1
|
Yan S, Li M, Jiang Q, Li M, Hu M, Shi X, Liang P, Yin M, Gao X, Shen J, Zhang L. Self-assembled co-delivery nanoplatform for increasing the broad-spectrum susceptibility of fall armyworm toward insecticides. J Adv Res 2025; 67:93-104. [PMID: 38286302 DOI: 10.1016/j.jare.2024.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 01/31/2024] Open
Abstract
INTRODUCTION Unscientific application of insecticides has led to severe resistance of pests to almost all classes of insecticides. Enhanced detoxification is the most common mechanism for this kind of resistance. OBJECT Fall armyworm (FAW) has developed insecticide resistance, which is often linked to the overexpression of detoxification genes. Herein, a multicomponent nano-pesticide is designed to increase its broad-spectrum susceptibility toward insecticides. METHOD Regulatory function of nuclear factor erythroid 2-related factor 2 (Nrf2) in detoxification was confirmed using transcriptome sequencing, quantitative real-time PCR and enzyme activity measurement. A star polycation (SPc) was adopted to construct the pesticide/SPc/complex, whose self-assembly mechanism and characterization were examined using isothermal titration calorimetry, dynamic light scattering and transmission electron microscope. The delivery efficiency of SPc-loaded dsRNA was examined in vitro and in vivo using fluorescent tracer technique. A multicomponent nano-pesticide was created through the integration of bacterial expression system and nano-delivery system, and its bioactivity was tested in laboratory and field. RESULTS We confirmed the crucial role of Nrf2 in regulating the detoxification in FAW, and silencing Nrf2 could decrease detoxification gene expression and increase insecticide susceptibility. We then applied the SPc to self-assemble a nanoplatform for delivering Nrf2 double-stranded RNA (dsRNA) and pesticide simultaneously. Nano-sized pesticide/SPc/dsRNA complex exhibited high delivery efficiency in vitro and in vivo. Excitingly, the insecticidal activities of pesticide/SPc/dsNrf2 complexes were remarkably improved with the normalized synergistic ratios of 5.43-6.25 for chlorantraniliprole, 4.45-15.00 for emamectin benzoate, and 6.75-15.00 for spinetoram. Finally, we developed a multicomponent nano-pesticide (pesticide/SPc/dsNrf2 complex) using a bacterial expression system and nano-delivery system. This approach exhibited excellent leaf protection and pest control efficacy. CONCLUSION The integration between the pesticide nanometerization and insecticide susceptibility improvement offers a promising strategy to increase insecticidal activity. Our study provides a revolutionary and universal strategy to increase insecticidal activity and decease application doses.
Collapse
Affiliation(s)
- Shuo Yan
- College of Plant Protection, China Agricultural University, 100193 Beijing, PR China.
| | - Mingjian Li
- College of Plant Protection, China Agricultural University, 100193 Beijing, PR China
| | - Qinhong Jiang
- College of Plant Protection, China Agricultural University, 100193 Beijing, PR China
| | - Mingshan Li
- College of Plant Protection, China Agricultural University, 100193 Beijing, PR China
| | - Mengfan Hu
- College of Plant Protection, China Agricultural University, 100193 Beijing, PR China
| | - Xueyan Shi
- College of Plant Protection, China Agricultural University, 100193 Beijing, PR China
| | - Pei Liang
- College of Plant Protection, China Agricultural University, 100193 Beijing, PR China
| | - Meizhen Yin
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029 Beijing, PR China
| | - Xiwu Gao
- College of Plant Protection, China Agricultural University, 100193 Beijing, PR China
| | - Jie Shen
- College of Plant Protection, China Agricultural University, 100193 Beijing, PR China.
| | - Lei Zhang
- College of Plant Protection, China Agricultural University, 100193 Beijing, PR China.
| |
Collapse
|
2
|
Shi Z, Lei JT, Elizarraras JM, Zhang B. Mapping the functional network of human cancer through machine learning and pan-cancer proteogenomics. NATURE CANCER 2024:10.1038/s43018-024-00869-z. [PMID: 39663389 DOI: 10.1038/s43018-024-00869-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 10/25/2024] [Indexed: 12/13/2024]
Abstract
Large-scale omics profiling has uncovered a vast array of somatic mutations and cancer-associated proteins, posing substantial challenges for their functional interpretation. Here we present a network-based approach centered on FunMap, a pan-cancer functional network constructed using supervised machine learning on extensive proteomics and RNA sequencing data from 1,194 individuals spanning 11 cancer types. Comprising 10,525 protein-coding genes, FunMap connects functionally associated genes with unprecedented precision, surpassing traditional protein-protein interaction maps. Network analysis identifies functional protein modules, reveals a hierarchical structure linked to cancer hallmarks and clinical phenotypes, provides deeper insights into established cancer drivers and predicts functions for understudied cancer-associated proteins. Additionally, applying graph-neural-network-based deep learning to FunMap uncovers drivers with low mutation frequency. This study establishes FunMap as a powerful and unbiased tool for interpreting somatic mutations and understudied proteins, with broad implications for advancing cancer biology and informing therapeutic strategies.
Collapse
Affiliation(s)
- Zhiao Shi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - John M Elizarraras
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
3
|
Messingschlager M, Bartel-Steinbach M, Mackowiak SD, Denkena J, Bieg M, Klös M, Seegebarth A, Straff W, Süring K, Ishaque N, Eils R, Lehmann I, Lermen D, Trump S. Genome-wide DNA methylation sequencing identifies epigenetic perturbations in the upper airways under long-term exposure to moderate levels of ambient air pollution. ENVIRONMENTAL RESEARCH 2023; 233:116413. [PMID: 37343754 DOI: 10.1016/j.envres.2023.116413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/30/2023] [Accepted: 06/11/2023] [Indexed: 06/23/2023]
Abstract
While the link between exposure to high levels of ambient particulate matter (PM) and increased incidences of respiratory and cardiovascular diseases is widely recognized, recent epidemiological studies have shown that low PM concentrations are equally associated with adverse health effects. As DNA methylation is one of the main mechanisms by which cells regulate and stabilize gene expression, changes in the methylome could constitute early indicators of dysregulated signaling pathways. So far, little is known about PM-associated DNA methylation changes in the upper airways, the first point of contact between airborne pollutants and the human body. Here, we focused on cells of the upper respiratory tract and assessed their genome-wide DNA methylation pattern to explore exposure-associated early regulatory changes. Using a mobile epidemiological laboratory, nasal lavage samples were collected from a cohort of 60 adults that lived in districts with records of low (Simmerath) or moderate (Stuttgart) PM10 levels in Germany. PM10 concentrations were verified by particle measurements on the days of the sample collection and genome-wide DNA methylation was determined by enzymatic methyl sequencing at single-base resolution. We identified 231 differentially methylated regions (DMRs) between moderately and lowly PM10 exposed individuals. A high proportion of DMRs overlapped with regulatory elements, and DMR target genes were involved in pathways regulating cellular redox homeostasis and immune response. In addition, we found distinct changes in DNA methylation of the HOXA gene cluster whose methylation levels have previously been linked to air pollution exposure but also to carcinogenesis in several instances. The findings of this study suggest that regulatory changes in upper airway cells occur at PM10 levels below current European thresholds, some of which may be involved in the development of air pollution-related diseases.
Collapse
Affiliation(s)
- Marey Messingschlager
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Charitéplatz 1, 10117, Berlin, Germany; Freie Universität Berlin, Institute for Biology, Königin-Luise-Strasse 12-16, 14195, Berlin, Germany
| | - Martina Bartel-Steinbach
- Fraunhofer Institute for Biomedical Engineering IBMT, Josef-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany
| | - Sebastian D Mackowiak
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Johanna Denkena
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Matthias Bieg
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Matthias Klös
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Charitéplatz 1, 10117, Berlin, Germany
| | - Anke Seegebarth
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Charitéplatz 1, 10117, Berlin, Germany
| | - Wolfgang Straff
- Environmental Medicine and Health Effects Assessment, German Environment Agency, Corrensplatz 1, 14195, Berlin, Germany
| | - Katrin Süring
- Environmental Medicine and Health Effects Assessment, German Environment Agency, Corrensplatz 1, 14195, Berlin, Germany
| | - Naveed Ishaque
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Roland Eils
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Charitéplatz 1, 10117, Berlin, Germany; German Center for Lung Research (DZL), Germany; Health Data Science Unit, Heidelberg University Hospital and BioQuant, University of Heidelberg, Germany; Freie Universität Berlin, Department of Mathematics and Computer Science, Arnimallee 14, 14195, Berlin, Germany
| | - Irina Lehmann
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Charitéplatz 1, 10117, Berlin, Germany; German Center for Lung Research (DZL), Germany.
| | - Dominik Lermen
- Fraunhofer Institute for Biomedical Engineering IBMT, Josef-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany
| | - Saskia Trump
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
4
|
Li ET, Wu HJ, Wang ZM, Li KB, Zhang S, Cao YZ, Yin J. PI3K/Akt/CncC signaling pathway mediates the response to EPN-Bt infection in Holotrichia parallela larvae. PEST MANAGEMENT SCIENCE 2023; 79:1660-1673. [PMID: 36565065 DOI: 10.1002/ps.7337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 11/28/2022] [Accepted: 12/24/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Combining the entomopathogenic nematode (EPN), Heterorhabditis beicherriana LF strain, and Bacillus thuringiensis (Bt) HBF-18 strain is a practical strategy to manage the larvae of Holotrichia parallela Motschulsky (white grubs). However, the mechanisms underlying the larval defense response to this combined biocontrol strategy are unknown. RESULTS The activities of some antioxidant enzymes (SOD, POD, CAT) and some detoxifying enzymes (AChE, P-450, CarE, GST) in grubs showed an activation-inhibition trend throughout the EPN-Bt exposure time course. Eight potentially key antioxidant and detoxifying enzyme genes in response to EPN-Bt infection were identified from the midgut of grubs through RNA sequencing. After silencing CAT, CarE18, and GSTs1, the enzyme activities were significantly decreased by 30.29%, 68.80%, and 34.63%, respectively. Meanwhile, the mortality of grubs was increased by 18.40%, 46.30%, and 42.59% after exposure to EPN-Bt for 1 day. Interestingly, the PI3K/Akt signaling pathway was significantly enriched in KEGG enrichment analysis, and the expression levels of phosphatidylinositol 3-kinase (PI3K), protein kinase B (Akt), cap 'n' collar isoform-C (CncC), kelch-like ECH-associated protein 1 (Keap1), and CarE18 were all up-regulated when exposed to EPN-Bt for 1 day. Furthermore, RNAi-mediated PI3K silencing showed a similar down-regulated trend between PI3K/Akt/CncC and CarE18. Moreover, silencing PI3K rendered grubs more susceptible to EPN-Bt and accelerated symbiotic bacteria multiplication in grubs. CONCLUSION These results suggest that the PI3K/Akt/CncC pathway mediates the expression of CarE18 and participates in the defense response of H. parallela larvae against EPN-Bt infection. Our data provide valuable insights into the design of appropriate management strategies for this well-known agricultural pest. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Er-Tao Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Han-Jia Wu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhi-Min Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ke-Bin Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shuai Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ya-Zhong Cao
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiao Yin
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
5
|
Collins JM, Lu R, Wang X, Zhu HJ, Wang D. Transcriptional Regulation of Carboxylesterase 1 in Human Liver: Role of the Nuclear Receptor Subfamily 1 Group H Member 3 and Its Splice Isoforms. Drug Metab Dispos 2022; 50:43-48. [PMID: 34697082 PMCID: PMC8969197 DOI: 10.1124/dmd.121.000649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023] Open
Abstract
Carboxylesterase 1 (CES1) is the predominant carboxylesterase in the human liver, involved in metabolism of both xenobiotics and endogenous substrates. Genetic or epigenetic factors that alter CES1 activity or expression are associated with changes in drug response, lipid, and glucose homeostasis. However, the transcriptional regulation of CES1 in the human liver remains uncertain. By applying both the random forest and Sobol's Sensitivity Indices (SSI) to analyze existing liver RNA expression microarray data (GSE9588), we identified nuclear receptor subfamily 1 group H member 3 (NR1H3) liver X receptor (LXR)α as a key factor regulating constitutive CES1 expression. This model prediction was validated using small interfering RNA (siRNA) knockdown and CRISPR-mediated transcriptional activation of NR1H3 in Huh7 and HepG2 cells. We found that NR1H3's activation of CES1 is splice isoform-specific, namely that increased expression of the NR1H3-211 isoform increased CES1 expression whereas NR1H3-201 did not. Also, in human liver samples, expression of NR1H3-211 and CES1 are correlated, whereas NR1H3-201 and CES1 are not. This trend also occurs during differentiation of induced pluripotent stem cells (iPSCs) to hepatocytes, where only expression of the NR1H3-211 isoform parallels expression of CES1 Moreover, we found that treatment with the NR1H3 agonist T0901317 in HepG2 cells had no effect on CES1 expression. Overall, our results demonstrate a key role of NR1H3 in maintaining the constitutive expression of CES1 in the human liver. Furthermore, our results support that the effect of NR1H3 is splice isoform-specific and appears to be ligand independent. SIGNIFICANCE STATEMENT: Despite the central role of carboxylesterase 1 (CES1) in metabolism of numerous medications, little is known about its transcriptional regulation. This study identifies nuclear receptor subfamily 1 group H member 3 as a key regulator of constitutive CES1 expression and therefore is a potential target for future studies to understand interperson variabilities in CES1 activity and drug metabolism.
Collapse
Affiliation(s)
- Joseph M Collins
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.C., D.W.); The Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California (R.L.); Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, (X.W.); and Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan (H.-J.Z.)
| | - Rong Lu
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.C., D.W.); The Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California (R.L.); Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, (X.W.); and Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan (H.-J.Z.)
| | - Xinwen Wang
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.C., D.W.); The Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California (R.L.); Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, (X.W.); and Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan (H.-J.Z.)
| | - Hao-Jie Zhu
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.C., D.W.); The Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California (R.L.); Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, (X.W.); and Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan (H.-J.Z.)
| | - Danxin Wang
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.C., D.W.); The Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California (R.L.); Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, (X.W.); and Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan (H.-J.Z.)
| |
Collapse
|
6
|
Yan D, Ra OH, Yan B. The nucleoside antiviral prodrug remdesivir in treating COVID-19 and beyond with interspecies significance. ANIMAL DISEASES 2021; 1:15. [PMID: 34778881 PMCID: PMC8422062 DOI: 10.1186/s44149-021-00017-5] [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: 06/07/2021] [Accepted: 07/25/2021] [Indexed: 01/18/2023] Open
Abstract
Infectious pandemics result in hundreds and millions of deaths, notable examples of the Spanish Flu, the Black Death and smallpox. The current pandemic, caused by SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), is unprecedented even in the historical term of pandemics. The unprecedentedness is featured by multiple surges, rapid identification of therapeutic options and accelerated development of vaccines. Remdesivir, originally developed for Ebola viral disease, is the first treatment of COVID-19 (Coronavirus disease 2019) approved by the United States Food and Drug Administration. As demonstrated by in vitro and preclinical studies, this therapeutic agent is highly potent with a broad spectrum activity against viruses from as many as seven families even cross species. However, randomized controlled trials have failed to confirm the efficacy and safety. Remdesivir improves some clinical signs but not critical parameters such as mortality. This antiviral agent is an ester/phosphorylation prodrug and excessive hydrolysis which increases cellular toxicity. Remdesivir is given intravenously, leading to concentration spikes and likely increasing the potential of hydrolysis-based toxicity. This review has proposed a conceptual framework for improving its efficacy and minimizing toxicity not only for the COVID-19 pandemic but also for future ones caused by remdesivir-sensitive viruses.
Collapse
Affiliation(s)
- Daisy Yan
- Sidney Kimmel Medical College, Thomas Jefferson University, 1025 Walnut St, Philadelphia, PA 19107 USA
| | - One Hyuk Ra
- Department of Anesthesiology, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115 USA
| | - Bingfang Yan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229 USA
| |
Collapse
|
7
|
Crinelli R, Zara C, Galluzzi L, Buffi G, Ceccarini C, Smietana M, Mari M, Magnani M, Fraternale A. Activation of NRF2 and ATF4 Signaling by the Pro-Glutathione Molecule I-152, a Co-Drug of N-Acetyl-Cysteine and Cysteamine. Antioxidants (Basel) 2021; 10:antiox10020175. [PMID: 33530504 PMCID: PMC7911873 DOI: 10.3390/antiox10020175] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 11/19/2022] Open
Abstract
I-152 combines two pro-glutathione (GSH) molecules, namely N-acetyl-cysteine (NAC) and cysteamine (MEA), to improve their potency. The co-drug efficiently increases/replenishes GSH levels in vitro and in vivo; little is known about its mechanism of action. Here we demonstrate that I-152 not only supplies GSH precursors, but also activates the antioxidant kelch-like ECH-associated protein 1/nuclear factor E2-related factor 2 (KEAP1/NRF2) pathway. The mechanism involves disulfide bond formation between KEAP1 cysteine residues, NRF2 stabilization and enhanced expression of the γ-glutamil cysteine ligase regulatory subunit. Accordingly, a significant increase in GSH levels, not reproduced by treatment with NAC or MEA alone, was found. Compared to its parent compounds, I-152 delivered NAC more efficiently within cells and displayed increased reactivity to KEAP1 compared to MEA. While at all the concentrations tested, I-152 activated the NRF2 pathway; high doses caused co-activation of activating transcription factor 4 (ATF4) and ATF4-dependent gene expression through a mechanism involving Atf4 transcriptional activation rather than preferential mRNA translation. In this case, GSH levels tended to decrease over time, and a reduction in cell proliferation/survival was observed, highlighting that there is a concentration threshold which determines the transition from advantageous to adverse effects. This body of evidence provides a molecular framework for the pro-GSH activity and dose-dependent effects of I-152 and shows how synergism and cross reactivity between different thiol species could be exploited to develop more potent drugs.
Collapse
Affiliation(s)
- Rita Crinelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Carolina Zara
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Luca Galluzzi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Gloria Buffi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Chiara Ceccarini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Michael Smietana
- Institut des Biomolécules Max Mousseron, Université de Montpellier UMR 5247 CNRS, ENSCM, 34095 Montpellier, France
| | - Michele Mari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Alessandra Fraternale
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| |
Collapse
|
8
|
Qian Y, Markowitz JS. Natural Products as Modulators of CES1 Activity. Drug Metab Dispos 2020; 48:993-1007. [PMID: 32591414 DOI: 10.1124/dmd.120.000065] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/12/2020] [Indexed: 12/30/2022] Open
Abstract
Carboxylesterase (CES) 1 is the predominant esterase expressed in the human liver and is capable of catalyzing the hydrolysis of a wide range of therapeutic agents, toxins, and endogenous compounds. Accumulating studies have demonstrated associations between the expression and activity of CES1 and the pharmacokinetics and/or pharmacodynamics of CES1 substrate medications (e.g., methylphenidate, clopidogrel, oseltamivir). Therefore, any perturbation of CES1 by coingested xenobiotics could potentially compromise treatment. Natural products are known to alter drug disposition by modulating cytochrome P450 and UDP-glucuronosyltransferase enzymes, but this issue is less thoroughly explored with CES1. We report the results of a systematic literature search and discuss natural products as potential modulators of CES1 activity. The majority of research reports reviewed were in vitro investigations that require further confirmation through clinical study. Cannabis products (Δ 9-tetrahydrocannabinol, cannabidiol, cannabinol); supplements from various plant sources containing naringenin, quercetin, luteolin, oleanolic acid, and asiatic acid; and certain traditional medicines (danshen and zhizhuwan) appear to pose the highest inhibition potential. In addition, ursolic acid, gambogic acid, and glycyrrhetic acid, if delivered intravenously, may attain high enough systemic concentrations to significantly inhibit CES1. The provision of a translational interpretation of in vitro assessments of natural product actions and interactions is limited by the dearth of basic pharmacokinetic data of the natural compounds exhibiting potent in vitro influences on CES1 activity. This is a major impediment to assigning even potential clinical significance. The modulatory effects on CES1 expression after chronic exposure to natural products warrants further investigation. SIGNIFICANCE STATEMENT: Modulation of CES1 activity by natural products may alter the course of treatment and clinical outcome. In this review, we have summarized the natural products that can potentially interact with CES1 substrate medications. We have also noted the limitations of existing reports and outlined challenges and future directions in this field.
Collapse
Affiliation(s)
- Yuli Qian
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - John S Markowitz
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| |
Collapse
|
9
|
Shen Y, Shi Z, Yan B. Carboxylesterases: Pharmacological Inhibition Regulated Expression and Transcriptional Involvement of Nuclear Receptors and other Transcription Factors. NUCLEAR RECEPTOR RESEARCH 2019. [DOI: 10.32527/2019/101435] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Yuanjun Shen
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA
| | - Zhanquan Shi
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Bingfang Yan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
10
|
He W, Xu Y, Ren X, Xiang D, Lei K, Zhang C, Liu D. Vitamin E Ameliorates Lipid Metabolism in Mice with Nonalcoholic Fatty Liver Disease via Nrf2/CES1 Signaling Pathway. Dig Dis Sci 2019; 64:3182-3191. [PMID: 31076985 DOI: 10.1007/s10620-019-05657-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 05/03/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Vitamin E has been reported to have a beneficial effect on nonalcoholic fatty liver disease (NAFLD); however, the underlying mechanism of action has not yet been clearly defined. AIM We aimed to evaluate the effects and mechanisms of vitamin E on lipid and glucose homeostasis both in vivo and in vitro. METHODS An NAFLD model was established in C57BL/6 mice fed a 30% fructose solution for 8 weeks. Subsequently, NAFLD mice were given vitamin E (70 mg/kg) for 2 weeks. In addition, L02 cells were treated with 5 mM fructose and 100 nM vitamin E to explore the potential mechanisms of action. RESULTS Vitamin E reversed the impaired glucose tolerance of fructose-treated mice. Histopathological examination showed that liver steatosis was significantly relieved in vitamin E-treated mice. These effects may be attributed to the upregulation of nuclear factor erythroid-2-related factor 2 (Nrf2), carboxylesterase 1 (CES1), and downregulated proteins involved in lipid synthesis by vitamin E treatment. In vivo, vitamin E also significantly reduced lipid accumulation in fructose-treated L02 cells, and the Nrf2 inhibitor ML385 reversed the protective effects of vitamin E. CONCLUSION These data indicated that the therapeutic effects of vitamin E on lipid and glucose homeostasis may be associated with activation of the Nrf2/CES1 signaling pathway.
Collapse
Affiliation(s)
- Wenxi He
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanjiao Xu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuhua Ren
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Xiang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Lei
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengliang Zhang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
11
|
Shaw P, Chattopadhyay A. Nrf2–ARE signaling in cellular protection: Mechanism of action and the regulatory mechanisms. J Cell Physiol 2019; 235:3119-3130. [PMID: 31549397 DOI: 10.1002/jcp.29219] [Citation(s) in RCA: 262] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 09/03/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Pallab Shaw
- Department of Zoology, Toxicology and Cancer Biology Laboratory Visva‐Bharati Santiniketan West Bengal India
| | - Ansuman Chattopadhyay
- Department of Zoology, Toxicology and Cancer Biology Laboratory Visva‐Bharati Santiniketan West Bengal India
| |
Collapse
|
12
|
Mao T, Li F, Fang Y, Wang H, Chen J, Li M, Lu Z, Qu J, Li J, Hu J, Cheng X, Ni M, Li B. Effects of chlorantraniliprole exposure on detoxification enzyme activities and detoxification-related gene expression in the fat body of the silkworm, Bombyx mori. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 176:58-63. [PMID: 30921697 DOI: 10.1016/j.ecoenv.2019.03.074] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 06/09/2023]
Abstract
Chlorantraniliprole (CAP) can induce excessive calcium release from muscle of insects, causing muscle paralysis until death, and its residues in farmland can cause poisoning in Bombyx mori (B. mori), resulting in the failure of cocooning. No reports have investigated the effects of CAP exposure on detoxification enzyme activities and detoxification-related gene expression in B. mori. In the present study, we treated mulberry leaves with CAP by the leaf-dipping method, and then B. mori larvae were continuously fed with the polluted mulberry leaves. Moreover, the detoxification enzyme activities and the expressions of detoxification-related genes in the fat body of B. mori were examined. The results showed that at 24 h after CAP exposure, the activities of P450 and GST enzymes were all significantly increased, with P450 enzymes responding fastest. CarE enzyme activity was up-regulated in 24 h, and then it was decreased compared with the control group. Furthermore, the expressions of the key genes in the PI3K/Akt/CncC signaling pathway (PI3K, PDK, Akt, CncC and Keap1) at the mRNA were significantly increased. Western blotting analysis revealed that Akt was inhibited at the protein level, resulting in decreased expression of Keap1 and increased expression of CncC. These results indicated that the PI3K/Akt/CncC signaling pathway in the fat body of B. mori responded to CAP exposure and regulated the expressions of downstream detoxification enzymes, thus enhancing the detoxifying capability of B. mori.
Collapse
Affiliation(s)
- Tingting Mao
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Fanchi Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China; Sericulture Institute of Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Yilong Fang
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Hui Wang
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Jian Chen
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Mengxue Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Zhengting Lu
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Jianwei Qu
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Jinxin Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Jiahuan Hu
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Xiaoyu Cheng
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China
| | - Min Ni
- Jiangsu Key Laboratory of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, Jiangsu, 215009, PR China
| | - Bing Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, Jiangsu, 215123, PR China; Sericulture Institute of Soochow University, Suzhou, Jiangsu, 215123, PR China.
| |
Collapse
|
13
|
Xu J, Qiu JC, Ji X, Guo HL, Wang X, Zhang B, Wang T, Chen F. Potential Pharmacokinetic Herb-Drug Interactions: Have we Overlooked the Importance of Human Carboxylesterases 1 and 2? Curr Drug Metab 2019; 20:130-137. [PMID: 29600756 DOI: 10.2174/1389200219666180330124050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/02/2018] [Accepted: 02/10/2018] [Indexed: 12/11/2022]
Abstract
Background:
Herbal products have grown steadily across the globe and have increasingly been incorporated
into western medicine for healthcare aims, thereby causing potential pharmacokinetic Herb-drug Interactions
(HDIs) through the inhibition or induction of drug-metabolizing enzymes and transporters. Human Carboxylesterases
1 (CES1) and 2 (CES2) metabolize endogenous and exogenous chemicals including many important therapeutic
medications. The growing number of CES substrate drugs also underscores the importance of the enzymes. Herein,
we summarized those potential inhibitors and inducers coming from herbal constituents toward CES1 and CES2. We
also reviewed the reported HDI studies focusing on herbal products and therapeutic agents metabolized by CES1 or
CES2.
Methods:
We searched in PubMed for manuscript published in English after Jan 1, 2000 combining terms “carboxylesterase
1”, “carboxylesterase 2”, “inhibitor”, “inducer”, “herb-drug interaction”, “inhibitory”, and “herbal supplement”.
We also searched specific websites including FDA and EMA. The data of screened papers were analyzed and
summarized.
Results:
The results showed that more than 50 natural inhibitors of CES1 or CES2, including phenolic chemicals,
triterpenoids, and tanshinones were found from herbs, whereas only few inducers of CES1 and CES2 were reported.
Systemic exposure to some commonly used drugs including oseltamivir, irinotecan, and clopidogrel were changed
when they were co-administered with herb products such as goldenseal, black cohosh, ginger, St. John’s Wort, curcumin,
and some Chinese compound formula in animals.
Conclusion:
Nonclinical and clinical studies on HDIs are warranted in the future to provide safety information toward
better clinical outcomes for the combination of herbal products and conventional drugs.
Collapse
Affiliation(s)
- Jing Xu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jin-Chun Qiu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xing Ji
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hong-Li Guo
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan Wang
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Bo Zhang
- College of Food Science and Engineering, Jinzhou Medical University, Jinzhou, China
| | - Tengfei Wang
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Feng Chen
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Wu S, Lu H, Bai Y. Nrf2 in cancers: A double-edged sword. Cancer Med 2019; 8:2252-2267. [PMID: 30929309 PMCID: PMC6536957 DOI: 10.1002/cam4.2101] [Citation(s) in RCA: 314] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/21/2019] [Accepted: 02/26/2019] [Indexed: 12/11/2022] Open
Abstract
The Nrf2/Keap1 pathway is an important signaling cascade responsible for the resistance of oxidative damage induced by exogenous chemicals. It maintains the redox homeostasis, exerts anti-inflammation and anticancer activity by regulating its multiple downstream cytoprotective genes, thereby plays a vital role in cell survival. Interestingly, in recent years, accumulating evidence suggests that Nrf2 has a contradictory role in cancers. Aberrant activation of Nrf2 is associated with poor prognosis. The constitutive activation of Nrf2 in various cancers induces pro-survival genes and promotes cancer cell proliferation by metabolic reprogramming, repression of cancer cell apoptosis, and enhancement of self-renewal capacity of cancer stem cells. More importantly, Nrf2 is proved to contribute to the chemoresistance and radioresistance of cancer cells as well as inflammation-induced carcinogenesis. A number of Nrf2 inhibitors discovered for cancer treatment were reviewed in this report. These provide a new strategy that targeting Nrf2 could be a promising therapeutic approach against cancer. This review aims to summarize the dual effects of Nrf2 in cancer, revealing its function both in cancer prevention and inhibition, to further discover novel anticancer treatment.
Collapse
Affiliation(s)
- Shijia Wu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hong Lu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
15
|
Human carboxylesterases: a comprehensive review. Acta Pharm Sin B 2018; 8:699-712. [PMID: 30245959 PMCID: PMC6146386 DOI: 10.1016/j.apsb.2018.05.005] [Citation(s) in RCA: 315] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 12/12/2022] Open
Abstract
Mammalian carboxylesterases (CEs) are key enzymes from the serine hydrolase superfamily. In the human body, two predominant carboxylesterases (CES1 and CES2) have been identified and extensively studied over the past decade. These two enzymes play crucial roles in the metabolism of a wide variety of endogenous esters, ester-containing drugs and environmental toxicants. The key roles of CES in both human health and xenobiotic metabolism arouse great interest in the discovery of potent CES modulators to regulate endobiotic metabolism or to improve the efficacy of ester drugs. This review covers the structural and catalytic features of CES, tissue distributions, biological functions, genetic polymorphisms, substrate specificities and inhibitor properties of CES1 and CES2, as well as the significance and recent progress on the discovery of CES modulators. The information presented here will help pharmacologists explore the relevance of CES to human diseases or to assign the contribution of certain CES in xenobiotic metabolism. It will also facilitate medicinal chemistry efforts to design prodrugs activated by a given CES isoform, or to develop potent and selective modulators of CES for potential biomedical applications.
Collapse
|
16
|
Wilding CS. Regulating resistance: CncC:Maf, antioxidant response elements and the overexpression of detoxification genes in insecticide resistance. CURRENT OPINION IN INSECT SCIENCE 2018; 27:89-96. [PMID: 30025640 DOI: 10.1016/j.cois.2018.04.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 03/12/2018] [Accepted: 04/09/2018] [Indexed: 05/24/2023]
Abstract
Although genetic and genomic tools have greatly furthered our understanding of resistance-associated mutations in molecular target sites of insecticides, the genomic basis of transcriptional regulation of detoxification loci in insect pests and vectors remains relatively unexplored. Recent work using RNAi, reporter assays and comparative genomics are beginning to reveal the molecular architecture of this response, identifying critical transcription factors and their binding sites. Central to this is the insect ortholog of the mammalian transcription factor Nrf2, Cap 'n' Collar isoform-C (CncC) which as a heterodimer with Maf-S regulates the transcription of phase I, II and III detoxification loci in a range of insects, with CncC knockdown or upregulation directly affecting phenotypic resistance. CncC:Maf binds to specific antioxidant response element sequences upstream of detoxification genes to initiate transcription. Recent work is now identifying these binding sites for resistance-associated loci and, coupled with genome sequence data and reporter assays, enabling identification of polymorphisms in the CncC:Maf binding site which regulate the insecticide resistance phenotype.
Collapse
Affiliation(s)
- Craig S Wilding
- School of Natural Sciences and Psychology, Liverpool John Moores University, Liverpool L3 3AF, UK.
| |
Collapse
|
17
|
Wang H, Xu YS, Wang ML, Cheng C, Bian R, Yuan H, Wang Y, Guo T, Zhu LL, Zhou H. Protective effect of naringin against the LPS-induced apoptosis of PC12 cells: Implications for the treatment of neurodegenerative disorders. Int J Mol Med 2017; 39:819-830. [PMID: 28260042 PMCID: PMC5360435 DOI: 10.3892/ijmm.2017.2904] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/10/2017] [Indexed: 01/01/2023] Open
Abstract
Several studies have demonstrated that increased apoptosis plays an essential role in neurodegenerative disorders. It has been demonstrated that lipopolysaccharide (LPS) induces apoptosis largely through the production of intracellular reactive oxygen species (ROS) and inflammatory mediators. In this study, we investigated the potential protective mechanisms of naringin (Nar), a pummelo peel extract, on LPS-induced PC12 cell apoptosis. Nar pre-conditioning prior to stimulation with LPS for 18 h was a prerequisite for evaluating PC12 cell viability and the protective mechanisms of Nar. Nar significantly improved cell survival in a time- and concentration-dependent manner. On the one hand, Nar downregulated cytochrome P450 2E1 (CYP2E1), inhibited the release of ROS, mitigated the stimulation of oxidative stress, and rectified the antioxidant protein contents of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), superoxide dismutase (SOD)2 and glutathione synthetase (GSS). On the other hand, Nar down-regulated inflammatory gene and protein expression, including interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, HMGB1, high mobility group box 1 protein (HMGB1), cyclo-oxygenase-2 (COX-2), the Toll-like receptor 4 (TLR4)-myeloid differentiation factor 88 (MyD88)-TNF receptor-associated factor 6 (TRAF6) path way and downstream mitogen activated protein kinase (MAPK) phosphorylation, activator protein transcription factor-1 (AP-1) and nuclear factor (NF)-κB. Moroever, Nar markedly attenuated the cytochrome c shift from the mitochondria to the cytosol and regulated caspase-3-related protein expression. To the best of our knowledge, this is the first study to report the antioxidant, anti-inflammatory and anti-apoptotic effects of Nar in neuronal-like PC12 cells. These results suggest that Nar can be utilized as a potential drug for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Hui Wang
- Department of Neurosurgery, Traffic Hospital of Shandong Province, Jinan, Shandong 250031, P.R. China
| | - You Song Xu
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Miao Lin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Chao Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Rui Bian
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Hao Yuan
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yi Wang
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Ting Guo
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Lin Lin Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Hang Zhou
- Department of Neurosurgery, The 2nd Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| |
Collapse
|
18
|
Wang DD, Jin Q, Hou J, Feng L, Li N, Li SY, Zhou Q, Zou LW, Ge GB, Wang JG, Yang L. Highly sensitive and selective detection of human carboxylesterase 1 activity by liquid chromatography with fluorescence detection. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1008:212-218. [DOI: 10.1016/j.jchromb.2015.11.046] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/20/2015] [Accepted: 11/25/2015] [Indexed: 11/29/2022]
|
19
|
Wang DD, Jin Q, Zou LW, Hou J, Lv X, Lei W, Cheng HL, Ge GB, Yang L. A bioluminescent sensor for highly selective and sensitive detection of human carboxylesterase 1 in complex biological samples. Chem Commun (Camb) 2016; 52:3183-6. [DOI: 10.1039/c5cc09874b] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A highly selective and sensitive bioluminescent sensor (DME) for real-time monitoring of human carboxylesterase 1 (hCE1) activities in complex biological samples and bio-imaging of endogenous hCE1 in living cells.
Collapse
Affiliation(s)
- Dan-Dan Wang
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| | - Qiang Jin
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| | - Li-Wei Zou
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| | - Jie Hou
- Dalian Medical University
- Dalian
- China
| | - Xia Lv
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| | - Wei Lei
- Dalian Medical University
- Dalian
- China
| | - Hai-Ling Cheng
- Cancer Institute
- The Second Hospital of Dalian Medical University
- Dalian
- China
| | - Guang-Bo Ge
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| | - Ling Yang
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian
- China
| |
Collapse
|
20
|
Cheng L, Li F, Ma R, Hu X. Forsythiaside inhibits cigarette smoke-induced lung inflammation by activation of Nrf2 and inhibition of NF-κB. Int Immunopharmacol 2015; 28:494-9. [PMID: 26209933 DOI: 10.1016/j.intimp.2015.07.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/28/2015] [Accepted: 07/10/2015] [Indexed: 12/13/2022]
Abstract
Cigarette smoke has been reported to be the major cause of chronic obstructive pulmonary disease (COPD). It causes persistent inflammation by regulating the redox-sensitive pathways. Forsythiaside, an active constituent isolated from the Chinese medicinal herb Forsythia suspensa, has been reported to have anti-inflammatory and anti-oxidant effects. Thus, in this study, we investigated the protective effects of forsythiaside against cigarette smoke-induced lung inflammation in mice. COPD mice model was established by cigarette smoke. Forsythiaside was given 2h before cigarette smoke exposure for five consecutive days. Bronchoalveolar lavage fluid and lung tissues were collected to assess pathological changes, lipid peroxidation, inflammatory cytokine production, Nrf-2, and NF-κB expression. Our results showed that forsythiaside attenuated the infiltration of inflammatory cells, NO and inflammatory cytokines TNF-α, IL-6 and IL-1β production, and reversed the CS-induced decrease of glutathione/glutathione disulfide (GSH/GSSG) ratio. Western blot analysis showed that forsythiaside inhibited cigarette smoke-induced NF-κB activation. In addition, forsythiaside dose-dependently up-regulated the expression of Nrf2 and HO-1. In conclusion, forsythiaside protected against cigarette smoke-induced lung injury through activating Nrf2 and inhibiting NF-κB signaling pathway.
Collapse
Affiliation(s)
- Li Cheng
- Department of Comprehensive Medical, Affiliated Dongfeng Hospital of Hubei Medical College, Shiyan 442008, China
| | - Fan Li
- Department of Comprehensive Medical, Affiliated Dongfeng Hospital of Hubei Medical College, Shiyan 442008, China
| | - Rui Ma
- Department of Comprehensive Medical, Affiliated Dongfeng Hospital of Hubei Medical College, Shiyan 442008, China
| | - Xianping Hu
- Department of Comprehensive Medical, Affiliated Dongfeng Hospital of Hubei Medical College, Shiyan 442008, China.
| |
Collapse
|
21
|
Zhang Z, Leir SH, Harris A. Oxidative stress regulates CFTR gene expression in human airway epithelial cells through a distal antioxidant response element. Am J Respir Cell Mol Biol 2015; 52:387-96. [PMID: 25259561 DOI: 10.1165/rcmb.2014-0263oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cystic fibrosis transmembrane conductance regulator gene (CFTR) expression in human airway epithelial cells involves the recruitment of distal cis-regulatory elements, which are associated with airway-selective DNase hypersensitive sites at -44 kb and -35 kb from the gene. The -35-kb site encompasses an enhancer that is regulated by the immune mediators interferon regulatory factor 1 and 2 and by nuclear factor Y. Here we investigate the -44-kb element, which also has enhancer activity in vitro in airway epithelial cells but is inactive in intestinal epithelial cells. This site contains an antioxidant response element (ARE) that plays a critical role in its function in airway cell lines and primary human bronchial epithelial cells. The natural antioxidant sulforaphane (SFN) induces nuclear translocation of nuclear factor, erythroid 2-like 2 (Nrf2), a transcription factor that regulates genes with AREs in their promoters, many of which are involved in response to injury. Under normal conditions, the -44-kb ARE is occupied by the repressor BTB and CNC homology 1, basic leucine zipper transcription factor (Bach1), and v-Maf avian musculoaponeurotic fibrosarcoma oncogene homolog K (MafK) heterodimers. After 2 hours of SFN treatment, Nrf2 displaces these repressive factors and activates CFTR expression. Site-directed mutagenesis shows that both the ARE and an adjacent NF-κB binding site are required for activation of the -44-kb element in airway epithelial cells. Moreover, this element is functionally linked to the -35-kb enhancer in modulating CFTR expression in response to environmental stresses in the airway.
Collapse
Affiliation(s)
- Zhaolin Zhang
- Human Molecular Genetics Program, Lurie Children's Research Center, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | |
Collapse
|
22
|
Yin L, Zheng L, Xu L, Dong D, Han X, Qi Y, Zhao Y, Xu Y, Peng J. In-silico prediction of drug targets, biological activities, signal pathways and regulating networks of dioscin based on bioinformatics. Altern Ther Health Med 2015; 15:41. [PMID: 25879470 PMCID: PMC4354738 DOI: 10.1186/s12906-015-0579-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 02/21/2015] [Indexed: 11/25/2022]
Abstract
Background Inverse docking technology has been a trend of drug discovery, and bioinformatics approaches have been used to predict target proteins, biological activities, signal pathways and molecular regulating networks affected by drugs for further pharmacodynamic and mechanism studies. Methods In the present paper, inverse docking technology was applied to screen potential targets from potential drug target database (PDTD). Then, the corresponding gene information of the obtained drug-targets was applied to predict the related biological activities, signal pathways and processes networks of the compound by using MetaCore platform. After that, some most relevant regulating networks were considered, which included the nodes and relevant pathways of dioscin. Results 71 potential targets of dioscin from humans, 7 from rats and 8 from mice were screened, and the prediction results showed that the most likely targets of dioscin were cyclin A2, calmodulin, hemoglobin subunit beta, DNA topoisomerase I, DNA polymerase lambda, nitric oxide synthase and UDP-N-acetylhexosamine pyrophosphorylase, etc. Many diseases including experimental autoimmune encephalomyelitis of human, temporal lobe epilepsy of rat and ankylosing spondylitis of mouse, may be inhibited by dioscin through regulating immune response alternative complement pathway, G-protein signaling RhoB regulation pathway and immune response antiviral actions of interferons, etc. The most relevant networks (5 from human, 3 from rat and 5 from mouse) indicated that dioscin may be a TOP1 inhibitor, which can treat cancer though the cell cycle– transition and termination of DNA replication pathway. Dioscin can down regulate EGFR and EGF to inhibit cancer, and also has anti-inflammation activity by regulating JNK signaling pathway. Conclusions The predictions of the possible targets, biological activities, signal pathways and relevant regulating networks of dioscin provide valuable information to guide further investigation of dioscin on pharmacodynamics and molecular mechanisms, which also suggests a practical and effective method for studies on the mechanism of other chemicals.
Collapse
|
23
|
Oda S, Fukami T, Yokoi T, Nakajima M. A comprehensive review of UDP-glucuronosyltransferase and esterases for drug development. Drug Metab Pharmacokinet 2015; 30:30-51. [DOI: 10.1016/j.dmpk.2014.12.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/24/2014] [Accepted: 12/02/2014] [Indexed: 01/24/2023]
|
24
|
Vachirayonsti T, Ho KW, Yang D, Yan B. Suppression of the pregnane X receptor during endoplasmic reticulum stress is achieved by down-regulating hepatocyte nuclear factor-4α and up-regulating liver-enriched inhibitory protein. Toxicol Sci 2015; 144:382-92. [PMID: 25616597 DOI: 10.1093/toxsci/kfv008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is recognized as a common theme in the development of metabolic syndrome and other diseases. Chronic liver diseases develop ER stress and also show decreased capacity of drug metabolism. The pregnane X receptor (PXR) is a master regulator of genes involved in drug elimination. This study was performed to determine whether ER stress condition decreases the expression of PXR and whether the decrease alters the induction of cytochrome P450 3A4 (CYP3A4). Human primary hepatocytes and HepG2 cell line (human hepatocellular carcinoma) were treated with brefeldin A and thapsigargin, 2 well-established ER stressors. Without exceptions, both stressors significantly decreased the expression of PXR. The decrease led to reduced induction of CYP3A4. Reporter dissection study, electrophoretic mobility shift assay, and chromatin immunoprecipitation located in the PXR promoter region 2 adjacent elements recognized by hepatocyte nuclear factor-4α (HNF-4α) and cytidine-cytidine-adenosine-adenosine-thymidine enhanced binding proteins (C/EBPs), respectively. Additional studies demonstrated that HNF-4α was down-regulated during ER stress but the expression of C/EBPβ varied depending on a particular form of C/EBPβ. Liver-enriched activator protein (LAP) was down-regulated but liver-enriched inhibitory protein (LIP) was highly induced. Nevertheless, over-expression of HNF-4α or LAP restored the expression of PXR. Interestingly, the very same sequence also responded to interleukin-6 (IL-6), and primary hepatocytes treated with thapsigargin significantly increased the level of IL-6 mRNA. These findings establish a functional interconnection between ER stress and signaling of proinflammatory cytokines in the regulation of PXR expression.
Collapse
Affiliation(s)
- Thaveechai Vachirayonsti
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, University of Rhode Island, Kingston, Rhode Island 02881
| | - Karen W Ho
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, University of Rhode Island, Kingston, Rhode Island 02881
| | - Dongfang Yang
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, University of Rhode Island, Kingston, Rhode Island 02881
| | - Bingfang Yan
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, University of Rhode Island, Kingston, Rhode Island 02881
| |
Collapse
|