-art mechanistic considerations on NRF2 signaling are discussed in detail, e.g. classical concepts like KEAP1 oxidation/electrophilic modification, NRF2 ubiquitination and degradation. Specific focus is also laid on NRF2-dependent heme oxygenase-1 induction with detailed presentation of the protective down-stream pathways of heme oxygenase-1, including interaction with BACH1 system. The significant impact of all environmental stressors on the circadian rhythm and the interactions of NRF2 with the circadian clock will also be considered here. A broad range of NRF2 activators is discussed in relation to environmental stressor-induced health side effects, thereby suggesting promising new mitigation strategies (e.g. by nutraceuticals) to fight the negative effects of the environment on our health.
Collapse
Affiliation(s)
- Maria Teresa Bayo Jimenez
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Katie Frenis
- Department of Hematology and Oncology, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Omar Hahad
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Leibniz Insitute for Resilience Research (LIR), Mainz, Germany
| | - Sebastian Steven
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Guy Cohen
- The Skin Research Institute, The Dead Sea and Arava Science Center, Masada, 86910, Israel; Ben Gurion University of the Negev, Eilat Campus, Eilat, 8855630, Israel
| | - Antonio Cuadrado
- Departamento de Bioquímica, Facultad de Medicina, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Instituto de Investigaciones Biomédicas 'Alberto Sols' UAM-CSIC, Universidad Autónoma de Madrid, Madrid, Spain
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| |
Collapse
|
102
|
Zhang S, Duan S, Xie Z, Bao W, Xu B, Yang W, Zhou L. Epigenetic Therapeutics Targeting NRF2/KEAP1 Signaling in Cancer Oxidative Stress. Front Pharmacol 2022; 13:924817. [PMID: 35754474 PMCID: PMC9218606 DOI: 10.3389/fphar.2022.924817] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/18/2022] [Indexed: 02/05/2023] Open
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) and its negative regulator kelch-like ECH-associated protein 1 (KEAP1) regulate various genes involved in redox homeostasis, which protects cells from stress conditions such as reactive oxygen species and therefore exerts beneficial effects on suppression of carcinogenesis. In addition to their pivotal role in cellular physiology, accumulating innovative studies indicated that NRF2/KEAP1-governed pathways may conversely be oncogenic and cause therapy resistance, which was profoundly modulated by epigenetic mechanism. Therefore, targeting epigenetic regulation in NRF2/KEAP1 signaling is a potential strategy for cancer treatment. In this paper, the current knowledge on the role of NRF2/KEAP1 signaling in cancer oxidative stress is presented, with a focus on how epigenetic modifications might influence cancer initiation and progression. Furthermore, the prospect that epigenetic changes may be used as therapeutic targets for tumor treatment is also investigated.
Collapse
Affiliation(s)
- Shunhao Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sining Duan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhuojun Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wanlin Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Xu
- Department of Stomatology, Panzhihua Central Hospital, Panzhihua, China
| | - Wenbin Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, Department of Medical Affairs, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lingyun Zhou
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
103
|
Datta S, Ghosh S, Bishayee A, Sinha D. Flexion of Nrf2 by tea phytochemicals: A review on the chemopreventive and chemotherapeutic implications. Pharmacol Res 2022; 182:106319. [PMID: 35732198 DOI: 10.1016/j.phrs.2022.106319] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 01/11/2023]
Abstract
Nuclear factor erythroid 2 [NF-E2]-related factor 2 (Nrf2), the redox-sensitive transcription factor, plays a key role in stress-defense and detoxification. Nrf2 is tightly controlled by its negative regulator cum sensor Kelch-[ECH]-associated protein 1 (Keap1). Nrf2 is well known for its dual nature owing to its cancer preventive and cancer promoting abilities. Modulation of this biphasic nature of Nrf2 signaling by phytochemicals may be a potential cancer preventive and anticancer therapeutic strategy. Phytocompounds may either act as Nrf2-activator or Nrf2-inhibitor depending on their differential concentration and varied cellular environment. Tea is not just the most popular global beverage with innumerable health-benefits but has well-established chemopreventive and chemotherapeutic effects. Various types of tea infusions contain a wide range of bioactive compounds, such as polyphenolic catechins and flavonols, which are endowed with potent antioxidant properties. Despite of their rapid biotransformation and poor bioavailability, regular tea consumption is risk-reductive for several cancer forms. Tea catechins show their dual Nrf2-modulatory effect by directly acting on Nrf2-Keap1 or their upstream regulators and downstream effectors in a highly case-specific manner. In this review, we have tried to present a comprehensive evaluation of the Nrf2-mediated chemopreventive and chemotherapeutic applications of tea in various preclinical cancer models, the Nrf2-modulatory mechanisms, and the limitations which need to be addressed in future research.
Collapse
Affiliation(s)
- Suchisnigdha Datta
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata - 700 026, West Bengal, India
| | - Sukanya Ghosh
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata - 700 026, West Bengal, India
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| | - Dona Sinha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata - 700 026, West Bengal, India.
| |
Collapse
|
104
|
Suzen S, Tucci P, Profumo E, Buttari B, Saso L. A Pivotal Role of Nrf2 in Neurodegenerative Disorders: A New Way for Therapeutic Strategies. Pharmaceuticals (Basel) 2022; 15:ph15060692. [PMID: 35745610 PMCID: PMC9227112 DOI: 10.3390/ph15060692] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/22/2022] [Accepted: 05/28/2022] [Indexed: 02/04/2023] Open
Abstract
Clinical and preclinical research indicates that neurodegenerative diseases are characterized by excess levels of oxidative stress (OS) biomarkers and by lower levels of antioxidant protection in the brain and peripheral tissues. Dysregulations in the oxidant/antioxidant balance are known to be a major factor in the pathogenesis of neurodegenerative diseases and involve mitochondrial dysfunction, protein misfolding, and neuroinflammation, all events that lead to the proteostatic collapse of neuronal cells and their loss. Nuclear factor-E2-related factor 2 (Nrf2) is a short-lived protein that works as a transcription factor and is related to the expression of many cytoprotective genes involved in xenobiotic metabolism and antioxidant responses. A major emerging function of Nrf2 from studies over the past decade is its role in resistance to OS. Nrf2 is a key regulator of OS defense and research supports a protective and defending role of Nrf2 against neurodegenerative conditions. This review describes the influence of Nrf2 on OS and in what way Nrf2 regulates antioxidant defense for neurodegenerative conditions. Furthermore, we evaluate recent research and evidence for a beneficial and potential role of specific Nrf2 activator compounds as therapeutic agents.
Collapse
Affiliation(s)
- Sibel Suzen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Tandogan, 06100 Ankara, Turkey
- Correspondence: ; Tel.: +90-533-391-5844
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy;
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (E.P.); (B.B.)
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (E.P.); (B.B.)
| | - Luciano Saso
- Department of Physiology and Pharmacology ‘‘Vittorio Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy;
| |
Collapse
|
105
|
Zhou N, Tian Y, Liu W, Tu B, Xu W, Gu T, Zou K, Lu L. Protective Effects of Resveratrol and Apigenin Dietary Supplementation on Serum Antioxidative Parameters and mRNAs Expression in the Small Intestines of Diquat-Challenged Pullets. Front Vet Sci 2022; 9:850769. [PMID: 35711792 PMCID: PMC9196582 DOI: 10.3389/fvets.2022.850769] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Poultry as a large-scale intensive farming is vulnerable to oxidative stress. Resveratrol and apigenin are recognized to have many beneficial bioactive functions. This study tested the hypothesis that dietary resveratrol and apigenin supplementation alleviates oxidative stress in the small intestine of diquat-challenged pullets. A total of 200 healthy pullets were randomly divided into four treatment groups: control group fed with a basal diet (CON), diquat group fed with a basal diet (DIQ), resveratrol group fed with a basal diet containing 500 mg/kg resveratrol (RES), and an apigenin group fed with a basal diet containing 500 mg/kg apigenin (API) and injected intraperitoneally with either 1 ml of saline (CON) or 8 mg/kg body weight of diquat (DIQ, RES, and API) to induce oxidative stress. The day of the injection was considered as day 0. The results indicated that resveratrol and apigenin were able to decrease the malondialdehyde (MDA) level and upregulate total antioxidant capacity (T-AOC), superoxide dismutase (SOD), and glutathione peroxidase (GSH-PX) levels in serum on day 1 and 10 after being diquat-challenged. In addition, resveratrol increased mRNA expression of NQO1 (NAD(P)H dehydrogenase quinone 1) and HO-1 (heme oxygenase-1) in ileum and jejunum on day 10, while apigenin upregulated nuclear factor erythroid 2-related factor 2 (NRF2), NQO1, and HO-1 in ileum and jejunum on day 10. Both resveratrol and apigenin increased the mRNA expression of CLAUDIN-1 in ileum and jejunum on day 1 and that of ZO-1 (zonula occludens-1) in ileum on day 10 post-diquat-injection. These findings indicate that dietary supplementation with resveratrol and apigenin attenuates oxidative stress involving NRF2 signaling pathways in diquat-challenged pullets to some extent. These observations are valuable for the chicken industry and resveratrol and apigenin applications in animal husbandry.
Collapse
Affiliation(s)
- Ning Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Wenchao Liu
- Huzhou Lvchang Ecoagriculture Co., Ltd., Huzhou, China
| | - Bingjiang Tu
- Animal Disease Prevention and Control Center, Huzhou Wuxing District Agricultural and Rural Bureau, Huzhou, China
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Kang Zou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Kang Zou
| | - Lizhi Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science and Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
- Lizhi Lu
| |
Collapse
|
106
|
Zhang L, Hou N, Chen B, Kan C, Han F, Zhang J, Sun X. Post-Translational Modifications of p53 in Ferroptosis: Novel Pharmacological Targets for Cancer Therapy. Front Pharmacol 2022; 13:908772. [PMID: 35685623 PMCID: PMC9171069 DOI: 10.3389/fphar.2022.908772] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/10/2022] [Indexed: 12/21/2022] Open
Abstract
The tumor suppressor p53 is a well-known cellular guardian of genomic integrity that blocks cell cycle progression or induces apoptosis upon exposure to cellular stresses. However, it is unclear how the remaining activities of p53 are regulated after the abrogation of these routine activities. Ferroptosis is a form of iron- and lipid-peroxide-mediated cell death; it is particularly important in p53-mediated carcinogenesis and corresponding cancer prevention. Post-translational modifications have clear impacts on the tumor suppressor function of p53. Here, we review the roles of post-translational modifications in p53-mediated ferroptosis, which promotes the elimination of tumor cells. A thorough understanding of the p53 functional network will be extremely useful in future strategies to identify pharmacological targets for cancer therapy.
Collapse
Affiliation(s)
- Le Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Bing Chen
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
107
|
Regulation of BDNF transcription by Nrf2 and MeCP2 ameliorates MPTP-induced neurotoxicity. Cell Death Dis 2022; 8:267. [PMID: 35595779 PMCID: PMC9122988 DOI: 10.1038/s41420-022-01063-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 05/06/2022] [Accepted: 05/12/2022] [Indexed: 12/31/2022]
Abstract
Mounting evidence suggests the key role of brain-derived neurotrophic factor (BDNF) in the dopaminergic neurotoxicity of Parkinson’s disease (PD). Activation of NF-E2-related factor-2 (Nrf2) and inhibition of methyl CpG-binding protein 2 (MeCP2) can regulate BDNF upregulation. However, the regulation of BDNF by Nrf2 and MeCP2 in the PD pathogenesis has not been reported. Here, we revealed that Nrf2/MeCP2 coordinately regulated BDNF transcription, reversing the decreased levels of BDNF expression in 1-methyl-4-phenylpyridinium (MPP+)-treated SH-SY5Y cells and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. Repeated administration of sulforaphane (SFN, an Nrf2 activator) attenuated dopaminergic neurotoxicity in MPTP-treated mice through activation of BDNF and suppression of MeCP2 expression. Furthermore, intracerebroventricular injection of MeCP2-HDO, a DNA/RNA heteroduplex oligonucleotide (HDO) silencing MeCP2 expression, ameliorated dopaminergic neurotoxicity in MPTP-treated mice via activation of Nrf2 and BDNF expression. Moreover, we found decreased levels of Nrf2 and BDNF, and increased levels of MeCP2 protein expression in the striatum of patients with dementia with Lewy bodies (DLB). Interesting, there were correlations between BDNF and Nrf2 (or MeCP2) expression in the striatum from DLB patients. Therefore, it is likely that the activation of BDNF transcription by activation of Nrf2 and/or suppression of MeCP2 could be a new therapeutic approach for PD.
Collapse
|
108
|
Wang L, Liu X, Kang Q, Pan C, Zhang T, Feng C, Chen L, Wei S, Wang J. Nrf2 Overexpression Decreases Vincristine Chemotherapy Sensitivity Through the PI3K-AKT Pathway in Adult B-Cell Acute Lymphoblastic Leukemia. Front Oncol 2022; 12:876556. [PMID: 35646695 PMCID: PMC9134735 DOI: 10.3389/fonc.2022.876556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/19/2022] [Indexed: 01/08/2023] Open
Abstract
Uncontrolled proliferation is an important cancer cell biomarker, which plays a critical role in carcinogenesis, progression and development of resistance to chemotherapy. An improved understanding of novel genes modulating cancer cell proliferation and mechanism will help develop new therapeutic strategies. The nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor, decreases apoptosis when its expression is upregulated. However, the relationship between Nrf2 and Vincristine (VCR) chemotherapy resistance in B-cell acute lymphoblastic leukemia (B-ALL) is not yet established. Our results showed that Nrf2 levels could sufficiently modulate the sensitivity of B-ALL cells to VCRby regulating an apoptotic protein, i.e., the Bcl-2 agonist of cell death (BAD). Chemotherapeutic agents used for the treatment of B-ALL induced Nrf2 overactivation and PI3K-AKT pathway activation in the cells, independent of the resistance to chemotherapy; thus, a potential resistance loop during treatment for B-ALL with a drug combination is established. Therefore, B-ALL patients with a high expression of Nrf2 might mean induction chemotherapy with VCR effective little.
Collapse
Affiliation(s)
- Li Wang
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Xin Liu
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Qian Kang
- Department of Hematology, Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Haematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chengyun Pan
- Department of Hematology, Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Haematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tianzhuo Zhang
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Cheng Feng
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Lu Chen
- Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Sixi Wei
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jishi Wang
- Department of Hematology, Guizhou Province Institute of Hematology, Guizhou Province Laboratory of Haematopoietic Stem Cell Transplantation Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Jiangsu, China
| |
Collapse
|
109
|
Cao D, Wang Y, Li W, Ji J, Guo J, Zhang D, Liu J. 3,4‑Dihydroxyacetophenone attenuates oxidative stress‑induced damage to HUVECs via regulation of the Nrf2/HO‑1 pathway. Mol Med Rep 2022; 25:199. [PMID: 35475506 PMCID: PMC9073850 DOI: 10.3892/mmr.2022.12715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/04/2022] [Indexed: 11/08/2022] Open
Abstract
It has been reported that oxidative stress plays a prominent role in diabetic macrovascular diseases. 3,4-Dihydroxyacetophenone (3,4-DHAP) has been found to have a variety of biological activities. However, few studies have assessed the antioxidant capacity of 3,4-DHAP and the underlying mechanisms. Thus, the aim of the present study was to explore the effects of 3,4-DHAP on oxidative stress in human umbilical vein endothelial cells (HUVECs). HUVECs were pre-treated with 3,4-DHAP and then exposed to high glucose conditions. Cell viability and cytotoxicity were measured using an MTT assay. Reactive oxygen species (ROS) levels were measured using an inverted fluorescence microscope and a fluorescent enzyme labeling instrument. Protein expression levels of nuclear factor E2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), microtubule-associated protein 1A/1B-light chain 3 (LC3) and poly ADP-ribose polymerase-1 (PARP-1) were measured using western blotting, and mRNA expression of Nrf2 and HO-1 were measured through reverse transcription-quantitative PCR (RT-qPCR). Nrf2 nuclear translocation was evaluated using immunofluorescence analysis and autophagosomes were observed using transmission electron microscope (TEM). The results of the present study demonstrated that compared with the control group, cell viability of the high glucose group was reduced and cell cytotoxicity of the high glucose group was increased. ROS production in the high glucose group was clearly enhanced. In addition, high glucose upregulated Nrf2 and HO-1 protein and mRNA expression levels. Nuclear translocation of Nrf2 in the high glucose group was also increased. The formation of autophagosomes in the high glucose group was also higher than that in the control group. Furthermore, LC3-II/LC3-I and PARP-1 protein expression levels were increased after treatment with high glucose. However, compared to the high glucose group, 3,4-DHAP (10 µmol/l) significantly enhanced cell viability. 3,4-DHAP markedly decreased the production of ROS, increased Nrf2 and HO-1 protein and mRNA expression levels, and promoted nuclear translocation of Nrf2 in HUVECs. In addition, 3,4-DHAP promoted the formation of autophagosomes, and notably increased the protein expression levels of LC3-II/LC3-I and PARP-1. Moreover, it was determined that compared to the 3,4-DHAP group, treatment with 3,4-DHAP and ML385 enhanced cell viability, and decreased ROS production, Nrf2 and HO-1 protein and mRNA expression levels, nuclear translocation of Nrf2, and LC3-II/LC3-I and PARP-1 protein expression levels. Collectively, the results of the present study showed that 3,4-DHAP protected HUVECs against oxidative stress via regulation of the Nrf2/HO-1 pathway, by increasing autophagy and promoting DNA damage repair.
Collapse
Affiliation(s)
- Daihong Cao
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Yunhan Wang
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Wentao Li
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Jiafen Ji
- Department of Pediatrics, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Juntang Guo
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Daijuan Zhang
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Jiangyue Liu
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
110
|
Liu H, Zhao L, Wang M, Yang K, Jin Z, Zhao C, Shi G. FNDC5 Causes Resistance to Sorafenib by Activating the PI3K/Akt/Nrf2 Pathway in Hepatocellular Carcinoma Cells. Front Oncol 2022; 12:852095. [PMID: 35392237 PMCID: PMC8980859 DOI: 10.3389/fonc.2022.852095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/28/2022] [Indexed: 01/16/2023] Open
Abstract
In this study, we aimed to reveal the resistance mechanism of hepatocellular carcinoma (HCC) cells to sorafenib by exploring the effect of FNDC5 on sorafenib-induced ferroptosis in HCC cells. We compared the expression level of FNDC5 between sorafenib-resistant and sorafenib-sensitive HCC cell lines and the level of ferroptosis between the groups after treatment with sorafenib. We knocked down FNDC5 in drug-resistant cell lines and overexpressed it in sorafenib-sensitive HCC cell lines to further demonstrate the role of FNDC5 in sorafenib-induced ferroptosis. Using PI3K inhibitors, we revealed the specific mechanism by which FNDC5 functions. In addition, we verified our findings obtained in in vitro experiments using a subcutaneous tumorigenic nude mouse model. The findings revealed that FNDC5 inhibits sorafenib-induced ferroptosis in HCC cells. In addition, FNDC5 activated the PI3K/Akt pathway, which in turn promoted the nuclear translocation of Nrf2 and increased the intracellular antioxidant response, thereby conferring resistance to ferroptosis. Our study provides novel insights for improving the efficacy of sorafenib.
Collapse
Affiliation(s)
- Huayuan Liu
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Lei Zhao
- Department of Infection Management, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Mengya Wang
- Department of Physiology, School of Basic Medicine, Institute of Brain Science and Disorders, Qingdao University, Qingdao, China
| | - Kexin Yang
- Department of Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Zhipeng Jin
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Chengjian Zhao
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Guangjun Shi
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
111
|
Sun Y, Wan W, Zhao X, Han X, Ye T, Chen X, Ran Q, Wang X, Liu X, Qu C, Shi S, Zhang C, Yang B. Chronic Sigma 1 receptor activation alleviates right ventricular dysfunction secondary to pulmonary arterial hypertension. Bioengineered 2022; 13:10843-10856. [PMID: 35473584 PMCID: PMC9208487 DOI: 10.1080/21655979.2022.2065953] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Sigma 1 receptor (S1R) has shown a preferable protective effect on left ventricular function, but whether it protects right ventricular (RV) function is still elusive.This study aimed to determine the effects of S1R on RV dysfunction secondary to pulmonary arterial hypertension.Sixty wild-type male Sprague–Dawley rats were randomly divided into the control group, the fluvoxamine group, the pulmonary arterial hypertension group and the pulmonary arterial hypertension combined with fluvoxamine group. Monocrotaline (60 mg/kg) was administered to induce pulmonary arterial hypertension, and fluvoxamine was given for 21 consecutive days to activate S1R after one week of monocrotaline administration. Echocardiographic, serologic, and histologic parameters, qRT-PCR, and western blotting were conducted after 4 weeks of monocrotaline administration.The expression of S1R was decreased in the right ventricle in pulmonary arterial hypertension. TAPSE, and the FAC of the right ventricle were significantly decreased, and RV EDP and the plasma concentration of N-terminal pro-B-type natriuretic peptide was increased in the pulmonary arterial hypertension group, but fluvoxamine partly restored those abnormalities (all P < 0.05). Moreover, pulmonary arteriole remodeling, and fibrosis and hypertrophy in the RV were shown in the pulmonary arterial hypertension group; interestingly, fluvoxamine recovered RV structural remodeling (all P < 0.05) but neither alleviated pulmonary arteriole remodeling nor reduced pulmonary artery pressure. Furthermore, S1R activation protects RV function by upgrading the NRF 2/HO 1-mediated antioxidant stress pathway. In conclusion, chronic S1R activation ameliorates structural remodeling and RV dysfunction secondary to pulmonary arterial hypertension without altering pulmonary artery pressure.
Collapse
Affiliation(s)
- Yazhou Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Weiguo Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xin Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Tianxin Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xiaoli Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Qian Ran
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Cui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| |
Collapse
|
112
|
Yamaguchi A, Maeshige N, Ma X, Uemura M, Noguchi H, Matsuda M, Nishimura Y, Hasunuma T, Kondo H, Fujino H. Pulsed-Ultrasound Irradiation Induces the Production of Itaconate and Attenuates Inflammatory Responses in Macrophages. J Inflamm Res 2022; 15:2387-2395. [PMID: 35444446 PMCID: PMC9013924 DOI: 10.2147/jir.s361609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/07/2022] [Indexed: 12/30/2022] Open
Abstract
Background Itaconate is a key metabolite in the innate immune system and exerts strong anti-inflammatory effects in macrophages. For the production of itaconate in macrophages, immune-responsive gene 1 (IRG1) is an imperative enzyme, and activating the IRG1-itaconate pathway is reported to alleviate inflammatory diseases by upregulating nuclear factor-erythroid 2-related factor 2 (NRF2). However, there are very few reports on strategies to increase itaconate production. Ultrasound therapy is a widely used intervention for anti-inflammatory and soft-tissue regeneration purposes. Here we show the effect of ultrasound irradiation on the production of itaconate in macrophages. Methods Murine bone marrow-derived macrophages (BMDMs) were exposed to pulsed ultrasound (3.0 W/cm2) for 5 minutes. Three hours after irradiation, the intracellular levels of metabolites and mRNA expression levels of Irg1 and Nrf2 were measured using CE/MS and qPCR, respectively. To evaluate macrophage inflammation status, 3 h after irradiation, the cells were stimulated with 100 ng/mL lipopolysaccharide (LPS) for 1.5 h and the mRNA expression levels of pro-inflammatory factors (Il-1β, Il-6, and Tnf-α) were measured. Student’s t-test, one-way ANOVA and Tukey’s multiple comparison test were used for statistical processing, and the significance level was set to less than 5%. Results Ultrasound irradiation significantly increased the intracellular itaconate level and the expression levels of Irg1 and Nrf2 in BMDMs. Upregulation of Il-1β, Il-6, and Tnf-α by LPS was significantly suppressed in BMDMs treated with ultrasound. Ultrasound irradiation did not affect cell viability and apoptosis. Conclusion Ultrasound irradiation induces the production of itaconate by upregulating Irg1 expression and attenuates inflammatory responses in macrophages via Nrf2. These results suggest that ultrasound is a potentially useful method to increase itaconate production in macrophages.
Collapse
Affiliation(s)
- Atomu Yamaguchi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Noriaki Maeshige
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
- Correspondence: Noriaki Maeshige, Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Kobe, 654-0142, Hyogo, Japan, Tel +81 78 796 4582, Email
| | - Xiaoqi Ma
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Mikiko Uemura
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Hikari Noguchi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Mami Matsuda
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
| | - Yuya Nishimura
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
| | - Tomohisa Hasunuma
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
- Engineering Biology Research Center, Kobe University, Kobe, Japan
| | - Hiroyo Kondo
- Department of Food Science and Nutrition, Nagoya Women’s University, Nagoya, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
113
|
Liu B, Li X, Wang D, Yu Y, Lu D, Chen L, Lv F, Li Y, Cheng L, Song Y, Xing Y. CEMIP promotes extracellular matrix-detached prostate cancer cells survival by inhibiting ferroptosis. Cancer Sci 2022; 113:2056-2070. [PMID: 35363929 PMCID: PMC9207355 DOI: 10.1111/cas.15356] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 11/28/2022] Open
Abstract
Cells detached from the extracellular matrix (ECM) can trigger different modes of cell death, and the survival of ECM‐detached cells is one of the prerequisites for the metastatic cascade. Ferroptosis, a form of iron‐dependent programmed cell death, has recently been found to be involved in matrix‐detached cancer cells. However, the molecular mechanisms by which ECM‐detached cells escape ferroptosis are not fully understood. Here, we observed that cell migration‐inducing protein (CEMIP) upregulation facilitates ferroptosis resistance during ECM detachment by promoting cystine uptake in prostate cancer (PCa) cells. Meanwhile, silencing CEMIP causes it to lose its ability to promote cystine uptake and inhibit ferroptosis. Mechanistically, the interaction of CEMIP with inositol 1,4,5‐trisphosphate receptor type 3 (ITPR3) modulates calcium ion (Ca2+) leakage from the endoplasmic reticulum, activating calcium/calmodulin‐dependent protein kinase II (CaMKII), which further facilitates nuclear factor erythroid 2‐related factor 2 (NRF2) phosphorylation and nuclear localization, leading to elevated transcription of solute carrier family 7 member 11 (SLC7A11), a glutamate/cystine antiporter, in PCa cells. Our findings delineate a novel role of CEMIP in ferroptosis resistance during ECM detachment and provide new insights into therapeutic strategies for metastatic PCa.
Collapse
Affiliation(s)
- Bing Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuexiang Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Decai Wang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ying Yu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430061, China
| | - Dingheng Lu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liang Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang Lv
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yunxue Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lulin Cheng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yarong Song
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yifei Xing
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
114
|
Baicalein and Αlpha-Tocopherol Inhibit Toll-like Receptor Pathways in Cisplatin-Induced Nephrotoxicity. Molecules 2022; 27:molecules27072179. [PMID: 35408581 PMCID: PMC9000769 DOI: 10.3390/molecules27072179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/13/2022] [Accepted: 03/14/2022] [Indexed: 02/05/2023] Open
Abstract
Cisplatin (CP) is a conventional chemotherapeutic agent with serious adverse effects. Its toxicity was linked to the stimulation of oxidative stress and inflammation. As a result, this study explored the protective effect of baicalein and alpha-tocopherol in nephrotoxicity induced by cisplatin. Until receiving an intraperitoneal injection of CP (3 mg/kg BW), rats were given baicalein orally 100 mg/kg for seven days or/and a single intraperitoneal injection of α-tocopherol 250 mg/kg. Renal function was tested to explore whether baicalein and α-tocopherol have any beneficial effects; blood urea nitrogen (BUN), serum creatinine, malondialdehyde (MDA) content, antioxidant activity biomarkers and histopathology of renal tissue, oxidative stress biomarkers, inflammatory response markers, and histopathological features of kidney architecture were measured. Cisplatin treatment resulted in extreme renal failure, as measured by high serum creatinine and BUN levels and severe renal changes. Cisplatin therapy resulted in increased lipid peroxidation and decreased glutathione and superoxide dismutase levels, reflecting oxidative stress. Upon treatment with α-tocopherol, baicalein, and combined therapy, there was augmentation in the antioxidant status as well as a reduction in IL-6, NF-κB, TNF, TLR2, and TLR4 and a significant increase in Keap-1 and NRF-2. The combined treatment was the most effective and the nearest to the normal status. These findings suggest that baicalein and α-tocopherol may be useful in preventing cisplatin-induced nephrotoxicity.
Collapse
|
115
|
Wu X, Zhang Q, Guo Y, Zhang H, Guo X, You Q, Wang L. Methods for the Discovery and Identification of Small Molecules Targeting Oxidative Stress-Related Protein–Protein Interactions: An Update. Antioxidants (Basel) 2022; 11:antiox11040619. [PMID: 35453304 PMCID: PMC9025695 DOI: 10.3390/antiox11040619] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
The oxidative stress response pathway is one of the hotspots of current pharmaceutical research. Many proteins involved in these pathways work through protein–protein interactions (PPIs). Hence, targeting PPI to develop drugs for an oxidative stress response is a promising strategy. In recent years, small molecules targeting protein–protein interactions (PPIs), which provide efficient methods for drug discovery, are being investigated by an increasing number of studies. However, unlike the enzyme–ligand binding mode, PPIs usually exhibit large and dynamic binding interfaces, which raise additional challenges for the discovery and optimization of small molecules and for the biochemical techniques used to screen compounds and study structure–activity relationships (SARs). Currently, multiple types of PPIs have been clustered into different classes, which make it difficult to design stationary methods for small molecules. Deficient experimental methods are plaguing medicinal chemists and are becoming a major challenge in the discovery of PPI inhibitors. In this review, we present current methods that are specifically used in the discovery and identification of small molecules that target oxidative stress-related PPIs, including proximity-based, affinity-based, competition-based, structure-guided, and function-based methods. Our aim is to introduce feasible methods and their characteristics that are implemented in the discovery of small molecules for different types of PPIs. For each of these methods, we highlight successful examples of PPI inhibitors associated with oxidative stress to illustrate the strategies and provide insights for further design.
Collapse
Affiliation(s)
- Xuexuan Wu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yuqi Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hengheng Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoke Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (X.G.); (Q.Y.); (L.W.); Tel.: +86-025-83271351 (Q.Y.); +86-15261483858 (L.W.)
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (X.G.); (Q.Y.); (L.W.); Tel.: +86-025-83271351 (Q.Y.); +86-15261483858 (L.W.)
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (X.G.); (Q.Y.); (L.W.); Tel.: +86-025-83271351 (Q.Y.); +86-15261483858 (L.W.)
| |
Collapse
|
116
|
Multifaceted Roles of the KEAP1–NRF2 System in Cancer and Inflammatory Disease Milieu. Antioxidants (Basel) 2022; 11:antiox11030538. [PMID: 35326187 PMCID: PMC8944524 DOI: 10.3390/antiox11030538] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
In a multicellular environment, many different types of cells interact with each other. The KEAP1–NRF2 system defends against electrophilic and oxidative stresses in various types of cells. However, the KEAP1–NRF2 system also regulates the expression of genes involved in cell proliferation and inflammation, indicating that the system plays cell type-specific roles. In this review, we introduce the multifarious roles of the KEAP1–NRF2 system in various types of cells, especially focusing on cancer and inflammatory diseases. Cancer cells frequently hijack the KEAP1–NRF2 system, and NRF2 activation confers cancer cells with a proliferative advantage and therapeutic resistance. In contrast, the activation of NRF2 in immune cells, especially in myeloid cells, suppresses tumor development. In chronic inflammatory diseases, such as sickle cell disease, NRF2 activation in myeloid and endothelial cells represses the expression of proinflammatory cytokine and adherent molecule genes, mitigating inflammation and organ damage. Based on these cell-specific roles played by the KEAP1–NRF2 system, NRF2 inducers have been utilized for the treatment of inflammatory diseases. In addition, the use of NRF2 inducers and/or inhibitors with canonical antineoplastic drugs is an emerging approach to cancer treatment.
Collapse
|
117
|
Li S, Yuan J, Che S, Zhang L, Ruan Z, Sun X. Decabromodiphenyl ether induces ROS-mediated intestinal toxicity through the Keap1-Nrf2 pathway. J Biochem Mol Toxicol 2022; 36:e22995. [PMID: 35266255 DOI: 10.1002/jbt.22995] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 11/30/2021] [Accepted: 01/28/2022] [Indexed: 12/22/2022]
Abstract
Polybrominated diphenyl ethers (PBDEs) are widely used brominated flame retardants as commercial products. PBDEs have been demonstrated to induce hepatic, reproductive, neural, and thyroid toxicity effects. This study aimed to clarify the potential intestinal toxicity effects of decabrominated diphenyl ether (PBDE-209) in vivo and in vitro. First, we investigated the change of PBDE-209 on oxidative stress in the intestine of mice. Subsequently, the potential toxicity mechanism of PBDE-209 in vitro was investigated. Caco-2 cells were treated with different concentrations of PBDE-209 (1, 5, and 25 μmol/L) for 24 and 48 h. We determined the cell viability, reactive oxygen species (ROS) level, multiple cellular parameters, and relative mRNA expressions. The results showed that PBDE-209 significantly injured the colon of mice, increased the intestinal levels of malondialdehyde (MDA), and changed the antioxidant enzyme activities. PBDE-209 inhibited the proliferation and induced cytotoxicity of Caco-2 cells. The change in ROS production and mitochondrial membrane potential (MMP) revealed that PBDE-209 caused oxidative stress in Caco-2 cells. The real-time PCR assays revealed that PBDE-209 inhibited the mRNA expression level of antioxidative defense factor, nuclear factor erythroid 2-related factor 2 (Nrf2). Furthermore, the FAS and Cytochrome P450 1A1 (CYP1A1) mRNA expression levels were increased in Caco-2 cells. These results suggested that PBDE-209 exerts intestinal toxicity effects in vivo and in vitro and inhibits the antioxidative defense gene expression in Caco-2 cells. This study provides an opportunity to advance the understanding of toxicity by the persistent environmental pollutant PBDE-209 to the intestine.
Collapse
Affiliation(s)
- Shiqi Li
- State Key Laboratory of Food Science and Technology, Institute of Nutrition and School of Food Science, Nanchang University, Nanchang, China
| | - Jinwen Yuan
- State Key Laboratory of Food Science and Technology, Institute of Nutrition and School of Food Science, Nanchang University, Nanchang, China
| | - Siyan Che
- State Key Laboratory of Food Science and Technology, Institute of Nutrition and School of Food Science, Nanchang University, Nanchang, China
| | - Li Zhang
- State Key Laboratory of Food Science and Technology, Institute of Nutrition and School of Food Science, Nanchang University, Nanchang, China
| | - Zheng Ruan
- State Key Laboratory of Food Science and Technology, Institute of Nutrition and School of Food Science, Nanchang University, Nanchang, China
| | - Xiaoming Sun
- State Key Laboratory of Food Science and Technology, Institute of Nutrition and School of Food Science, Nanchang University, Nanchang, China
| |
Collapse
|
118
|
Zhou N, Tian Y, Liu W, Tu B, Gu T, Xu W, Zou K, Lu L. Effects of quercetin and coated sodium butyrate dietary supplementation in diquat-challenged pullets. Anim Biosci 2022; 35:1434-1443. [PMID: 35240016 PMCID: PMC9449397 DOI: 10.5713/ab.21.0493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/02/2022] [Indexed: 11/27/2022] Open
Abstract
Objective This study was designed to investigate the hypothesis that dietary quercetin (QUE) and coated sodium butyrate (SB) supplementation alleviate oxidative stress in the small intestine of diquat (DIQ)-challenged pullets. Methods A total of 200 13-week-old pullets were divided into four groups: the control group (CON), the DIQ group, the QUE group, and the coated SB group, and injected intraperitoneally with either saline (CON) or diquat (DIQ, QUE, and SB) to induce oxidative stress on day 0. Results On the first day, the malondialdehyde and superoxide dismutase (SOD) concentrations in the SB group were significantly different from those in the DIQ and QUE groups (p<0.05), and dietary supplementation with SB increased serum glutathione peroxidase (GSH-PX) levels compared with the DIQ group (p<0.05). Quercetin and SB increased the levels of CLAUDIN-1 and zonula occludens-1 (ZO-1) in the jejunum. On the tenth day of treatment, QUE attenuated the decrease in GSH-PX levels compared to those of the CON group (p<0.05), while SB increased SOD, GSH-PX, and total antioxidant capacity levels compared to those of the DIQ group. Nuclear factor erythroid 2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1) mRNA levels in the QUE and SB groups increased (p<0.05) and CLAUDIN-1 mRNA levels in the QUE and SB groups were upregulated compared to those in the DIQ group ileum tissue. Conclusion Supplementation of QUE and SB demonstrated the ability to relieve oxidative stress in pullets post DIQ-injection with a time-dependent manner and QUE and SB may be potential antioxidant additives for relieving oxidative stress and protecting the intestinal barrier of pullets.
Collapse
Affiliation(s)
- Ning Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.,State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310000, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310000, China
| | - Wenchao Liu
- Huzhou Lvchang Ecoagriculture Co., Ltd. Huzhou, 313025, China
| | - Bingjiang Tu
- Huzhou Wuxing District Animal Disease Prevention and Control Center, Huzhou, 313000, China
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310000, China
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310000, China
| | - Kang Zou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Lizhi Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.,State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310000, China
| |
Collapse
|
119
|
Ahmadi R, Emami S. Recent applications of vinyl sulfone motif in drug design and discovery. Eur J Med Chem 2022; 234:114255. [DOI: 10.1016/j.ejmech.2022.114255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/20/2022] [Accepted: 03/03/2022] [Indexed: 01/10/2023]
|
120
|
Zhou Y, Lin W, Rao T, Zheng J, Zhang T, Zhang M, Lin Z. Ferroptosis and Its Potential Role in the Nervous System Diseases. J Inflamm Res 2022; 15:1555-1574. [PMID: 35264867 PMCID: PMC8901225 DOI: 10.2147/jir.s351799] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/09/2022] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis is a novel regulated cell death characterized by metabolic disorders and iron-dependent oxidative destruction of the lipid bilayer. It is primarily caused by the imbalance of oxidation and anti-oxidation in the body and is precisely regulated by numerous factors and pathways inside and outside the cell. Recent studies have indicated that ferroptosis plays a vital role in the pathophysiological process of multiple systems of the body including the nervous system. Ferroptosis may be closely linked to the occurrence and development of neurodegenerative diseases, strokes, and brain tumors. It may also be involved in the development, maturation, and aging of the nervous system. Therefore, this study aims to investigate ferroptosis’s occurrence and regulatory mechanism and summarize its research progress in the pathogenesis and treatment of neurological diseases. This would allow for novel ideas for basic and clinical research of neurological diseases.
Collapse
Affiliation(s)
- Yiyang Zhou
- Department of Pediatrics, The Second School of Medicine, Taizhou Women and Children’s Hospital of Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People’s Republic of China
| | - Wei Lin
- Department of Pediatrics, The Second School of Medicine, Taizhou Women and Children’s Hospital of Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People’s Republic of China
| | - Tian Rao
- Department of Pediatrics, The Second School of Medicine, Taizhou Women and Children’s Hospital of Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People’s Republic of China
| | - Jinyu Zheng
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People’s Republic of China
| | - Tianlei Zhang
- Department of Pediatrics, The Second School of Medicine, Taizhou Women and Children’s Hospital of Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People’s Republic of China
| | - Min Zhang
- Department of Pediatrics, The Second School of Medicine, Taizhou Women and Children’s Hospital of Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People’s Republic of China
| | - Zhenlang Lin
- Department of Pediatrics, The Second School of Medicine, Taizhou Women and Children’s Hospital of Wenzhou Medical University, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People’s Republic of China
- Correspondence: Zhenlang Lin, Email
| |
Collapse
|
121
|
Exercise and Oxidative Stress Biomarkers among Adult with Cancer: A Systematic Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2097318. [PMID: 35222792 PMCID: PMC8881118 DOI: 10.1155/2022/2097318] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/07/2021] [Accepted: 01/20/2022] [Indexed: 01/07/2023]
Abstract
Evidence shows that exercise can have a favourable effect in cancer patients. The exercise’s clinical benefits are likely to concern multiple interrelated biological pathways, among which oxidative stress plays a key role. Regular training can induce an adaptive response that strengthens the antioxidative status of the body. To formulate public health recommendations regarding the optimal exercise prescription for cancer patients, a detailed understanding is needed regarding the effect of exercise on variables linked to oxidative stress and antioxidant status of patients. The goal of this systematic review, based on PRISMA, was to explore and critically analyse the evidence regarding the efficacy of exercise on oxidative stress biomarkers among people with cancer. Study search was conducted in the following databases: PubMed, Cochrane, CINAHL, Embase, PEDro, and SPORTDiscus. The studies’ quality was assessed with the Cochrane risk-of-bias tool and STROBE scale. After identification and screening steps, 10 articles were included. The findings provide an encouraging picture of exercise, including resistance training and aerobic activities, in people with cancer. The exercise improved the indicators of the total antioxidant capacity, increased the antioxidant enzymes’ activity, or reduced the biomarkers of oxidative damage in various forms of cancer such as breast, lung, head, and neck. Regarding oxidative DNA damage, the role of exercise intervention has been difficult to assess. The heterogeneity of study design and the plethora of biomarkers measured hampered the comparison of the articles. This limited the possibility of establishing a comprehensive conclusion on the sensitivity of biomarkers to estimate the exercise’s benefits. Further high-quality studies are required to provide data regarding oxidative stress biomarkers responding to exercise. This information will be useful to assess the efficacy of exercise in people with cancer and support the appropriate prescription of exercise in anticancer strategy.
Collapse
|
122
|
Miao B, Zheng J, Zheng G, Tian X, Zhang W, Yuan F, Yang Z. Using Collagen Peptides From the Skin of Monkfish (Lophius litulon) to Ameliorate Kidney Damage in High-Fat Diet Fed Mice by Regulating the Nrf2 Pathway and NLRP3 Signaling. Front Nutr 2022; 9:798708. [PMID: 35223948 PMCID: PMC8866304 DOI: 10.3389/fnut.2022.798708] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Background Oxidative stress and inflammation play important roles in high-fat diet (HFD) induced kidney damage. Previous studies show that the collagen extracted from the skin of monkfish (Lophius litulon) with pepsin (pepsin-solubilized collagen, PSC) exhibits good biological activities. This study investigates the protective effect of PSCP against chronic kidney injury in HFD-fed mice. Methods Pepsin-solubilized collagen was further hydrolyzed into collagen peptides, and the compound with the best 2,2-diphenyl-1-picrylhydrazyl (DPPH) clearance rate was named pepsin-solubilized collagen peptide (PSCP). A group of mice were fed an HFD for 4 weeks, and then for another 6 weeks PSCP was added to their diet at the amount of either 100 or 200 mg/kg. Results Pepsin-solubilized collagen peptide treatment (200 mg/kg) reduced the mice's serum levels of uric acid (UA), creatinine (CRE), and blood urea nitrogen (BUN) by 27, 20, and 37%, respectively. This treatment also remarkably improved renal histopathology. Moreover, the activities of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) were increased by 96, 52, and 74%, respectively, and decreased the malondialdehyde (MDA) level by 36%. Additionally, PSCP activated the Nrf2 pathway and inhibited NLRP3 signaling to significantly reduce the levels of inflammatory cytokines IL-1β, IL-6, and TNF-α. Conclusions Our results indicate that compound PSCP has the potential to prevent or control chronic kidney damage.
Collapse
Affiliation(s)
- Bingtao Miao
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Jiawen Zheng
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Guoping Zheng
- Zhoushan Institute for Food and Drug Control, Zhoushan, China
| | - Xiaoxiao Tian
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Wen Zhang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Falei Yuan
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
- *Correspondence: Falei Yuan
| | - Zuisu Yang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
- Zuisu Yang
| |
Collapse
|
123
|
Sun Y, Liang C, Zheng L, Liu L, Li Z, Yang G, Li Y. Anti-fatigue effect of hypericin in a chronic forced exercise mouse model. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114767. [PMID: 34710555 DOI: 10.1016/j.jep.2021.114767] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hypericum perforatum L. is a traditional Chinese medicine used to sooth the liver, relieve depression, reduce body temperature, reduce sweating, and stimulate lactation. HP was extracted from Hypericum perforatum L. AIM OF STUDY The antifatigue effects of hypericin were assessed in a series of experiments. MATERIALS AND METHODS Six-to eight-week-old male ICR mice were raised in our lab. Mice were subjected to swimming training for 2 h, 6 days/week for 6 weeks. One hour prior to each swimming session, intraperitoneal injection of saline or HP (2 or 4 mg/kg) was performed. RESULTS Compared with the fatigue model control group, HP was found to significantly increase the swimming time in forced swimming tests. The molecular mechanisms underlying the antifatigue effects were further revealed by analysing energy metabolism, the oxidant-antioxidant system and the inflammatory response. HP normalized changes in BLA, LDH, BUN, and CK, LG in the liver. In addition, multiple assays have confirmed that HP improved the MDA, T-AOC, GSH-PX and SOD activity, and the relevant signalling pathways involved in the antifatigue effects were clarified. Furthermore, HP improves the expression of pro- and anti-inflammatory cytokines in skeletal muscle. CONCLUSION These results suggested that the anti-chronic fatigue effects of HP are likely achieved by normalizing energy metabolism and attenuating oxidative and inflammatory responses. Consequently, this study supports HP use in the clinic to alleviate chronic fatigue.
Collapse
Affiliation(s)
- Yang Sun
- School of Physical Education, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Chen Liang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Lihua Zheng
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Lei Liu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, 130024, China.
| | - Zhijin Li
- Xiamen Health and Medical Big Data Center (Xiamen Medicine Research Institute), Xiamen Key Laboratory of Natural Medicine Research and Development, Xiamen, Fujian, 361008, China.
| | - Guang Yang
- School of Physical Education, Northeast Normal University, Changchun, Jilin, 130024, China.
| | - Yuxin Li
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin, 130024, China
| |
Collapse
|
124
|
So B, Park J, Jang J, Lim W, Imdad S, Kang C. Effect of Aerobic Exercise on Oxidative Stress and Inflammatory Response During Particulate Matter Exposure in Mouse Lungs. Front Physiol 2022; 12:773539. [PMID: 35185596 PMCID: PMC8850364 DOI: 10.3389/fphys.2021.773539] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/03/2021] [Indexed: 11/22/2022] Open
Abstract
Regular exercise provides several health benefits that can improve the cardiovascular and musculoskeletal systems, but clear evidence on the effect of exercise-induced hyperventilation in particulate matter (PM) exposure is still lacking. This study aimed to investigate the effects of exercise in PM exposure on reactive oxygen species (ROS) generation, inflammatory response, and mitochondrial integrity in human lung epithelial cells (A549), as well as in mouse lung tissue. In in vitro experiments, PM treatment was shown to significantly increased ROS production, and reduced cell viability and mitochondrial function in A549 cells. The mice were divided into four groups for an in vivo exercise experiment: control (CON), PM inhalation (PI), PM inhalation during exercise (PIE), and exercise (EX) groups. The PI and PIE groups were exposed to 100 μg/m3 of PM for 1 h per day for a week. The PIE and EX groups performed treadmill exercises every day for 1 h at 20 m/min for a week. The levels of pro-inflammatory markers (IL-6 and TNF-α) were significantly higher in the PI group than in the CON group (P < 0.001 and P < 0.01, respectively). The carbonyl protein level was decreased in EX vs. PI (P < 0.001). Mitochondrial fission (Drp1) content was significantly decreased in the EX vs. CON group (P < 0.01), but anti-mitochondrial fission (P-Drp1 Ser637) was increased in the EX vs. PI group (P < 0.05). Mitochondrial autophagy (mitophagy), which is an assessment of mitochondrial integrity, was markedly increased in PI vs. CON (P < 0.001), but the level was reversed in PIE (P < 0.05). Lung fibrosis was increased in PI vs. CON group (P < 0.001), however, the cells were rescued in the PIE (P < 0.001). The number of apoptotic cells was remarkably increased in the PI vs. CON group (P < 0.001), whereas the level was decreased in the PIE (P < 0.001). Taken together, these results showed that short-term exposure to PM triggers oxidative stress, pro-inflammatory responses, and apoptosis in the lungs, but the PM-induced adverse effects on the lung tissue are not exacerbated by exercise-induced PM hyperventilation but rather has a protective effect.
Collapse
Affiliation(s)
- Byunghun So
- Molecular Metabolism in Health and Disease, Exercise Physiology Laboratory, Inha University, Incheon, South Korea
| | - Jinhan Park
- Molecular Metabolism in Health and Disease, Exercise Physiology Laboratory, Inha University, Incheon, South Korea
| | - Junho Jang
- Molecular Metabolism in Health and Disease, Exercise Physiology Laboratory, Inha University, Incheon, South Korea
| | - Wonchung Lim
- Department of Sports Medicine, College of Health Science, Cheongju University, Cheongju, South Korea
| | - Saba Imdad
- Molecular Metabolism in Health and Disease, Exercise Physiology Laboratory, Inha University, Incheon, South Korea
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju, South Korea
| | - Chounghun Kang
- Molecular Metabolism in Health and Disease, Exercise Physiology Laboratory, Inha University, Incheon, South Korea
- Department of Physical Education, College of Education, Inha University, Incheon, South Korea
- *Correspondence: Chounghun Kang,
| |
Collapse
|
125
|
Gao W, Guo L, Yang Y, Wang Y, Xia S, Gong H, Zhang BK, Yan M. Dissecting the Crosstalk Between Nrf2 and NF-κB Response Pathways in Drug-Induced Toxicity. Front Cell Dev Biol 2022; 9:809952. [PMID: 35186957 PMCID: PMC8847224 DOI: 10.3389/fcell.2021.809952] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Nrf2 and NF-κB are important regulators of the response to oxidative stress and inflammation in the body. Previous pharmacological and genetic studies have confirmed crosstalk between the two. The deficiency of Nrf2 elevates the expression of NF-κB, leading to increased production of inflammatory factors, while NF-κB can affect the expression of downstream target genes by regulating the transcription and activity of Nrf2. At the same time, many therapeutic drug-induced organ toxicities, including hepatotoxicity, nephrotoxicity, cardiotoxicity, pulmonary toxicity, dermal toxicity, and neurotoxicity, have received increasing attention from researchers in clinical practice. Drug-induced organ injury can destroy body function, reduce the patients’ quality of life, and even threaten the lives of patients. Therefore, it is urgent to find protective drugs to ameliorate drug-induced injury. There is substantial evidence that protective medications can alleviate drug-induced organ toxicity by modulating both Nrf2 and NF-κB signaling pathways. Thus, it has become increasingly important to explore the crosstalk mechanism between Nrf2 and NF-κB in drug-induced toxicity. In this review, we summarize the potential molecular mechanisms of Nrf2 and NF-κB pathways and the important effects on adverse effects including toxic reactions and look forward to finding protective drugs that can target the crosstalk between the two.
Collapse
Affiliation(s)
- Wen Gao
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Lin Guo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shuang Xia
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bi-Kui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Miao Yan,
| |
Collapse
|
126
|
Liang W, Greven J, Fragoulis A, Horst K, Bläsius F, Wruck C, Pufe T, Kobbe P, Hildebrand F, Lichte P. Sulforaphane-Dependent Up-Regulation of NRF2 Activity Alleviates Both Systemic Inflammatory Response and Lung Injury After Hemorrhagic Shock/Resuscitation in Mice. Shock 2022; 57:221-229. [PMID: 34559743 DOI: 10.1097/shk.0000000000001859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Hemorrhagic shock/resuscitation (HS/R) is closely associated with overwhelming oxidative stress and systemic inflammation. As an effective activator of the nuclear factor-erythroid factor 2 related factor 2 (Nrf2) pathway, sulforaphane (SFN) exerts antioxidant and anti-inflammatory effects. We explored SFN's effects on alveolar macrophages (AMs), systemic inflammation, and pulmonary damage in an isolated murine HS/R model. Male C57/BL6 wild type and transgenic antioxidant response element (ARE)-luciferase (luc) mice (both n = 6 per group) were exposed to either pressure-controlled HS/R (mean arterial pressure 35-45 mm Hg for 90 min) or sham procedure (surgery without HS/R) or were sacrificed without intervention (control group). Fluid resuscitation was performed via the reinfusion of withdrawn blood and 0.9% saline. Sulforaphane or 0.9% saline (vehicle) was administrated intraperitoneally. Mice were sacrificed 6, 24, or 72 h after resuscitation. Bioluminescence imaging of ARE-luc mice was conducted to measure pulmonary Nrf2 activity. Plasma was collected to determine systemic cytokine levels. Alveolar macrophages were isolated before measuring cytokines in the supernatant and performing immunofluorescence staining, as well as Western blot for intracellular Nrf2. Histological damage was assessed via the acute lung injury score and wet/dry ratio.Hemorrhagic shock/resuscitation was associated with pulmonary Nrf2 activation. Sulforaphane enhanced pulmonary Nrf2 activity and the Nrf2 activation of AM, while it decreased lung damage. Sulforaphane exerted down-regulatory effects on AM-generated and systemic pro-inflammatory mediators, while it did not have such effects on IL-10.In conclusion, SFN beneficially enhances pulmonary Nrf2 activity and promotes Nrf2 accumulation in AMs' nuclei. This may exert not only local protective effects but also systemic effects via the down-regulation of pro-inflammatory cytokines. The administration of Nrf2 activator post-HS/R may represent an innovative treatment strategy.
Collapse
Affiliation(s)
- Weiqiang Liang
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan City, Shandong Province, PR China
| | - Johannes Greven
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Athanassios Fragoulis
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, Aachen, Germany
| | - Klemens Horst
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Felix Bläsius
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Wruck
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, Aachen, Germany
| | - Philipp Kobbe
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Frank Hildebrand
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp Lichte
- Department of Orthopedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
127
|
The multifaceted role of ferroptosis in liver disease. Cell Death Differ 2022; 29:467-480. [PMID: 35075250 PMCID: PMC8901678 DOI: 10.1038/s41418-022-00941-0] [Citation(s) in RCA: 379] [Impact Index Per Article: 126.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
Ferroptosis is an iron-dependent form of non-apoptotic cell death characterized by excessive lipid peroxidation and associated with a plethora of pathological conditions in the liver. Emerging evidence supports the notion that dysregulated metabolic pathways and impaired iron homeostasis play a role in the progression of liver disease via ferroptosis. Although the molecular mechanisms by which ferroptosis causes disease are poorly understood, several ferroptosis-associated genes and pathways have been implicated in liver disease. Here, we review the physiological role of the liver in processing nutrients, our current understanding of iron metabolism, the characteristics of ferroptosis, and the mechanisms that regulate ferroptosis. In addition, we summarize the role of ferroptosis in the pathogenesis of liver disease, including liver injury, non-alcoholic steatohepatitis, liver fibrosis, liver cirrhosis, and hepatocellular carcinoma. Finally, we discuss the therapeutic potential of targeting ferroptosis for managing liver disease.
Collapse
|
128
|
Wang L, Wu R, Sargsyan D, Su S, Kuo HC, Li S, Chou P, Sarwar MS, Phadnis A, Wang Y, Su X, Kong AN. Nfe2l2 Regulates Metabolic Rewiring and Epigenetic Reprogramming in Mediating Cancer Protective Effect by Fucoxanthin. AAPS J 2022; 24:30. [PMID: 35043283 DOI: 10.1208/s12248-022-00679-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
Fucoxanthin (FX) is a carotenoid with many pharmaceutical properties due to its antioxidant/anti-inflammatory and epigenetic effects. NFE2L2 is involved in the defense against oxidative stress/inflammation-mediated diseases, like anticancer effects elicited by phytochemicals including FX. However, the role of FX and NFE2L2 in metabolic rewiring, epigenomic reprogramming, and transcriptomic network in blocking pro-tumorigenic signaling and eliciting cancer-protective effects remains unknown. Herein, we utilized multi-omics approaches to evaluate the role of NFE2L2 and the impact of FX on tumor promoter TPA-induced skin cell transformation. FX blocked TPA-induced ROS and oxidized GSSG/reduced GSH in Nfe2l2wild-type(WT) but not Nfe2l2-knockdown (KD) cells. Both Nfe2l2 KD and TPA altered cellular metabolisms and metabolites which are tightly coupled to epigenetic machinery. The suppressive effects of FX on TPA-enhancedSAM/SAH was abrogated by Nfe2l2 KD indicating Nfe2l2 plays a critical role in FX-mediated metabolic rewiring and its potential consequences on epigenetic reprogramming. Epigenomic CpG methyl-seq revealed that FX attenuated TPA-induced differentially methylated regions (DMRs) of Uhrf1 and Dnmt1 genes. Transcriptomic RNA-seq showed that FX abrogated TPA-induced differentially expressed genes (DEGs) of Nfe2l2-related genes Nqo1, Ho1, and Keap1. Associative analysis of DEGs and DMRs identified that the mRNA expressions of Uhrf1 and Dnmt1 were correlated with the promoter CpG methylation status. Chromatin immunoprecipitation assay showed that FX restored Uhrf1 expression by regulating H3K27Me3 enrichment in the promoter region. In this context, FX/Nfe2l2's redox signaling drives metabolic rewiring causing epigenetic and transcriptomic reprogramming potentially contributing to the protection of TPA-induced JB6 cellular transformation skin cancer model. Graphical abstract.
Collapse
Affiliation(s)
- Lujing Wang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA
- Graduate Program of Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, USA
| | - Renyi Wu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA
| | - Davit Sargsyan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA
- Graduate Program of Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, USA
| | - Shan Su
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA
| | - Hsiao-Chen Kuo
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA
- Graduate Program of Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, USA
| | - Shanyi Li
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA
| | - Pochung Chou
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA
- Graduate Program of Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, USA
| | - Md Shahid Sarwar
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA
| | - Ameya Phadnis
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA
| | - Yujue Wang
- Metabolomics Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, 08903, USA
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, 08903, USA
| | - Xiaoyang Su
- Metabolomics Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, 08903, USA
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, 08903, USA
| | - Ah-Ng Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, 08854, USA.
| |
Collapse
|
129
|
Mechanism of Neonatal Intestinal Injury Induced by Hyperoxia Therapy. J Immunol Res 2022; 2022:2316368. [PMID: 35071607 PMCID: PMC8769871 DOI: 10.1155/2022/2316368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/16/2021] [Indexed: 11/18/2022] Open
Abstract
High concentration oxygen is widely used in the treatment of neonates, which has a significant effect on improving blood oxygen concentration in neonates with respiratory distress. The adverse effects of hyperoxia therapy on the lung, retina, and neurodevelopment of newborns have been extensively studied, but less attention has been paid to intestinal damage caused by hyperoxia therapy. In this review, we focus on the physical, immune, and microorganism barriers of the intestinal tract and discuss neonatal intestinal tract damage caused by hyperoxia therapy and analyze the molecular mechanism of intestinal damage caused by hyperoxia in combination with necrotizing enterocolitis.
Collapse
|
130
|
Nrf2 in the Field of Dentistry with Special Attention to NLRP3. Antioxidants (Basel) 2022; 11:antiox11010149. [PMID: 35052653 PMCID: PMC8772975 DOI: 10.3390/antiox11010149] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/06/2022] [Accepted: 01/09/2022] [Indexed: 02/06/2023] Open
Abstract
The aim of this review article was to summarize the functional implications of the nuclear factor E2-related factor or nuclear factor (erythroid-derived 2)-like 2 (Nrf2), with special attention to the NACHT (nucleotide-binding oligomerization), LRR (leucine-rich repeat), and PYD (pyrin domain) domains-containing protein 3 (NLRP3) inflammasome in the field of dentistry. NLRP3 plays a crucial role in the progression of inflammatory and adaptive immune responses throughout the body. It is already known that this inflammasome is a key regulator of several systemic diseases. The initiation and activation of NLRP3 starts with the oral microbiome and its association with the pathogenesis and progression of several oral diseases, including periodontitis, periapical periodontitis, and oral squamous cell carcinoma (OSCC). The possible role of the inflammasome in oral disease conditions may involve the aberrant regulation of various response mechanisms, not only in the mouth but in the whole body. Understanding the cellular and molecular biology of the NLRP3 inflammasome and its relationship to Nrf2 is necessary for the rationale when suggesting it as a potential therapeutic target for treatment and prevention of oral inflammatory and immunological disorders. In this review, we highlighted the current knowledge about NLRP3, its likely role in the pathogenesis of various inflammatory oral processes, and its crosstalk with Nrf2, which might offer future possibilities for disease prevention and targeted therapy in the field of dentistry and oral health.
Collapse
|
131
|
Kang JS, Nam LB, Yoo OK, Lee K, Suh YA, Kim D, Kim WK, Lim CY, Lee H, Keum YS. BAP1 Downregulates NRF2 Target Genes and Exerts Anti-Tumorigenic Effects by Deubiquitinating KEAP1 in Lung Adenocarcinoma. Antioxidants (Basel) 2022; 11:antiox11010114. [PMID: 35052618 PMCID: PMC8773298 DOI: 10.3390/antiox11010114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/31/2021] [Accepted: 01/01/2022] [Indexed: 12/30/2022] Open
Abstract
KELCH-ECH-associated protein 1 (KEAP1) is an adaptor protein of Cullin 3 (CUL3) E3 ubiquitin ligase that targets a redox sensitive transcription factor, NF-E2-related factor 2 (NRF2). BRCA1-associated protein 1 (BAP1) is a tumor suppressor and deubiquitinase whose mutations increase the risk of several types of familial cancers. In the present study, we have identified that BAP1 deubiquitinates KEAP1 by binding to the BTB domain. Lentiviral transduction of BAP1 decreased the expression of NRF2 target genes, suppressed the migration and invasion, and sensitized cisplatin-induced apoptosis in human lung adenocarcinoma (LUAD) A549 cells. Examination of the lung tissues in KrasG12D/+ mice demonstrated that the level of Bap1 and Keap1 mRNAs progressively decreases during lung tumor progression, and it is correlated with NRF2 activation and the inhibition of oxidative stress. Supporting this observation, lentiviral transduction of BAP1 decreased the growth of A549 xenografts in athymic nude mice. Transcriptome analysis of human lung tissues showed that the levels of Bap1 mRNA are significantly higher in normal samples than LUAD samples. Moreover, the expression of Bap1 mRNA is associated with a better survival of LUAD patients. Together, our study demonstrates that KEAP1 deubiquitination by BAP1 is novel tumor suppressive mechanism of LUAD.
Collapse
Affiliation(s)
- Jong-Su Kang
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, 32 Dongguk-ro, Goyang 10326, Gyeonggi-do, Korea; (J.-S.K.); (L.B.N.); (O.-K.Y.); (K.L.)
| | - Le Ba Nam
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, 32 Dongguk-ro, Goyang 10326, Gyeonggi-do, Korea; (J.-S.K.); (L.B.N.); (O.-K.Y.); (K.L.)
| | - Ok-Kyung Yoo
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, 32 Dongguk-ro, Goyang 10326, Gyeonggi-do, Korea; (J.-S.K.); (L.B.N.); (O.-K.Y.); (K.L.)
| | - Kyeong Lee
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, 32 Dongguk-ro, Goyang 10326, Gyeonggi-do, Korea; (J.-S.K.); (L.B.N.); (O.-K.Y.); (K.L.)
| | - Young-Ah Suh
- Department of Biomedical Sciences, Asan Medical Center, The University of Ulsan College of Medicine, Seoul 05505, Gyeonggi-do, Korea;
| | - Dalyong Kim
- Department of Internal Medicine, School of Medicine, Dongguk University, Goyang 10326, Gyeonggi-do, Korea; (D.K.); (W.K.K.)
| | - Woo Kyung Kim
- Department of Internal Medicine, School of Medicine, Dongguk University, Goyang 10326, Gyeonggi-do, Korea; (D.K.); (W.K.K.)
| | - Chi-Yeon Lim
- Department of Biostatistics, School of Medicine, Dongguk University, Goyang 10326, Gyeonggi-do, Korea;
| | - Haeseung Lee
- College of Pharmacy, Pusan National University, Busan 46241, Gyeongsangnam-do, Korea;
| | - Young-Sam Keum
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, 32 Dongguk-ro, Goyang 10326, Gyeonggi-do, Korea; (J.-S.K.); (L.B.N.); (O.-K.Y.); (K.L.)
- Correspondence: ; Tel.: +82-31-961-5215; Fax: +82-31-961-52
| |
Collapse
|
132
|
Mokrane N, Snabi Y, Cens T, Guiramand J, Charnet P, Bertaud A, Menard C, Rousset M, de Jesus Ferreira MC, Thibaud JB, Cohen-Solal C, Vignes M, Roussel J. Manipulations of Glutathione Metabolism Modulate IP 3-Mediated Store-Operated Ca 2+ Entry on Astroglioma Cell Line. Front Aging Neurosci 2022; 13:785727. [PMID: 34975458 PMCID: PMC8719003 DOI: 10.3389/fnagi.2021.785727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/01/2021] [Indexed: 02/03/2023] Open
Abstract
The regulation of the redox status involves the activation of intracellular pathways as Nrf2 which provides hormetic adaptations against oxidative stress in response to environmental stimuli. In the brain, Nrf2 activation upregulates the formation of glutathione (GSH) which is the primary antioxidant system mainly produced by astrocytes. Astrocytes have also been shown to be themselves the target of oxidative stress. However, how changes in the redox status itself could impact the intracellular Ca2+ homeostasis in astrocytes is not known, although this could be of great help to understand the neuronal damage caused by oxidative stress. Indeed, intracellular Ca2+ changes in astrocytes are crucial for their regulatory actions on neuronal networks. We have manipulated GSH concentration in astroglioma cells with selective inhibitors and activators of the enzymes involved in the GSH cycle and analyzed how this could modify Ca2+ homeostasis. IP3-mediated store-operated calcium entry (SOCE), obtained after store depletion elicited by Gq-linked purinergic P2Y receptors activation, are either sensitized or desensitized, following GSH depletion or increase, respectively. The desensitization may involve decreased expression of the proteins STIM2, Orai1, and Orai3 which support SOCE mechanism. The sensitization process revealed by exposing cells to oxidative stress likely involves the increase in the activity of Calcium Release-Activated Channels (CRAC) and/or in their membrane expression. In addition, we observe that GSH depletion drastically impacts P2Y receptor-mediated changes in membrane currents, as evidenced by large increases in Ca2+-dependent K+ currents. We conclude that changes in the redox status of astrocytes could dramatically modify Ca2+ responses to Gq-linked GPCR activation in both directions, by impacting store-dependent Ca2+-channels, and thus modify cellular excitability under purinergic stimulation.
Collapse
Affiliation(s)
- Nawfel Mokrane
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Yassin Snabi
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Thierry Cens
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Janique Guiramand
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Pierre Charnet
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Anaïs Bertaud
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Claudine Menard
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Matthieu Rousset
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Marie-Céleste de Jesus Ferreira
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | | | - Catherine Cohen-Solal
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Michel Vignes
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Julien Roussel
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| |
Collapse
|
133
|
Sarmah S, Roy AS. A review on prevention of glycation of proteins: Potential therapeutic substances to mitigate the severity of diabetes complications. Int J Biol Macromol 2022; 195:565-588. [DOI: 10.1016/j.ijbiomac.2021.12.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 12/21/2022]
|
134
|
Hei G, Smith RC, Li R, Ou J, Song X, Zheng Y, He Y, Arriaza J, Fahey JW, Cornblatt B, Kang D, Yang Y, Huang J, Wang X, Cadenhead K, Zhang M, Davis JM, Zhao J, Jin H, Wu R. Sulforaphane Effects on Cognition and Symptoms in First and Early Episode Schizophrenia: A Randomized Double-Blind Trial. SCHIZOPHRENIA BULLETIN OPEN 2022; 3:sgac024. [PMID: 39144775 PMCID: PMC11205988 DOI: 10.1093/schizbullopen/sgac024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Objective Cognitive symptoms are associated with significant dysfunction in schizophrenia. Oxidative stress and inflammation involving histone deacetylase (HDAC) have been implicated in the pathophysiology of schizophrenia. Sulforaphane has antioxidant properties and is an HDAC inhibitor. The objective of this study was to determine the efficacy of sulforaphane on cognition dysfunction for patients with schizophrenia. Methods This double-blind randomized 22-week trial of patients with first-episode schizophrenia was conducted in four psychiatric institutions in China. Patients were randomized to three groups (two doses of sulforaphane vs. placebo) and symptomatic and cognitive assessments were completed at multiple times. The primary outcome measure was change in the MATRICS Composite score. The secondary outcomes were change in MATRICS Domain scores, PANSS Total Scores and change in side-effects. Results A total of 172 patients were randomized and 151 patients had at least one follow up evaluation. There were no significant effects of sulforaphane, on the primary outcome, MATRICS overall composite score. However, on secondary outcomes, sulforaphane did significantly improve performance scores on MATRICS battery Domains of spatial working memory (F = 5.68, P = 0.004), reasoning-problem solving (F = 2.82, P = 0.063), and verbal learning (F = 3.56, P = 0.031). There were no effects on PANSS symptom scores. Sulforaphane was well tolerated. Conclusion Although the primary outcome was not significant, improvement in three domains of the MATRICS battery, suggests a positive cognitive effect on some cognitive functions, which warrants further clinical trials to further assess whether sulforaphane may be a useful adjunct for treating some types of cognitive deficits in schizophrenia.
Collapse
Affiliation(s)
- Gangrui Hei
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Robert C Smith
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Ranran Li
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Jianjun Ou
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Xueqing Song
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yingjun Zheng
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, China
| | - Yiqun He
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Jen Arriaza
- School of Professional Studies, New York University, New York, NY, USA
| | - Jed W Fahey
- Center for Human Nutrition, International Health. School of Medicine, John Hopkins University, Baltimore, Maryland, USA
| | | | - Dongyu Kang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Ye Yang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Jing Huang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Xiaoyi Wang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Kristin Cadenhead
- Department of Psychiatry, University of California San Diego and Psychiatric Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Mimei Zhang
- Columbia University Mailman School of Public Health, New York City, NY, USA
| | - John M Davis
- Department of Psychiatry, University of Illinois, Chicago, IL, USA
| | - Jingping Zhao
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Hua Jin
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
- Department of Psychiatry, University of California San Diego and Psychiatric Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Renrong Wu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| |
Collapse
|
135
|
Chiang YW, Su CH, Sun HY, Chen SP, Chen CJ, Chen WY, Chang CC, Chen CM, Kuan YH. Bisphenol A induced apoptosis via oxidative stress generation involved Nrf2/HO-1 pathway and mitochondrial dependent pathways in human retinal pigment epithelium (ARPE-19) cells. ENVIRONMENTAL TOXICOLOGY 2022; 37:131-141. [PMID: 34664771 DOI: 10.1002/tox.23384] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/23/2021] [Accepted: 10/03/2021] [Indexed: 05/21/2023]
Abstract
Bisphenol A (BPA) is an estrogen-like compound, and an environmental hormone, that is commonly used in daily life. Therefore, it may enter the human body through food or direct contact, causing BPA residues in blood and urine. Because most studies focused on the analysis of BPA in reproductive cells or tissues, regarding evidence the effect of BPA on human retinal pigment epithelium (ARPE-19) cells unavailable. Accordingly, the present study explored the cytotoxicity of BPA on ARPE-19 cells. After BPA treatment, the expression of Bcl-XL an antiapoptotic protein, in the mitochondria decreased, and the expression of Bax, a proapoptotic protein increased. Then the mitochondrial membrane potential was affected. BPA changed in mitochondrial membrane potential led to the release of cytochrome C, which activated caspase-9 to promote downstream caspase-3 leading to cytotoxicity. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and heme oxygenase 1 (HO-1) pathway play a major role in age-related macular degeneration. Our results showed that expression of HO-1 and Nrf2 suppressed by BPA. Superoxide dismutase and catalase, which Nrf2 downstream antioxidants, were degraded by BPA. AMP-activated kinase (AMPK), which can regulate the phosphorylation of Nrf2, and the phosphorylation of AMPK expression was reduced by BPA. Finally, BPA-induced ROS generation and cytotoxicity were reduced by N-acetyl-l-cysteine. Taken together, these results suggest that BPA induced ARPE-19 cells via oxidative stress, which was associated with down regulated Nrf2/HO-1 pathway, and the mitochondria dependent apoptotic signaling pathway.
Collapse
Affiliation(s)
- Yun-Wei Chiang
- Department of Life Sciences, National Chung-Hsing University, Taichung, Taiwan
- Department of Optometry, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Chun-Hung Su
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
- Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Han-Yin Sun
- Department of Optometry, Chung Shan Medical University, Taichung, Taiwan
- Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shih-Pin Chen
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
- Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Jung Chen
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chia-Che Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Yu-Hsiang Kuan
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
136
|
Cecerska-Heryć E, Polikowska A, Serwin N, Roszak M, Grygorcewicz B, Heryć R, Michalczyk A, Dołęgowska B. Importance of oxidative stress in the pathogenesis, diagnosis, and monitoring of patients with neuropsychiatric disorders, a review. Neurochem Int 2021; 153:105269. [PMID: 34971747 DOI: 10.1016/j.neuint.2021.105269] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/29/2021] [Accepted: 12/21/2021] [Indexed: 12/31/2022]
Abstract
Oxidative stress is defined as the persistent imbalance between the activity of toxic reactive forms of both oxygen and nitrogen and the antioxidant defense. In low concentrations, they are essential for the proper functioning of the body. Still, their excessive amount contributes to the damage of the biomolecules, consequently leading to various pathologies of the organism. Due to the lipid-rich brain structure, enormous oxygen consumption, and the lack of a sufficient antioxidant barrier make it highly susceptible to oxidative imbalance. Hence, oxidative stress has been linked to various psychiatric disorders. These diseases include all behavioral, emotional, and cognitive abnormalities associated with a significant impediment to social life. Each of the diseases in question: Alzheimer's disease, schizophrenia, depression, and bipolar disorder, is characterized by excessive oxidative stress. Considerable damages to DNA, RNA, proteins, lipids, and mitochondrial dysfunction, are observed. All conditions show increased lipid peroxidation, which appears to be typical of psychiatric disorders because the brain contains large amounts of these types of molecules. In addition, numerous abnormalities in the antioxidant defense are noted, but the results of studies on the activity of antioxidant enzymes differ significantly. The most promising biomarkers seem to be GSH in Alzheimer's disease as an early-stage marker of the disease and thioredoxin in schizophrenia as a marker for therapy monitoring. Data from the literature are consistent with the decrease in antioxidants such as vitamin C, E, uric acid, albumin, etc. Despite these numerous inconsistencies, it seems that oxidative stress is present in the course of psychiatric diseases. Still, it cannot be conclusively determined whether it is the direct cause of development, a consequence of other abnormalities at the biochemical or molecular level, or the result of the disease itself.
Collapse
Affiliation(s)
- Elżbieta Cecerska-Heryć
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111, Szczecin, Poland.
| | - Aleksandra Polikowska
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Natalia Serwin
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Marta Roszak
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Bartłomiej Grygorcewicz
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Rafał Heryć
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Anna Michalczyk
- Department of Psychiatry, Pomeranian Medical University of Szczecin, Broniewskiego 26, 71-460, Szczecin, Poland
| | - Barbara Dołęgowska
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111, Szczecin, Poland
| |
Collapse
|
137
|
Crosstalk between Neuron and Glial Cells in Oxidative Injury and Neuroprotection. Int J Mol Sci 2021; 22:ijms222413315. [PMID: 34948108 PMCID: PMC8709409 DOI: 10.3390/ijms222413315] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/30/2022] Open
Abstract
To counteract oxidative stress and associated brain diseases, antioxidant systems rescue neuronal cells from oxidative stress by neutralizing reactive oxygen species and preserving gene regulation. It is necessary to understand the communication and interactions between brain cells, including neurons, astrocytes and microglia, to understand oxidative stress and antioxidant mechanisms. Here, the role of glia in the protection of neurons against oxidative injury and glia–neuron crosstalk to maintain antioxidant defense mechanisms and brain protection are reviewed. The first part of this review focuses on the role of glia in the morphological and physiological changes required for brain homeostasis under oxidative stress and antioxidant defense mechanisms. The second part focuses on the essential crosstalk between neurons and glia for redox balance in the brain for protection against oxidative stress.
Collapse
|
138
|
Hellmuth N, Brat C, Awad O, George S, Kahnt A, Bauer T, Huynh Phuoc HP, Steinhilber D, Angioni C, Hassan M, Hock KJ, Manolikakes G, Zacharowski K, Roos J, Maier TJ. Structural Modifications Yield Novel Insights Into the Intriguing Pharmacodynamic Potential of Anti-inflammatory Nitro-Fatty Acids. Front Pharmacol 2021; 12:715076. [PMID: 34867322 PMCID: PMC8637440 DOI: 10.3389/fphar.2021.715076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Endogenous nitro-fatty acids (NFA) are potent electrophilic lipid mediators that exert biological effects in vitro and in vivo via selective covalent modification of thiol-containing target proteins. The cytoprotective, anti-inflammatory, and anti-tumorigenic effects of NFA in animal models of disease caused by targeted protein nitroalkylation are a valuable basis for the development of future anti-phlogistic and anti-neoplastic drugs. Considering the complexity of diseases and accompanying comorbidities there is an urgent need for clinically effective multifunctional drugs. NFA are composed of a fatty acid backbone containing a nitroalkene moiety triggering Michael addition reactions. However, less is known about the target-specific structure–activity relationships and selectivities comparing different NFA targets. Therefore, we analyzed 15 NFA derivatives and compared them with the lead structure 9-nitro-oleic acid (9NOA) in terms of their effect on NF-κB (nuclear factor kappa B) signaling inhibition, induction of Nrf-2 (nuclear factor erythroid 2-related factor 2) gene expression, sEH (soluble epoxide hydrolase), LO (lipoxygenase), and COX-2 (cyclooxygenase-2) inhibition, and their cytotoxic effects on colorectal cancer cells. Minor modifications of the Michael acceptor position and variation of the chain length led to drugs showing increased target preference or enhanced multi-targeting, partly with higher potency than 9NOA. This study is a significant step forward to better understanding the biology of NFA and their enormous potential as scaffolds for designing future anti-inflammatory drugs.
Collapse
Affiliation(s)
- Nadine Hellmuth
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Camilla Brat
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Omar Awad
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany.,Paul-Ehrlich Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Sven George
- Institute of Pharmaceutical Chemistry, Goethe-University, Frankfurt, Germany
| | - Astrid Kahnt
- Institute of Pharmaceutical Chemistry, Goethe-University, Frankfurt, Germany
| | - Tom Bauer
- Paul-Ehrlich Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Hai Phong Huynh Phuoc
- Paul-Ehrlich Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe-University, Frankfurt, Germany
| | - Carlo Angioni
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University, Frankfurt, Germany
| | - Mohamed Hassan
- Department of Chemistry, TU Kaiserslautern, Kaiserslautern, Germany.,Department of Chemistry, Faculty of Science, Aswan University, Aswan, Egypt
| | - Katharina J Hock
- Department of Chemistry, TU Kaiserslautern, Kaiserslautern, Germany
| | | | - Kai Zacharowski
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Jessica Roos
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany.,Paul-Ehrlich Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Thorsten J Maier
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany.,Paul-Ehrlich Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| |
Collapse
|
139
|
Yu S, Jia J, Zheng J, Zhou Y, Jia D, Wang J. Recent Progress of Ferroptosis in Lung Diseases. Front Cell Dev Biol 2021; 9:789517. [PMID: 34869391 PMCID: PMC8635032 DOI: 10.3389/fcell.2021.789517] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/31/2021] [Indexed: 01/08/2023] Open
Abstract
Ferroptosis is a new form of programmed cell death due to iron-dependent excess accumulation of lipid peroxides and differs from other programmed cell deaths in morphological and biochemical characteristics. The process of ferroptosis is precisely regulated by iron metabolism, lipid metabolism, amino acid metabolism, and numerous signaling pathways, and plays a complex role in many pathophysiological processes. Recent studies have found that ferroptosis is closely associated with the development and progression of many lung diseases, including acute lung injury, pulmonary ischemia-reperfusion injury, lung cancer, chronic obstructive pulmonary disease, and pulmonary fibrosis. Here, we present a review of the main regulatory mechanisms of ferroptosis and its research progress in the pathogenesis and treatment of lung diseases, with the aim of providing new ideas for basic and clinical research of lung-related diseases.
Collapse
Affiliation(s)
- Shangjiang Yu
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinqiu Jia
- Department of Pediatrics, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou, China
| | - Jinyu Zheng
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiyang Zhou
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Danyun Jia
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
140
|
Valashedi MR, Nikoo A, Najafi-Ghalehlou N, Tomita K, Kuwahara Y, Sato T, Roushandeh AM, Roudkenar MH. Pharmacological Targeting of Ferroptosis in Cancer Treatment. Curr Cancer Drug Targets 2021; 22:108-125. [PMID: 34856903 DOI: 10.2174/1568009621666211202091523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 01/17/2023]
Abstract
Ferroptosis is a non-apoptotic mode of Regulated Cell Death (RCD) driven by excessive accumulation of toxic lipid peroxides and iron overload. Ferroptosis could be triggered by inhibiting the antioxidant defense system and accumulating iron-dependent Reactive Oxygen Species (ROS) that react with polyunsaturated fatty acids in abundance. Emerging evidence over the past few years has revealed that ferroptosis is of great potential in inhibiting growth and metastasis and overcoming tumor cell resistance. Thus, targeting this form of cell death could be perceived as a potentially burgeoning approach in cancer treatment. This review briefly presents the underlying mechanisms of ferroptosis and further aims to discuss various types of existing drugs and natural compounds that could be potentially repurposed for targeting ferroptosis in tumor cells. This, in turn, will provide critical perspectives on future studies concerning ferroptosis-based cancer therapy.
Collapse
Affiliation(s)
- Mehdi Rabiee Valashedi
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht. Iran
| | - Amirsadegh Nikoo
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht. Iran
| | - Nima Najafi-Ghalehlou
- Department of Medical Laboratory Sciences, Faculty of Paramedicine, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Kazuo Tomita
- Department of Applied Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima. Japan
| | - Yoshikazu Kuwahara
- Department of Applied Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima. Iran
| | - Tomoaki Sato
- Department of Applied Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima. Iran
| | - Amaneh Mohammadi Roushandeh
- Department of Applied Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima. Iran
| | - Mehryar Habibi Roudkenar
- Department of Applied Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima. Iran
| |
Collapse
|
141
|
Iron Overload, Oxidative Stress, and Ferroptosis in the Failing Heart and Liver. Antioxidants (Basel) 2021; 10:antiox10121864. [PMID: 34942967 PMCID: PMC8698778 DOI: 10.3390/antiox10121864] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Iron accumulation is a key mediator of several cytotoxic mechanisms leading to the impairment of redox homeostasis and cellular death. Iron overload is often associated with haematological diseases which require regular blood transfusion/phlebotomy, and it represents a common complication in thalassaemic patients. Major damages predominantly occur in the liver and the heart, leading to a specific form of cell death recently named ferroptosis. Different from apoptosis, necrosis, and autophagy, ferroptosis is strictly dependent on iron and reactive oxygen species, with a dysregulation of mitochondrial structure/function. Susceptibility to ferroptosis is dependent on intracellular antioxidant capacity and varies according to the different cell types. Chemotherapy-induced cardiotoxicity has been proven to be mediated predominantly by iron accumulation and ferroptosis, whereas there is evidence about the role of ferritin in protecting cardiomyocytes from ferroptosis and consequent heart failure. Another paradigmatic organ for transfusion-associated complication due to iron overload is the liver, in which the role of ferroptosis is yet to be elucidated. Some studies report a role of ferroptosis in the initiation of hepatic inflammation processes while others provide evidence about an involvement in several pathologies including immune-related hepatitis and acute liver failure. In this manuscript, we aim to review the literature to address putative common features between the response to ferroptosis in the heart and liver. A better comprehension of (dys)similarities is pivotal for the development of future therapeutic strategies that can be designed to specifically target this type of cell death in an attempt to minimize iron-overload effects in specific organs.
Collapse
|
142
|
Dhar R, Rana MN, Zhang L, Li Y, Li N, Hu Z, Yan C, Wang X, Zheng X, Liu H, Cui H, Li Z, Tang H. Phosphodiesterase 4B is required for NLRP3 inflammasome activation by positive feedback with Nrf2 in the early phase of LPS- induced acute lung injury. Free Radic Biol Med 2021; 176:378-391. [PMID: 34644617 DOI: 10.1016/j.freeradbiomed.2021.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/03/2021] [Accepted: 10/08/2021] [Indexed: 12/22/2022]
Abstract
Acute lung injury (ALI) is associated with overproduction of inflammatory mediators in lung tissue. Previous studies have revealed that inflammation induces activation of phosphodiesterase 4B (PDE4B) accompanied by the production of inflammatory mediators, but the detailed mechanism remains unclear. Here, we focused on the NOD-, LRR- and pyrin domain-containing protein 3(NLRP3) inflammasome complexes to study the crosstalk between PDE4B and NF-E2-related factor 2 (Nrf2). We used global knockout PDE4B or Nrf2 mice to prepare LPS induced acute lung injury model by intratracheally administration, and LPS primed bone marrow-derived macrophages (BMDMs), following overexpression of PDE4B or Nrf2, luciferase activity analysis, and chIP-qPCR analyses. We found that deficiency of PDE4B could potently attenuate the lung histopathological changes, suppress the secretion of pro-inflammatory mediators such as tumor necrosis factor α (TNF-α), interleukin (IL)-1β, IL-6, IL-18, and cleaved caspase-1, 8, and GSDMD accompanied with defective activation of the ROS/Nrf2/NLRP3. Meanwhile deficiency of Nrf2 showed the similar results. Furtherly, overexpression by PDE4B or Nrf2 plasmid transfection in MH-S cells could enhance the Nrf2 or PDE4B expression. Luciferase analysis suggested that Nrf2 activated PDE4B promoter activity, while PDE4B could increase Nrf2 substrate ARE activity in MH-S cells in dose dependent manners. ChIP-qPCR analyses showed that Nrf2 bound to the PDE4B promoter region at ̴ 1532 to ̴1199 position in macrophages. Altogether, deficiency of PDE4B inhibit the inflammasome activation and pyroptosis in LPS stimulated lung injury model and macrophages by regulating ROS/Nrf2/NLRP3 activation. The study provides new insight that PDE4B is required for NLRP3 inflammasome activation by positive feedback with Nrf2.
Collapse
Affiliation(s)
- Rana Dhar
- Department of Pharmacology, Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Mohammad Nasiruddin Rana
- Department of Pharmacology, Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Lejun Zhang
- Department of Pharmacology, Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yajun Li
- Department of Pharmacology, Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Ning Li
- Department of Pharmacology, Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zhengqiang Hu
- Department of Pharmacology, Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Chungunag Yan
- Department of Pathogenic Biology and Immunology, Medical School of Southeast University, Nanjing 210009, China
| | - Xuefeng Wang
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310005, China
| | - Xuyang Zheng
- Department of Pediatrics, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Hongyun Liu
- Institute of Dairy Science, MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Huashun Cui
- Department of Acupuncture and Moxibustion, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, China.
| | - Zigang Li
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, China.
| | - Huifang Tang
- Department of Pharmacology, Zhejiang Respiratory Drugs Research Laboratory, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
143
|
Di Fino L, Arruebarrena Di Palma A, Perk EA, García-Mata C, Schopfer FJ, Laxalt AM. Nitro-fatty acids: electrophilic signaling molecules in plant physiology. PLANTA 2021; 254:120. [PMID: 34773515 PMCID: PMC10704571 DOI: 10.1007/s00425-021-03777-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/30/2021] [Indexed: 06/13/2023]
Abstract
MAIN CONCLUSION Nitro fatty acids (NO2-FA)have relevant physiological roles as signaling molecules in biotic and abiotic stress, growth, and development, but the mechanism of action remains controversial. The two main mechanisms involving nitric oxide release and thiol modification are discussed. Fatty acids (FAs) are major components of membranes and contribute to cellular energetic demands. Besides, FAs are precursors of signaling molecules, including oxylipins and other oxidized fatty acids derived from the activity of lipoxygenases. In addition, non-canonical modified fatty acids, such as nitro-fatty acids (NO2-FAs), are formed in animals and plants. The synthesis NO2-FAs involves a nitration reaction between unsaturated fatty acids and reactive nitrogen species (RNS). This review will focus on recent findings showing that, in plants, NO2-FAs such as nitro-linolenic acid (NO2-Ln) and nitro-oleic acid (NO2-OA) have relevant physiological roles as signaling molecules in biotic and abiotic stress, growth, and development. Moreover, since there is controversy on mechanisms of action of NO2-FAs as signaling molecules, we will provide evidence showing why this aspect needs further evaluation.
Collapse
Affiliation(s)
- Luciano Di Fino
- Instituto de Investigaciones Biológicas, CONICET-Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Andrés Arruebarrena Di Palma
- Instituto de Investigaciones Biológicas, CONICET-Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Enzo A Perk
- Instituto de Investigaciones Biológicas, CONICET-Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Carlos García-Mata
- Instituto de Investigaciones Biológicas, CONICET-Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Francisco J Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana M Laxalt
- Instituto de Investigaciones Biológicas, CONICET-Universidad Nacional de Mar del Plata, Mar del Plata, Argentina.
| |
Collapse
|
144
|
ROS as Regulators of Cellular Processes in Melanoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1208690. [PMID: 34725562 PMCID: PMC8557056 DOI: 10.1155/2021/1208690] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022]
Abstract
In this review, we examine the multiple roles of ROS in the pathogenesis of melanoma, focusing on signal transduction and regulation of gene expression. In recent years, different studies have analyzed the dual role of ROS in regulating the redox system, with both negative and positive consequences on human health, depending on cell concentration of these agents. High ROS levels can result from an altered balance between oxidant generation and intracellular antioxidant activity and can produce harmful effects. In contrast, low amounts of ROS are considered beneficial, since they trigger signaling pathways involved in physiological activities and programmed cell death, with protective effects against melanoma. Here, we examine these beneficial roles, which could have interesting implications in melanoma treatment.
Collapse
|
145
|
Cui Q, Yan L. Tripartite motif-containing protein 16 protects against myocardial ischemia/reperfusion injury by affecting the Keap1/Nrf2 axis. Cell Tissue Res 2021; 386:349-363. [PMID: 34436665 DOI: 10.1007/s00441-021-03518-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
Tripartite motif-containing protein (TRIM16) is a newly identified oxidative-stress-responsive protein. Oxidative stress is a hallmark of myocardial ischemia/reperfusion (I/R) injury and contributes to the cardiac injury. To date, whether TRIM16 plays a role in mediating oxidative stress during myocardial I/R injury is undetermined. The work is devoted to evaluate the possible relevance of TRIM16 in myocardial I/R injury. TRIM16 induction by myocardial hypoxia/reoxygenation (H/R) injury in vitro or myocardial I/R injury in vivo was observed. TRIM16 overexpression alleviated H/R-induced injury of rat cardiomyocytes. TRIM16 overexpression markedly attenuated cardiac injury, infarct size, and myocardial apoptosis induced by myocardial I/R injury. Further research revealed that TRIM16 was capable of enhancing Nrf2 activation via the regulation of Keap1. The inhibition of Nrf2 diminished TRIM16-overexpression-mediated cardioprotective effects. Overall, this work demonstrates that TRIM16 protects against myocardial I/R injury via affecting the Keap1/Nrf2 axis. This work offers new insights into the molecular mechanism underlying myocardial I/R injury and proposes TRIM16 as an attractive candidate target for cardioprotection.
Collapse
Affiliation(s)
- Qianwei Cui
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, Xi'an 710068, China
| | - Li Yan
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, Xi'an 710068, China.
| |
Collapse
|
146
|
Afshari-Kaveh M, Abbasalipourkabir R, Nourian A, Ziamajidi N. The Protective Effects of Vitamins A and E on Titanium Dioxide Nanoparticles (nTiO2)-Induced Oxidative Stress in the Spleen Tissues of Male Wistar Rats. Biol Trace Elem Res 2021; 199:3677-3687. [PMID: 33210191 DOI: 10.1007/s12011-020-02487-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
Titanium dioxide nanoparticles (nTiO2) can accumulate in different tissues and damage them with oxidative stress induction. Different components with antioxidant capacity can protect the tissues. So in this study, the protective effects of vitamin A and E on the nTiO2-induced oxidative stress in rats' spleen tissues were examined. Thirty-six male Wistar rats were randomly divided into 6 groups: Control 1 (received water), nTiO2, nTiO2 + vitamin E, nTiO2 + vitamin A, nTiO2 + vitamin A and E, and Control 2 (received olive oil). To investigate the status of oxidative stress, total antioxidant capacity (TAC), total oxidant status (TOS), and lipid peroxidation (LPO) were determined in spleen tissue as well as the activities of antioxidant enzymes, including glutathione peroxidase (GPx) and superoxide dismutase (SOD). Also, the gene expression of GPx, SOD, and nuclear factor-E2-related factor-2 (Nrf-2) were determined by qRT-PCR. To evaluate the spleen histopathological changes, H&E staining was carried out. nTiO2 significantly increased TOS and LPO levels, whereas it decreased TAC level, GPx and SOD activities, and gene expression of GPx, SOD, and Nrf-2 in spleen tissues of rats compared with controls (p < 0.05). In vitamin-treated rats, the levels of TOS and LPO significantly decreased, and the level of TAC, the activities of GPx and SOD, and the gene expression of GPx, SOD, and Nrf-2 increased compared to nTiO2 group (p < 0.05). These parameters are maintained near to normal levels. Histological findings confirmed the protective effects of these vitamins on tissue damage caused by nTiO2. Vitamin A and E can protect the spleen tissues from nTiO2-induced oxidative stress.
Collapse
Affiliation(s)
- Mozhgan Afshari-Kaveh
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roghayeh Abbasalipourkabir
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Nourian
- Department of Pathobiology, Faculty of Paraveterinary Medicine, Bu-Ali Sina University, Hamedan, Iran
| | - Nasrin Ziamajidi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
147
|
Ouyang J, Sun L, Pan J, Zeng Z, Zeng C, Zeng F, Tian M, Wu S. A Targeted Nanosystem for Detection of Inflammatory Diseases via Fluorescent/Optoacoustic Imaging and Therapy via Modulating Nrf2/NF-κB Pathways. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102598. [PMID: 34523220 DOI: 10.1002/smll.202102598] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/06/2021] [Indexed: 05/05/2023]
Abstract
Inflammatory diseases are sometimes devastating and notoriously difficult to treat. Precisely modulating inflammatory signaling pathways is a promising approach for treating inflammatory diseases. Herein, a multifunctional nanosystem is developed for active targeting, activatable imaging and on-demand therapy against inflammatory diseases through modulating inflammatory pathways. A chromophore-drug dyad (QBS-FIS) is synthesized by linking a chromophore and a Nrf2 (nuclear factor E2-related factor) activator fisetin through boronate bond which serves as fluorescence quencher and ROS (reactive oxygen species)-responsive linker. QBS-FIS molecules form nanoparticles in water and are coated with macrophage cell membrane to ensure active targeting toward inflammation site. To further improve therapeutic efficacy, a NF-kB (nuclear-factor kappa-light-chain-enhancer of activated B cells) inhibitor thalidomide is co-encapsulated to afford the nanosystem (QBS-FIS&Thd@MM). Upon administration into mice, the nanosystem migrates to inflammatory site and pathological ROS therein cleaves the boronate bonds, thereby activating the chromophore for imaging liver/kidney inflammatory diseases for disease diagnosis and recovery evaluation via fluorescence and optoacoustic imaging as well as releasing the active drugs for treating acute liver inflammation through activating Nrf2 pathway and inhibiting NF-kB pathway. The 3D multispectral optoacoustic tomography imaging is applied to precisely locate the inflammatory foci in a spatiotemporal manner.
Collapse
Affiliation(s)
- Juan Ouyang
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Lihe Sun
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Jiayue Pan
- Medical Center, Zhejiang University, Hangzhou, 310009, China
| | - Zhuo Zeng
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Cheng Zeng
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Fang Zeng
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Mei Tian
- Medical Center, Zhejiang University, Hangzhou, 310009, China
| | - Shuizhu Wu
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| |
Collapse
|
148
|
Chen J, Chen Y, Zheng Y, Zhao J, Yu H, Zhu J, Li D. Protective Effects and Mechanisms of Procyanidins on Parkinson's Disease In Vivo and In Vitro. Molecules 2021; 26:5558. [PMID: 34577027 PMCID: PMC8464719 DOI: 10.3390/molecules26185558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022] Open
Abstract
This research assessed the molecular mechanism of procyanidins (PCs) against neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and its metabolite 1-methyl-4-phenylpyridinium (MPP+) induced Parkinson's disease (PD) models. In vitro, PC12 cells were incubated with PCs or deprenyl for 24 h, and then exposed to 1.5 mM MPP+ for 24 h. In vivo, zebrafish larvae (AB strain) 3 days post-fertilization (dpf) were incubated with deprenyl or PCs in 400 μM MPTP for 4 days. Compared with MPP+/MPTP alone, PCs significantly improved antioxidant activities (e.g., glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), catalase (CAT)), and decreased levels of reactive oxygen species (ROS) and malondialdehyde (MDA). Furthermore, PCs significantly increased nuclear Nrf2 accumulation in PC12 cells and raised the expression of NQO1, HO-1, GCLM, and GCLC in both PC12 cells and zebrafish compared to MPP+/MPTP alone. The current study shows that PCs have neuroprotective effects, activate the nuclear factor-erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway and alleviate oxidative damage in MPP+/MPTP-induced PD models.
Collapse
Affiliation(s)
- Juan Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Yixuan Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Yangfan Zheng
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Jiawen Zhao
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Huilin Yu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Jiajin Zhu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Duo Li
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
- Institute of Nutrition & Health, Qingdao University, Qingdao 266000, China;
| |
Collapse
|
149
|
Sanches LJ, Marinello PC, da Silva Brito WA, Lopes NMD, Luiz RC, Cecchini R, Cecchini AL. Metformin pretreatment reduces effect to dacarbazine and suppresses melanoma cell resistance. Cell Biol Int 2021; 46:73-82. [PMID: 34506671 DOI: 10.1002/cbin.11700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/24/2021] [Accepted: 09/05/2021] [Indexed: 11/11/2022]
Abstract
Oxidative stress role on metformin process of dacarbazine (DTIC) inducing resistance of B16F10 melanoma murine cells are investigated. To induce resistance to DTIC, murine melanoma cells were exposed to increasing concentrations of dacarabazine (DTIC-res group). Metformin was administered before and during the induction of resistance to DTIC (MET-DTIC). The oxidative stress parameters of the DTIC-res group showed increased levels of malondialdehyde (MDA), thiol, and reduced nuclear p53, 8-hydroxy-2'-deoxyguanosine (8-OH-DG), nuclear factor kappa B (NF-ĸB), and Nrf2. In presence of metformin in the resistant induction process to DTIC, (MET-DTIC) cells had increased antioxidant thiols, MDA, nuclear p53, 8-OH-DG, Nrf2, and reducing NF-ĸB, weakening the DTIC-resistant phenotype. The exclusive administration of metformin (MET group) also induced the cellular resistance to DTIC. The MET group presented high levels of total thiols, MDA, and reduced percentage of nuclear p53. It also presented reduced nuclear 8-OH-DG, NF-ĸB, and Nrf2 when compared with the control. Oxidative stress and the studied biomarkers seem to be part of the alterations evidenced in DTIC-resistant B16F10 cells. In addition, metformin administration is able to play a dual role according to the experimental protocol, preventing or inducing a DTIC-resistant phenotype. These findings should help future research with the aim of investigating DTIC resistance in melanoma.
Collapse
Affiliation(s)
- Larissa J Sanches
- Department of Pathological Sciences, Laboratory of Molecular Pathology, Londrina State University, UEL, Londrina, Parana, Brazil
| | - Poliana C Marinello
- Department of Pathological Sciences, Laboratory of Molecular Pathology, Londrina State University, UEL, Londrina, Parana, Brazil
| | - Walison A da Silva Brito
- Department of Pathological Sciences, Laboratory of Molecular Pathology, Londrina State University, UEL, Londrina, Parana, Brazil.,Leibniz-Institute for Plasma Science and Technology (INP Greifswald), ZIK plasmatis "Plasma Redox Effects", Greifswald, Germany
| | - Natália M D Lopes
- Department of Pathological Sciences, Laboratory of Molecular Pathology, Londrina State University, UEL, Londrina, Parana, Brazil
| | - Rodrigo C Luiz
- Department of Pathological Sciences, Laboratory of Pathophysiology and Free radicals, Londrina State University, UEL, Londrina, Parana, Brazil
| | - Rubens Cecchini
- Department of Pathological Sciences, Laboratory of Pathophysiology and Free radicals, Londrina State University, UEL, Londrina, Parana, Brazil
| | - Alessandra L Cecchini
- Department of Pathological Sciences, Laboratory of Molecular Pathology, Londrina State University, UEL, Londrina, Parana, Brazil
| |
Collapse
|
150
|
An Overview of the Nrf2/ARE Pathway and Its Role in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22179592. [PMID: 34502501 PMCID: PMC8431732 DOI: 10.3390/ijms22179592] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 12/20/2022] Open
Abstract
Nrf2 is a basic region leucine-zipper transcription factor that plays a pivotal role in the coordinated gene expression of antioxidant and detoxifying enzymes, promoting cell survival in adverse environmental or defective metabolic conditions. After synthesis, Nrf2 is arrested in the cytoplasm by the Kelch-like ECH-associated protein 1 suppressor (Keap1) leading Nrf2 to ubiquitin-dependent degradation. One Nrf2 activation mechanism relies on disconnection from the Keap1 homodimer through the oxidation of cysteine at specific sites of Keap1. Free Nrf2 enters the nucleus, dimerizes with small musculoaponeurotic fibrosarcoma proteins (sMafs), and binds to the antioxidant response element (ARE) sequence of the target genes. Since oxidative stress, next to neuroinflammation and mitochondrial dysfunction, is one of the hallmarks of neurodegenerative pathologies, a molecular intervention into Nrf2/ARE signaling and the enhancement of the transcriptional activity of particular genes are targets for prevention or delaying the onset of age-related and inherited neurogenerative diseases. In this study, we review evidence for the Nrf2/ARE-driven pathway dysfunctions leading to various neurological pathologies, such as Alzheimer’s, Parkinson’s, and Huntington’s diseases, as well as amyotrophic lateral sclerosis, and the beneficial role of natural and synthetic molecules that are able to interact with Nrf2 to enhance its protective efficacy.
Collapse
|