1
|
Prieto LA, Khiar-Fernández N, Calderón-Montaño JM, López-Lázaro M, Lucía-Tamudo J, Nogueira JJ, León R, Moreno N, Valdivia V, Recio R, Fernández I. Exploring the broad-spectrum activity of carbohydrate-based Iberin analogues: From anticancer effect to antioxidant properties. Eur J Med Chem 2025; 289:117469. [PMID: 40058183 DOI: 10.1016/j.ejmech.2025.117469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025]
Abstract
Iberin is a lower homologue of sulforaphane (SFN) which has shown effectiveness in addressing various pathologies, including its anti-inflammatory properties, antitumor activity against various cancers, and antimicrobial effects. Building on this activity, a series of carbohydrate-based analogues of the natural isothiocyanate (ITC) iberin were synthesized, and their anticancer and antioxidant activities were evaluated. Cytotoxicity studies on three cancer cell lines using Resazurin assay demonstrated significant cytotoxic activity, particularly against bladder cancer. The sulfonyl derivatives exhibited the most potent effects, with IC50 values comparable to those of reference natural isothiocyanates (from 10 to 20 μM). Computational simulations support the hypothesis that carbohydrate-based ITCs can interact with STAT3's SH2 domain in a manner similar to SFN, laying the groundwork for their potential development as STAT3-targeted anticancer agents. The antioxidant potential of these compounds was assessed by their ability to activate the Nrf2 factor, yielding CD values (concentration required to double luciferase activity compared to basal conditions) between 1.55 and 10.36 μM, without cytotoxicity at these concentrations. Notably, the phenylsulfone derivative 22β displayed slightly higher or comparable antioxidant activity to that of natural isothiocyanates. Based on these findings, this phenylsulfone analogue was selected as the optimal compound due to its dual anticancer and antioxidant activities. An additional advantage of this carbohydrate-based ITC is that it is a solid compound, making it easier to handle than natural isothiocyanates, which are typically liquids.
Collapse
Affiliation(s)
- L A Prieto
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012, Sevilla, Spain
| | - N Khiar-Fernández
- Department of Organic Chemistry, School of Chemistry, Universidad Complutense de Madrid, Plaza de las Ciencias s/n, 28040, Madrid, Spain
| | - J M Calderón-Montaño
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012, Seville, Spain
| | - M López-Lázaro
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012, Seville, Spain
| | - J Lucía-Tamudo
- Faculty of Chemistry and Pharmacy, Institute of Physical and Theoretical Chemistry, University of Regensburg, 93040, Regensburg, Germany
| | - J J Nogueira
- Department of Chemistry, Universidad Autónoma de Madrid, Calle Francisco Tomás y Valiente, 7, 28049, Madrid, Spain; Institute for Advanced Research in Chemistry (IAdChem), Universidad Autónoma de Madrid, Calle Francisco Tomás y Valiente, 7, 28049, Madrid, Spain
| | - R León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), C/Juan de la Cierva 3, Madrid, 28006, Spain
| | - N Moreno
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012, Sevilla, Spain
| | - V Valdivia
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012, Sevilla, Spain.
| | - R Recio
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012, Sevilla, Spain
| | - I Fernández
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012, Sevilla, Spain.
| |
Collapse
|
2
|
Luo Y, Melhem S, Feelisch M, Chatre L, Morton NM, Dolga AM, van Goor H. Thiosulphate sulfurtransferase: Biological roles and therapeutic potential. Redox Biol 2025; 82:103595. [PMID: 40107018 PMCID: PMC11957799 DOI: 10.1016/j.redox.2025.103595] [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: 01/07/2025] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Mitochondria are central to eukaryotic cell function, driving energy production, intermediary metabolism, and cellular homeostasis. Dysregulation of mitochondrial function often results in oxidative stress, a hallmark of numerous diseases, underscoring the critical need for maintaining mitochondrial integrity. Among mitochondrial enzymes, thiosulfate sulfurtransferase (TST) has emerged as a key regulator of sulfur metabolism, redox balance, and Fe-S protein maintenance. Beyond its well-known role in cyanide detoxification, TST facilitates hydrogen sulfide (H2S) metabolism by catalyzing the transfer of sulfur from persulfides (R-SSH) to thiosulfate (S2O32-), promoting H2S oxidation and preventing its toxic accumulation. Additionally, TST contributes to the thiol-dependent antioxidant system by regulating reactive sulfur species and sustaining mitochondrial functionality through its role in sulfide-driven bioenergetics. This review highlights the biochemical and therapeutic significance of TST in mitochondrial and cellular health, emphasizing its protective roles in diseases associated with oxidative stress and mitochondrial dysfunction. Dysregulation of TST has been implicated in diverse pathologies, including specific metabolic disorders, neurological diseases, cardiovascular conditions, kidney dysfunction, inflammatory bowel disease, and cancer. These associations underline TST's potential as a biomarker and therapeutic target. Therapeutic strategies to activate the TST pathway are explored, with a focus on sodium thiosulfate (STS), novel small molecule (Hit 2), and recombinant hTST protein. STS, an FDA-approved compound, has demonstrated antioxidant and anti-inflammatory effects across multiple preclinical models, mitigating oxidative damage and improving mitochondrial integrity. A slow-release oral formulation of STS is under development, offering promise for expanding its clinical applications. Small molecule activators like Hit 2 and hTST protein have shown efficacy in enhancing mitochondrial respiration and reducing oxidative stress, though both reagents need further in vitro and in vivo investigations. Despite promising advancements, TST-based therapies remain underexplored. Future research should focus on leveraging TST's interplay with pathways like NRF2 signaling, investigating its broader protective roles in cellular health, and developing targeted interventions. Enhancing TST activity represents an innovative therapeutic approach for addressing mitochondrial dysfunction, oxidative stress, and their associated pathologies, offering new hope for the treatment of diseases associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Yang Luo
- University of Groningen, Dept. of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Faculty of Science and Engineering, Groningen, the Netherlands; University Medical Center Groningen, Dept. of Pathology and Medical Biology, Groningen, the Netherlands
| | - Shaden Melhem
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Laurent Chatre
- Université de Caen Normandie, CNRS, Normandie Univ, ISTCT, UMR6030, GIP Cyceron, Caen, F-14000, France
| | - Nicholas M Morton
- Centre for Systems Health and Integrated Metabolic Research, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Amalia M Dolga
- University of Groningen, Dept. of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Faculty of Science and Engineering, Groningen, the Netherlands
| | - Harry van Goor
- University Medical Center Groningen, Dept. of Pathology and Medical Biology, Groningen, the Netherlands.
| |
Collapse
|
3
|
Qi R, Fei Y. Auraptene Mitigates Cigarette Smoke and Lipopolysaccharide-Induced Chronic Obstructive Pulmonary Disease in Mice and BEAS-2B Cells via Regulating Keap1/Nrf2/HO-1 Pathway. J Biochem Mol Toxicol 2025; 39:e70253. [PMID: 40269617 DOI: 10.1002/jbt.70253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/24/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is a most common respiratory condition characterized by airflow limitation, airway inflammation, and lung injury. The present study was undertaken to unveil the therapeutic potentials of the auroptene against lipopolysaccharide (LPS) and cigarette smoke (CS)-induced COPD in mice. The CS along with LPS was exposed to healthy C57BL/6 mice through the intranasal route to induce COPD. The exposure to CS was continued for 12 weeks. The LPS challenge was occurred on weeks 2, 4, 6, and 8. The auraptene was treated orally by gavage route 1 h before to CS exposure for last 4 weeks. After the completion of treatment, the respiratory function was assessed using a pulmonary function test equipment. The levels of mucin proteins, extracellular matrix (ECM) proteins, proliferative cytokine markers, epithelial marker protein E-cadherin, oxidative stress-related biomarkers, and inflammation-associated markers were assessed using respective commercial assay kits. An analysis of histopathology and histo-morphology was conducted on the pulmonary tissues. An in vitro assays were conducted on the CS condensate (CSC) and LPS-challenged BEAS-2B cells. The expressions of Keap1/Nrf2/HO-1 pathway associated proteins were assessed using assay kits. The findings of the current work has clearly proved that auraptene at 25 mg/kg concentrations significantly increased the pulmonary functions in the mice with COPD. The treatment of auraptene effectively reduced the ECM protein levels, proliferative cytokine marker levels, and inflammation-related cytokine levels in the COPD mice. In addition, the auraptene treatment effectively increased the antioxidants and mitigated the lung tissue injuries in the COPD mice. The Keap1/Nrf2/HO-1 signaling pathway expressions successfully regulated by the auraptene treatment in the CSC and LPS-induced BEAS-2B cells. Therefore, the current findings has highlighted that auraptene has the capability to be a beneficial intervention to treat COPD.
Collapse
Affiliation(s)
- Rui Qi
- TCM Department, XD Group Hospital, Xi'an, China
| | - Yuwen Fei
- TCM Department, XD Group Hospital, Xi'an, China
| |
Collapse
|
4
|
Zhao Z, Lu H, Wang J, Wu T, Xu S, Ge Y, You Q, Jiang Z, Lu M. Discovery of β-amino acid substituted naphthalene sulfonamide derivatives as potent Kelch-like ECH-associated protein 1-nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2) protein-protein interaction inhibitors for ulcerative colitis management. Eur J Med Chem 2025; 288:117384. [PMID: 39965408 DOI: 10.1016/j.ejmech.2025.117384] [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: 12/20/2024] [Revised: 01/24/2025] [Accepted: 02/02/2025] [Indexed: 02/20/2025]
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a key regulator of cellular defense system against oxidative insults. Directly inhibiting the Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 protein-protein interaction (PPI) has emerged as a promising approach to activate Nrf2 for the treatment of diseases associated with oxidative stress. Herein, we identified β-amino acids as privileged structural fragments for designing novel naphthalene sulfonamide-based Keap1-Nrf2 PPI inhibitors. Comprehensive structure-activity relationship (SAR) exploration identified compound 19 as the optimal inhibitor with an IC50 of 0.55 μM for disrupting the Keap1-Nrf2 interaction and a Kd of 0.50 μM for binding to Keap1. Further studies demonstrated that 19 effectively activated the Nrf2-regulated cytoprotective system and provided protective effects against dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in both in vitro and in vivo models. These findings highlight the potential of β-amino acid substituted naphthalene sulfonamide Keap1-Nrf2 inhibitor 19 as a prospective therapeutic agent for UC via Keap1 targeting.
Collapse
Affiliation(s)
- Ziquan Zhao
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hongjin Lu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Junjie Wang
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Tingting Wu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Shicheng Xu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuxin Ge
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University Medical College, Suzhou, 215123, China.
| | - Zhengyu Jiang
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Mengchen Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University Medical College, Suzhou, 215123, China.
| |
Collapse
|
5
|
Cuadrado A, Cazalla E, Bach A, Bathish B, Naidu SD, DeNicola GM, Dinkova-Kostova AT, Fernández-Ginés R, Grochot-Przeczek A, Hayes JD, Kensler TW, León R, Liby KT, López MG, Manda G, Shivakumar AK, Hakomäki H, Moerland JA, Motohashi H, Rojo AI, Sykiotis GP, Taguchi K, Valverde ÁM, Yamamoto M, Levonen AL. Health position paper and redox perspectives - Bench to bedside transition for pharmacological regulation of NRF2 in noncommunicable diseases. Redox Biol 2025; 81:103569. [PMID: 40059038 PMCID: PMC11970334 DOI: 10.1016/j.redox.2025.103569] [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: 12/21/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a redox-activated transcription factor regulating cellular defense against oxidative stress, thereby playing a pivotal role in maintaining cellular homeostasis. Its dysregulation is implicated in the progression of a wide array of human diseases, making NRF2 a compelling target for therapeutic interventions. However, challenges persist in drug discovery and safe targeting of NRF2, as unresolved questions remain especially regarding its context-specific role in diseases and off-target effects. This comprehensive review discusses the dualistic role of NRF2 in disease pathophysiology, covering its protective and/or destructive roles in autoimmune, respiratory, cardiovascular, and metabolic diseases, as well as diseases of the digestive system and cancer. Additionally, we also review the development of drugs that either activate or inhibit NRF2, discuss main barriers in translating NRF2-based therapies from bench to bedside, and consider the ways to monitor NRF2 activation in vivo.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Eduardo Cazalla
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Gina M DeNicola
- Department of Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Raquel Fernández-Ginés
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28007, Madrid, Spain
| | - Karen T Liby
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Manuela G López
- Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain; Instituto Teófilo Hernando, Madrid, Spain
| | - Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | | | - Henriikka Hakomäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jessica A Moerland
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Hozumi Motohashi
- Department of Medical Biochemistry, Graduate School of Medicine Tohoku University, Sendai, Japan; Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Keiko Taguchi
- Laboratory of Food Chemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan; Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas "Sols-Morreale" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
6
|
Ozleyen A, Duran GN, Donmez S, Ozbil M, Doveston RG, Tumer TB. Identification and inhibition of PIN1-NRF2 protein-protein interactions through computational and biophysical approaches. Sci Rep 2025; 15:8907. [PMID: 40087364 PMCID: PMC11909128 DOI: 10.1038/s41598-025-89342-0] [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: 06/30/2024] [Accepted: 02/04/2025] [Indexed: 03/17/2025] Open
Abstract
NRF2 is a transcription factor responsible for coordinating the expression of over a thousand cytoprotective genes. Although NRF2 is constitutively expressed, its stability is modulated by the redox-sensitive protein KEAP1 and other conditional binding partner regulators. The new era of NRF2 research has highlighted the cooperation between NRF2 and PIN1 in modifying its cytoprotective effect. Despite numerous studies, the understanding of the PIN1-NRF2 interaction remains limited. Herein, we described the binding interaction of PIN1 and three different 14-mer long phospho-peptides mimicking NRF2 protein using computer-based, biophysical, and biochemical approaches. According to our computational analyses, the residues positioned in the WW domain of PIN1 (Ser16, Arg17, Ser18, Tyr23, Ser32, Gln33, and Trp34) were found to be crucial for PIN1-NRF2 interactions. Biophysical FP assays were used to verify the computational prediction. The data demonstrated that Pintide, a peptide predominantly interacting with the PIN1 WW-domain, led to a significant reduction in the binding affinity of the NRF2 mimicking peptides. Moreover, we evaluated the impact of known PIN1 inhibitors (juglone, KPT-6566, and EGCG) on the PIN1-NRF2 interaction. Among the inhibitors, KPT-6566 showed the most potent inhibitory effect on PIN1-NRF2 interaction within an IC50 range of 0.3-1.4 µM. Furthermore, our mass spectrometry analyses showed that KPT-6566 appeared to covalently modify PIN1 via conjugate addition, rather than disulfide exchange of the sulfonyl-acetate moiety. Altogether, such inhibitors would also be highly valuable molecular probes for further investigation of PIN1 regulation of NRF2 in the cellular context and potentially pave the way for drug molecules that specifically inhibit the cytoprotective effects of NRF2 in cancer.
Collapse
Affiliation(s)
- Adem Ozleyen
- Leicester Institute for Structural and Chemical Biology, University of Leicester, Leicester, LE1 7RH, UK
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK
- Health Institutes of Türkiye, Türkiye Biotechnology Institute, 06270, Ankara, Turkey
| | - Gizem Nur Duran
- Institute of Biotechnology, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
| | - Serhat Donmez
- Graduate Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University, 17020, Canakkale, Turkey
- Institute of Science and Technology Austria (ISTA), 3400, Klosterneuburg, Austria
| | - Mehmet Ozbil
- Institute of Biotechnology, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
| | - Richard G Doveston
- Leicester Institute for Structural and Chemical Biology, University of Leicester, Leicester, LE1 7RH, UK.
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK.
| | - Tugba Boyunegmez Tumer
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Canakkale Onsekiz Mart University, 17020, Canakkale, Turkey.
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
7
|
Zheng Z, Zhang P, Fu Y, Jiang Y, Zhu J, Wang F, Li S, Zhang Z, Chang T, Li T, Zhang M, Ruan B, Wang X. The potential role of the SIRT1-Nrf2 signaling pathway in alleviating hidden hearing loss via antioxidant stress. Cell Biol Int 2025; 49:262-276. [PMID: 39618038 PMCID: PMC11811746 DOI: 10.1002/cbin.12264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 02/12/2025]
Abstract
Hidden hearing loss (HHL) is characterized by normal audiometric thresholds but impaired auditory function, particularly in noisy environments. In vivo, we employed auditory brainstem response (ABR) testing and ribbon synapses counting to assess changes in mouse hearing function, and observed the morphology of hair cells through scanning electron microscopy. SRT1720 was administered to the cochlea via round window injection. In vitro, western blot analysis and RT-qPCR were used, and Lenti-shNrf2 was used to knockdown Nrf2 expression. In addition, various oxidative stress indicators were detected by immunofluorescence, kit-based assays, and flow cytometry. ABR measurement of HHL mouse showed a significant increase in hearing threshold, as well as a decrease and delay in the I wave amplitude and latency on the first day after noise exposure. Histological observation showed a significant loss of ribbon synapses and stereocilia lodging. HHL mice exhibited oxidative stress, which was reduced by pretreatment with SRT1720. Additionally, SRT1720 could reduce hydrogen peroxide-induced oxidative stress in HEI-OC1 cells through activating the SIRT1/Nrf2 pathway. Subsequent experiments with Nrf2 knockdown confirmed the importance of this pathway. findings highlight oxidative stress as the primary contributor to HHL, with the SIRT1/Nrf2 signaling pathway emerging as a promising therapeutic target for alleviating HHL.
Collapse
Affiliation(s)
- Zeyu Zheng
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of EducationAir Force Medical UniversityXi'anShaanxiChina
- Department of Aviation MedicineXijing HospitalXi'anChina
| | - Peng Zhang
- Department of OtolaryngologyXijing HospitalXi'anChina
| | - Yang Fu
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of EducationAir Force Medical UniversityXi'anShaanxiChina
- Department of Aviation MedicineXijing HospitalXi'anChina
| | - Yihong Jiang
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of EducationAir Force Medical UniversityXi'anShaanxiChina
- Department of Aviation MedicineXijing HospitalXi'anChina
| | - Jing Zhu
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of EducationAir Force Medical UniversityXi'anShaanxiChina
- Department of Aviation MedicineXijing HospitalXi'anChina
| | - Fei Wang
- School of Basic MedicineAir Force Medical UniveristyXi'anChina
| | - Shaoheng Li
- Department of Ophthalmology, Eye Institute of Chinese PLAXijing HospitalXi'anChina
| | - Zhuoru Zhang
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of EducationAir Force Medical UniversityXi'anShaanxiChina
- Department of Aviation MedicineXijing HospitalXi'anChina
| | - Tong Chang
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of EducationAir Force Medical UniversityXi'anShaanxiChina
- Department of Aviation MedicineXijing HospitalXi'anChina
| | - Tian Li
- Tianjin Medical UniversityTianjinChina
| | - Min Zhang
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of EducationAir Force Medical UniversityXi'anShaanxiChina
- Department of OtolaryngologyXijing HospitalXi'anChina
| | - Bai Ruan
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of EducationAir Force Medical UniversityXi'anShaanxiChina
- Department of Aviation MedicineXijing HospitalXi'anChina
| | - Xiaocheng Wang
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of EducationAir Force Medical UniversityXi'anShaanxiChina
- Department of Aviation MedicineXijing HospitalXi'anChina
| |
Collapse
|
8
|
Salman S, Paulet V, Hardonnière K, Kerdine‐Römer S. The role of NRF2 transcription factor in inflammatory skin diseases. Biofactors 2025; 51:e70013. [PMID: 40207460 PMCID: PMC11983367 DOI: 10.1002/biof.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
The skin is the body's largest organ and performs several vital functions, such as controlling the movement of essential substances while protecting against external threats. Although mainly composed of keratinocytes (KCs), the skin also contains a complex network of immune cells that play a critical role in host defense and maintaining skin homeostasis. KCs proliferate in the basal layer of the epidermis and undergo differentiation, altering their functional and phenotypic characteristics. These differentiation steps are crucial for the stratification of the epidermis and the formation of the stratum corneum, ensuring the skin barrier's functions. Exposure to UV, environmental pollutants, or chemicals can lead to an overproduction of reactive species of oxygen (ROS), leading to oxidative stress. To ensure redox homeostasis and prevent damage resulting from the formation of ROS, the skin has an extensive network of antioxidant defense systems, mainly orchestrated by the nuclear factor erythroid-2-related factor 2 (Nrf2) pathway. Indeed, Nrf2 induces the expression of detoxification and antioxidant enzymes and suppresses inductions of pro-inflammatory cytokine genes. In this context, Nrf2 is critical in preserving skin functions such as epidermal differentiation, regulating skin immunity, and managing environmental stresses. Besides, this pathway plays an important role in the pathogenesis of common inflammatory skin diseases such as allergic contact dermatitis, atopic dermatitis, and psoriasis. Therefore, the present review highlights the crucial role of Nrf2 in KCs for maintaining skin homeostasis and regulating skin immunity, as well as its contribution to the pathophysiology of inflammatory skin diseases. Finally, a particular emphasis will be placed on the therapeutic potential of targeting the Nrf2 pathway to alleviate symptoms of these inflammatory skin disorders.
Collapse
Affiliation(s)
- Sara Salman
- Université Paris‐Saclay, INSERM, Inflammation, Microbiome and ImmunosurveillanceOrsayFrance
| | - Virginie Paulet
- Université Paris‐Saclay, INSERM, Inflammation, Microbiome and ImmunosurveillanceOrsayFrance
| | - Kévin Hardonnière
- Université Paris‐Saclay, INSERM, Inflammation, Microbiome and ImmunosurveillanceOrsayFrance
| | - Saadia Kerdine‐Römer
- Université Paris‐Saclay, INSERM, Inflammation, Microbiome and ImmunosurveillanceOrsayFrance
| |
Collapse
|
9
|
Wang W, Zhang L, Liu X, Guo Q, Jiang X, Wu J, Zhu Y, Gu Y, Chen L, Xi K. Punicalagin inhibits neuron ferroptosis and secondary neuroinflammation to promote spinal cord injury recovery. Int Immunopharmacol 2025; 148:114048. [PMID: 39823794 DOI: 10.1016/j.intimp.2025.114048] [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: 11/02/2024] [Revised: 12/27/2024] [Accepted: 01/06/2025] [Indexed: 01/20/2025]
Abstract
Spinal cord injury (SCI) represents a severe type of central nervous system damage, with no effective treatment currently available, partly due to neuronal ferroptosis and subsequent neuroinflammation. Punicalagin, an anti-inflammatory compound extracted from pomegranate peel, has exhibited therapeutic potential for inflammatory diseases. In this study, we present evidence that punicalagin facilitates the recovery of neurological function following SCI by mitigating neuronal ferroptosis. Mechanistically, this effect involves the upregulation of nuclear factor E2-related factor 2 (Nrf2) and the activation of the Nrf2- Solute Carrier Family 7 Member 11 (SLC7A11)- Glutathione Peroxidase 4 (GPX4) signaling pathway. Furthermore, punicalagin aids in the resolution of secondary neuroinflammation by modulating the M1/M2 polarization of microglia, thereby promoting SCI recovery. Collectively, these findings suggest that punicalagin enhances functional recovery after SCI by inhibiting neuronal ferroptosis and reducing microglial M1 polarization. Consequently, punicalagin may represent a promising therapeutic agent for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Wei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006 PR China
| | - Lichen Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006 PR China
| | - Xin Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006 PR China
| | - Qiangqiang Guo
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006 PR China
| | - Xinzhao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006 PR China
| | - Jie Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006 PR China
| | - Yiwei Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006 PR China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006 PR China.
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006 PR China.
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006 PR China.
| |
Collapse
|
10
|
Ma X, Xue S, Ma H, Saeed S, Zhang Y, Meng Y, Chen H, Yu H, Wang H, Hu S, Cai M. Esketamine alleviates LPS-induced depression-like behavior by activating Nrf2-mediated anti-inflammatory response in adolescent mice. Neuroscience 2025; 567:294-307. [PMID: 39755229 DOI: 10.1016/j.neuroscience.2024.12.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/07/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND The mechanisms underlying esketamine's therapeutic effects remain elusive. The study aimed to explore the impact of single esketamine treatment on LPS-induced adolescent depressive-like behaviors and the role of Nrf2 regulated neuroinflammatory response in esketamine-produced rapid antidepressant efficacy. METHODS Adolescent male C57BL/6J mice were randomly assigned to three groups: control, LPS, and LPS + esketamine (15 mg/kg, i.p.). Depressive-like behaviors were evaluated via the OFT, NFST, and TST. Protein expression of Nrf2 and inflammatory cytokines, including TNF-α, IL-1β, and iNOS in the hippocampus and mPFC, were measured by western blot. Moreover, the Nrf2 inhibitor, ML385, was also applied in the current study. The depressive-like behaviors and the protein expression of Nrf2, TNF-α, IL-1β, and iNOS in mPFC and hippocampus were also measured. Additionally, the plasma's pro-inflammatory cytokines and anti-inflammatory cytokines were assessed using ELISA methods with or without ML385. RESULTS A single administration of esketamine treatment alleviated the LPS-induced depressive-like behaviors. Esketamine increased the expression of Nrf2 and reduced the expression of the inflammatory cytokines, including TNF-α, IL-1β, and iNOS, in the mPFC and hippocampus. Notably, pharmacological inhibition of Nrf2 via ML385 administration abrogated the antidepressive-like behaviors and anti-inflammatory effects induced by esketamine. In the periphery, esketamine mitigated the LPS-induced elevation of pro-inflammatory cytokines, and the reduction of anti-inflammatory cytokines, and this effect was reversed by Nrf2 inhibition. CONCLUSION Esketamine treatment exerts rapid antidepressant effects and attenuates neuroinflammation in LPS-induced adolescent depressive-like behaviors, potentially through the activation of Nrf2-mediated anti-inflammatory signaling.
Collapse
Affiliation(s)
- Xinxu Ma
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China; Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shanshan Xue
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hongzhe Ma
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Saboor Saeed
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yunyun Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yumeng Meng
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haixia Chen
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huan Yu
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huaizhi Wang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; The Zhejiang Key Laboratory of Precision Psychiatry, Hangzhou 310003, China; MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310003, China; Brain Research Institute of Zhejiang University, Hangzhou, 310003, China; The State Key Lab of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China; Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou 310003, China.
| | - Min Cai
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
11
|
Rojo AI, Buttari B, Cadenas S, Carlos AR, Cuadrado A, Falcão AS, López MG, Georgiev MI, Grochot-Przeczek A, Gumeni S, Jimenez-Villegas J, Horbanczuk JO, Konu O, Lastres-Becker I, Levonen AL, Maksimova V, Michaeloudes C, Mihaylova LV, Mickael ME, Milisav I, Miova B, Rada P, Santos M, Seabra MC, Strac DS, Tenreiro S, Trougakos IP, Dinkova-Kostova AT. Model organisms for investigating the functional involvement of NRF2 in non-communicable diseases. Redox Biol 2025; 79:103464. [PMID: 39709790 PMCID: PMC11733061 DOI: 10.1016/j.redox.2024.103464] [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: 10/25/2024] [Revised: 11/26/2024] [Accepted: 12/08/2024] [Indexed: 12/24/2024] Open
Abstract
Non-communicable chronic diseases (NCDs) are most commonly characterized by age-related loss of homeostasis and/or by cumulative exposures to environmental factors, which lead to low-grade sustained generation of reactive oxygen species (ROS), chronic inflammation and metabolic imbalance. Nuclear factor erythroid 2-like 2 (NRF2) is a basic leucine-zipper transcription factor that regulates the cellular redox homeostasis. NRF2 controls the expression of more than 250 human genes that share in their regulatory regions a cis-acting enhancer termed the antioxidant response element (ARE). The products of these genes participate in numerous functions including biotransformation and redox homeostasis, lipid and iron metabolism, inflammation, proteostasis, as well as mitochondrial dynamics and energetics. Thus, it is possible that a single pharmacological NRF2 modulator might mitigate the effect of the main hallmarks of NCDs, including oxidative, proteostatic, inflammatory and/or metabolic stress. Research on model organisms has provided tremendous knowledge of the molecular mechanisms by which NRF2 affects NCDs pathogenesis. This review is a comprehensive summary of the most commonly used model organisms of NCDs in which NRF2 has been genetically or pharmacologically modulated, paving the way for drug development to combat NCDs. We discuss the validity and use of these models and identify future challenges.
Collapse
Affiliation(s)
- Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain.
| | - Brigitta Buttari
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161, Rome, Italy
| | - Susana Cadenas
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), Cantoblanco, Madrid, Spain
| | - Ana Rita Carlos
- CE3C-CHANGE, Department of Animal Biology, Faculty of Sciences, University of Lisbon, 1749-016, Lisbon, Portugal
| | - Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Ana Sofia Falcão
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Hospital Universitario de la Princesa, Madrid, Spain
| | - Milen I Georgiev
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000, Plovdiv, Bulgaria
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - José Jimenez-Villegas
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Jarosław Olav Horbanczuk
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, 36A Postępu, Jastrzębiec, 05-552, Poland
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; Department of Neuroscience, Bilkent University, Ankara, Turkey; UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Isabel Lastres-Becker
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute Teófilo Hernando for Drug Discovery, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Viktorija Maksimova
- Department of Applied Pharmacy, Division of Pharmacy, Faculty of Medical Sciences, Goce Delcev University, Stip, Krste Misirkov Str., No. 10-A, P.O. Box 201, 2000, Stip, Macedonia
| | | | - Liliya V Mihaylova
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000, Plovdiv, Bulgaria
| | - Michel Edwar Mickael
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, 36A Postępu, Jastrzębiec, 05-552, Poland
| | - Irina Milisav
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, 1000, Ljubljana, Slovenia; Laboratory of oxidative stress research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, 1000, Ljubljana, Slovenia
| | - Biljana Miova
- Department of Experimental Physiology and Biochemistry, Institute of Biology, Faculty of Natural Sciences and Mathematics, University "St Cyril and Methodius", Skopje, Macedonia
| | - Patricia Rada
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Marlene Santos
- REQUIMTE/LAQV, Escola Superior de Saúde (E2S), Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 400, 4200-072, Porto, Portugal; Molecular Oncology & Viral Pathology, IPO-Porto Research Center (CI-IPOP), Portuguese Institute of Oncology, 4200-072, Porto, Portugal
| | - Miguel C Seabra
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, 10 000, Zagreb, Croatia
| | - Sandra Tenreiro
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, UK; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Tork MAB, Fotouhi S, Roozi P, Negah SS. Targeting NLRP3 Inflammasomes: A Trojan Horse Strategy for Intervention in Neurological Disorders. Mol Neurobiol 2025; 62:1840-1881. [PMID: 39042218 DOI: 10.1007/s12035-024-04359-2] [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: 02/05/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Recently, a growing focus has been on identifying critical mechanisms in neurological diseases that trigger a cascade of events, making it easier to target them effectively. One such mechanism is the inflammasome, an essential component of the immune response system that plays a crucial role in disease progression. The NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3) inflammasome is a subcellular multiprotein complex that is widely expressed in the central nervous system (CNS) and can be activated by a variety of external and internal stimuli. When activated, the NLRP3 inflammasome triggers the production of proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) and facilitates rapid cell death by assembling the inflammasome. These cytokines initiate inflammatory responses through various downstream signaling pathways, leading to damage to neurons. Therefore, the NLRP3 inflammasome is considered a significant contributor to the development of neuroinflammation. To counter the damage caused by NLRP3 inflammasome activation, researchers have investigated various interventions such as small molecules, antibodies, and cellular and gene therapy to regulate inflammasome activity. For instance, recent studies indicate that substances like micro-RNAs (e.g., miR-29c and mR-190) and drugs such as melatonin can reduce neuronal damage and suppress neuroinflammation through NLRP3. Furthermore, the transplantation of bone marrow mesenchymal stem cells resulted in a significant reduction in the levels of pyroptosis-related proteins NLRP3, caspase-1, IL-1β, and IL-18. However, it would benefit future research to have an in-depth review of the pharmacological and biological interventions targeting inflammasome activity. Therefore, our review of current evidence demonstrates that targeting NLRP3 inflammasomes could be a pivotal approach for intervention in neurological disorders.
Collapse
Affiliation(s)
- Mohammad Amin Bayat Tork
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soroush Fotouhi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Roozi
- Department of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajad Sahab Negah
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran.
| |
Collapse
|
13
|
Salah TM, Rabie MA, El Sayed NS. Renoprotective effect of berberine in cisplatin-induced acute kidney injury: Role of Klotho and the AMPK/mtor/ULK1/Beclin-1 pathway. Food Chem Toxicol 2025; 196:115179. [PMID: 39645019 DOI: 10.1016/j.fct.2024.115179] [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: 10/13/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Cisplatin (Cisp) is a potent cancer drug, but its use is limited by acute kidney injury (AKI). Autophagy, a process that removes damaged proteins and maintains cellular homeostasis, has been shown to alleviate Cisp-induced AKI. The balance between autophagy and apoptosis is crucial to kidney protection. Treatment with Berberine, known for its antioxidant and anti-inflammatory effects in nephrotoxicity models, was studied for its potential to enhance autophagy in Cisp-induced AKI. Treatment with Berberine (Berb) upregulated Klotho gene expression, enhancing autophagy as indicated by elevated protein levels of pS486-AMPK, pS638-ULK1, and Beclin-1, accompanied by a decrease in pS248-mTOR protein expression. Also, Berb mitigated oxidative stress by reducing elevated MDA levels and boosting SOD activity, which in turn suppressed inflammation by down-regulating HMGB1 and RAGE gene expression, as well as reducing pS536-NF-κB and IL-6 protein contents. Additionally, Berb reduced apoptosis by increasing Bcl-2 and decreasing Bax. This coordinated action preserved kidney function, evidenced by reductions in early injury markers (cystatin C, KIM-1, NGAL) and late markers (creatinine, BUN), along with attenuation of histopathological alterations. The use 3-MA, autophagy inhibitor, nullified these protective effects, highlighting Berb's role in promoting autophagy, reducing oxidative stress, inflammation, and apoptosis, and preserving renal health in Cisp-induced AKI.
Collapse
Affiliation(s)
- Tasneem M Salah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| |
Collapse
|
14
|
Buttari B, Tramutola A, Rojo AI, Chondrogianni N, Saha S, Berry A, Giona L, Miranda JP, Profumo E, Davinelli S, Daiber A, Cuadrado A, Di Domenico F. Proteostasis Decline and Redox Imbalance in Age-Related Diseases: The Therapeutic Potential of NRF2. Biomolecules 2025; 15:113. [PMID: 39858508 PMCID: PMC11764413 DOI: 10.3390/biom15010113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/20/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a master regulator of cellular homeostasis, overseeing the expression of a wide array of genes involved in cytoprotective processes such as antioxidant and proteostasis control, mitochondrial function, inflammation, and the metabolism of lipids and glucose. The accumulation of misfolded proteins triggers the release, stabilization, and nuclear translocation of NRF2, which in turn enhances the expression of critical components of both the proteasomal and lysosomal degradation pathways. This process facilitates the clearance of toxic protein aggregates, thereby actively maintaining cellular proteostasis. As we age, the efficiency of the NRF2 pathway declines due to several factors including increased activity of its repressors, impaired NRF2-mediated antioxidant and cytoprotective gene expression, and potential epigenetic changes, though the precise mechanisms remain unclear. This leads to diminished antioxidant defenses, increased oxidative damage, and exacerbated metabolic dysregulation and inflammation-key contributors to age-related diseases. Given NRF2's role in mitigating proteotoxic stress, the pharmacological modulation of NRF2 has emerged as a promising therapeutic strategy, even in aged preclinical models. By inducing NRF2, it is possible to mitigate the damaging effects of oxidative stress, metabolic dysfunction, and inflammation, thus reducing protein misfolding. The review highlights NRF2's therapeutic implications for neurodegenerative diseases and cardiovascular conditions, emphasizing its role in improving proteostasis and redox homeostasis Additionally, it summarizes current research into NRF2 as a therapeutic target, offering hope for innovative treatments to counteract the effects of aging and associated diseases.
Collapse
Affiliation(s)
- Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.B.); (E.P.)
| | - Antonella Tramutola
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University, 00185 Rome, Italy;
| | - Ana I. Rojo
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), National Institute of Health Carlos III (ISCIII), Instituto de Investigación Sanitaria La Paz (IdiPaz), 28049 Madrid, Spain; (A.I.R.); (A.C.)
| | - Niki Chondrogianni
- Institute of Chemical Biology, National Hellenic Research Foundation, 116 35 Athens, Greece;
| | - Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura 00185, Uttar Pradesh, India;
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (L.G.)
| | - Letizia Giona
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (L.G.)
- PhD Program in Science of Nutrition, Metabolism, Aging and Gender-Related Diseases, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Joana P. Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.B.); (E.P.)
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Andreas Daiber
- Department for Cardiology 1, University Medical Center Mainz, Molecular Cardiology, Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Antonio Cuadrado
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), National Institute of Health Carlos III (ISCIII), Instituto de Investigación Sanitaria La Paz (IdiPaz), 28049 Madrid, Spain; (A.I.R.); (A.C.)
| | - Fabio Di Domenico
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University, 00185 Rome, Italy;
| |
Collapse
|
15
|
Sarkar S, Pandey A, Kumar Yadav S, Haris Siddiqui M, Pant AB, Yadav S. Differentiated and mature neurons are more responsive to neurotoxicant exposure at both transcriptional and translational levels. Neuroscience 2025; 564:110-125. [PMID: 39571964 DOI: 10.1016/j.neuroscience.2024.11.017] [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: 07/16/2024] [Revised: 10/13/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024]
Abstract
SH-SY5Y human neuroblastoma cells have been extensively used as an in vitro model system in a diverse range of studies involving neurodevelopment, neurotoxicity, neurodegeneration, and neuronal ageing. Both naïve and differentiated phenotypes of SH-SY5Y cells are utilized to model human neurons under in vitro conditions. The process of differentiation causes extensive remodeling of neuronal cells at multiple omic levels, including the epigenome and proteome. In the present investigation, the miRNAome and proteome profiles of arsenic-treated naïve and differentiated SH-SY5Y cells were generated using the miRNA OpenArray technology and high-resolution mass spectrometry. Our findings demonstrated that differentiation dramatically affected the response of SH-SY5Y cells to toxicant exposure, as indicated by increased tolerance of differentiated cells against arsenic exposure compared to naïve cells in cell viability assay. Arsenic-exposed naïve and differentiated SH-SY5Y cells possess distinct miRNA and protein profiles with few similarities. Compared to naïve cells, differentiated cells have undergone higher deregulation in the expression of brain-enriched miRNAs and proteins and have shown a more drastic decrease in oxygen consumption rate, which is a measure of mitochondrial respiration after exposure to arsenic. Proteins identified in arsenic-treated differentiated SH-SY5Y cells were more enriched in pathways underlying multifactorial neurotoxic events. Additionally, more functional regulatory modules have been identified between the miRNAs and proteins differentially expressed in arsenic-treated differentiated SH-SY5Y cells relative to naïve cells. Collectively, our studies have shown that differentiated SH-SY5Y cells displayed alterations in the expression of a greater number of miRNAs and proteins following neurotoxicant exposure, indicating their higher responsivity.
Collapse
Affiliation(s)
- Sana Sarkar
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India; Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, India.
| | - Anuj Pandey
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India.
| | - Sanjeev Kumar Yadav
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India.
| | | | - A B Pant
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India.
| | - Sanjay Yadav
- All India Institute of Medical Sciences (AIIMS), Raebareli, Uttar Pradesh, India.
| |
Collapse
|
16
|
Sivasuriyan KS, Namasivayam SKR, Pandian A. Molecular insights into the anti-cancer activity of chitosan-okra mucilage polymeric nanocomposite doped with nano zero-valent iron against multi-drug-resistant oral carcinoma cells. Int J Biol Macromol 2025; 286:138495. [PMID: 39644860 DOI: 10.1016/j.ijbiomac.2024.138495] [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: 08/16/2024] [Revised: 11/12/2024] [Accepted: 12/05/2024] [Indexed: 12/09/2024]
Abstract
Recent advances in nanotechnology, particularly those utilizing polymeric nanocomposites, have garnered significant attention for their effectiveness and biocompatibility in cancer diagnosis and treatment. In this study, a chitosan-okra mucilage polymeric nanocomposite doped with nano zero-valent iron (CS-OM-nZVI), synthesized using green chemistry principles, was evaluated for its anti-cancer activity against drug-resistant oral carcinoma cells (KBChR). The nanocomposite was created from chitosan, mucilage derived from okra biomass, and nano zerovalent iron particles synthesized through chemical reduction. The resulting nanocomposite exhibited a highly stable, crystalline nanoscale structure with excellent stability. Anti-cancer activity was assessed by measuring cell viability, apoptosis induction, oxidative stress markers, DNA fragmentation, and performing in silico docking studies between the components of the polymeric nanocomposite (CS-OM-nZVI) and key proteins involved in carcinoma pathogenesis. The nanocomposite demonstrated significant anticancer activity, with an IC50 of 600 μg/mL, indicating notable effects on cell viability. It also induced significant morphological changes associated with apoptosis, such as chromatin condensation and nuclear fragmentation. Additionally, the nanocomposite had a marked effect on oxidative stress markers, particularly catalase and superoxide dismutase activity. In silico docking studies revealed that the polymeric composite modulates and enhances both intrinsic and extrinsic apoptotic pathways, confirmed by chitosan's binding to Caspase-3. This study suggests that the prepared nanocomposite is a promising anti-cancer agent against drug-resistant oral carcinoma cells, demonstrating a significant impact on cancer cell viability.
Collapse
Affiliation(s)
- Krithika Shree Sivasuriyan
- Centre for Applied Research, Saveetha School of Engineering, Saveetha institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 602105, India
| | - S Karthick Raja Namasivayam
- Centre for Applied Research, Saveetha School of Engineering, Saveetha institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 602105, India.
| | - Arjun Pandian
- Centre for Applied Research, Saveetha School of Engineering, Saveetha institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 602105, India
| |
Collapse
|
17
|
Sharma V, Sharma P, Singh TG. Emerging role of Nrf2 in Parkinson's disease therapy: a critical reassessment. Metab Brain Dis 2024; 40:70. [PMID: 39699763 DOI: 10.1007/s11011-024-01452-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 09/30/2024] [Indexed: 12/20/2024]
Abstract
Parkinson's disease (PD) is the neurodegenerative disorder characterized by the progressive degeneration of nigrostriatal dopaminergic neurons, leading to the range of motor and non-motor symptoms. There is mounting evidence suggesting that oxidative stress, neuroinflammation and mitochondrial dysfunction play pivotal roles in the pathogenesis of PD. Current therapies only alleviate perturbed motor symptoms. Therefore, it is essential to find out new therapies that allow us to improve not only motor symptoms, but non-motor symptoms like cognitive impairment and modulate disease progression. Nuclear factor erythroid 2-related factor 2 (Nrf2) is transcription factor that regulates the expression of numerous anti-oxidants and cytoprotective genes can counteract oxidative stress, neuroinflammation and mitochondrial dysfunction, thereby potentially ameliorating PD-associated pathology. The current review discusses about the Nrf2 structure and function with special emphasis on various molecular signalling pathways involved in positive and negative modulation of Nrf2, namely Glycogen synthase kinase-3β, Phosphoinositide-3-kinase, AMP-activated protein kinase, Mitogen activated protein kinase, nuclear factor-κB and P62. Furthermore, this review highlights the various Nrf2 activators as promising therapeutic agents for slowing down the progression of PD.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Prateek Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
18
|
Lv B, Xing S, Wang Z, Zhang A, Wang Q, Bian Y, Pei Y, Sun H, Chen Y. NRF2 inhibitors: Recent progress, future design and therapeutic potential. Eur J Med Chem 2024; 279:116822. [PMID: 39241669 DOI: 10.1016/j.ejmech.2024.116822] [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: 08/05/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a crucial transcription factor involved in oxidative stress response, which controls the expression of various cytoprotective genes. Recent research has indicated that constitutively activated NRF2 can enhance patients' resistance to chemotherapy drugs, resulting in unfavorable prognosis. Therefore, the development of NRF2 inhibitors has emerged as a promising approach for overcoming drug resistance in cancer treatment. However, there are limited reports and reviews focusing on NRF2 inhibitors. This review aims to provide a comprehensive analysis of the structure and regulation of the NRF2 signaling pathway, followed by a comprehensive review of reported NRF2 inhibitors. Moreover, the current design strategies and future prospects of NRF2 inhibitors will be discussed, aiming to establish a foundation for the development of more effective NRF2 inhibitors.
Collapse
Affiliation(s)
- Bingbing Lv
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Zhiqiang Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Ao Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Qinjie Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yaoyao Bian
- Jiangsu Provincial Engineering Center of TCM External Medication Researching and Industrializing, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yuqiong Pei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
19
|
Luo G, Kumar H, Aldridge K, Rieger S, Han E, Jiang E, Chan ER, Soliman A, Mahdi H, Letterio JJ. A Core NRF2 Gene Set Defined Through Comprehensive Transcriptomic Analysis Predicts Selective Drug Resistance and Poor Multicancer Prognosis. Antioxid Redox Signal 2024; 41:1031-1050. [PMID: 39028025 DOI: 10.1089/ars.2023.0409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Aims: The nuclear factor erythroid 2-related factor 2-Kelch-like ECH-associated protein 1 (NRF2-KEAP1) pathway plays an important role in the cellular response to oxidative stress but may also contribute to metabolic changes and drug resistance in cancer. However, despite its pervasiveness and important role, most of nuclear factor erythroid 2-related factor 2 (NRF2) target genes are defined in context-specific experiments and analysis, making it difficult to translate from one situation to another. Our study investigates whether a core NRF2 gene signature can be derived and used to represent NRF2 activation in various contexts, allowing better reproducibility and understanding of NRF2. Results: We define a core set of 14 upregulated NRF2 target genes from 7 RNA-sequencing datasets that we generated and analyzed. This NRF2 gene signature was validated using analyses of published datasets and gene sets. An NRF2 activity score based on expression of these core target genes correlates with resistance to drugs such as PX-12 and necrosulfonamide but not to paclitaxel or bardoxolone methyl. We validated these findings in our Kelch-like ECH-associated protein 1 (KEAP1) knockout cancer cell lines. Finally, our NRF2 score is prognostic for cancer survival and validated in additional independent cohorts for lung adenocarcinoma and also novel cancer types not associated with NRF2-KEAP1 mutations such as clear cell renal carcinoma, hepatocellular carcinoma, and acute myeloid leukemia. Innovation and Conclusions: These analyses define a core NRF2 gene signature that is robust, versatile, and useful for evaluating NRF2 activity and for predicting drug resistance and cancer prognosis. Using this gene signature, we uncovered novel selective drug resistance and cancer prognosis associated with NRF2 activation. Antioxid. Redox Signal. 41, 1031-1050.
Collapse
Affiliation(s)
- George Luo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Harshita Kumar
- Department of Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Stevie Rieger
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - EunHyang Han
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Ethan Jiang
- Booth School of Business, University of Chicago, Chicago, Illinois, USA
| | - Ernest R Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ahmed Soliman
- Department of Pediatrics, SUNY Downstate Hospital, Brooklyn, New York, USA
| | - Haider Mahdi
- Magee Women's Research Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Magee Women's Hospital, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John J Letterio
- The Angie Fowler Adolescent and Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, Ohio, USA
- The Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
20
|
Sanchez-Guzman X, Alvarez-Domínguez L, Ramírez-Torres MF, Montes-Alvarado JB, Garcia-Ibañez P, Moreno DA, Domínguez F, Maycotte P. Cruciferous Plant Extracts, Their Isothyocianate or Indol Derivatives, and Their Effect on Cellular Viability of Breast Cancer Cell Lines. J Med Food 2024; 27:1183-1192. [PMID: 39382485 DOI: 10.1089/jmf.2023.0199] [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] [Indexed: 10/10/2024] Open
Abstract
Brassicaceaes are rich in glucosinolates (GSL), whose derivatives, the isothyocianates sulforaphane (SFN), iberine (IB), or indole derivatives as indole-3-carbinol (I3C), have anticancer activities. We evaluated the effects of a broccoli sprout (Brassica oleracea var italica) and red cabbage (B. oleracea L. var capitata f. rubra) extracts and their GSL derivatives on breast cancer cells. Broccoli sprout aqueous extract (BSE) and red cabbage aqueous (RCA) or ethanolic (RCE) extracts were high in SFN, IB, and/or I3C. BSE and RCA decreased proliferation at doses of 2.5-5 mg/mL but induced proliferation at lower doses. RCE decreased proliferation starting at 10 µg/mL with selectivity toward cancer cells. SFN, IB, or I3C alone or in combination did not decrease proliferation similarly, suggesting synergistic effects with other phytochemicals in the extract. RCE showed selectivity toward breast cancer cells, but the effect of the individual metabolites or their combination did not reduce proliferation to the same extent. It will be important to determine the combination responsible for this effect to characterize their use for breast cancer treatment.
Collapse
Affiliation(s)
- Xochiquetzal Sanchez-Guzman
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, OOAD, Puebla, Mexico
- Universidad Politécnica Metropolitana de Puebla, Puebla, Mexico
| | - Lilian Alvarez-Domínguez
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, OOAD, Puebla, Mexico
- Facultad de Biología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - María Fernanda Ramírez-Torres
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, OOAD, Puebla, Mexico
- Universidad Politécnica de Puebla, Puebla, Mexico
| | | | | | - Diego A Moreno
- Phytochemistry and Healthy Food Lab (LabFAS), CEBAS, CSIC, Murcia, Spain
| | - Fabiola Domínguez
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, OOAD, Puebla, Mexico
| | - Paola Maycotte
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, OOAD, Puebla, Mexico
| |
Collapse
|
21
|
Meyer F, Bitsch A, Forman HJ, Fragoulis A, Ghezzi P, Henschenmacher B, Kellner R, Kuhne J, Ludwig T, Sachno D, Schmid G, Tsaioun K, Verbeek J, Wright R. The effects of radiofrequency electromagnetic field exposure on biomarkers of oxidative stress in vivo and in vitro: A systematic review of experimental studies. ENVIRONMENT INTERNATIONAL 2024; 194:108940. [PMID: 39566441 DOI: 10.1016/j.envint.2024.108940] [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: 12/14/2023] [Revised: 07/25/2024] [Accepted: 08/04/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Oxidative stress is thought to be related to many diseases. Furthermore, it is hypothesized that radiofrequency electromagnetic fields (RF-EMF) may induce excessive oxidative stress in various cell types and thereby have the potential to compromise human and animal health. The objective of this systematic review (SR) is to summarize and evaluate the literature on the relation between the exposure to RF-EMF in the frequency range from 100 kHz to 300 GHz and biomarkers of oxidative stress. METHODS The SR framework was developed following the guidelines established in the WHO Handbook for Guideline Development and NTP/OHAT's Handbook for Conducting a Literature-Based Health Assessment. We used the latter handbook's methodology for implementing the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach for environmental health assessments. We searched the following databases up until June 30, 2023: PubMed, Embase, Web of Science Core Collection, Scopus, and the EMF-Portal. The reference lists of included studies and retrieved review articles were also manually searched. We rated Risk of Bias (RoB) using the OHAT RoB Rating Tool and assessed publication bias using funnel plots of included studies. We assessed the certainty of the evidence (high, moderate, low, or very low) for an association between RF-EMF and oxidative stress using an adapted version of the GRADE framework. Data were extracted according to a predefined set of forms developed in DistillerSR. Data were analysed after grouping them first as in vitro or in vivo and then according to outcome category, species category, and exposed tissue. We synthesized study results using a random effects meta-analysis when study characteristics were judged sufficiently similar to be combined and heterogeneity (I2) was lower than 75 %, otherwise we describe the findings narratively. RESULTS Fifty-six (56) studies, 45 in vivo and 11 in vitro, in which cells (in vitro) or animals (in vivo) were exposed to frequencies in the range 800-2450 MHz, were included in the systematic review after eliminating 12,353 publications because they did not meet the criteria defined in the published protocol (Henschenmacher et al., 2022). Of 56 studies 52 studies with 169 individual results were included in the meta-analysis. Together, these studies examined six human in vitro samples and fifty animal samples, including rodents (mice, rats, hamsters, and guinea pigs, (n = 46)) and rabbits (n = 4). RF-EMF were predominantly applied as continuous wave exposures in these studies. The outcome biomarkers for modified proteins and amino acids were measured in n = 30 studies, for oxidized DNA bases in n = 26 studies, for oxidized lipids in n = 3 studies and hydrogen peroxide production in 2 studies. Outcomes were mostly measured in the brain (n = 22), liver (n = 9), cells (n = 9), blood (n = 6), and testis (n = 2). RoB in studies was high, mainly due to biases in exposure and outcome assessment. IN VIVO STUDIES Brain: The effect on biomarkers for oxidized DNA bases in the rodent brain (five studies, n = 98) had an inconsistent effect, varying from a large decrease with a standardized mean difference (SMD) of -3.40 (95 % CI [-5.15, -1.64]) to a large increase with an SMD of 2.2 (95 % CI [0.78, 3.62]). In the brain of rabbits (two studies, n = 44), the effect sizes also varied, from an SMD of -1.06 (95 % CI [-2.13, 0.00]) to an SMD of 5.94 (95 % CI [3.14, 8.73]). The effect on biomarkers for modified proteins and amino acids in the rodent brain (15 studies, n = 328) also varied from a large decrease with an SMD of -6.11 (95 % CI [-8.16, -4.06]) to a large increase with an SMD of 5.33 (95 % CI [2.49, 8.17]). The effect on biomarkers for oxidized lipids in the brain of rodents (one study, n = 56) also varied from a large decrease with SMD = -4.10 (95 % CI [-5.48, -2.73]) to SMD = 1.27 (95 % CI [0.45, 2.10]). Liver: The effect on biomarkers for oxidized DNA bases in the rodent liver (two studies, n = 26) was inconsistent with effect sizes in both directions: SMD = -0.71 (95 % CI [-1.80, 0.38]) and SMD = 1.56 (95 % CI [0.19, 2.92]). The effect on biomarkers for oxidized DNA bases in the rabbits' liver (two studies, n = 60) was medium with a pooled SMD of 0.39 (95 % CI [-0.79, 1.56]). Biomarkers for modified proteins and amino acids in the liver of rodents (six studies, n = 159) increased with a pooled SMD of 0.55 (95 % CI [0.06, 1.05]). Blood: The effect of RF-EMF on biomarkers for oxidized DNA bases in rodent blood (four studies, n = 104) was inconsistent, with SMDs ranging from -1.14 (95 % CI [-2.23, -0.06]) to 1.71 (95 % CI [-0.10, 3.53]). RF-EMF had no effect on biomarkers for modified proteins and amino acids in rodent blood (three studies, n = 40), with a pooled SMD of -0.08 (95 % CI [-1.32, 1.16]). There was a large increase in biomarkers for oxidized DNA bases in rodent plasma (two studies, n = 38) with a pooled SMD of 2.25 (95 % CI [1.27, 3.24]). Gonads: There was an increase in biomarkers for oxidized DNA bases in the rodent testis (two studies, n = 24) with a pooled SMD of 1.60 (95 % CI [0.62, 2.59]). The effect of RF-EMF on biomarkers for modified proteins and amino acids in the ovary of rodents (two studies, n = 52) was inconsistent with a medium effect, SMD = 0.24 (95 % CI [-0.74, 1.23])) and a large effect (SMD = 2.08 (95 % CI [1.22, 2.94])). Thymus: RF-EMF increased biomarkers for modified proteins and amino acids in the thymus of rodents (one study, n = 42) considerably with a pooled SMD of 6.16 (95 % CI [3.55, 8.76]). Cells: RF-EMF increased oxidized DNA bases in rodent cells with SMD of 2.49 (95 % CI [1.30, 3.67]) (one study, n = 27). There was a medium effect in oxidized lipids (one study, n = 18) but not statistically significant with SMD = 0.34 (95 % CI [-0.62, 1.29]). IN VITRO STUDIES In in vitro studies in human cells (three studies, n = 110), there were inconsistent increases in biomarkers for oxidized DNA bases, where the SMDs varied between 0.01 (95 % CI [-0.59, 0.62]) and 7.12 (95% CI [0.06, 14.18]) in 4 results (2 of them statistically significant). In rodent cells (three studies, n = 24), there was a not statistically significant large effect in biomarkers for oxidized DNA bases with SMD = 2.07 (95 % CI [-1.38, 5.52]). The RF-EMF biomarkers for modified proteins and amino acids in human cells (one study, n = 18) showed a large effect with SMD = 1.07 (95 % CI [-0.05, 2.19]). In rodent cells (two studies, n = 24) a medium effect of SMD = 0.56 (95 % CI [-0.29, 1.41]) was observed. DISCUSSION The evidence on the relation between the exposure to RF-EMF and biomarkers of oxidative stress was of very low certainty, because a majority of the included studies were rated with a high RoB level and provided high heterogeneity. This is due to inaccurate measurements of exposure and/or of measurement of oxidative stress biomarkers and missing information on the blinding of research personnel to exposure conditions or outcome measurements. There may be no or an inconsistent effect of RF-EMF on biomarkers of oxidative stress in the brain, liver, blood, plasma and serum, and in the female reproductive system in animal experiments but the evidence is of very low certainty. There may be an increase in biomarkers of oxidative stress in testes, serum and thymus of rodents but the evidence is of very low certainty. Future studies should improve experimental designs and characterization of exposure systems as well as the use of validated biomarker measurements with positive controls. Other: This review was partially funded by the World Health Organization. The protocol for this review is registered in PROSPERO (https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021235573) and published in Environment International (https://doi.org/10.1016/j.envint.2021.106932) (Henschenmacher et al., 2022).
Collapse
Affiliation(s)
- Felix Meyer
- Federal Office for Radiation Protection, Competence Centre EMF, Karl-Liebknecht-Strasse 33, 03046 Cottbus, Germany.
| | - Annette Bitsch
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - Henry Jay Forman
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089, USA; University of California Merced, 5200 Lake Road, Merced, CA 95343, USA
| | - Athanassios Fragoulis
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - Pietro Ghezzi
- Brighton and Sussex Medical School, University of Sussex, Trafford Centre, Falmer BN1 9RY, United Kingdom; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Bernd Henschenmacher
- Federal Office for Radiation Protection, Ingolstädter Landstrasse 1, 85764 Oberschleißheim, Germany
| | - Rupert Kellner
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - Jens Kuhne
- Federal Office for Radiation Protection, Competence Centre EMF, Karl-Liebknecht-Strasse 33, 03046 Cottbus, Germany
| | - Tonia Ludwig
- Federal Office for Radiation Protection, Ingolstädter Landstrasse 1, 85764 Oberschleißheim, Germany
| | - Dmitrij Sachno
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - Gernot Schmid
- Seibersdorf Laboratories, Campus Seibersdorf, 2444 Seibersdorf, Austria
| | - Katya Tsaioun
- Evidence-based Toxicology Collaboration (EBTC), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Jos Verbeek
- University Medical Center Amsterdam, Cochrane Work, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Robert Wright
- Welch Medical Library, Johns Hopkins University School of Medicine, 1900 E. Monument Street, Baltimore, MD 21205, USA
| |
Collapse
|
22
|
Rosina M, Veltri F, Nesci V, Bissacco J, Bovenzi R, Mascioli D, Simonetta C, Zenuni H, Maftei D, Marano M, Pierantozzi M, Stefani A, Chiurchiù V, Longone P, Valle C, Mercuri NB, Ferri A, Schirinzi T. Immunometabolic Signature and Tauopathy Markers in Blood Cells of Progressive Supranuclear Palsy. Mov Disord 2024; 39:2211-2219. [PMID: 39283273 DOI: 10.1002/mds.30009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/30/2024] [Accepted: 08/20/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Peripheral immune cells critically contribute to the clinical-pathological progression of neurodegenerative diseases and also represent a reliable frame for translational applications. However, data on progressive supranuclear palsy (PSP) are almost scarce in this regard. OBJECTIVE Our goal is to provide a broad biological characterization of peripheral immune cells in a selected PSP cohort. METHODS Seventy-one PSP patients scored on the PSP Rating Scale (PSPRS), and 59 controls were enrolled. The blood cell count was collected, together with the neutrophil-to-lymphocyte ratio (NLR) calculation. In a subgroup of patients and controls, the peripheral blood mononuclear cells (PBMCs) were analyzed by the mitochondrial bioenergetic performance and the western blot assay of the nuclear factor erythroid 2-related factor (NRF2)/heme oxygenase 1 (HO-1) pathway and the total tau (t-tau) and phosphorylated tau (p-tau) proteins. Case-control comparison and correlation analyses were performed. RESULTS PSP patients had a NLR higher than controls, with increased circulating neutrophils. The leukocyte metabolism was also globally increased and the NRF2/HO-1 pathway activated in patients. P-tau, but not t-tau, significantly accumulated in PSP PBMCs and inversely correlated with the PSPRS. CONCLUSIONS PSP displays a systemic inflammatory shift of the peripheral immunity, which may justify a metabolic reprogramming of the blood leukocytes. Consistently, the NRF2/HO-1 pathway, a master regulator of inflammatory and metabolic response, was activated. PBMCs also engulf tau proteins, especially p-tau, in a way inverse to the disease severity, allowing for a peripheral tracking of tauopathy in patients. Immunometabolic targets may, therefore, gain relevance to PSP in biomarker or therapeutic purposes. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Marco Rosina
- Neurology Unit, Fondazione PTV - Tor Vergata University Hospital, Rome, Italy
- IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Federica Veltri
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Valentina Nesci
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Jacopo Bissacco
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Roberta Bovenzi
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Davide Mascioli
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Clara Simonetta
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Henri Zenuni
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Daniela Maftei
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Massimo Marano
- Unit of Neurology, Neurophysiology, Neurobiology and Psychiatry, Campus Bio-Medico University of Rome, Rome, Italy
- Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Mariangela Pierantozzi
- Neurology Unit, Fondazione PTV - Tor Vergata University Hospital, Rome, Italy
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Alessandro Stefani
- Neurology Unit, Fondazione PTV - Tor Vergata University Hospital, Rome, Italy
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Valerio Chiurchiù
- National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | | | - Cristiana Valle
- IRCCS Fondazione Santa Lucia, Rome, Italy
- National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, Fondazione PTV - Tor Vergata University Hospital, Rome, Italy
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Alberto Ferri
- IRCCS Fondazione Santa Lucia, Rome, Italy
- National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Tommaso Schirinzi
- Neurology Unit, Fondazione PTV - Tor Vergata University Hospital, Rome, Italy
- Neurology Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| |
Collapse
|
23
|
Cazalla E, Cuadrado A, García-Yagüe ÁJ. Role of the transcription factor NRF2 in maintaining the integrity of the Blood-Brain Barrier. Fluids Barriers CNS 2024; 21:93. [PMID: 39574123 PMCID: PMC11580557 DOI: 10.1186/s12987-024-00599-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND The Blood-Brain Barrier (BBB) is a complex and dynamic interface that regulates the exchange of molecules and cells between the blood and the central nervous system. It undergoes structural and functional throughout oxidative stress and inflammation, which may compromise its integrity and contribute to the pathogenesis of neurodegenerative diseases. MAIN BODY Maintaining BBB integrity is of utmost importance in preventing a wide range of neurological disorders. NRF2 is the main transcription factor that regulates cellular redox balance and inflammation-related gene expression. It has also demonstrated a potential role in regulating tight junction integrity and contributing to the inhibition of ECM remodeling, by reducing the expression of several metalloprotease family members involved in maintaining BBB function. Overall, we review current insights on the role of NRF2 in addressing protection against the effects of BBB dysfunction, discuss its involvement in BBB maintenance in different neuropathological diseases, as well as, some of its potential activators that have been used in vitro and in vivo animal models for preventing barrier dysfunction. CONCLUSIONS Thus, emerging evidence suggests that upregulation of NRF2 and its target genes could suppress oxidative stress, and neuroinflammation, restore BBB integrity, and increase its protection.
Collapse
Affiliation(s)
- Eduardo Cazalla
- Department of Biochemistry, School of Medicine, Autonomous University of Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Sols-Morreale" (CSIC-UAM), C/ Arturo Duperier, 4, Madrid, 28029, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Antonio Cuadrado
- Department of Biochemistry, School of Medicine, Autonomous University of Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Sols-Morreale" (CSIC-UAM), C/ Arturo Duperier, 4, Madrid, 28029, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ángel Juan García-Yagüe
- Department of Biochemistry, School of Medicine, Autonomous University of Madrid (UAM), Madrid, Spain.
- Instituto de Investigaciones Biomédicas "Sols-Morreale" (CSIC-UAM), C/ Arturo Duperier, 4, Madrid, 28029, Spain.
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
24
|
Wang T, Liu M, Li X, Zhang S, Gu H, Wei X, Wang X, Xu Z, Shen T. Naturally-derived modulators of the Nrf2 pathway and their roles in the intervention of diseases. Free Radic Biol Med 2024; 225:560-580. [PMID: 39368519 DOI: 10.1016/j.freeradbiomed.2024.09.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024]
Abstract
Cumulative evidence has verified that persistent oxidative stress is involved in the development of various chronic diseases, including pulmonary, neurodegenerative, kidney, cardiovascular, and liver diseases, as well as cancers. Nuclear factor erythroid 2-related factor 2 (Nrf2) plays a pivotal role in regulating cellular oxidative stress and inflammatory reactions, making it a focal point for disease prevention and treatment strategies. Natural products are essential resources for discovering leading molecules for new drug research and development. In this review, we comprehensively outlined the progression of the knowledge on the Nrf2 pathway, Nrf2 activators in clinical trials, the naturally-derived Nrf2 modulators (particularly from 2014-present), as well as their effects on the pathogenesis of chronic diseases.
Collapse
Affiliation(s)
- Tian Wang
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Mingjie Liu
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xinyu Li
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Sen Zhang
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Haoran Gu
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xuan Wei
- Shandong Center for Food and Drug Evaluation and Inspection, Jinan, Shandong, PR China
| | - Xiaoning Wang
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Zhenpeng Xu
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China.
| | - Tao Shen
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China.
| |
Collapse
|
25
|
Angelis NV, Paronis E, Sarikaki G, Kyriakopoulos A, Agapaki A, Niotopoulou PM, Knai CC, Alexakos P, Liagkas O, Mavreas KF, Baxevanis CN, Skaltsounis AL, Tsitsilonis OE, Kostakis IK. Ole-Oxy, a Semi-Synthetic Analog of Oleuropein, Ameliorates Acute Skin and Colon Inflammation in Mice. Antioxidants (Basel) 2024; 13:1422. [PMID: 39594563 PMCID: PMC11590887 DOI: 10.3390/antiox13111422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Inflammation is a key process in the pathophysiology of various diseases, with macrophages playing a central role in the inflammatory response. This study investigates the anti-inflammatory potential of a newly synthesized analog of oleuropein (OP), the major olive tree (Olea europaea) metabolite. This derivative of OP, named Ole-Oxy, was designed by introducing an oxygen atom between the aromatic ring and the aliphatic chain of OP, to enhance interaction with proteins and improve bioactivity. Ole-Oxy demonstrated notable anti-inflammatory effects in vitro, particularly in phorbol 12-myristate 13-acetate-differentiated THP-1 macrophages, where it markedly reduced interleukin-6, tumor necrosis factor-α, and reactive oxygen species (ROS) levels, surpassing the effects of OP. In vivo, Ole-Oxy was evaluated in mouse models of acute skin and colon inflammation, showing significant efficacy in C57BL/6J mice, likely due to their Th1-biased immune response. Our results suggest that Ole-Oxy modulates inflammation through ROS scavenging and differential macrophage activation, underscoring the need for further research to fully elucidate its mechanism of action and optimize its pharmacokinetic properties for future therapeutic applications.
Collapse
Affiliation(s)
- Nikolaos V. Angelis
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, Ilisia, 15784 Athens, Greece; (N.V.A.); (E.P.); (P.-M.N.); (C.C.K.); (O.L.); (C.N.B.)
| | - Efthymios Paronis
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, Ilisia, 15784 Athens, Greece; (N.V.A.); (E.P.); (P.-M.N.); (C.C.K.); (O.L.); (C.N.B.)
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece; (A.A.); (P.A.)
| | - Georgia Sarikaki
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Ilisia, 15774 Athens, Greece; (G.S.); (A.-L.S.)
| | | | - Anna Agapaki
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece; (A.A.); (P.A.)
| | - Pigi-Maria Niotopoulou
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, Ilisia, 15784 Athens, Greece; (N.V.A.); (E.P.); (P.-M.N.); (C.C.K.); (O.L.); (C.N.B.)
| | - Christina C. Knai
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, Ilisia, 15784 Athens, Greece; (N.V.A.); (E.P.); (P.-M.N.); (C.C.K.); (O.L.); (C.N.B.)
| | - Pavlos Alexakos
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece; (A.A.); (P.A.)
| | - Odyssefs Liagkas
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, Ilisia, 15784 Athens, Greece; (N.V.A.); (E.P.); (P.-M.N.); (C.C.K.); (O.L.); (C.N.B.)
| | | | - Constantin N. Baxevanis
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, Ilisia, 15784 Athens, Greece; (N.V.A.); (E.P.); (P.-M.N.); (C.C.K.); (O.L.); (C.N.B.)
| | - Alexios-Leandros Skaltsounis
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Ilisia, 15774 Athens, Greece; (G.S.); (A.-L.S.)
| | - Ourania E. Tsitsilonis
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, Ilisia, 15784 Athens, Greece; (N.V.A.); (E.P.); (P.-M.N.); (C.C.K.); (O.L.); (C.N.B.)
| | - Ioannis K. Kostakis
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Ilisia, 15771 Athens, Greece
| |
Collapse
|
26
|
Jin WJ, Zhu XX, Luo KT, Wang S, Li JA, Qian LF, Xu GX. Enhancement of Cognitive Function in Rats with Vascular Dementia Through Modulation of the Nrf2/GPx4 Signaling Pathway by High-Frequency Repetitive Transcranial Magnetic Stimulation. Physiol Res 2024; 73:857-868. [PMID: 39560194 PMCID: PMC11629951 DOI: 10.33549/physiolres.935330] [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: 01/29/2024] [Accepted: 05/16/2024] [Indexed: 12/13/2024] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) represents a non-invasive therapeutic modality acknowledged for augmenting neurological function recovery following stroke. Nonetheless, uncertainties remain regarding its efficacy in promoting cognitive function recovery in patients diagnosed with vascular dementia (VD). In this study, VD was experimentally induced in a rat model utilizing the bilateral common carotid artery occlusion method. Following a recuperation period of seven days, rats were subjected to high-frequency repetitive transcranial magnetic stimulation (HF-rTMS) at a frequency of 10 Hz. Cognitive function was assessed utilizing the Morris water maze test, and the levels of IL-6, TNF-alpha, SOD, GSH, MDA, and Fe2+ in cerebral tissue were quantitatively analyzed through enzyme-linked immunosorbent assay. Moreover, the gene and protein expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and glutathione peroxidase 4 (GPx4) were meticulously investigated via quantitative polymerase chain reaction (qPCR) and Western blotting techniques. The use of HF-rTMS notably augmented cognitive function in rats with VD, concomitantly reducing neuroinflammation, oxidative stress, and ferroptosis within the brain. The group subjected to HF-rTMS demonstrated an increase in the levels of both proteins and genes associated with Nrf2 and GPx4, in comparison to the VD group. These results highlight the potential of HF-rTMS treatment in enhancing cognitive function in rats diagnosed with VD through the modulation of the Nrf2/GPx4 signaling pathway. This modulation, in turn, mitigates processes linked with neuroinflammation, oxidative stress, and ferroptosis. Nevertheless, additional studies are essential to comprehensively elucidate the underlying mechanisms and clinical implications of HF-rTMS treatment in the treatment of VD.
Collapse
Affiliation(s)
- W-J Jin
- Department of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China; Rehabilitation Medicine Center, Zhejiang Chinese Medical University Affiliated Jiaxing TCM Hospital, Jiaxing, China.
| | | | | | | | | | | | | |
Collapse
|
27
|
Hunt M, Torres M, Bachar-Wikstrom E, Wikstrom JD. Cellular and molecular roles of reactive oxygen species in wound healing. Commun Biol 2024; 7:1534. [PMID: 39562800 DOI: 10.1038/s42003-024-07219-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024] Open
Abstract
Wound healing is a highly coordinated spatiotemporal sequence of events involving several cell types and tissues. The process of wound healing requires strict regulation, and its disruption can lead to the formation of chronic wounds, which can have a significant impact on an individual's health as well as on worldwide healthcare expenditure. One essential aspect within the cellular and molecular regulation of wound healing pathogenesis is that of reactive oxygen species (ROS) and oxidative stress. Wounding significantly elevates levels of ROS, and an array of various reactive species are involved in modulating the wound healing process, such as through antimicrobial activities and signal transduction. However, as in many pathologies, ROS play an antagonistic pleiotropic role in wound healing, and can be a pathogenic factor in the formation of chronic wounds. Whilst advances in targeting ROS and oxidative stress have led to the development of novel pre-clinical therapeutic methods, due to the complex nature of ROS in wound healing, gaps in knowledge remain concerning the specific cellular and molecular functions of ROS in wound healing. In this review, we highlight current knowledge of these functions, and discuss the potential future direction of new studies, and how these pathways may be targeted in future pre-clinical studies.
Collapse
Affiliation(s)
- Matthew Hunt
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Monica Torres
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden.
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
28
|
Wu G, Li B, Wei X, Chen Y, Zhao Y, Peng Y, Su J, Hu Z, Zhuo L, Tian Y, Wang Z, Peng X. Design, synthesis and biological evaluation of N-salicyloyl tryptamine derivatives as multifunctional neuroprotectants for the treatment of ischemic stroke. Eur J Med Chem 2024; 278:116795. [PMID: 39216381 DOI: 10.1016/j.ejmech.2024.116795] [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: 03/28/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Ischemic stroke (IS) is a disease of high death and disability worldwide with few medications in clinical treatment. Neuroinflammation and oxidative stress are considered as crucial factors in the progression of IS. In our previous studies, N-salicyloyl tryptamine derivative (NST) L7 exhibited promising anti-inflammatory properties and is considered a potential clinical therapy for IS but had limited antioxidant capacity. Here, we have designed, synthesized, and biologically evaluated 30 novel NSTs for their neuroprotective effects against cerebral ischemia-reperfusion (CI/R) injury. To identify a multifunctional neuroprotectant with enhanced antioxidant and anti-inflammatory capacity, as well as an effective therapeutic agent for CI/R damage. Among them, M11 exhibited synergistic highly anti-oxidant, anti-inflammatory, anti-ferroptosis, and anti-apoptosis effects and surpassed the parent compound L7. Further studies demonstrated that the synergistic and efficient neuroprotective role of M11 was mainly achieved by activating Nrf2 and stimulating its downstream target HO-1/GCLC/NQO1/GPX4. In addition, M11 possessed good blood-brain barrier permeability. Moreover, M11 effectively reduced cerebral infarct volume and improved neurological deficits in MCAO/R mice. Its hydrochloride form, M11·HCl, exhibited better pharmacokinetic properties, high safety, and a significant reduction in infarct volume, which is comparable to Edaravone. In conclusion, our findings suggested that M11 capable of activating Nrf2, could represent a promising candidate agent for IS.
Collapse
Affiliation(s)
- Genping Wu
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Bo Li
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiuzhen Wei
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yaxin Chen
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yuting Zhao
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yan Peng
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jianhui Su
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zecheng Hu
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Linsheng Zhuo
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Ying Tian
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhen Wang
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China; MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, China.
| | - Xue Peng
- The Affiliated Nanhua Hospital, School of Pharmaceutical Science, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
29
|
Xu Q, Li Y, Qin X, Xin Y, Wang J, Zhang Y, Xu K, Yang X, Wang X. osa-miR168a, a Plant miRNA That Survives the Process of In Vivo Food Digestion, Attenuates Dextran Sulfate Sodium-Induced Colitis in Mice by Oral Administration. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25146-25160. [PMID: 39480689 DOI: 10.1021/acs.jafc.4c07283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Previous studies showed that osa-miR168a, a plant miRNA rich in fruits and vegetables, had cross-kingdom biological effects on immunocytes, silkworms, and rodents. In this study, the effects of miR168a on mouse colitis induced by dextran sulfate sodium (DSS) were investigated. The results showed that miR168a oligomers were resistant during the process of food digestion, ending up with a residual concentration of 67.8 ± 11.2 fM in mouse intestines 4 h after oral gavage. More importantly, direct oral administration of the miRNA to the colitis mice significantly ameliorated the progression of the disease, as evidenced by the reduction in DAI score, histopathological lesions, and proinflammatory cytokines. Repairing intestinal barrier function by promoting the regeneration of TJ proteins and the mucus layer, suppressing oxidative stress and colonic inflammation via modulating Nrf2 and NF-κB signaling pathways, and restoring the imbalanced gut microbiota caused by DSS are proposed mechanisms behind the anticolitis activity of miR168a. This study provided new evidence of the cross-kingdom regulatory effects of dietary miRNAs, suggesting the potential of the plant miRNA for the prevention and treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Qin Xu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Ying Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Xinshu Qin
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yirao Xin
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Jianing Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Yi Zhang
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ke Xu
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Xingbin Yang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Xingyu Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| |
Collapse
|
30
|
Hashiesh HM, Azimullah S, Nagoor Meeran MF, Saraswathiamma D, Arunachalam S, Jha NK, Sadek B, Adeghate E, Sethi G, Albawardi A, Al Marzooqi S, Ojha S. Cannabinoid 2 Receptor Activation Protects against Diabetic Cardiomyopathy through Inhibition of AGE/RAGE-Induced Oxidative Stress, Fibrosis, and Inflammasome Activation. J Pharmacol Exp Ther 2024; 391:241-257. [PMID: 38955492 DOI: 10.1124/jpet.123.002037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
Oxidative stress, fibrosis, and inflammasome activation from advanced glycation end product (AGE)-receptor of advanced glycation end product (RAGE) interaction contribute to diabetic cardiomyopathy (DCM) formation and progression. Our study revealed the impact of β-caryophyllene (BCP) on activating cannabinoid type 2 receptors (CB2Rs) against diabetic complication, mainly cardiomyopathy and investigated the underlying cell signaling pathways in mice. The murine model of DCM was developed by feeding a high-fat diet with streptozotocin injections. After the development of diabetes, the animals received a 12-week oral BCP treatment at a dose of 50 mg/kg/body weight. BCP treatment showed significant improvement in glucose tolerance and insulin resistance and enhanced serum insulin levels in diabetic animals. BCP treatment effectively reversed the heart remodeling and restored the phosphorylated troponin I and sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a expression. Ultrastructural examination showed reduced myocardial cell injury in DCM mice treated with BCP. The preserved myocytes were found to be associated with reduced expression of AGE/RAGE in DCM mice hearts. BCP treatment mitigated oxidative stress by inhibiting expression of NADPH oxidase 4 and activating phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. Also, BCP suppressed cardiac fibrosis and endothelial-to-mesenchymal transition in DCM mice by inhibiting transforming growth factor β (TGF-β)/suppressor of mothers against decapentaplegic (Smad) signaling. Further, BCP treatment suppressed nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome activation in DCM mice and alleviated cellular injury to the pancreatic tissues evidenced by significant elevation of the number of insulin-positive cells. To demonstrate a CB2R-dependent mechanism of BCP, another group of DCM mice were pretreated with AM630, a CB2R antagonist. AM630 was observed to abrogate the beneficial effects of BCP in DCM mice. Taken together, BCP demonstrated the potential to protect the myocardium and pancreas of DCM mice mediating CB2R-dependent mechanisms. SIGNIFICANCE STATEMENT: BCP, a CB2R agonist, shows protection against DCM. BCP attenuates oxidative stress, inflammation, and fibrosis in DCM via activating CB2Rs. BCP mediating CB2R activation favorably modulates AGE/RAGE, PI3K/AKT/Nrf2β and TGF-β/Smad and (NLRP3) inflammasome in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Hebaallah Mamdouh Hashiesh
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Sheikh Azimullah
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Mohamed Fizur Nagoor Meeran
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Dhanya Saraswathiamma
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Seenipandi Arunachalam
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Niraj Kumar Jha
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Bassem Sadek
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Ernest Adeghate
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Gautam Sethi
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Alia Albawardi
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Saeeda Al Marzooqi
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Shreesh Ojha
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| |
Collapse
|
31
|
Maluleke TT, Manilall A, Shezi N, Baijnath S, Millen AME. Acute exposure to LPS induces cardiac dysfunction via the activation of the NLRP3 inflammasome. Sci Rep 2024; 14:24378. [PMID: 39420211 PMCID: PMC11487256 DOI: 10.1038/s41598-024-76066-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
Systemic inflammation contributes to left ventricular (LV) dysfunction, however the role of the NLRP3 inflammasome in LV dysfunction in acute inflammatory conditions is unclear. This study investigated the role of the NLRP3 inflammasome in acute (24 h) cardiac structural and functional changes in vivo and in vitro in lipopolysaccharide (LPS)-induced inflammation. LPS-treated Sprague-Dawley (SD) rats showed increased LPS metabolite abundance in their LVs as measured by atmospheric pressure matrix-assisted laser desorption ionisation (AP-MALDI) mass spectrometry imaging (MSI). Echocardiography and histology showed that in LPS-exposed rats, LV internal diameter was decreased, with evidence of macrophage infiltration and oedema. However, there were no changes in LV wall thickness or collagen volume. Additionally, LPS-exposed rats exhibited impaired LV relaxation, potentially contributing to decreased stroke volume. While global systolic function was preserved, LPS exposure in SD rats resulted in impaired myocardial deformation assessed by speckle-tracking echocardiography. Exposure to LPS resulted in upregulation of the expression of components of the NLRP3 inflammasome in rodents. In vitro LPS exposure resulted in increased gene expression of NLRP3 and downstream cytokines IL-1β and IL-18, antioxidant SOD2, and elevated markers of pyroptosis (GSDMD) which were inhibited by treatment with a NLRP3 antagonist. However, LPS-induced increases in the gene expression of apoptosic markers (BAX/Bcl2) were not impacted by NLRP3 antagonism. These findings suggest that inflammation induced adverse cardiac structural and functional changes is, at least in part, mediated by the NLRP3 inflammasome in acute, high-grade inflammatory states. In addition, in vitro findings suggest that while the NLRP3 inflammasome mediates pyroptotic pathways, regulation of apoptosis that is independent of the inflammasome.
Collapse
Affiliation(s)
- Tshiamo T Maluleke
- Wits Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, Wits Health Consortium (PTY) Ltd, University of The Witwatersrand, Johannesburg, South Africa
| | - Ashmeetha Manilall
- Wits Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, Wits Health Consortium (PTY) Ltd, University of The Witwatersrand, Johannesburg, South Africa
| | - Nandi Shezi
- Wits Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, Wits Health Consortium (PTY) Ltd, University of The Witwatersrand, Johannesburg, South Africa
| | - Sooraj Baijnath
- Wits Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, Wits Health Consortium (PTY) Ltd, University of The Witwatersrand, Johannesburg, South Africa
| | - Aletta M E Millen
- Wits Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, Wits Health Consortium (PTY) Ltd, University of The Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
32
|
Dewanjee S, Bhattacharya H, Bhattacharyya C, Chakraborty P, Fleishman J, Alexiou A, Papadakis M, Jha SK. Nrf2/Keap1/ARE regulation by plant secondary metabolites: a new horizon in brain tumor management. Cell Commun Signal 2024; 22:497. [PMID: 39407193 PMCID: PMC11476647 DOI: 10.1186/s12964-024-01878-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Brain cancer is regarded as one of the most life-threatening forms of cancer worldwide. Oxidative stress acts to derange normal brain homeostasis, thus is involved in carcinogenesis in brain. The Nrf2/Keap1/ARE pathway is an important signaling cascade responsible for the maintenance of redox homeostasis, and regulation of anti-inflammatory and anticancer activities by multiple downstream pathways. Interestingly, Nrf2 plays a somewhat, contradictory role in cancers, including brain cancer. Nrf2 has traditionally been regarded as a tumor suppressor since its cytoprotective functions are considered to be the principle cellular defense mechanism against exogenous and endogenous insults, such as xenobiotics and oxidative stress. However, hyperactivation of the Nrf2 pathway supports the survival of normal as well as malignant cells, protecting them against oxidative stress, and therapeutic agents. Plants possess a pool of secondary metabolites with potential chemotherapeutic/chemopreventive actions. Modulation of Nrf2/ARE and downstream activities in a Keap1-dependant manner, with the aid of plant-derived secondary metabolites exhibits promise in the management of brain tumors. Current article highlights the effects of Nrf2/Keap1/ARE cascade on brain tumors, and the potential role of secondary metabolites regarding the management of the same.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India.
| | - Hiranmoy Bhattacharya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Chiranjib Bhattacharyya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Joshua Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, 11741, Greece
- Department of Research & Development, AFNP Med, Wien, 1030, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, 110008, India.
| |
Collapse
|
33
|
Yan KX, Ge BJ, Sang R, Zhao P, Liu XM, Yu MH, Liu XT, Qiu Q, Zhang XM. Taraxasterol attenuates zearalenone-induced kidney damage in mice by modulating oxidative stress and endoplasmic reticulum stress. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117093. [PMID: 39317070 DOI: 10.1016/j.ecoenv.2024.117093] [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: 07/25/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
Taraxasterol is one of the bioactive ingredients from traditional Chinese herb Taraxacum, which exhibits multiple pharmacological activities and protective effects. However, the underlying influence and mechanism of its use against kidney damage caused from zearalenone (ZEA) remain unexplored. The ZEA-induced kidney damage model of mice was established by feeding diets containing ZEA (2 mg/kg), and taraxasterol (5 and 10 mg/kg) was administered by gavage for 28 days. Results demonstrated taraxasterol increased average daily gain (ADG) and average daily feed intake (ADFI), reduced feed-to-gain ratio (F/G) and kidney index of mice induced by ZEA. Taraxasterol alleviated histopathological changes of kidney, reduced ZEA residue and the levels of blood urea nitrogen (BUN), uric acid (UA), and creatinine (CRE). Concurrently, taraxasterol reduced the contents of oxidative stress indicator reactive oxygen species (ROS) and malondialdehyde (MDA), and increased the activities of antioxidant enzymes catalase (CAT), total superoxide dismutase (T-SOD), and glutathione peroxidase (GSH-Px). Further, taraxasterol up-regulated the mRNA and protein expression of nuclear factor erythroid-2-related factor 2 (Nrf2), GSH-Px, NAD(P)H quinone oxidoreductase 1 (NQO1), and heme oxygenase-1 (HO-1), and down-regulated the mRNA and protein expression of KELCH like ECH associated protein (Keap1) in Nrf2/Keap1 pathway. Taraxasterol down-regulated the mRNA and protein expression of immunoglobulin binding protein (Bip), C/EBP homologous protein (CHOP), Bcl-2 associated X (Bax), cysteine protease (Caspase)-12, and Caspase-3, and up-regulated B-cell lymphoma 2 (Bcl-2) expression in endoplasmic reticulum stress pathway. This study suggests that taraxasterol attenuates ZEA-induced mouse kidney damage through the modulation of Nrf2/Keapl pathway to play antioxidant role and endoplasmic reticulum stress pathway to enhance anti-apoptotic ability. It will provide a basis for taraxasterol as a potential drug to prevent and treat ZEA-induced kidney damage.
Collapse
Affiliation(s)
- Ke-Xin Yan
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Bing-Jie Ge
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Rui Sang
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Peng Zhao
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Xin-Man Liu
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Ming-Hong Yu
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Xiao-Tong Liu
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Qian Qiu
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Xue-Mei Zhang
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| |
Collapse
|
34
|
Liu L, de Leeuw K, van Goor H, Westra J. The Role of Antioxidant Transcription Factor Nrf2 and Its Activating Compounds in Systemic Lupus Erythematosus. Antioxidants (Basel) 2024; 13:1224. [PMID: 39456477 PMCID: PMC11504041 DOI: 10.3390/antiox13101224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease in which kidney involvement, so-called lupus nephritis (LN), is common and one of the most severe manifestations. Oxidative stress (OS) may play a role in the pathogenesis of LN through the exacerbation of inflammation and immune cell dysfunction/dysregulation. Nuclear factor erythroid 2-related factor 2 (Nrf2), also known as nuclear factor erythroid-derived 2-like 2, is a transcription factor that in humans is encoded by the NFE2L2 gene and is regarded as a central regulator of the antioxidative response. Nrf2-activating compounds have been shown to alleviate oxidative stress in cells and tissues of lupus-prone mice. Although the precise mechanisms of Nrf2 activation on the immune system in SLE remain to be elucidated, Nrf2-activating compounds are considered novel therapeutical options to suppress OS and thereby might alleviate disease activity in SLE, especially in LN. This review therefore summarizes the role of the Nrf2 signaling pathway in the pathogenesis of SLE with LN and describes compounds modulating this pathway as potential additional clinical interventions.
Collapse
Affiliation(s)
- Lu Liu
- Department of Rheumatology and Clinical Immunology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Karina de Leeuw
- Department of Rheumatology and Clinical Immunology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Johanna Westra
- Department of Rheumatology and Clinical Immunology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
35
|
Kaur T, Sidana P, Kaur N, Choubey V, Kaasik A. Unraveling neuroprotection in Parkinson's disease: Nrf2-Keap1 pathway's vital role amidst pathogenic pathways. Inflammopharmacology 2024; 32:2801-2820. [PMID: 39136812 DOI: 10.1007/s10787-024-01549-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/01/2024] [Indexed: 10/11/2024]
Abstract
Parkinson's disease (PD) is an age-related chronic neurological condition characterized by progressive degeneration of dopaminergic neurons and the presence of Lewy bodies, primarily composed of alpha-synuclein and ubiquitin. The pathophysiology of PD encompasses alpha-synuclein aggregation, oxidative stress, neuroinflammation, mitochondrial dysfunction, and impaired autophagy and ubiquitin-proteasome systems. Among these, the Keap1-Nrf2 pathway is a key regulator of antioxidant defense mechanisms. Nrf2 has emerged as a crucial factor in managing oxidative stress and inflammation, and it also influences ubiquitination through p62 expression. Keap1 negatively regulates Nrf2 by targeting it for degradation via the ubiquitin-proteasome system. Disruption of the Nrf2-Keap1 pathway in PD affects cellular responses to oxidative stress and inflammation, thereby playing a critical role in disease progression. In addition, the role of neuroinflammation in PD has gained significant attention, highlighting the interplay between immune responses and neurodegeneration. This review discusses the various mechanisms responsible for neuronal degeneration in PD, with a special emphasis on the neuroprotective role of the Nrf2-Keap1 pathway. Furthermore, it explores the implications of inflammopharmacology in modulating these pathways to provide therapeutic insights for PD.
Collapse
Affiliation(s)
- Tanzeer Kaur
- Department of Biophysics, Panjab University, Chandigarh, India.
| | - Palak Sidana
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Navpreet Kaur
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Vinay Choubey
- Department of Pharmacology, University of Tartu, Tartu, Estonia
| | - Allen Kaasik
- Department of Pharmacology, University of Tartu, Tartu, Estonia
| |
Collapse
|
36
|
Zhang X, Zhang D, Fan A, Zhou X, Yang C, Zhou J, Shen M, Liu H, Zou K, Tao J. A novel effect of sulforaphane on promoting mouse granulosa cells proliferation via the NRF2-TKT pathway. J Adv Res 2024:S2090-1232(24)00422-3. [PMID: 39341455 DOI: 10.1016/j.jare.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/15/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
INTRODUCTION Granulosa cells (GCs) is essential for maintaining follicular development. Follicle-stimulating Hormone (FSH) has been demonstrated to effectively promote GCs proliferation, driving the establishment of various superovulation techniques for animal husbandry. However, these techniques face challenges, such as high costs, hormonal imbalances, and an increased risk of early ovarian dysfunction. Therefore, it is important to investigate new methods to improve GCs proliferation. OBJECTIVES This study aimed to investigate the effect of sulforaphane (SFN) on ovarian GCs proliferation and the underlying mechanisms. METHODS A comparative transcriptomic analysis of ovaries from the control, SFN, and FSH groups was conducted to identify the primary factors contributing to high proliferative capacity. The role of SFN in the regulation of cell proliferation has been examined in mouse ovarian GCs. Gene interference, overexpression, CUT&TAG technology, and transcriptome analyses were performed to elucidate the underlying mechanisms of the nuclear factor E2-related factor 2 (NRF2)-transketolase (TKT) axis in mediating GCs proliferation. RESULTS Our research revealed a previously unknown function of SFN, an isothiocyanate of plant origin that is prevalent in cruciferous vegetables, in facilitating the proliferation of mouse ovarian GCs. The efficacy of SFN in enhancing GCs proliferation is similar to that of FSH. At the mechanistic level, SFN promotes NRF2 to transport to the nucleus, which subsequently activates the key enzyme of the non-oxidative pentose phosphate pathway TKT. This activation is instrumental in generating ribose 5-phosphate, a critical precursor for amino acid and nucleotide biosynthesis that underpins the proliferation of GCs. CONCLUSION Collectively, our findings delineate a novel pathway by which SFN, through the NRF2-TKT axis, enhances the nucleotide pool and thereby supports the proliferation of mouse GCs, presenting novel avenues for exploration in reproductive biology and agricultural sciences.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Dingding Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Aoyun Fan
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xinyi Zhou
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Caixia Yang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jiaqi Zhou
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ming Shen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Honglin Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Kang Zou
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Jingli Tao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
37
|
Sheng W, Yue Y, Qi T, Qin H, Liu P, Wang D, Zeng H, Yu F. The Multifaceted Protective Role of Nuclear Factor Erythroid 2-Related Factor 2 in Osteoarthritis: Regulation of Oxidative Stress and Inflammation. J Inflamm Res 2024; 17:6619-6633. [PMID: 39329083 PMCID: PMC11424688 DOI: 10.2147/jir.s479186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by the degradation of joint cartilage, subchondral bone sclerosis, synovitis, and structural changes in the joint. Recent research has highlighted the role of various genes in the pathogenesis and progression of OA, with nuclear factor erythroid 2-related factor 2 (NRF2) emerging as a critical player. NRF2, a vital transcription factor, plays a key role in regulating the OA microenvironment and slowing the disease's progression. It modulates the expression of several antioxidant enzymes, such as Heme oxygenase-1 (HO-1) and NAD(P)H oxidoreductase 1 (NQO1), among others, which help reduce oxidative stress. Furthermore, NRF2 inhibits the nuclear factor kappa-B (NF-κB) signaling pathway, thereby decreasing inflammation, joint pain, and the breakdown of cartilage extracellular matrix, while also mitigating cell aging and death. This review discusses NRF2's impact on oxidative stress, inflammation, cell aging, and various cell death modes (such as apoptosis, necroptosis, and ferroptosis) in OA-affected chondrocytes. The role of NRF2 in OA macrophages, and synovial fibroblasts was also discussed. It also covers NRF2's role in preserving the cartilage extracellular matrix and alleviating joint pain. The purpose of this review is to provide a comprehensive understanding of NRF2's protective mechanisms in OA, highlighting its potential as a therapeutic target and underscoring its significance in the development of novel treatment strategies for OA.
Collapse
Affiliation(s)
- Weibei Sheng
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Yaohang Yue
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Tiantian Qi
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Haotian Qin
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Peng Liu
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Deli Wang
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Hui Zeng
- Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, People’s Republic of China
| | - Fei Yu
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| |
Collapse
|
38
|
Sood R, Anoopkumar-Dukie S, Rudrawar S, Hall S. Neuromodulatory effects of leukotriene receptor antagonists: A comprehensive review. Eur J Pharmacol 2024; 978:176755. [PMID: 38909933 DOI: 10.1016/j.ejphar.2024.176755] [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: 02/19/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/25/2024]
Abstract
Cysteinyl leukotrienes (CysLTs) are central to the pathophysiology of asthma and various inflammatory disorders. Leukotriene receptor antagonists (LTRAs) effectively treat respiratory conditions by targeting cysteinyl leukotriene receptors, CysLT1 and CysLT2 subtypes. This review explores the multifaceted effects of LTs, extending beyond bronchoconstriction. CysLT receptors are not only present in the respiratory system but are also crucial in neuronal signaling pathways. LTRAs modulate these receptors, influencing downstream signaling, calcium levels, inflammation, and oxidative stress (OS) within neurons hinting at broader implications. Recent studies identify novel molecular targets, sparking interest in repurposing LTRAs for therapeutic use. Clinical trials are investigating their potential in neuroinflammation control, particularly in Alzheimer's disease (AD) and Parkinson's diseases (PD). However, montelukast, a long-standing LTRA since 1998, raises concerns due to neuropsychiatric adverse drug reactions (ADRs). Despite widespread use, understanding montelukast's metabolism and underlying ADR mechanisms remains limited. This review comprehensively examines LTRAs' diverse biological effects, emphasizing non-bronchoconstrictive activities. It also analyses plausible mechanisms behind LTRAs' neuronal effects, offering insights into their potential as neurodegenerative disease modulators. The aim is to inform clinicians, researchers, and pharmaceutical developers about LTRAs' expanding roles, particularly in neuroinflammation control and their promising repurposing for neurodegenerative disease management.
Collapse
Affiliation(s)
- Radhika Sood
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia
| | | | - Santosh Rudrawar
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia; Institute for Glycomics, Griffith University, Queensland, 4222, Australia
| | - Susan Hall
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia.
| |
Collapse
|
39
|
Hassanein EHM, Althagafy HS, Baraka MA, Abd-Alhameed EK, Ibrahim IM, Abd El-Maksoud MS, Mohamed NM, Ross SA. The promising antioxidant effects of lignans: Nrf2 activation comes into view. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6439-6458. [PMID: 38695909 PMCID: PMC11422461 DOI: 10.1007/s00210-024-03102-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/11/2024] [Indexed: 09/25/2024]
Abstract
Lignans are biologically active compounds widely distributed, recognized, and identified in seeds, fruits, and vegetables. Lignans have several intriguing bioactivities, including anti-inflammatory, antioxidant, and anticancer activities. Nrf2 controls the expression of many cytoprotective genes. Activation of Nrf2 is a promising therapeutic approach for treating and preventing diseases resulting from oxidative injury and inflammation. Lignans have been demonstrated to stimulate Nrf2 signaling in a variety of in vitro and experimental animal models. The review summarizes the findings of fourteen lignans (Schisandrin A, Schisandrin B, Schisandrian C, Magnolol, Honokiol, Sesamin, Sesamol, Sauchinone, Pinoresinol, Phyllanthin, Nectandrin B, Isoeucommin A, Arctigenin, Lariciresinol) as antioxidative and anti-inflammatory agents, affirming how Nrf2 activation affects their pharmacological effects. Therefore, lignans may offer therapeutic candidates for the treatment and prevention of various diseases and may contribute to the development of effective Nrf2 modulators.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohammad A Baraka
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Esraa K Abd-Alhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mostafa S Abd El-Maksoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Nesma M Mohamed
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt.
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Assiut, Assiut, 77771, Egypt.
| | - Samir A Ross
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, 38677, USA
- Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| |
Collapse
|
40
|
Xu WD, Yang C, Huang AF. The role of Nrf2 in immune cells and inflammatory autoimmune diseases: a comprehensive review. Expert Opin Ther Targets 2024; 28:789-806. [PMID: 39256980 DOI: 10.1080/14728222.2024.2401518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Nrf2 regulates mild stress, chronic inflammation, and metabolic changes by regulating different immune cells via downstream signaling. Collection of information about the role of Nrf2 in inflammatory autoimmune diseases will better understand the therapeutic potential of targeting Nrf2 in these diseases. AREAS COVERED In this review, we comprehensively discussed biological function of Nrf2 in different immune cells, including Nrf2 preventing oxidative tissue injury, affecting apoptosis of immune cells and inflammatory cytokine production. Moreover, we discussed the role of Nrf2 in the development of inflammatory autoimmune diseases. EXPERT OPINION Nrf2 binds to downstream signaling molecules and then provides durable protection against different cellular and organ stress. It has emerged as an important target for inflammatory autoimmune diseases. Development of Nrf2 modulator drugs needs to consider factors such as target specificity, short/long term safety, disease indication identification, and the extent of variation in Nrf2 activity. We carefully discussed the dual role of Nrf2 in some diseases, which helps to better target Nrf2 in the future.
Collapse
Affiliation(s)
- Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Chan Yang
- Preventive Health Center, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
41
|
Zhang Q, Chen Z, Li J, Huang K, Ding Z, Chen B, Ren T, Xu P, Wang G, Zhang H, Zhang XD, Zhang J, Wang H. The deubiquitinase OTUD1 stabilizes NRF2 to alleviate hepatic ischemia/reperfusion injury. Redox Biol 2024; 75:103287. [PMID: 39079388 PMCID: PMC11340619 DOI: 10.1016/j.redox.2024.103287] [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: 07/03/2024] [Accepted: 07/26/2024] [Indexed: 08/09/2024] Open
Abstract
Hepatic ischemia/reperfusion (I/R) injury is an important cause of liver function impairment following liver surgery. The ubiquitin-proteasome system (UPS) plays a crucial role in protein quality control and has substantial impact on the hepatic I/R process. Although OTU deubiquitinase 1 (OTUD1) is involved in diverse biological processes, its specific functional implications in hepatic I/R are not yet fully understood. This study demonstrates that OTUD1 alleviates oxidative stress, apoptosis, and inflammation induced by hepatic I/R injury. Mechanistically, OTUD1 deubiquitinates and activates nuclear factor erythroid 2-related factor 2 (NRF2) through its catalytic site cysteine 320 residue and ETGE motif, thereby attenuating hepatic I/R injury. Additionally, administration of a short peptide containing the ETGE motif significantly mitigates hepatic I/R injury in mice. Overall, our study elucidates the mechanism and role of OTUD1 in ameliorating hepatic I/R injury, providing a theoretical basis for potential treatment using ETGE-peptide.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China
| | - Zihan Chen
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinglei Li
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kunpeng Huang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhihao Ding
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Biao Chen
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tianxing Ren
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Peng Xu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guoliang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongji Zhang
- Department of Surgery, University of Virginia, Charlottesville, VA, 22903, USA
| | - Xiao-Dong Zhang
- Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jinxiang Zhang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
42
|
Yu H, Lou J, Ni L, Yan M, Zhu K, Mao S, Zhu J. Isoquercetin Ameliorates Osteoarthritis via Nrf2/NF-κB Axis: An In Vitro and In Vivo Study. Chem Biol Drug Des 2024; 104:e14620. [PMID: 39251394 DOI: 10.1111/cbdd.14620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/20/2024] [Accepted: 07/09/2024] [Indexed: 09/11/2024]
Abstract
Osteoarthritis (OA) is a progressive joint disease characterized by extracellular matrix (ECM) degradation and inflammation, which is involved with pathological microenvironmental alterations induced by damaged chondrocytes. However, current therapies are not effective in alleviating the progression of OA. Isoquercetin is a natural flavonoid glycoside compound that has various pharmacological effects including anticancer, anti-diabetes and blood lipid regulation. Previous evidence suggests that isoquercetin has anti-inflammatory properties in various diseases, but its effect on OA has not been investigated yet. In this study, through western bolt, qRT-PCR and ELISA, it was found that isoquercetin could reduce the increase of ADAMTS5, MMP13, COX-2, iNOS and IL-6 induced by IL-1β, suggesting that isoquercetin could inhibit the inflammation and ECM degradation of chondrocytes. Through nuclear-plasma separation technique, western blot and immunocytochemistry, it can be found that Nrf2 and NF-κB pathways are activated in this process, and isoquercetin may rely on this process to play its protective role. In vivo, the results of X-ray and SO staining show that intra-articular injection of isoquercetin reduces the degradation of cartilage in the mouse OA model. In conclusion, the present work suggests that isoquercetin may benefit chondrocytes by regulating the Nrf2/NF-κB signaling axis, which supports isoquercetin as a potential drug for the treatment of OA.
Collapse
Affiliation(s)
- He Yu
- Department of Orthopaedics, Zhejiang Hospital, Zhejiang, Hangzhou, China
| | - Junsheng Lou
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Libin Ni
- Department of Orthopaedics, Zhejiang Hospital, Zhejiang, Hangzhou, China
| | - Minwei Yan
- Department of Orthopaedics, Zhejiang Hospital, Zhejiang, Hangzhou, China
| | - Kewu Zhu
- Center for Drug Delivery System Research, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Su Mao
- Department of Orthopaedics, Zhejiang Hospital, Zhejiang, Hangzhou, China
| | - Jungao Zhu
- Department of Orthopaedics, Zhejiang Hospital, Zhejiang, Hangzhou, China
| |
Collapse
|
43
|
Liu T, Li J, Sun L, Zhu C, Wei J. The role of ACE2 in RAS axis on microglia activation in Parkinson's disease. Neuroscience 2024; 553:128-144. [PMID: 38986737 DOI: 10.1016/j.neuroscience.2024.06.024] [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: 03/26/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND The classic renin-angiotensin system (RAS) induces organ damage, while the ACE2/Ang-(1-7)/MasR axis opposes it. However, the role of ACE2 in the brain is unclear. We studied ACE2's role in the brain. METHOD We used male C57BL/6J (WT) mice, ACE2 knockout (KO) mice, and MPTP-induced mice. Behavioral tests confirmed successful modeling. We assessed the impact of ACE2 KO on the RAS axis and PD index, including ACE, ACE2, AT1, AT2, MasR, TH, α-syn, and Iba1. We investigated ACE2 and MasR's involvement in microglial activation via western blot and immunofluorescence. GSE10867 and GSE26532 datasets were used to analyze the effects of AT1 antagonists and in vitro PD models on microglia. RESULT Behavioral tests revealed that MPTP mice displayed motor deficits, depression, anxiety, and increased inflammatory markers in the SN and CPU, with reduced antioxidant capacity. ACE2 KO worsened these symptoms and exacerbated inflammation and oxidative stress. LPS-induced ACE2/MasR activation in BV2 cells demonstrated anti-inflammatory and neuroprotective effects by modulating microglial polarization. Antagonists inhibited microglial activation via inflammation and ROS processes. CONCLUSION The RAS axis regulates inflammation and oxidative stress to maintain CNS function, suggesting potential targets for neurologic disease treatment. Understanding microglial RAS activation can offer new therapeutic strategies.
Collapse
Affiliation(s)
- Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Jingwen Li
- Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Lin Sun
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475000, China.
| | - Chaoyang Zhu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China.
| |
Collapse
|
44
|
Młynarska E, Hajdys J, Czarnik W, Fularski P, Leszto K, Majchrowicz G, Lisińska W, Rysz J, Franczyk B. The Role of Antioxidants in the Therapy of Cardiovascular Diseases-A Literature Review. Nutrients 2024; 16:2587. [PMID: 39203723 PMCID: PMC11357572 DOI: 10.3390/nu16162587] [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: 06/25/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Antioxidants are endogenous and exogenous substances with the ability to inhibit oxidation processes by interacting with reactive oxygen species (ROS). ROS, in turn, are small, highly reactive substances capable of oxidizing a wide range of molecules in the human body, including nucleic acids, proteins, lipids, carbohydrates, and even small inorganic compounds. The overproduction of ROS leads to oxidative stress, which constitutes a significant factor contributing to the development of disease, not only markedly diminishing the quality of life but also representing the most common cause of death in developed countries, namely, cardiovascular disease (CVD). The aim of this review is to demonstrate the effect of selected antioxidants, such as coenzyme Q10 (CoQ10), flavonoids, carotenoids, and resveratrol, as well as to introduce new antioxidant therapies utilizing miRNA and nanoparticles, in reducing the incidence and progression of CVD. In addition, new antioxidant therapies in the context of the aforementioned diseases will be considered. This review emphasizes the pleiotropic effects and benefits stemming from the presence of the mentioned substances in the organism, leading to an overall reduction in cardiovascular risk, including coronary heart disease, dyslipidaemia, hypertension, atherosclerosis, and myocardial hypertrophy.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland; (J.H.); (W.C.); (P.F.); (K.L.); (G.M.); (W.L.)
| | - Joanna Hajdys
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland; (J.H.); (W.C.); (P.F.); (K.L.); (G.M.); (W.L.)
| | - Witold Czarnik
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland; (J.H.); (W.C.); (P.F.); (K.L.); (G.M.); (W.L.)
| | - Piotr Fularski
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland; (J.H.); (W.C.); (P.F.); (K.L.); (G.M.); (W.L.)
| | - Klaudia Leszto
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland; (J.H.); (W.C.); (P.F.); (K.L.); (G.M.); (W.L.)
| | - Gabriela Majchrowicz
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland; (J.H.); (W.C.); (P.F.); (K.L.); (G.M.); (W.L.)
| | - Wiktoria Lisińska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland; (J.H.); (W.C.); (P.F.); (K.L.); (G.M.); (W.L.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland;
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland; (J.H.); (W.C.); (P.F.); (K.L.); (G.M.); (W.L.)
| |
Collapse
|
45
|
Kluknavsky M, Balis P, Liskova S, Micurova A, Skratek M, Manka J, Bernatova I. Dimethyl Fumarate Prevents the Development of Chronic Social Stress-Induced Hypertension in Borderline Hypertensive Rats. Antioxidants (Basel) 2024; 13:947. [PMID: 39199192 PMCID: PMC11351876 DOI: 10.3390/antiox13080947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
This study investigated the effects of chronic crowding-induced social stress and dimethyl fumarate (DMF) on borderline hypertensive rats, focusing on the transcription nuclear factor (erythroid-derived 2)-like 2 (NRF2) gene Nfe2l2, on the expression of selected NFR2-mediated gene expressions in the heart, and on vascular function. Rats were exposed to chronic crowding, DMF treatment (30 mg/kg/day, p.o.), or a combination of both for six weeks. Blood pressure (BP) was measured non-invasively, gene expressions were analysed using RT-qPCR, and vascular function was assessed by measuring noradrenaline (NA)-induced vasoconstriction and endothelium-dependent and -independent relaxations in the femoral arteries using a wire myograph. Chronic stress increased BP, Nfe2l2 expression, and NA-induced vasoconstriction, though it did not affect relaxation responses nor the left heart ventricle-to-body weight (LHV/BW) ratio. DMF elevated Nfe2l2 expression (as the main effect) in the heart but did not alter BP and vascular functions vs. control when administered alone. Interestingly, DMF increased the LHV/BW ratio, supposedly due to reductive stress induced by continuous NRF2 activation. When combined with stress, DMF treatment prevented stress-induced hypertension and mitigated NA-induced vasoconstriction without altering relaxation functions. In addition, the combination of stress and DMF increased Tnf and Nos2 expression and the expressions of several genes involved in iron metabolism. In conclusion, these findings suggest that DMF can prevent chronic stress-induced hypertension by reducing vascular contractility. Moreover, DMF itself may produce reductive stress in the heart and induce inflammation when combined with stress. This indicates a need for the careful consideration of long-term DMF treatment considering its impact on the heart.
Collapse
Affiliation(s)
- Michal Kluknavsky
- Centre of Experimental Medicine, Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, 813 71 Bratislava, Slovakia; (M.K.); (P.B.); (S.L.); (A.M.)
| | - Peter Balis
- Centre of Experimental Medicine, Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, 813 71 Bratislava, Slovakia; (M.K.); (P.B.); (S.L.); (A.M.)
| | - Silvia Liskova
- Centre of Experimental Medicine, Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, 813 71 Bratislava, Slovakia; (M.K.); (P.B.); (S.L.); (A.M.)
- Institute of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Andrea Micurova
- Centre of Experimental Medicine, Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, 813 71 Bratislava, Slovakia; (M.K.); (P.B.); (S.L.); (A.M.)
| | - Martin Skratek
- Institute of Measurement Science, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (M.S.); (J.M.)
| | - Jan Manka
- Institute of Measurement Science, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (M.S.); (J.M.)
| | - Iveta Bernatova
- Centre of Experimental Medicine, Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, 813 71 Bratislava, Slovakia; (M.K.); (P.B.); (S.L.); (A.M.)
| |
Collapse
|
46
|
Luo H, Tang L, Zeng M, Yin R, Ding P, Luo L, Li M. BertSNR: an interpretable deep learning framework for single-nucleotide resolution identification of transcription factor binding sites based on DNA language model. Bioinformatics 2024; 40:btae461. [PMID: 39107889 PMCID: PMC11310455 DOI: 10.1093/bioinformatics/btae461] [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: 03/14/2024] [Revised: 06/07/2024] [Indexed: 08/10/2024] Open
Abstract
MOTIVATION Transcription factors are pivotal in the regulation of gene expression, and accurate identification of transcription factor binding sites (TFBSs) at high resolution is crucial for understanding the mechanisms underlying gene regulation. The task of identifying TFBSs from DNA sequences is a significant challenge in the field of computational biology today. To address this challenge, a variety of computational approaches have been developed. However, these methods face limitations in their ability to achieve high-resolution identification and often lack interpretability. RESULTS We propose BertSNR, an interpretable deep learning framework for identifying TFBSs at single-nucleotide resolution. BertSNR integrates sequence-level and token-level information by multi-task learning based on pre-trained DNA language models. Benchmarking comparisons show that our BertSNR outperforms the existing state-of-the-art methods in TFBS predictions. Importantly, we enhanced the interpretability of the model through attentional weight visualization and motif analysis, and discovered the subtle relationship between attention weight and motif. Moreover, BertSNR effectively identifies TFBSs in promoter regions, facilitating the study of intricate gene regulation. AVAILABILITY AND IMPLEMENTATION The BertSNR source code can be found at https://github.com/lhy0322/BertSNR.
Collapse
Affiliation(s)
- Hanyu Luo
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
- School of Computer Science, University of South China, Hengyang, Hunan 421001, China
| | - Li Tang
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Min Zeng
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Rui Yin
- Department of Health Outcome and Biomedical Informatics, University of Florida, Gainesville, FL 32611, United States
| | - Pingjian Ding
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Lingyun Luo
- School of Computer Science, University of South China, Hengyang, Hunan 421001, China
| | - Min Li
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| |
Collapse
|
47
|
He L, Chen Q, Wang L, Pu Y, Huang J, Cheng CK, Luo JY, Kang L, Lin X, Xiang L, Fang L, He B, Xia Y, Lui KO, Pan Y, Liu J, Zhang CL, Huang Y. Activation of Nrf2 inhibits atherosclerosis in ApoE -/- mice through suppressing endothelial cell inflammation and lipid peroxidation. Redox Biol 2024; 74:103229. [PMID: 38870781 PMCID: PMC11247299 DOI: 10.1016/j.redox.2024.103229] [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: 04/04/2024] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Nuclear erythroid 2-related factor 2 (Nrf2), a transcription factor, is critically involved in the regulation of oxidative stress and inflammation. However, the role of endothelial Nrf2 in atherogenesis has yet to be defined. In addition, how endothelial Nrf2 is activated and whether Nrf2 can be targeted for the prevention and treatment of atherosclerosis is not explored. METHODS RNA-sequencing and single-cell RNA sequencing analysis of mouse atherosclerotic aortas were used to identify the differentially expressed genes. In vivo endothelial cell (EC)-specific activation of Nrf2 was achieved by injecting adeno-associated viruses into ApoE-/- mice, while EC-specific knockdown of Nrf2 was generated in Cdh5CreCas9floxed-stopApoE-/- mice. RESULTS Endothelial inflammation appeared as early as on day 3 after feeding of a high cholesterol diet (HCD) in ApoE-/- mice, as reflected by mRNA levels, immunostaining and global mRNA profiling, while the immunosignal of the end-product of lipid peroxidation (LPO), 4-hydroxynonenal (4-HNE), started to increase on day 10. TNF-α, 4-HNE, and erastin (LPO inducer), activated Nrf2 signaling in human ECs by increasing the mRNA and protein expression of Nrf2 target genes. Knockdown of endothelial Nrf2 resulted in augmented endothelial inflammation and LPO, and accelerated atherosclerosis in Cdh5CreCas9floxed-stopApoE-/- mice. By contrast, both EC-specific and pharmacological activation of Nrf2 inhibited endothelial inflammation, LPO, and atherogenesis. CONCLUSIONS Upon HCD feeding in ApoE-/- mice, endothelial inflammation is an earliest event, followed by the appearance of LPO. EC-specific activation of Nrf2 inhibits atherosclerosis while EC-specific knockdown of Nrf2 results in the opposite effect. Pharmacological activators of endothelial Nrf2 may represent a novel therapeutic strategy for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Lei He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China; School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, PR China
| | - Qinghua Chen
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - Yujie Pu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - Juan Huang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, PR China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, PR China
| | - Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - Jiang-Yun Luo
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, PR China
| | - Lijing Kang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - Xiao Lin
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Li Xiang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, PR China
| | - Liang Fang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yin Xia
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, PR China
| | - Kathy O Lui
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Yong Pan
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, PR China
| | - Jie Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, PR China
| | - Cheng-Lin Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, PR China.
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China; School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, PR China.
| |
Collapse
|
48
|
Lin H, Wang L, Jiang X, Wang J. Glutathione dynamics in subcellular compartments and implications for drug development. Curr Opin Chem Biol 2024; 81:102505. [PMID: 39053236 PMCID: PMC11722958 DOI: 10.1016/j.cbpa.2024.102505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/30/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
Glutathione (GSH) is a pivotal tripeptide antioxidant essential for maintaining cellular redox homeostasis and regulating diverse cellular processes. Subcellular compartmentalization of GSH underscores its multifaceted roles across various organelles including the cytosol, mitochondria, endoplasmic reticulum, and nucleus, each exhibiting distinct regulatory mechanisms. Perturbations in GSH dynamics contribute to pathophysiological conditions, emphasizing the clinical significance of understanding its intricate regulation. This review consolidates current knowledge on subcellular GSH dynamics, highlighting its implications in drug development, particularly in covalent drug design and antitumor strategies targeting intracellular GSH levels. Challenges and future directions in deciphering subcellular GSH dynamics are discussed, advocating for innovative methodologies to advance our comprehension and facilitate the development of precise therapeutic interventions based on GSH modulation.
Collapse
Affiliation(s)
- Hanfeng Lin
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; Center for NextGen Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lingfei Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiqian Jiang
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jin Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; Center for NextGen Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
49
|
Borozdina NA, Dyachenko IA, Popkova DV. Promising Directions for Regulating Signaling Pathways Involved in the Type 2 Diabetes Mellitus Development (A Review). RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2024; 50:1263-1284. [DOI: 10.1134/s1068162024040137] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/11/2023] [Accepted: 11/12/2023] [Indexed: 01/04/2025]
|
50
|
Hamblet C, Björhall K, Busch S, Gehrmann U, Öberg L, Kubisch-Dohmen R, Haas S, Aneja MK, Geiger J, Rudolph C, Hornberg E. Transcriptional Dynamics of NRF2 Overexpression and KEAP1-NRF2 Inhibitors in Human Cell Line and Primary Lung Cells. Antioxidants (Basel) 2024; 13:924. [PMID: 39199170 PMCID: PMC11351141 DOI: 10.3390/antiox13080924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Oxidative stress in the human lung is caused by both internal (e.g., inflammation) and external stressors (smoking, pollution, and infection) to drive pathology in a number of lung diseases. Cellular damage caused by oxidative damage is reversed by several pathways, one of which is the antioxidant response. This response is regulated by the transcriptional factor NRF2, which has the ability to regulate the transcription of more than 250 genes. In disease, this balance is overwhelmed, and the cells are unable to return to homeostasis. Several pharmacological approaches aim to improve the antioxidant capacity by inhibiting the interaction of NRF2 with its key cytosolic inhibitor, KEAP1. Here, we evaluate an alternative approach by overexpressing NRF2 from chemically modified RNAs (cmRNAs). Our results demonstrate successful expression of functional NRF2 protein in human cell lines and primary cells. We establish a kinetic transcriptomic profile to compare antioxidant response gene expression after treatment of primary human bronchial epithelial cells with either KEAP1 inhibitors or cmRNAs. The key gene signature is then applied to primary human lung fibroblasts and alveolar macrophages to uncover transcriptional preferences in each cell system. This study provides a foundation for the understanding of NRF2 dynamics in the human lung and provides initial evidence of alternative ways for pharmacological interference.
Collapse
Affiliation(s)
- Corinne Hamblet
- Bioscience Chronic Obstructive Pulmonary Disease & Idiopathic Pulmonary Fibrosis, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, 431 83 Mölndal, Sweden
| | - Karin Björhall
- Bioscience Chronic Obstructive Pulmonary Disease & Idiopathic Pulmonary Fibrosis, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, 431 83 Mölndal, Sweden
| | - Susann Busch
- Respiratory & Immunology, Neuroscience, Vaccines and Immune Therapies Safety, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 431 83 Mölndal, Sweden
| | - Ulf Gehrmann
- Translational Science and Experimental Medicine Research, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, 431 83 Mölndal, Sweden
| | - Lisa Öberg
- Translational Science and Experimental Medicine Research, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, 431 83 Mölndal, Sweden
| | | | | | | | | | | | - Ellinor Hornberg
- Projects and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, 431 83 Mölndal, Sweden
| |
Collapse
|