Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 572
Filter
1.
Cell Chem Biol ; 31(6): 1047-1049, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906110

ABSTRACT

In this issue of Cell Chemical Biology, Lu et al.1 report the discovery of a bivalent KEAP1 inhibitor (biKEAP1), which more rapidly activates NRF2 compared to previously reported monovalent KEAP1 inhibitors. biKEAP1suppresses acute inflammation in animal models.


Subject(s)
Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/antagonists & inhibitors , Animals , Humans , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , Inflammation/drug therapy , Inflammation/metabolism , Mice
2.
Eur J Histochem ; 68(2)2024 May 22.
Article in English | MEDLINE | ID: mdl-38779782

ABSTRACT

Osteoarthritis (OA) is a common degenerative joint disease in the elderly, while oxidative stress-induced chondrocyte degeneration plays a key role in the pathologic progression of OA. One possible reason is that the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), which acts as the intracellular defense factor against oxidative stress, is significantly inhibited in chondrocytes. Spinosin (SPI) is a potent Nrf2 agonist, but its effect on OA is still unknown. In this study, we found that SPI can alleviate tert-Butyl hydroperoxide (TBHP)-induced extracellular matrix degradation of chondrocytes. Additionally, SPI can effectively activate Nrf2, heme oxygenase-1 (HO-1), and NADPH quinone oxidoreductase 1 (NQO1) in chondrocytes under the TBHP environment. When Nrf2 was silenced by siRNA, the cartilage protective effect of SPI was also weakened. Finally, SPI showed good alleviative effects on OA in mice. Thus, SPI can ameliorate oxidative stress-induced chondrocyte dysfunction and exhibit a chondroprotective effect through activating the Nrf2/HO-1 pathway, which may provide a novel and promising option for the treatment of OA.


Subject(s)
Chondrocytes , Heme Oxygenase-1 , NF-E2-Related Factor 2 , Osteoarthritis , Signal Transduction , Animals , Male , Mice , Chondrocytes/drug effects , Chondrocytes/pathology , Heme Oxygenase-1/metabolism , Membrane Proteins , Mice, Inbred C57BL , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Osteoarthritis/chemically induced , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Oxidative Stress/drug effects , Signal Transduction/drug effects , tert-Butylhydroperoxide
3.
Acta Pharmacol Sin ; 45(7): 1506-1519, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38480835

ABSTRACT

Combining radiotherapy with Nrf-2 inhibitor holds promise as a potential therapeutic strategy for radioresistant lung cancer. Here, the radiosensitizing efficacy of a synthetic glucocorticoid clobetasol propionate (CP) in A549 human lung cancer cells was evaluated. CP exhibited potent radiosensitization in lung cancer cells via inhibition of Nrf-2 pathway, leading to elevation of oxidative stress. Transcriptomic studies revealed significant modulation of pathways related to ferroptosis, fatty acid and glutathione metabolism. Consistent with these findings, CP treatment followed by radiation exposure showed characteristic features of ferroptosis in terms of mitochondrial swelling, rupture and loss of cristae. Ferroptosis is a form of regulated cell death triggered by iron-dependent ROS accumulation and lipid peroxidation. In combination with radiation, CP showed enhanced iron release, mitochondrial ROS, and lipid peroxidation, indicating ferroptosis induction. Further, iron chelation, inhibition of lipid peroxidation or scavenging mitochondrial ROS prevented CP-mediated radiosensitization. Nrf-2 negatively regulates ferroptosis through upregulation of antioxidant defense and iron homeostasis. Interestingly, Nrf-2 overexpressing A549 cells were refractory to CP-mediated ferroptosis induction and radiosensitization. Thus, this study identified anti-psoriatic drug clobetasol propionate can be repurposed as a promising radiosensitizer for Keap-1 mutant lung cancers.


Subject(s)
Clobetasol , Ferroptosis , Lung Neoplasms , Mitochondria , NF-E2-Related Factor 2 , Reactive Oxygen Species , Humans , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , Ferroptosis/drug effects , Reactive Oxygen Species/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Clobetasol/pharmacology , Radiation-Sensitizing Agents/pharmacology , A549 Cells , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects
4.
J Ovarian Res ; 17(1): 49, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38396022

ABSTRACT

Ovarian cancer is a significant challenge in women's health due to the lack of effective screening and diagnostic methods, often leading to late detection and the highest mortality rate among all gynecologic tumors worldwide. Recent research has shown that ovarian cancer has an "iron addiction" phenotype which makes it vulnerable to ferroptosis inducers. We tested the combination of NRF2-targeted inhibitors with GPX4-targeted inhibitors in ovarian cancer through in vitro and in vivo experiment. The data showed that combination treatment effectively suppressed adherent cell growth, inhibited suspended cell spheroid formation, and restrained the ability of spheroid formation in 3D-culture. Mechanistically, the combination induced accumulation of ROS, 4-HNE, as well as activation of caspase-3 which indicates that this combination simultaneously increases cell ferroptosis and apoptosis. Notably, inhibition of GPX4 or NRF2 can suppress ovarian cancer spreading and growth in the peritoneal cavity of mice, while the combination of NRF2 inhibitor ML385 with GPX4 inhibitors showed a significant synergistic effect compared to individual drug treatment in a syngeneic mouse ovarian cancer model. Overall, these findings suggest that combining NRF2 inhibitors with GPX4 inhibitors results in a synergy suppression of ovarian cancer in vitro and in vivo, and maybe a promising therapeutic strategy for the treatment of ovarian cancer.


Subject(s)
Genital Neoplasms, Female , Ovarian Neoplasms , Animals , Female , Humans , Mice , Apoptosis , Cell Cycle , Genital Neoplasms, Female/drug therapy , NF-E2-Related Factor 2/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
5.
Cell Death Differ ; 31(4): 431-446, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38418695

ABSTRACT

Ferroptosis, a regulated form of cell death triggered by iron-dependent lipid peroxidation, has emerged as a promising therapeutic strategy for cancer treatment, particularly in hepatocellular carcinoma (HCC). However, the mechanisms underlying the regulation of ferroptosis in HCC remain to be unclear. In this study, we have identified a novel regulatory pathway of ferroptosis involving the inhibition of Apurinic/apyrimidinic endonuclease 1 (APE1), a key enzyme with dual functions in DNA repair and redox regulation. Our findings demonstrate that inhibition of APE1 leads to the accumulation of lipid peroxidation and enhances ferroptosis in HCC. At the molecular level, the inhibition of APE1 enhances ferroptosis which relies on the redox activity of APE1 through the regulation of the NRF2/SLC7A11/GPX4 axis. We have identified that both genetic and chemical inhibition of APE1 increases AKT oxidation, resulting in an impairment of AKT phosphorylation and activation, which leads to the dephosphorylation and activation of GSK3ß, facilitating the subsequent ubiquitin-proteasome-dependent degradation of NRF2. Consequently, the downregulation of NRF2 suppresses SLC7A11 and GPX4 expression, triggering ferroptosis in HCC cells and providing a potential therapeutic approach for ferroptosis-based therapy in HCC. Overall, our study uncovers a novel role and mechanism of APE1 in the regulation of ferroptosis and highlights the potential of targeting APE1 as a promising therapeutic strategy for HCC and other cancers.


Subject(s)
Carcinoma, Hepatocellular , DNA-(Apurinic or Apyrimidinic Site) Lyase , Ferroptosis , Liver Neoplasms , Humans , Ferroptosis/drug effects , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Liver Neoplasms/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Cell Line, Tumor , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Mice , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/genetics , Mice, Nude , Lipid Peroxidation/drug effects , Signal Transduction/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors
6.
Cell Chem Biol ; 31(6): 1188-1202.e10, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38157852

ABSTRACT

Most BTB-containing E3 ligases homodimerize to recognize a single substrate by engaging multiple degrons, represented by E3 ligase KEAP1 dimer and its substrate NRF2. Inactivating KEAP1 to hinder ubiquitination-dependent NRF2 degradation activates NRF2. While various KEAP1 inhibitors have been reported, all reported inhibitors bind to KEAP1 in a monovalent fashion and activate NRF2 in a lagging manner. Herein, we report a unique bivalent KEAP1 inhibitor, biKEAP1 (3), that engages cellular KEAP1 dimer to directly release sequestered NRF2 protein, leading to an instant NRF2 activation. 3 promotes the nuclear translocation of NRF2, directly suppressing proinflammatory cytokine transcription. Data from in vivo experiments showed that 3, with unprecedented potency, reduced acute inflammatory burden in several acute inflammation models in a timely manner. Our findings demonstrate that the bivalent KEAP1 inhibitor can directly enable sequestered substrate NRF2 to suppress inflammatory transcription response and dampen various acute inflammation injuries.


Subject(s)
Inflammation , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/antagonists & inhibitors , Humans , Animals , Inflammation/drug therapy , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Male
7.
ChemMedChem ; 18(24): e202300282, 2023 12 14.
Article in English | MEDLINE | ID: mdl-37871186

ABSTRACT

Drug repurposing has emerged as an attractive strategy for accelerating drug discovery for cancer treatment. In this study, we investigated combining Tranylcypromine (TCP) with a number of well-characterized drugs. Among these combinations, NRF2 inhibitor (ML385) exhibited synergistic effects in combination with TCP. Specifically, our results showed that the combination of TCP and ML385 resulted in a significant reduction in tumor proliferation while neither drug affected cancer cell growth meaningfully on its own. While further studies are needed to understand fully the extent of the synergistic efficacy, the underlying respective mechanisms and the potential side effects of this approach, our study has yielded a promising start for the development of an effective combination cancer therapy.


Subject(s)
Neoplasms , Tranylcypromine , Humans , Drug Repositioning/methods , Neoplasms/drug therapy , NF-E2-Related Factor 2/antagonists & inhibitors , Tranylcypromine/pharmacology , Tranylcypromine/therapeutic use
8.
Ecotoxicol Environ Saf ; 249: 114376, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36508821

ABSTRACT

Cadmium (Cd) is a ubiquitous toxic metal and environmental pollutant. Increasing studies have shown that Cd exposure increases the incidence of various endocrine system diseases, including thyrotoxicity reflected by thyroid structural damage and endocrine toxicity. However, the observed outcomes are complex and conflicting, leading to the mechanism of Cd-induced thyrotoxicity remaining obscure. In this study, 4-week-old male C57BL/6 mice were given 2 or 7 mg/kg Cadmium Chloride (CdCl2) intragastrically for 4 and 8 weeks, and the Cd-mediated thyrotoxicity was evaluated by determining alterations in thyroid structure and endocrine function, and alterations of oxidant stress, apoptosis, and pyroptosis. Our data showed that Cd exposure could reduce body weight and induce thyrotoxicity by impairing thyroid follicular morphology and endocrine function, accompanied by elevated oxidative stress and apoptosis, macrophage infiltration, and inflammatory cytokine secretion. Importantly, Cd significantly promoted thyroid follicular cell pyroptosis by increasing Nlrp3, Asc, Caspase-1, Gsdmd, IL-1ß, and IL-18 expression. Mechanistical analysis suggested that Cd treatment could inhibit antioxidant pathway by downregulating antioxidant response protein, Nrf2, and upregulating its negative feedback regulator, Keap1. Collectively, our in vivo findings suggest that Cd exposure could facilitate thyroid follicular cell pyroptosis by inhibiting Nrf2/Keap1 signaling, thereby disrupting thyroid tissue structure and endocrine function, which offers novel insights into the Cd-mediated detrimental consequences on thyroid homeostasis.


Subject(s)
Antioxidants , Cadmium , Environmental Exposure , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Pyroptosis , Thyroid Gland , Animals , Male , Mice , Antioxidants/metabolism , Cadmium/toxicity , Kelch-Like ECH-Associated Protein 1/antagonists & inhibitors , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Inbred C57BL , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Pyroptosis/drug effects , Thyroid Gland/drug effects , Thyroid Gland/pathology
9.
Med Res Rev ; 43(1): 237-287, 2023 01.
Article in English | MEDLINE | ID: mdl-36086898

ABSTRACT

The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is considered the master regulator of the phase II antioxidant response. It controls a plethora of cytoprotective genes related to oxidative stress, inflammation, and protein homeostasis, among other processes. Activation of these pathways has been described in numerous pathologies including cancer, cardiovascular, respiratory, renal, digestive, metabolic, autoimmune, and neurodegenerative diseases. Considering the increasing interest of discovering novel NRF2 activators due to its clinical application, initial efforts were devoted to the development of electrophilic drugs able to induce NRF2 nuclear accumulation by targeting its natural repressor protein Kelch-like ECH-associated protein 1 (KEAP1) through covalent modifications on cysteine residues. However, off-target effects of these drugs prompted the development of an innovative strategy, the search of KEAP1-NRF2 protein-protein interaction (PPI) inhibitors. These innovative activators are proposed to target NRF2 in a more selective way, leading to potentially improved drugs with the application for a variety of diseases that are currently under investigation. In this review, we summarize known KEAP1-NRF2 PPI inhibitors to date and the bases of their design highlighting the most important features of their respective interactions. We also discuss the preclinical pharmacological properties described for the most promising compounds.


Subject(s)
Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Neurodegenerative Diseases , Humans , Inflammation/drug therapy , Kelch-Like ECH-Associated Protein 1/antagonists & inhibitors , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Oxidative Stress
10.
Front Biosci (Landmark Ed) ; 27(7): 223, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35866405

ABSTRACT

Breast cancer is the most common malignancy among women worldwide. Several studies indicate that, in addition to established risk factors for breast cancer, other factors such as cortisol release related to psychological stress and drug treatment with high levels of glucocorticoids may also contribute significantly to the initiation of breast cancer. There are several possible mechanisms by which glucocorticoids might promote neoplastic transformation of breast tissue. Among these, the least known and studied is the inhibition of the nuclear erythroid factor 2-related (Nrf2)-antioxidant/electrophile response element (ARE/EpRE) pathway by high levels of glucocorticoids. Specifically, Nrf2 is a potent transcriptional activator that plays a central role in the basal and inducible expression of many cytoprotective genes that effectively protect mammalian cells from various forms of stress and reduce the propensity of tissues and organisms to develop disease or malignancy including breast cancer. Consequently, a loss of Nrf2 in response to high levels of gluco-corticoids may lead to a decrease in cellular defense against oxidative stress, which plays an important role in the initiation of human mammary carcinogenesis. In the present review, we provide a comprehensive overview of the current state of knowledge of the cellular mechanisms by which both glucocorticoid pharmacotherapy and endogenous GCs (cortisol in humans and corticosterone in rodents) may contribute to breast cancer development through inhibition of the Nrf2-ARE/EpRE pathway and the protective role of melatonin against glucocorticoid-induced apoptosis in the immune system.


Subject(s)
Breast Neoplasms , Carcinogenesis , Glucocorticoids , NF-E2-Related Factor 2 , Antioxidants/pharmacology , Breast Neoplasms/chemically induced , Carcinogenesis/chemically induced , Female , Glucocorticoids/adverse effects , Humans , Hydrocortisone , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Oxidative Stress
11.
Kidney360 ; 3(4): 687-699, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35721612

ABSTRACT

Background: Bardoxolone methyl activates nuclear factor erythroid 2-related factor 2 (Nrf2) via covalent binding and irreversible inhibition of Kelch-like ECH-associated protein 1 (Keap1), the negative regulator of Nrf2. Ongoing clinical trials of bardoxolone methyl show promising effects for patients with CKD. However, the direct inhibition of Keap1-Nrf2 protein-protein interaction (PPI) as an approach to activate Nrf2 is less explored. Methods: We developed a noncovalent Nrf2 activator UBE-1099, which highly selectively inhibits Keap1-Nrf2 PPI, and evaluated its efficacy on the progressive phenotype in an Alport syndrome mouse model (Col4a5-G5X). Results: Similar to bardoxolone methyl, UBE-1099 transiently increased proteinuria and reduced plasma creatinine in Alport mice. Importantly, UBE-1099 improved the glomerulosclerosis, renal inflammation, and fibrosis, and prolonged the life span of Alport mice. UBE-1099 ameliorated the dysfunction of Nrf2 signaling in the renal tissue of Alport mice. Moreover, transcriptome analysis in the glomerulus showed that UBE-1099 induced the expression of genes associated with the cell cycle and cytoskeleton, which may explain its unique mechanism of improvement such as glomerular morphologic change. Conclusions: UBE-1099 significantly ameliorates the progressive phenotype in Alport mice. Our results revealed the efficacy of Keap1-Nrf2 PPI inhibitor for glomerulosclerosis and present a potential therapeutic drug for CKD.


Subject(s)
Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Nephritis, Hereditary , Renal Insufficiency, Chronic , Animals , Disease Models, Animal , Kelch-Like ECH-Associated Protein 1/antagonists & inhibitors , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/metabolism , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Phenotype
12.
Ecotoxicol Environ Saf ; 238: 113561, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35489292

ABSTRACT

Zearalenone (ZEA) is a nonsteroidal estrogenic mycotoxin, which mainly contaminates grains and has estrogen-like effects on the reproductive system. Betulinic acid (BA), a natural lupane-type pentacyclic triterpene, has anti-oxidative and anti-inflammatory properties. This study aimed to investigate whether BA alleviates ZEA-induced testicular damage and explore the possible mechanism. Here, BA ameliorated testicular damage by mitigating the disordered arrangement of seminiferous tubules, the exfoliation of lumen cells, and the increase of cell apoptosis caused by ZEA. Meanwhile, BA alleviated ZEA-triggered testicular damage by restoring hormone levels and sperm motility, and reconstructing the blood-testis-barrier. Moreover, BA alleviated ZEA-exposed testicular oxidative stress by activating Nrf2 pathway. Furthermore, BA moderated ZEA-evoked testicular inflammation by inhibiting p38/ERK MAPK pathway. Overall, our results revealed that BA has a therapeutic protective effect on ZEA-induced testicular injury and oxidative stress via p38/ERK MAPK inhibition and Nrf2-mediated antioxidant defense activation, which provides a viable alternative to alleviate ZEA-induced male reproductive toxicology.


Subject(s)
MAP Kinase Signaling System , NF-E2-Related Factor 2 , Pentacyclic Triterpenes , Testis , Zearalenone , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis , MAP Kinase Signaling System/drug effects , Male , Mice , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Pentacyclic Triterpenes/pharmacology , Sperm Motility/drug effects , Testis/drug effects , Zearalenone/toxicity , p38 Mitogen-Activated Protein Kinases/metabolism , Betulinic Acid
13.
Pharm Biol ; 60(1): 638-651, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35298357

ABSTRACT

CONTEXT: Shengmai injection (SMI) has been used to treat heart failure. OBJECTIVE: This study determines the molecular mechanisms of SMI against cardiotoxicity caused by doxorubicin (DOX). MATERIALS AND METHODS: In vivo, DOX (15 mg/kg) was intraperitoneally injected in model, Dex (dexrazoxane), SMI-L (2.7 mL/kg), SMI-M (5.4 mL/kg), and SMI-H (10.8 mL/kg) for 7 consecutive days. Hematoxylin-eosin (HE) and Masson staining were used to evaluate histological changes, and cardiomyocyte apoptosis was identified using TdT-mediated dUTP nick-end labelling (TUNEL). Enzymatic indexes were determined. mRNA and protein expressions were analysed through RT-qPCR and Western blotting. In vitro, H9c2 cells were divided into control group, model group (2 mL 1 µM DOX), SMI group, ML385 group, and SMI + ML385 group, the intervention lasted for 24 h. mRNA and protein expressions were analysed. RESULTS: SMI markedly improved cardiac pathology, decreased cardiomyocyte apoptosis, increased creatine kinase (CK), lactate dehydrogenase (LDH), malondialdehyde (MDA), decreased superoxide dismutase (SOD). Compared with the model group, the protein expression of nuclear factor erythroid2-related factor 2 (Nrf2) (SMI-L: 2.42-fold, SMI-M: 2.67-fold, SMI-H: 3.07-fold) and haem oxygenase-1(HO-1) (SMI-L: 1.64-fold, SMI-M: 2.01-fold, SMI-H: 2.19-fold) was increased and the protein expression of kelch-like ECH-associated protein 1 (Keap1) (SMI-L: 0.90-fold, SMI-M: 0.77-fold, SMI-H: 0.66-fold) was decreased in SMI groups and Dex group in vivo. Additionally, SMI dramatically inhibited apoptosis, decreased CK, LDH and MDA levels, and enhanced SOD activity. Our results demonstrated that SMI reduced DOX-induced cardiotoxicity via activation of the Nrf2/Keap1 signalling pathway. CONCLUSIONS: This study revealed a new mechanism by which SMI alleviates DOX-induced 45 cardiomyopathy by modulating the Nrf2/Keap1 signal pathway.


Subject(s)
Doxorubicin/toxicity , Drugs, Chinese Herbal/pharmacology , Animals , Apoptosis/drug effects , Cardiotoxicity/prevention & control , Cells, Cultured , Drug Combinations , Drugs, Chinese Herbal/chemistry , Kelch-Like ECH-Associated Protein 1/chemistry , Kelch-Like ECH-Associated Protein 1/physiology , Molecular Docking Simulation , Myocardium/pathology , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
14.
Cell Death Dis ; 13(2): 166, 2022 02 21.
Article in English | MEDLINE | ID: mdl-35190529

ABSTRACT

The complex interaction between cancer cells and the immune microenvironment is a central regulator of tumor growth and the treatment response. Chemotherapy-induced senescence is accompanied by the senescence-associated secretion phenotype (SASP). However, the mechanisms underlying the regulation of the SASP remain the most poorly understood element of senescence. Here, we show that nuclear erythroid factor 2-like factor 2 (Nrf2), a master antioxidative transcription factor, accumulates upon doxorubicin-induced senescence. This is due to the increased cytoplasmic Inhibitor of Apoptosis Stimulating Protein of P53, iASPP, which binds with Keap1, interrupting Keap1/Nrf2 interaction and promoting Nrf2 stabilization and activation. Activated Nrf2 transactivates a novel target gene of SASP factor, macrophage colony-stimulating factor (M-CSF), which subsequently acts on macrophages and induces polarization from M1 to M2 via a paracrine mechanism. Genetic inhibition of iASPP-Nrf2 suppresses the growth of apoptosis-resistant xenografts, with further analysis revealing that M-CSF/M-CSFR-regulated macrophage polarization is critical for the functional outcomes delineated above. Overall, our data uncover a novel function of iASPP-Nrf2 in skewing the immune microenvironment under treatment-induced senescence. Targeting the iASPP-Nrf2 axis could be a powerful strategy for the implementation of new chemotherapy-based therapeutic opportunities.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Cell Polarity , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Macrophage Activation , Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Macrophage Colony-Stimulating Factor/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Microenvironment
15.
J Med Chem ; 65(4): 3473-3517, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35108001

ABSTRACT

Upregulation of the transcription factor Nrf2 by inhibition of the interaction with its negative regulator Keap1 constitutes an opportunity for the treatment of disease caused by oxidative stress. We report a structurally unique series of nanomolar Keap1 inhibitors obtained from a natural product-derived macrocyclic lead. Initial exploration of the structure-activity relationship of the lead, followed by structure-guided optimization, resulted in a 100-fold improvement in inhibitory potency. The macrocyclic core of the nanomolar inhibitors positions three pharmacophore units for productive interactions with key residues of Keap1, including R415, R483, and Y572. Ligand optimization resulted in the displacement of a coordinated water molecule from the Keap1 binding site and a significantly altered thermodynamic profile. In addition, minor reorganizations of R415 and R483 were accompanied by major differences in affinity between ligands. This study therefore indicates the importance of accounting both for the hydration and flexibility of the Keap1 binding site when designing high-affinity ligands.


Subject(s)
Kelch-Like ECH-Associated Protein 1/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , NF-E2-Related Factor 2/antagonists & inhibitors , Animals , Binding Sites , Hepatocytes/metabolism , Humans , Ligands , Microsomes, Liver/metabolism , Models, Molecular , Molecular Docking Simulation , Rats , Signal Transduction/drug effects , Structure-Activity Relationship
16.
Bioorg Med Chem ; 56: 116627, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35063896

ABSTRACT

As our ongoing work on lathyrane diterpenoid derivatization, three series of lathyrane diterpenoid derivatives were designed and synthesized based combination principles, including pyrazole, thiazole and furoxan moieties. Biological evaluation indicated that compound 23d exhibited excellently inhibitory activity on LPS-induced NO production in RAW264.7 cells (IC50 = 0.38 ± 0.18 µM). The preliminary structure-activity relationships (SARs) suggested that phenylsulfonyl substituted furoxan moiety had the strongest ability to improve anti-inflammatory activity of lathyrane diterpenoids. Furthermore, compound 23d significantly reduced the level of ROS. Its molecular mechanism was related to inhibiting the transcriptional activation of Nrf2/HO-1 pathway. Based on these considerations, 23d might be a promising anti-inflammatory agent, which is noteworthy for further exploration.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diterpenes/pharmacology , Heterocyclic Compounds/pharmacology , Nitrogen Compounds/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Diterpenes/chemical synthesis , Diterpenes/chemistry , Dose-Response Relationship, Drug , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/metabolism , Heterocyclic Compounds/chemistry , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Molecular Structure , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitrogen Compounds/chemistry , RAW 264.7 Cells , Structure-Activity Relationship
17.
ACS Appl Mater Interfaces ; 14(5): 6404-6416, 2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35077153

ABSTRACT

In situ oxygen generation is the most common strategy to boost reactive oxygen species (ROS) for enhancing the efficacy of phototherapy in cancer, including photodynamic therapy (PDT) and photothermal therapy (PTT). However, hyperoxidation or hyperthermia often triggers stress-defense pathways and promotes tumor cell survival, thus severely limiting the therapeutic efficacy. To overcome the tumor hypoxia and thermal resistance existing in phototherapy, we constructed a self-synergistic nanoplatform for tumors by incorporating brusatol, a nuclear factor erythroid 2-related factor (Nrf2) inhibitor, into the silica nanonetwork. It was then sequentially decorated with MnO2 and the photosensitizer chlorin e6 (Ce6) and then coated with poly(ethylene glycol)-folate (PEG-FA)-functionalized polydopamine (PDA) (designated as brusatol/silica@MnO2/Ce6@PDA-PEG-FA). As an oxygen generator, MnO2 can promote ROS production, which not only directly enhances Ce6-mediated PDT but also strengthens PDA-mediated PTT by attacking heat shock proteins (HSPs). Particularly, brusatol could efficiently inhibit the activation of Nrf2 defense pathway under hyperoxidation and hyperthermia and cause glutathione peroxidase 4 (GPX4) and ferritin heavy chain (FTH) inactivation, thereby inducing ferroptosis and ultimately enhancing the phototherapeutic effects. By exploiting these features, brusatol/silica@MnO2/Ce6@PDA-PEG-FA exhibited excellent antitumor efficacy with enhanced PDT and PTT both in in vitro and in vivo studies. Overall, our work highlights a promising strategy against hypoxia- and hyperthermia-associated resistance in phototherapy via suppressing stress-defense system and inducing ferroptosis.


Subject(s)
Ferroptosis , NF-E2-Related Factor 2/metabolism , Nanostructures/chemistry , Phototherapy/methods , Reactive Oxygen Species/metabolism , Animals , Cell Line, Tumor , Chlorophyllides/chemistry , Chlorophyllides/pharmacology , Chlorophyllides/therapeutic use , Ferroptosis/drug effects , Folic Acid/analogs & derivatives , Folic Acid/chemistry , Humans , Hyperthermia, Induced , Indoles/chemistry , Infrared Rays , Manganese Compounds/chemistry , Mice , NF-E2-Related Factor 2/antagonists & inhibitors , Nanostructures/therapeutic use , Nanostructures/toxicity , Oxides/chemistry , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Polyethylene Glycols/chemistry , Polymers/chemistry , Quassins/chemistry , Silicon Dioxide/chemistry
18.
Eur J Pharmacol ; 918: 174563, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-34942162

ABSTRACT

Oxidative stress plays a crucial role in fatigue, thus it is of significance to develop safe and efficient antioxidant to prevent fatigue. Phlorizin (PHZ) is a major active ingredient of dihydrochalcone from Lithocarpus polystachyus Rehd., which has already been approved as a new food material in China since 2017. The current study was designed to investigate the effect of PHZ on fatigue, and further to elucidate its possible underlying mechanism. Our results revealed that PHZ exerted beneficial effect on exhaustive exercise-induced fatigue in mice, as reflected by rotarod test and exhaustive swimming test. Moreover, PHZ also effectively decreased the levels of blood urea nitrogen, creatine kinase and plasma lactic acid, increased the liver glycogen and skeletal muscle glycogen of fatigued mice, as evidenced by enzyme linked immunosorbent assay. PHZ balanced the redox status through reducing generation of reactive oxygen species, enhancing the activities of antioxidative enzymes. Furthermore, PHZ not only increased the ratio of Bcl2/Bax, but also decreased the level of cleaved-caspase 3. Notably, PHZ facilitated nuclear factor erythroid 2-related factor 2 (Nrf2) translocated from cytoplasm to nucleus, and up-regulated its downstream antioxidant response element including heme oxygenase-1 and NADPH quinone oxidoreductase-1. Intriguingly, PHZ directly bound to Nrf2, as evidenced by molecular docking, and the anti-fatigue effects of PHZ were almost abolished in Nrf2 deficient mice. In summary, our findings suggest that PHZ might be a natural occurring antioxidant with safety profile to relieve fatigue via targeting Nrf2 to inhibit apoptosis.


Subject(s)
Muscle Fatigue/drug effects , NF-E2-Related Factor 2 , Oxidative Stress/drug effects , Phlorhizin/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Chalcones/pharmacology , Mice , Molecular Docking Simulation , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction/drug effects , Signal Transduction/drug effects
19.
Bioorg Chem ; 119: 105516, 2022 02.
Article in English | MEDLINE | ID: mdl-34856444

ABSTRACT

Both ruthenium (Ru) and isoquinoline (IQ) compounds are regarded as potential anticancer drug candidates. Here, we report the synthesis and characterization of three novel cyclometalated Ru(II)-isoquinoline complexes: RuIQ-3, RuIQ-4, and RuIQ-5, and evaluation of their in vitro cytotoxicities against a panel of cell lines including A549/DDP, a cisplatin-resistant human lung cancer cell line. A549/DDP 3D multicellular tumor spheroids (MCTSs) were also used to detect the drug resistance reversal effect of Ru(II)-IQ complexes. Our results indicated that the cytotoxic activities against cancer cells of Ru(II)-IQ complexes, especially RuIQ-5, were superior compared with cisplatin. In addition, RuIQ-5 exhibited low toxicity towards both normal HBE cells in vitro and zebrafish embryos in vivo. Further investigation on cellular mechanism of action indicated that after absorption by A549/DDP cells, RuIQ-5 was mainly distributed in the nucleus, which is different from cisplatin. Besides, RuIQ-5 could induce apoptosis through mitochondrial dysfunction, reactive oxygen species (ROS) accumulation, ROS-mediated DNA damage, and cycle arrest at both S and G2/M phases. Moreover, RuIQ-5 could inhibit the overexpression of Nrf2 through regulation of Akt/GSK-3ß/Fyn signaling pathway and hindering the nuclear translocation of Nrf2. Based on these findings, we firmly believe that the studied Ru(II)-IQ complexes hold great promise as anticancer therapeutics with high effectiveness and low toxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Coordination Complexes/pharmacology , Isoquinolines/pharmacology , Ruthenium/pharmacology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cisplatin/chemistry , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Isoquinolines/chemistry , Molecular Structure , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Ruthenium/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured , Zebrafish
20.
Oxid Med Cell Longev ; 2021: 1608586, 2021.
Article in English | MEDLINE | ID: mdl-34917229

ABSTRACT

Vitiligo is a cutaneous depigmentation disease due to loss of epidermal melanocytes. Accumulating evidence has indicated that oxidative stress plays a vital role in vitiligo via directly destructing melanocytes and triggering inflammatory response that ultimately undermines melanocytes. Folic acid (FA), an oxidized form of folate with high bioavailability, exhibits potent antioxidant properties and shows therapeutic potential in multiple oxidative stress-related diseases. However, whether FA safeguards melanocytes from oxidative damages remains unknown. In this study, we first found that FA relieved melanocytes from H2O2-induced abnormal growth and apoptosis. Furthermore, FA enhanced the activity of antioxidative enzymes and remarkably reduced intracellular ROS levels in melanocytes. Subsequently, FA effectively activated nuclear factor E2-related factor 2 (Nrf2) pathway, and Nrf2 knockdown blocked the protective effects of FA on H2O2-treated melanocytes. Additionally, FA inhibited the production of proinflammatory HMGB1 in melanocytes under oxidative stress. Taken together, our findings support the protective effects of FA on human melanocytes against oxidative injury via the activation of Nrf2 and the inhibition of HMGB1, thus indicating FA as a potential therapeutic agent for the treatment of vitiligo.


Subject(s)
Folic Acid/pharmacology , Gene Expression Regulation/drug effects , HMGB1 Protein/metabolism , Melanocytes/drug effects , NF-E2-Related Factor 2/antagonists & inhibitors , Oxidative Stress , Protective Agents/pharmacology , Antioxidants/pharmacology , Apoptosis , HMGB1 Protein/genetics , Humans , Melanocytes/metabolism , Melanocytes/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Vitamin B Complex/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL