Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 206
Filter
1.
Nutrients ; 16(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38999824

ABSTRACT

Parkinson's disease (PD) is a degenerative neurological disorder defined by the deterioration and loss of dopamine-producing neurons in the substantia nigra, leading to a range of motor impairments and non-motor symptoms. The underlying mechanism of this neurodegeneration remains unclear. This research examined the neuroprotective properties of Ecklonia cava polyphenols (ECPs) in mitigating neuronal damage induced by rotenone via the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway. Using human neuroblastoma SH-SY5Y cells and PD model mice, we found that ECP, rich in the antioxidant polyphenol phlorotannin, boosted the gene expression and functionality of the antioxidant enzyme NAD(P)H quinone oxidoreductase-1. ECP also promoted Nrf2 nuclear translocation and increased p62 expression, suggesting that p62 helps sustain Nrf2 activation via a positive feedback loop. The neuroprotective effect of ECP was significantly reduced by Compound C (CC), an AMP-activated protein kinase (AMPK) inhibitor, which also suppressed Nrf2 nuclear translocation. In PD model mice, ECPs improved motor functions impaired by rotenone, as assessed by the pole test and wire-hanging test, and restored intestinal motor function and colon tissue morphology. Additionally, ECPs increased tyrosine hydroxylase expression in the substantia nigra, indicating a protective effect on dopaminergic neurons. These findings suggest that ECP has a preventative effect on PD.


Subject(s)
NF-E2-Related Factor 2 , Neuroprotective Agents , Parkinson Disease , Polyphenols , Rotenone , NF-E2-Related Factor 2/metabolism , Animals , Polyphenols/pharmacology , Humans , Neuroprotective Agents/pharmacology , Mice , Male , Parkinson Disease/metabolism , Parkinson Disease/prevention & control , Parkinson Disease/drug therapy , Antioxidant Response Elements/drug effects , Signal Transduction/drug effects , Disease Models, Animal , Cell Line, Tumor , Antioxidants/pharmacology , Mice, Inbred C57BL , Plant Extracts/pharmacology , NAD(P)H Dehydrogenase (Quinone)/metabolism
2.
J Ethnopharmacol ; 333: 118497, 2024 Oct 28.
Article in English | MEDLINE | ID: mdl-38942156

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The traditional medicinal formulation, Qifu-yin (QFY), has been widely prescribed for Alzheimer's disease (AD) treatment in China, yet the comprehensive mechanisms through which QFY mitigates AD pathology remain to be fully delineated. AIM OF THE STUDY: This study aimed to explore the therapeutic implications of QFY on the synaptic injury and oxidative stress in the hippocampus of APPswe/PS1dE9 (APP/PS1) mice, with a concerted effort to elucidate the molecular mechanisms related to synaptic preservation and memory improvement. MATERIALS AND METHODS: The components of QFY were identified by ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). The neuroprotective effects of QFY was evaluated using six-month-old male APP/PS1 mice. Subsequent to a 15 days of QFY regimen, spatial memory was assessed utilizing the Morris water maze (MWM) test. Amyloid-beta (Aß) aggregation was detected via immunostaining, while the quantification of Aß1-40 and Aß1-42 was achieved through enzyme-linked immunosorbent assay (ELISA). Transmission electron microscopy (TEM) was used to investigate the synaptic structure and mitochondrial morphology. Golgi staining was applied to examine dendritic spine density. Reactive oxygen species (ROS), 3-nitrotyrosine (3-NT) and 4-hydroxy-nonenal (4-HNE) assays were employed to assess oxidative stress. The expression profiles of Aß metabolism-associated enzymes and the Keap1/Nrf2/ARE signaling pathway were determined by Western blot. RESULTS: A total of 20 principal compounds in QFY were identified. QFY mitigated memory deficits of APP/PS1 mice, including reducing escape latency and search distance and increasing the time and distance spent in the target quadrant. In addition, QFY increased platform crossings of APP/PS1 mice in the probe trial of MWM tests. TEM analysis showed that QFY increased synapse number in the CA1 region of APP/PS1 mice. Further studies indicated that QFY elevated the expression levels of Post synaptic density protein 95 (PSD95) and synaptophysin, and mitigated the loss of dendritic spine density in the hippocampus of APP/PS1 mice. QFY has been shown to ameliorated the structural abnormalities of mitochondria, including mitochondrial dissolution and degradation, up-regulate ATP synthesis and membrane potential in the hippocampus of APP/PS1 mice. Moreover, QFY activated the Keap1/Nrf2/ARE signaling pathway in the hippocampus of APP/PS1 mice, which might contribute to the neuroprotective effects of QFY. CONCLUSION: QFY activates the Keap1/Nrf2/ARE signaling, and protects against synaptic and mitochondrial dysfunction in APP/PS1 mice, proposing a potential alternative therapeutic strategy for AD management.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Drugs, Chinese Herbal , Kelch-Like ECH-Associated Protein 1 , Mice, Transgenic , NF-E2-Related Factor 2 , Neuroprotective Agents , Oxidative Stress , Signal Transduction , Animals , Kelch-Like ECH-Associated Protein 1/metabolism , Oxidative Stress/drug effects , NF-E2-Related Factor 2/metabolism , Male , Signal Transduction/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Mice , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Neuroprotective Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Presenilin-1/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Synapses/drug effects , Synapses/metabolism , Antioxidant Response Elements/drug effects , Disease Models, Animal
3.
Food Chem Toxicol ; 190: 114842, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942164

ABSTRACT

High levels of reactive oxygen species (ROS) have been associated with the progression of neurodegenerative diseases such as Alzheimer's disease. The activation of the NFE2-related factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathway may restore the neuron's redox balance and provide a therapeutic impact. Hydroxygenkwanin (HGK), a dominant flavone from Genkwa Flos, has received expanding attention due to its medicinal activities. Our investigation results demonstrated the ability of HGK to protect the PC12 cells from oxidative damage caused by an excessive hydrogen peroxide load. HGK also showed the ability to upregulate a panel of endogenous antioxidant proteins. Further investigations have demonstrated that the neuroprotection mechanism of HGK is dependent on the activation of the Nrf2/ARE signaling pathway. Activating the Nrf2/ARE pathway by HGK reveals a novel mechanism for understanding the pharmacological functions of HGK. These findings suggest that HGK could be considered for further development as an oxidative stress-related neurological pathologies potential therapeutic drug.


Subject(s)
Antioxidant Response Elements , NF-E2-Related Factor 2 , Neuroprotective Agents , Oxidative Stress , Signal Transduction , NF-E2-Related Factor 2/metabolism , Animals , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , PC12 Cells , Rats , Antioxidant Response Elements/drug effects , Oxidative Stress/drug effects , Hydrogen Peroxide , Flavones/pharmacology , Reactive Oxygen Species/metabolism
4.
Nephrology (Carlton) ; 29(8): 482-494, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38837564

ABSTRACT

BACKGROUND: Apoptosis and oxidative stress in kidneys are critical players in acute kidney injury (AKI). Rehmapicrogenin, a monomeric compound extracted from Rehmanniae radix, has been found to possess nitric oxide inhibitory and anti-inflammatory activities. Thus, this study aimed to investigate the roles and mechanisms of rehmapicrogenin in AKI. METHODS: Lipopolysaccharide (LPS) was used to induce AKI-like conditions. Cell survival conditions were detected by cell counting kit-8 assays and flow cytometry. Several renal function markers including blood urea nitrogen, proteinuria, creatinine, and albumin were measured. Apoptosis and reactive oxygen species (ROS) production were examined by TUNEL and dihydroethidium staining, respectively. Haematoxylin-eosin staining and periodic acid-Schiff staining were conducted to assess histopathological changes. Gene expression was evaluated by western blotting, commercially available kits and immunofluorescence staining. RESULTS: For in vitro analysis, rehmapicrogenin inhibited the LPS-induced podocyte apoptosis by activating the Nrf2/ARE pathway. For in vivo analysis, rehmapicrogenin improved renal functions in LPS-induced mice. Additionally, rehmapicrogenin suppressed LPS-induced podocyte apoptosis and oxidative stress in kidney tissues. Mechanistically, rehmapicrogenin activated the Nrf2/ARE pathway in LPS-induced mice. CONCLUSION: Rehmapicrogenin relieves the podocyte injury and renal dysfunctions through activating the Nrf2/ARE pathway to inhibit apoptosis and oxidative stress.


Subject(s)
Acute Kidney Injury , Apoptosis , Disease Models, Animal , Lipopolysaccharides , NF-E2-Related Factor 2 , Oxidative Stress , Podocytes , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Podocytes/drug effects , Podocytes/metabolism , Podocytes/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Apoptosis/drug effects , Signal Transduction/drug effects , Oxidative Stress/drug effects , Mice , Antioxidant Response Elements/drug effects , Male , Reactive Oxygen Species/metabolism , Cell Line , Antioxidants/pharmacology
5.
J Ethnopharmacol ; 332: 118352, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38762208

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Chrysanthemum morifolium Ramat. is a commonly used Chinese herb and food homologous plant with traditional effects such as anti-inflammatory, antifebrile, antibacterial and antiviral. AIM OF STUDY: Photoaging is one of the main causes of accelerated skin aging. Chrysanthemum morifolium Ramat. has reported to alleviate photodamage. In this study, we investigated the protective effect of the extract of buds of Chrysanthemum morifolium Ramat. (CE) on UVB-induced photoaging and further mechanism. MATERIALS AND METHODS: The extract of buds of chrysanthemum was analyzed by HPLC-Q-TOF-MS/MS. Antioxidant activity was assessed by DPPH and ABTS assay. Cell viability examined by cell counting kit-8 assay. The ROS level was detected by fluorescent probe DCFH-DA. Protein expression evaluated by Western blotting. The skin tissue investigated by immunohistochemistry. RESULTS: CE significantly reversed the decrease of cell viability that induced by UVB in HaCaT and HFF-1 cells. Further analysis showed that CE alleviated photoaging by inhibiting the expression of mitogen-activated protein kinase (MAPK) and activating the NF-E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway to promote the expression of antioxidant enzymes. Moreover, CE effectively improved the reduced skin hydration, disordered collagen and thickening epidermis caused by UVB in mice. CONCLUSIONS: All results demonstrated that CE had therapeutic effect on UVB-induced photoaging and provided theoretical basis for its further developing as a natural functional product with anti-photoaging effect.


Subject(s)
Chrysanthemum , NF-E2-Related Factor 2 , Plant Extracts , Skin Aging , Ultraviolet Rays , Chrysanthemum/chemistry , Skin Aging/drug effects , Skin Aging/radiation effects , Animals , NF-E2-Related Factor 2/metabolism , Ultraviolet Rays/adverse effects , Plant Extracts/pharmacology , Plant Extracts/chemistry , Humans , Mice , Antioxidants/pharmacology , Antioxidants/isolation & purification , Cell Survival/drug effects , Antioxidant Response Elements/drug effects , Skin/drug effects , Skin/radiation effects , Skin/pathology , Skin/metabolism , Flowers/chemistry , Mitogen-Activated Protein Kinases/metabolism , HaCaT Cells , Signal Transduction/drug effects , Cell Line
6.
Environ Toxicol ; 39(8): 4221-4230, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38738704

ABSTRACT

Chronic Prostatitis/Chronic Pelvic Pain Syndrome (CP/CPPS) is a common and serious disease with unclear pathogenesis and recurrent symptoms. Hedyotis diffusa Willd (HDW) has been recognized for its potential in managing various chronic inflammatory diseases. This research aimed to interrogate the mechanism of HDW in treating CP/CPPS. Complete Freund Adjuvant (CFA) and LPS were utilized to establish the rat and cell models of CP/CPPS. Results showed that HDW decreased levels of inflammation-related factors in CP rat prostate tissue and LPS-elicited RWPE-1 cell injury model. Moreover, HDW administration impaired oxidative stress in the prostate and RWPE-1 cells. In addition, HDW treatment activated the NRF2/ARE signaling in rat prostate tissue and cell models. Interestingly, NRF2/ARE pathway inhibitor ML385 reversed the inhibition effects of cell apoptosis, inflammation, and oxidative stress triggered by HDW. In summary, HDW alleviated inflammation and oxidative stress by activating NRF2/ARE signaling in CP/CPPS rat model and human prostate epithelial cell injury model.


Subject(s)
Hedyotis , Inflammation , NF-E2-Related Factor 2 , Oxidative Stress , Prostatitis , Signal Transduction , Male , Prostatitis/chemically induced , Prostatitis/pathology , Prostatitis/metabolism , Prostatitis/drug therapy , Animals , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Humans , Hedyotis/chemistry , Rats , Rats, Sprague-Dawley , Plant Extracts/pharmacology , Prostate/drug effects , Prostate/pathology , Prostate/metabolism , Cell Line , Antioxidant Response Elements/drug effects , Chronic Disease
7.
Ann Anat ; 254: 152260, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38521364

ABSTRACT

BACKGROUND: Oxidative stress plays a crucial role in the pathogenesis of many skeletal diseases by inducing osteocyte death. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulator of various antioxidant gene expressions through antioxidant response element (ARE) against cellular oxidative stress and can be induced by various stimulants, including the phytochemicals methysticin (MET) and L-sulforaphane (SFN). This study aimed to establish an osteocyte in vitro model to investigate the pharmacological effects of MET and SFN on the Nrf2/ARE pathway. METHODS: MLO-Y4 murine osteocytes and the stably transduced MLO-Y4-SIN-lenti-ARE reporter gene cell line were used. MET and SFN were used as Nrf2 inducers. The cytotoxicity of MET, SFN, and hydrogen peroxide (H2O2) was evaluated using the CytoTox-Glo™ Assay. Time- and dose-dependent ARE induction was examined by Monoluciferase Assay. The mRNA and protein expressions of Nrf2 target markers, such as heme-oxygenase 1 (Ho-1), NADPH quinone dehydrogenase 1 (Nqo1), and thioredoxin reductase 1 (Txnrd1), were detected by RT-qPCR, Western Blot, and immunofluorescence staining, respectively. Osteogenesis markers, osteopontin, and osteocalcin were compared with and without treatment by immunofluorescence staining. RESULTS: The experimental data showed that MET and SFN induced ARE activity in a time- and dose-dependent manner and increased the mRNA and protein expression of antioxidant markers compared to vehicle-treated controls. The protein expression of osteopontin and osteocalcin in the samples treated with SFN were significantly higher than without treatment, and the number of cell death treated with SFN was significantly lower than without treatment under H2O2-induced stress conditions. CONCLUSIONS: Nrf2 inducers MET and SFN increased the mRNA expression of antioxidant genes through the Nrf2/ARE pathway in osteocytes. Notably, SFN increased the protein expression of osteocyte-associated osteogenic markers and suppressed cell death under H2O2-induced stress condition. Thus, Nrf2 stimulators can exert stress-relieving and osteogenic effects on osteocytes.


Subject(s)
Antioxidant Response Elements , Isothiocyanates , NF-E2-Related Factor 2 , Osteocytes , Signal Transduction , Sulfoxides , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Mice , Osteocytes/drug effects , Osteocytes/metabolism , Signal Transduction/drug effects , Isothiocyanates/pharmacology , Sulfoxides/pharmacology , Antioxidant Response Elements/drug effects , Cell Line , Oxidative Stress/drug effects , Hydrogen Peroxide/pharmacology , Antioxidants/pharmacology , Osteopontin/metabolism , Osteopontin/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Thioredoxin Reductase 1/metabolism
8.
J Virol ; 97(4): e0016023, 2023 04 27.
Article in English | MEDLINE | ID: mdl-36939350

ABSTRACT

Host-derived cellular pathways can provide an unfavorable environment for virus replication. These pathways have been a subject of interest for herpesviruses, including the betaherpesvirus human cytomegalovirus (HCMV). Here, we demonstrate that a compound, ARP101, induces the noncanonical sequestosome 1 (SQSTM1)/p62-Keap1-Nrf2 pathway for HCMV suppression. ARP101 increased the levels of both LC3 II and SQSTM1/p62 and induced phosphorylation of p62 at the C-terminal domain, resulting in its increased affinity for Keap1. ARP101 treatment resulted in Nrf2 stabilization and translocation into the nucleus, binding to specific promoter sites and transcription of antioxidant enzymes under the antioxidant response element (ARE), and HCMV suppression. Knockdown of Nrf2 recovered HCMV replication following ARP101 treatment, indicating the role of the Keap1-Nrf2 axis in HCMV inhibition by ARP101. SQSTM1/p62 phosphorylation was not modulated by the mTOR kinase or casein kinase 1 or 2, indicating ARP101 engages other kinases. Together, the data uncover a novel antiviral strategy for SQSTM1/p62 through the noncanonical Keap1-Nrf2 axis. This pathway could be further exploited, including the identification of the responsible kinases, to define the biological events during HCMV replication. IMPORTANCE Antiviral treatment for human cytomegalovirus (HCMV) is limited and suffers from the selection of drug-resistant viruses. Several cellular pathways have been shown to modulate HCMV replication. The autophagy receptor sequestosome 1 (SQSTM1)/p62 has been reported to interact with several HCMV proteins, particularly with components of HCMV capsid, suggesting it plays a role in viral replication. Here, we report on a new and unexpected role for SQSTM1/p62, in HCMV suppression. Using a small-molecule probe, ARP101, we show SQSTM1/p62 phosphorylation at its C terminus domain initiates the noncanonical Keap1-Nrf2 axis, leading to transcription of genes under the antioxidant response element, resulting in HCMV inhibition in vitro. Our study highlights the dynamic nature of SQSTM1/p62 during HCMV infection and how its phosphorylation activates a new pathway that can be exploited for antiviral intervention.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Virus Replication , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Antiviral Agents/pharmacology , Transcription, Genetic/drug effects , Phosphorylation/drug effects , Antioxidant Response Elements/drug effects , Cell Line , Humans
9.
Int J Mol Sci ; 24(4)2023 Feb 12.
Article in English | MEDLINE | ID: mdl-36835090

ABSTRACT

Cancer is one of the leading causes of death worldwide. Chemotherapy and radiation therapy are currently providing the basis for cancer therapies, although both are associated with significant side effects. Thus, cancer prevention through dietary modifications has been receiving growing interest. The potential of selected flavonoids in reducing carcinogen-induced reactive oxygen species (ROS) and DNA damage through the activation of nuclear factor erythroid 2 p45 (NF-E2)-related factor (Nrf2)/antioxidant response element (ARE) pathway was studied in vitro. Dose-dependent effects of pre-incubated flavonoids on pro-carcinogen 4-[(acetoxymethyl)nitrosamino]-1-(3-pyridyl)-1-butanone (NNKAc)-induced ROS and DNA damage in human bronchial epithelial cells were studied in comparison to non-flavonoids. The most effective flavonoids were assessed for the activation of Nrf2/ARE pathway. Genistein, procyanidin B2 (PCB2), and quercetin significantly suppressed the NNKAc-induced ROS and DNA damage. Quercetin significantly upregulated the phosphorylated protein kinase B/Akt. PCB2 significantly upregulated the activation of Nrf2 and Akt through phosphorylation. Genistein and PCB2 significantly upregulated the phospho-Nrf2 nuclear translocation and catalase activity. In summary, genistein and PCB2 reduced the NNKAc-induced ROS and DNA damage through the activation of Nrf2. Further studies are required to understand the role of dietary flavonoids on the regulation of the Nrf2/ARE pathway in relation to carcinogenesis.


Subject(s)
Carcinogens , Epithelial Cells , Genistein , Proanthocyanidins , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species , Humans , Antioxidant Response Elements/drug effects , Carcinogens/pharmacology , DNA Damage/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Flavonoids/pharmacology , Genistein/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-akt/metabolism , Quercetin/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction , Proanthocyanidins/pharmacology
10.
Curr Med Sci ; 42(1): 56-67, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34881424

ABSTRACT

OBJECTIVE: Pulmonary hypertension (PH) is a severe pulmonary vascular disease that eventually leads to right ventricular failure and death. The purpose of this study was to investigate the mechanism by which pachymic acid (PA) pretreatment affects PH and pulmonary vascular remodeling in rats. METHODS: PH was induced via hypoxia exposure and administration of PA (5 mg/kg per day) in male Sprague-Dawley rats. Hemodynamic parameters were measured using a right ventricular floating catheter and pulmonary vascular morphometry was measured by hematoxylin-eosin (HE), α-SMA and Masson staining. MTT assays and EdU staining were used to detect cell proliferation, and apoptosis was analyzed by TUNEL staining. Western blotting and immunohistochemistry were used to detect the expression of proteins related to the Nrf2-Keap1-ARE pathway. RESULTS: PA significantly alleviated hypoxic PH and reversed right ventricular hypertrophy and pulmonary vascular remodeling. In addition, PA effectively inhibited proliferation and promoted apoptosis in hypoxia-induced pulmonary artery smooth muscle cells (PASMCs). Moreover, PA pretreatment inhibited the expression of peroxy-related factor (MDA) and promoted the expression of antioxidant-related factors (GSH-PX and SOD). Furthermore, hypoxia inhibited the Nrf2-Keap1-ARE signaling pathway, while PA effectively activated this pathway. Most importantly, addition of the Nrf2 inhibitor ML385 reversed the inhibitory effects of PA on ROS generation, proliferation, and apoptosis tolerance in hypoxia-induced PASMCs. CONCLUSION: Our study suggests that PA may reverse PH by regulating the Nrf2-Keap1-ARE signaling pathway.


Subject(s)
Antioxidant Response Elements/drug effects , Hypertension, Pulmonary/drug therapy , Kelch-Like ECH-Associated Protein 1/drug effects , NF-E2-Related Factor 2/drug effects , Phospholipase A2 Inhibitors/pharmacology , Triterpenes/pharmacology , Animals , Disease Models, Animal , Male , Phospholipase A2 Inhibitors/administration & dosage , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Triterpenes/administration & dosage
11.
In Vitro Cell Dev Biol Anim ; 57(7): 685-694, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34518994

ABSTRACT

The destruction of biological activity such as senescence and apoptosis caused by oxidative stress could play a pivotal role in the poor therapeutic efficiency of bone marrow mesenchymal stem cells (BMSCs) transplantation. Mitoquinone (MitoQ) has a highly effective mitochondrial antioxidant effect, and has been widely used in many oxidative damage models. This study aimed to investigate the protective effect of MitoQ on the oxidative stress-mediated senescence of canine BMSCs and the underlying mechanism. The senescence of BMSCs was determined by senescence-associated ß-galactosidase staining and quantitative real-time PCR. The expression of p-Nrf2 protein was detected by Western blotting. The results demonstrated that, as BMSCs were expanded in vitro, the senescent phenotype appeared. And the senescence of BMSCs may be caused by oxidative stress, manifested by increasing the level of ROS and decreasing the activity of antioxidant enzymes. Treatment of MitoQ down-regulated the mRNA levels of senescence-related and apoptosis-related genes, but up-regulated the mRNA levels of proliferation-related genes. Meanwhile, ROS generation and senescent activity were reduced in MitoQ-treated BMSCs. Further mechanism studies showed that MitoQ obviously promoted Nrf2 phosphorylation, and also facilitated the translocation of Nrf2 into the nucleus. Moreover, treatment of MitoQ increased the mRNA levels of downstream antioxidant genes and enhanced the activities of superoxide dismutase, catalase, and glutathione peroxidase. Thus, our study revealed that MitoQ, via the Nrf2/ARE signaling pathway, exerts an antioxidant effect as well as potentially delays OS-mediated senescence during BMSCs that were expanded in vitro, which may serve as a novel strategy to optimize the clinical application of BMSCs.


Subject(s)
Antioxidant Response Elements/physiology , Mesenchymal Stem Cells/drug effects , NF-E2-Related Factor 2/metabolism , Organophosphorus Compounds/pharmacology , Oxidative Stress/drug effects , Ubiquinone/analogs & derivatives , Animals , Antigens, CD/metabolism , Antioxidant Response Elements/drug effects , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Dogs , Enzymes/metabolism , Gene Expression Regulation/drug effects , Mesenchymal Stem Cells/metabolism , Oxidative Stress/physiology , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Ubiquinone/pharmacology
12.
Cells ; 10(8)2021 08 10.
Article in English | MEDLINE | ID: mdl-34440821

ABSTRACT

Systemic sclerosis (SSc) is an autoimmune connective tissue disease that leads to skin fibrosis. Altered metabolism has recently been described in autoimmune diseases and SSc. Itaconate is a product of the Krebs cycle intermediate cis-aconitate and is an immunomodulator. This work examines the role of the cell-permeable derivative of itaconate, 4-octyl itaconate (4-OI), in SSc. SSc and healthy dermal fibroblasts were exposed to 4-OI. The levels of collagen Nrf2-target genes and pro-inflammatory cytokines interleukin 6 (IL-6) and monocyte chemotactic protein 1 (MCP-1) were determined. Levels of reactive oxygen species (ROS) as well as the gene expression of collagen and Cellular Communication Network Factor 2 (CCN2) were measured after transforming growth factor beta 1 (TGF-ß1) stimulation in the presence or absence of 4-OI. Wild-type or Nrf2-knockout (Nrf2-KO) mouse embryonic fibroblasts (MEFs) were also treated with 4-OI to determine the role of Nrf2 in 4-OI-mediated effects. 4-OI reduced the levels of collagen in SSc dermal fibroblasts. Incubation with 4-OI led to activation of Nrf2 and its target genes heme oxygenase 1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (NQO1). 4-OI activated antioxidant response element (ARE)-dependent gene expression, reduced inflammatory cytokine release and reduced TGF-ß1-induced collagen and ROS production in dermal fibroblasts. The effects of 4-OI are dependent on Nrf2. The cell-permeable derivative of itaconate 4-OI is anti-fibrotic through upregulation of Nrf2 and could be a potential therapeutic option in an intractable disease.


Subject(s)
Down-Regulation/drug effects , Scleroderma, Systemic/pathology , Succinates/pharmacology , Up-Regulation/drug effects , Animals , Antioxidant Response Elements/drug effects , Antioxidant Response Elements/genetics , Collagen/metabolism , Connective Tissue Growth Factor/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Interleukin-6/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species/metabolism , Scleroderma, Systemic/metabolism , Transforming Growth Factor beta1/pharmacology
13.
Bioorg Chem ; 115: 105177, 2021 10.
Article in English | MEDLINE | ID: mdl-34303035

ABSTRACT

Ferroptosis is a new form of cell death, and inhibition of ferroptosis is a promising strategy to treat neurological diseases. In this work, sixteen compounds were isolated from Ajuga nipponensis and assayed for anti-ferroptosis activity in HT22 mouse hippocampal neuronal cells. Ajudecunoid C (1, ADC), a new neoclerodane diterpenoid, showed significant inhibitory activity against erastin and RSL3-induced ferroptosis with EC50 values of 4.1 ± 1.0 and 3.6 ± 0.3 µM, respectively. Experimental results demonstrated that ADC effectively prevented ferroptosis through scavenging free radical and activating NRF2-antioxidant response elements (AREs) pathway. This study reveals that ADC, as a new ferroptosis inhibitor, is a promising lead compound for the development of drugs against ferroptosis-related neurological diseases.


Subject(s)
Ajuga/chemistry , Ferroptosis/drug effects , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Animals , Antioxidant Response Elements/drug effects , Cell Line , Mice , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/isolation & purification , Signal Transduction/drug effects
14.
Eur J Med Chem ; 224: 113686, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34303079

ABSTRACT

Pathway activating mutations of the transcription factor NRF2 and its negative regulator KEAP1 are strongly correlative with poor clinical outcome with pemetrexed/carbo(cis)platin/pembrolizumab (PCP) chemo-immunotherapy in lung cancer. Despite the strong genetic support and therapeutic potential for a NRF2 transcriptional inhibitor, currently there are no known direct inhibitors of the NRF2 protein or its complexes with MAF and/or DNA. Herein we describe the design of a novel and high-confidence homology model to guide a medicinal chemistry effort that resulted in the discovery of a series of peptides that demonstrate high affinity, selective binding to the Antioxidant Response Element (ARE) DNA and thereby displace NRF2-MAFG from its promoter, which is an inhibitory mechanism that to our knowledge has not been previously described. In addition to their activity in electrophoretic mobility shift (EMSA) and TR-FRET-based assays, we show significant dose-dependent ternary complex disruption of NRF2-MAFG binding to DNA by SPR, as well as cellular target engagement by thermal destabilization of HiBiT-tagged NRF2 in the NCI-H1944 NSCLC cell line upon digitonin permeabilization, and SAR studies leading to improved cellular stability. We report the characterization and unique profile of lead peptide 18, which we believe to be a useful in vitro tool to probe NRF2 biology in cancer cell lines and models, while also serving as an excellent starting point for additional in vivo optimization toward inhibition of NRF2-driven transcription to address a significant unmet medical need in non-small cell lung cancer (NSCLC).


Subject(s)
DNA/chemistry , MafG Transcription Factor/antagonists & inhibitors , NF-E2-Related Factor 2/antagonists & inhibitors , Peptides/chemistry , Antioxidant Response Elements/drug effects , DNA/metabolism , Drug Design , Drug Stability , Electrophoretic Mobility Shift Assay , Half-Life , HeLa Cells , Humans , MafG Transcription Factor/metabolism , NF-E2-Related Factor 2/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Peptides/metabolism , Peptides/pharmacology , Peptides/therapeutic use , Structure-Activity Relationship
15.
Neurochem Res ; 46(9): 2439-2450, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34170454

ABSTRACT

Extensive studies have shown that oxidative stress is a crucial pathogenic factor in Alzheimer's disease (AD). Nuclear factor E2-related factor 2 (Nrf2) is a master cytoprotective regulator against oxidative stress, and thus represents an attractive therapeutic target in AD. The goal of our study is to investigate the contribution of Nrf2 in Rhynchophylline (Rhy)-induced neuroprotection in AD. The data showed that intraperitoneal administration of Rhy (10 or 20 mg/kg) could ameliorate Aß1-42-induced cognitive impairment, evidenced by performance improvement in memory tests. The result of Antioxidant response element (ARE)-luciferase activity assay indicated that Rhy treatment improved ARE promoter activity. The results of reactive oxygen species (ROS), malondialdehyde (MDA) and glutathione (GSH) assessment in the frontal cortex and hippocampus showed that Rhy treatment could attenuate Aß1-42-induced oxidative stress to some extent, evidenced by reversion of these cytokines compared to Aß1-42 + Veh group. Rhy treatment also restored expression of Nrf2 and its downstream protein heme oxygenase-1 (HO-1), NAD(P)H/quinone oxidoreductase 1 (NOQ1), and recombinant glutamate cysteine ligase, modifier subunit (GCLM) in the frontal cortex and hippocampus of Aß1-42-treated mice. In addition, to investigate whether activation of Nrf2-mediated pathway is responsible for the neuroprotection of Rhy, Nrf2 siRNA was used in human neuroblastoma cells (SH-SY5Y). Interestingly, the results showed that the protective effects of Rhy, including anti-oxidative, anti-apoptosis and elevation of Nrf2 and its downstream proteins, were abolished in Nrf2 siRNA-transfected cells. These findings indicate that Rhynchophylline is protective against Aß1-42-induced neurotoxicity via Nrf2-ARE activation, and suggest that Rhy may serve as a potential candidate and promising Nrf2 activator for management of AD.


Subject(s)
Alzheimer Disease/drug therapy , Memory/drug effects , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Oxindoles/therapeutic use , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Amyloid beta-Peptides , Animals , Antioxidant Response Elements/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Male , Mice, Inbred ICR , Peptide Fragments
16.
Biomed Pharmacother ; 141: 111834, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34153850

ABSTRACT

Fuchs endothelial corneal dystrophy is one of the most common indications for corneal transplantation, and impaired anti-oxidative function is observed in corneal endothelial cells (CECs). Curcumin is well-known for its anti-oxidative property; but, no study has examined the effect of curcumin on anti-oxidative therapeutic roles in corneal endothelial disease. In our experiments, oxidative stress 0.25 mM tert-butyl hydroperoxide for 2 h was induced in immortalized human CECs pretreated with curcumin. Cell behavior and viability, reactive oxygen species production, and the protein expression of the kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2(Nrf2)/antioxidant response element (ARE) pathway were examined; the Keap1/Nrf2/ARE pathway is crucial anti-oxidative pathway of curcumin. The results showed that pretreatment with 12.5 µM curcumin significantly reduced the ROS production and improved the survival of CECs under oxidative stress. In addition, curcumin pretreatment significantly increased the expression of nuclear Nrf2, and the productions of superoxide dismutase 1 and heme oxygenase-1, which were the target anti-oxidative enzymes of the Keap1/Nrf2/ARE pathway. Our findings showed that curcumin enhanced the growth and differentiation of CECs under oxidative stress. The activation of Keap1/Nrf2/ARE pathway by curcumin was crucial for CECs to improve their anti-oxidative capacity.


Subject(s)
Antioxidant Response Elements/drug effects , Antioxidants/pharmacology , Corneal Endothelial Cell Loss/prevention & control , Curcumin/pharmacology , Endothelial Cells/drug effects , Kelch-Like ECH-Associated Protein 1/agonists , NF-E2-Related Factor 2/agonists , Vesicular Transport Proteins/agonists , Cell Line/drug effects , Cornea/cytology , Cornea/drug effects , Humans , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
17.
Int J Mol Med ; 47(4)2021 04.
Article in English | MEDLINE | ID: mdl-33655321

ABSTRACT

Fine particulate matter (PM2.5) with an average aerodynamic diameter of <2.5 µm can cause severe lung injury. Oxidative stress and inflammation are considered the main outcomes of PM2.5 exposure. Curcumin is a well­known antioxidant; however, its effect on PM2.5­induced oxidative injury in airway epithelial cells remains unclear. In the present study, it was demonstrated that pre­treatment with curcumin significantly reduced the PM2.5­induced apoptosis of BEAS­2B human bronchial epithelial cells by decreasing the level of intercellular reactive oxygen species. Western blot analysis revealed that curcumin increased the expression of nuclear factor erythroid 2­related factor 2 (NRF2) and regulated the transcription of downstream genes, particularly those encoding antioxidant enzymes. Moreover, curcumin reduced the PM2.5­induced expression and production of inflammatory factors, and induced the expression of the anti­inflammatory factors, interleukin (IL)­5 and IL­13. Taken together, the present study demonstrates that curcumin protects BEAS­2B cells against PM2.5­induced oxidative damage and inflammation, and prevents cell apoptosis by increasing the activation of NRF2­related pathways. It is thus suggested that curcumin may be a potential compound for use in the prevention of PM2.5­induced tissue injury.


Subject(s)
Antioxidants/pharmacology , Curcumin/pharmacology , Lung Injury/prevention & control , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Particulate Matter/toxicity , Antioxidant Response Elements/drug effects , Apoptosis/drug effects , Environmental Exposure/adverse effects , Humans , Inflammation/prevention & control , Interleukin-13/metabolism , Interleukin-5/metabolism , Lung Injury/drug therapy , Reactive Oxygen Species/metabolism
18.
Hum Exp Toxicol ; 40(7): 1194-1207, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33530773

ABSTRACT

Methotrexate (MTX) is frequently used drug in treatment of cancer and autoimmune diseases. Unfortunately, MTX has many side effects including the hepato-renal toxicity. In this study, we hypothesized that Luteolin (Lut) exhibits protective effect against the MTX-induced hepato-renal toxicity. In order to investigate our hypothesis, the experiment was designed to examine the effect of exposure of male rats to MTX (20 mg/kg, i.p., at day 9) alone or together with Lut (50 mg/kg, oral for 14 days) compared to the control rats (received saline). The findings demonstrated that MTX treatment induced significant increases in the liver and kidney functions markers in serum samples including Aspartate transaminase (AST), Alanine transaminase (ALT), creatinine, urea and uric acid. MTX also mediated an oxidative stress expressed by elevated malondialdehyde (MDA) level and decreased level of reduced glutathione (GSH), antioxidant enzyme activities, and downregulation of the Nrf2 gene expression as an antioxidant trigger. Moreover, the inflammatory markers (NF-κB, TNF-α, and IL-1ß) were significantly elevated upon MTX treatment. In addition, MTX showed an apoptotic response mediated by elevating the pro-apoptotic (Bax) and lowering the anti-apoptotic (Bcl-2) proteins. All of these changes were confirmed by the observed alterations in the histopathological examination of the hepatic and renal tissues. Lut exposure significantly reversed all the MTX-induced changes in the measured parameters suggesting its potential protective role against the MTX-induced toxicity. Finally, our findings concluded the antioxidative, anti-inflammatory and anti-apoptotic effects of Lut as a mechanism of its protective role against the MTX-induced hepato-renal toxicity in rats.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chemical and Drug Induced Liver Injury/drug therapy , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Luteolin/pharmacology , Luteolin/therapeutic use , Methotrexate/toxicity , Animals , Antioxidant Response Elements/drug effects , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/physiopathology , Inflammation/chemically induced , Kidney/drug effects , Male , Oxidative Stress/drug effects , Protective Agents/pharmacology , Rats
19.
Sci Rep ; 11(1): 4773, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637843

ABSTRACT

Cytoprotection involving the nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathway is an important preventive strategy for normal cells against carcinogenesis. In our previous study, the chemopreventive potential of (E)-N-(2-(3, 5-Dimethoxystyryl) phenyl) furan-2-carboxamide (BK3C231) has been elucidated through its cytoprotective effects against DNA and mitochondrial damages in the human colon fibroblast CCD-18Co cell model. Therefore this study aimed to investigate the molecular mechanisms underlying BK3C231-induced cytoprotection and the involvement of the Nrf2/ARE pathway. The cells were pretreated with BK3C231 before exposure to carcinogen 4-nitroquinoline N-oxide (4NQO). BK3C231 increased the protein expression and activity of cytoprotective enzymes namely NAD(P)H:quinone oxidoreductase 1 (NQO1), glutathione S-transferase (GST) and heme oxygenase-1 (HO-1), as well as restoring the expression of glutamate-cysteine ligase catalytic subunit (GCLC) back to the basal level. Furthermore, dissociation of Nrf2 from its inhibitory protein, Keap1, and ARE promoter activity were upregulated in cells pretreated with BK3C231. Taken together, our findings suggest that BK3C231 exerts cytoprotection by activating the Nrf2 signaling pathway which leads to ARE-mediated upregulation of cytoprotective proteins. This study provides new mechanistic insights into BK3C231 chemopreventive activities and highlights the importance of stilbene derivatives upon development as a potential chemopreventive agent.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antioxidant Response Elements/drug effects , Cytoprotection , Fibroblasts/drug effects , Furans/pharmacology , NF-E2-Related Factor 2/metabolism , Cell Line , Colon/cytology , Colon/drug effects , Colon/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Signal Transduction/drug effects
20.
Bioengineered ; 12(1): 402-413, 2021 12.
Article in English | MEDLINE | ID: mdl-33356808

ABSTRACT

In this study, we examined the antitumor effects of Puerarin (PEU) on androgen-independent (DU145 and PC-3) and androgen-dependent (LNCaP) prostate cancer cells, and explored its potential mechanisms. Supplement with PEU (2.5 µM, 5 µM, and 10 µM) exhibited a marked inhibitory effect against the growth of DU145 and PC-3 cells, especially beyond 24 h, whereas there is only slight growth inhibitory effect on LNCaP cells at the high concentration of 10 µM at 72 h. This loss of cell viability in DU145 and PC-3 cells by PEU was mediated by the induction of apoptosis via up-regulation of Bax and cleaved-caspase-3, but downregulation of Bcl-2. Moreover, more intracellular ROS and LDH production were observed in DU145 and PC-3 cells upon PEU treatment. Meanwhile, the amount of pro-inflammatory cytokines (IL-1ß and IL-6) was increased, but the content of anti-inflammatory cytokines IL-10 was attenuated. Additionally, PEU pretreatment resulted in an increase of Keap1 protein expression, and a decline of Nrf2, HO-1 and NQO1 protein expression in DU145 and PC3 cells. The present findings indicated that PEU exerted its antitumor activities toward androgen-independent prostate cancer cells via inactivation of Keap1/NrF2/ARE signaling pathway.


Subject(s)
Apoptosis/drug effects , Isoflavones/pharmacology , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Prostatic Neoplasms/metabolism , Antioxidant Response Elements/drug effects , Cell Line, Tumor , Humans , Male , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...