Role of Nrf2 in Disease: Novel Molecular Mechanisms and Therapeutic Approaches

A special issue of Biomolecules (ISSN 2218-273X). This special issue belongs to the section "Cellular Biochemistry".

Deadline for manuscript submissions: closed (31 August 2020) | Viewed by 42775

Special Issue Editor


E-Mail Website
Guest Editor
1. Instituto de Investigaciones Biomédicas “Sols-Moreale” UAM-CSIC, c/Arturo Duperier 4, 28029 Madrid, Spain
2. Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain
3. Institute Teófilo Hernando for Drug Discovery, Universidad Autónoma de Madrid, 28029 Madrid, Spain
4. Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
Interests: neurodegenerative diseases; NRF2; oxidative stress; neuroinflammation; RNA-binding proteins; mitochondria
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Since it was cloned and characterized, the transcription factor (nuclear factor erythroid-2-related factor 2) NRF2 has been implicated in processes associated with redox balance, inflammation, proteostasis and lipids, purines and pentoses metabolism, becoming a pleiotropic transcription factor. For this reason, NRF2 has developed as a polyvalent target against various pathologies, where the signaling system of NRF2 is altered. Under normal conditions, basal NRF2 levels are low due to its interaction to KEAP1 (Kelch-like ECH-associated protein 1) that binds to and negatively regulates NRF2. Electrophiles or oxidative stress induce the inactivation of KEAP1 by direct modification of reactive cysteine residues, leading to the release and stabilization of NRF2, that translocates to the nucleus to bind to the antioxidant response element (ARE) sequence in the promoter regions of NRF2-dependent genes. This system makes it a good pharmacological target to modulate the activation of NRF2 and, therefore, its application in various pathologies.

This Special Issue on NRF2 should, on one hand, emphasize the importance of this transcription factor, and, on the other hand, it should also highlight existing pharmacological components that can modify the NRF2 signaling pathway. In this regard, we would like to invite review articles which address the above-mentioned issues from different perspectives. Alternatively, any original research papers contributing significantly to NRF2 signaling progress or advancing our understanding of biological implications are highly welcome. We look forward to reading your contributions.

Prof. Isabel Lastres-Becker
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Biomolecules is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • inflammation
  • autophagy
  • proteasome
  • proteostasis
  • oxidative stress
  • neurodegeneration
  • metabolism
  • KEAP1
  • redox balance

Related Special Issue

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

2 pages, 173 KiB  
Editorial
Special Issue “Role of NRF2 in Disease: Novel Molecular Mechanisms and Therapeutic Approaches”
by Isabel Lastres-Becker
Biomolecules 2021, 11(2), 202; https://doi.org/10.3390/biom11020202 - 02 Feb 2021
Cited by 1 | Viewed by 1446
Abstract
This Special Issue on NRF2 (https://www [...] Full article

Research

Jump to: Editorial, Review

16 pages, 2431 KiB  
Article
Gene Expression Profiling of Multiple Histone Deacetylases (HDAC) and Its Correlation with NRF2-Mediated Redox Regulation in the Pathogenesis of Diabetic Foot Ulcers
by Rajan Teena, Umapathy Dhamodharan, Daoud Ali, Kesavan Rajesh and Kunka Mohanram Ramkumar
Biomolecules 2020, 10(10), 1466; https://doi.org/10.3390/biom10101466 - 21 Oct 2020
Cited by 19 | Viewed by 3365
Abstract
Nuclear factor erythroid-2-related factor 2 (Nrf2) is a protein of the leucine zipper family, which mitigates inflammation and employs cytoprotective effects. Attempting to unravel the epigenetic regulation of type 2 diabetes mellitus (T2DM) and diabetic foot ulcer (DFU), we profiled the expression of [...] Read more.
Nuclear factor erythroid-2-related factor 2 (Nrf2) is a protein of the leucine zipper family, which mitigates inflammation and employs cytoprotective effects. Attempting to unravel the epigenetic regulation of type 2 diabetes mellitus (T2DM) and diabetic foot ulcer (DFU), we profiled the expression of eleven isoform-specific histone deacetylases (HDACs) and correlated them with NRF2 and cytokines. This study recruited a total of 60 subjects and categorized into DFU patients (n = 20), T2DM patients (n = 20), and healthy controls (n = 20). The DFU patients were subcategorized into uninfected and infected DFU (n = 10 each). We observed a progressive decline in the expression of NRF2 and its downstream targets among T2DM and DFU subjects. The inflammatory markers IL-6 and TNF-α were significantly upregulated, whereas anti-inflammatory marker IL-10 was significantly downregulated in DFU. Of note, a significant upregulation of HDAC1, 3, 4, 11, SIRT3 and downregulation of HDAC2,8, SIRT1, SIRT2, SIRT3, SIRT7 among DFU patients were observed. The significant positive correlation between NRF2 and SIRT1 in DFU patients suggested the vital role of NRF2/SIRT1 in redox homeostasis and angiogenesis. In contrast, the significant negative correlation between NRF2 and HDAC1, 3 and 4, implied an imbalance in NRF2-HDAC1, 3, 4 circuit. Furthermore, a significant positive correlation was observed between HDAC4 and IL-6, and the negative correlation between SIRT1 and IL-6 suggested the pro-inflammatory role of HDAC4 and the anti-inflammatory role of SIRT1 in NRF2 signaling. In conclusion, the epigenetic changes such as upregulation of HDAC1, 3, 4, 11, SIRT3 and downregulation of HDAC2, 8, SIRT1, SIRT2, SIRT6, SIRT7 and their association with NRF2 as well as inflammatory markers are suggestive of their roles in pathophysiology of T2DM and DFU. Full article
Show Figures

Graphical abstract

16 pages, 2809 KiB  
Communication
High NRF2 Levels Correlate with Poor Prognosis in Colorectal Cancer Patients and with Sensitivity to the Kinase Inhibitor AT9283 In Vitro
by Laura Torrente, Gunjit Maan, Asma Oumkaltoum Rezig, Jean Quinn, Angus Jackson, Andrea Grilli, Laura Casares, Ying Zhang, Evgeny Kulesskiy, Jani Saarela, Silvio Bicciato, Joanne Edwards, Albena T. Dinkova-Kostova and Laureano de la Vega
Biomolecules 2020, 10(10), 1365; https://doi.org/10.3390/biom10101365 - 25 Sep 2020
Cited by 22 | Viewed by 3539
Abstract
Aberrant hyperactivation of nuclear factor erythroid 2 (NF-E2) p45-related factor 2 (NRF2) is a common event in many tumour types and associates with resistance to therapy and poor patient prognosis; however, its relevance in colorectal tumours is not well-established. Measuring the expression of [...] Read more.
Aberrant hyperactivation of nuclear factor erythroid 2 (NF-E2) p45-related factor 2 (NRF2) is a common event in many tumour types and associates with resistance to therapy and poor patient prognosis; however, its relevance in colorectal tumours is not well-established. Measuring the expression of surrogate genes for NRF2 activity in silico, in combination with validation in patients’ samples, we show that the NRF2 pathway is upregulated in colorectal tumours and that high levels of nuclear NRF2 correlate with a poor patient prognosis. These results highlight the need to overcome the protection provided by NRF2 and present an opportunity to selectively kill cancer cells with hyperactive NRF2. Exploiting the CRISPR/Cas9 technology, we generated colorectal cancer cell lines with hyperactive NRF2 and used them to perform a drug screen. We identified AT9283, an Aurora kinase inhibitor, for its selectivity towards killing cancer cells with hyperactive NRF2 as a consequence to either genetic or pharmacological activation. Our results show that hyperactivation of NRF2 in colorectal cancer cells might present a vulnerability that could potentially be therapeutically exploited by using the Aurora kinase inhibitor AT9283. Full article
Show Figures

Figure 1

Review

Jump to: Editorial, Research

22 pages, 1545 KiB  
Review
Nrf2—A Molecular Target for Sepsis Patients in Critical Care
by Sandra Gunne, Ulrike Heinicke, Michael J. Parnham, Volker Laux, Kai Zacharowski and Andreas von Knethen
Biomolecules 2020, 10(12), 1688; https://doi.org/10.3390/biom10121688 - 17 Dec 2020
Cited by 15 | Viewed by 3213
Abstract
The transcription factor NF-E2 p45-related factor 2 (Nrf2) is an established master regulator of the anti-oxidative and detoxifying cellular response. Thus, a role in inflammatory diseases associated with the generation of large amounts of reactive oxygen species (ROS) seems obvious. In line with [...] Read more.
The transcription factor NF-E2 p45-related factor 2 (Nrf2) is an established master regulator of the anti-oxidative and detoxifying cellular response. Thus, a role in inflammatory diseases associated with the generation of large amounts of reactive oxygen species (ROS) seems obvious. In line with this, data obtained in cell culture experiments and preclinical settings have shown that Nrf2 is important in regulating target genes that are necessary to ensure cellular redox balance. Additionally, Nrf2 is involved in the induction of phase II drug metabolizing enzymes, which are important both in degrading and converting drugs into active forms, and into putative carcinogens. Therefore, Nrf2 has also been implicated in tumorigenesis. This must be kept in mind when new therapy approaches are planned for the treatment of sepsis. Therefore, this review highlights the function of Nrf2 in sepsis with a special focus on the translation of rodent-based results into sepsis patients in the intensive care unit (ICU). Full article
Show Figures

Figure 1

38 pages, 10270 KiB  
Review
NRF2 Regulation Processes as a Source of Potential Drug Targets against Neurodegenerative Diseases
by Ángel Cores, Marta Piquero, Mercedes Villacampa, Rafael León and J. Carlos Menéndez
Biomolecules 2020, 10(6), 904; https://doi.org/10.3390/biom10060904 - 14 Jun 2020
Cited by 51 | Viewed by 5295
Abstract
NRF2 acts by controlling gene expression, being the master regulator of the Phase II antioxidant response, and also being key to the control of neuroinflammation. NRF2 activity is regulated at several levels, including protein degradation by the proteasome, transcription, and post-transcription. The purpose [...] Read more.
NRF2 acts by controlling gene expression, being the master regulator of the Phase II antioxidant response, and also being key to the control of neuroinflammation. NRF2 activity is regulated at several levels, including protein degradation by the proteasome, transcription, and post-transcription. The purpose of this review is to offer a concise and critical overview of the main mechanisms of NRF2 regulation and their actual or potential use as targets for the treatment of neurodegenerative diseases. Full article
Show Figures

Figure 1

28 pages, 1235 KiB  
Review
The NRF2/KEAP1 Axis in the Regulation of Tumor Metabolism: Mechanisms and Therapeutic Perspectives
by Emiliano Panieri, Pelin Telkoparan-Akillilar, Sibel Suzen and Luciano Saso
Biomolecules 2020, 10(5), 791; https://doi.org/10.3390/biom10050791 - 20 May 2020
Cited by 52 | Viewed by 5609
Abstract
The NRF2/KEAP1 pathway is a fundamental signaling cascade that controls multiple cytoprotective responses through the induction of a complex transcriptional program that ultimately renders cancer cells resistant to oxidative, metabolic and therapeutic stress. Interestingly, accumulating evidence in recent years has indicated that metabolic [...] Read more.
The NRF2/KEAP1 pathway is a fundamental signaling cascade that controls multiple cytoprotective responses through the induction of a complex transcriptional program that ultimately renders cancer cells resistant to oxidative, metabolic and therapeutic stress. Interestingly, accumulating evidence in recent years has indicated that metabolic reprogramming is closely interrelated with the regulation of redox homeostasis, suggesting that the disruption of NRF2 signaling might represent a valid therapeutic strategy against a variety of solid and hematologic cancers. These aspects will be the focus of the present review. Full article
Show Figures

Figure 1

32 pages, 1642 KiB  
Review
Oxidative Stress in DNA Repeat Expansion Disorders: A Focus on NRF2 Signaling Involvement
by Piergiorgio La Rosa, Sara Petrillo, Enrico Silvio Bertini and Fiorella Piemonte
Biomolecules 2020, 10(5), 702; https://doi.org/10.3390/biom10050702 - 01 May 2020
Cited by 18 | Viewed by 4715
Abstract
DNA repeat expansion disorders are a group of neuromuscular and neurodegenerative diseases that arise from the inheritance of long tracts of nucleotide repetitions, located in the regulatory region, introns, or inside the coding sequence of a gene. Although loss of protein expression and/or [...] Read more.
DNA repeat expansion disorders are a group of neuromuscular and neurodegenerative diseases that arise from the inheritance of long tracts of nucleotide repetitions, located in the regulatory region, introns, or inside the coding sequence of a gene. Although loss of protein expression and/or the gain of function of its transcribed mRNA or translated product represent the major pathogenic effect of these pathologies, mitochondrial dysfunction and imbalance in redox homeostasis are reported as common features in these disorders, deeply affecting their severity and progression. In this review, we examine the role that the redox imbalance plays in the pathological mechanisms of DNA expansion disorders and the recent advances on antioxidant treatments, particularly focusing on the expression and the activity of the transcription factor NRF2, the main cellular regulator of the antioxidant response. Full article
Show Figures

Figure 1

20 pages, 2139 KiB  
Review
Electrophiles against (Skin) Diseases: More Than Nrf2
by Paulina Hennig, Gabriele Fenini, Michela Di Filippo and Hans-Dietmar Beer
Biomolecules 2020, 10(2), 271; https://doi.org/10.3390/biom10020271 - 11 Feb 2020
Cited by 20 | Viewed by 4046
Abstract
The skin represents an indispensable barrier between the organism and the environment and is the first line of defense against exogenous insults. The transcription factor NRF2 is a central regulator of cytoprotection and stress resistance. NRF2 is activated in response to oxidative stress [...] Read more.
The skin represents an indispensable barrier between the organism and the environment and is the first line of defense against exogenous insults. The transcription factor NRF2 is a central regulator of cytoprotection and stress resistance. NRF2 is activated in response to oxidative stress by reactive oxygen species (ROS) and electrophiles. These electrophiles oxidize specific cysteine residues of the NRF2 inhibitor KEAP1, leading to KEAP1 inactivation and, subsequently, NRF2 activation. As oxidative stress is associated with inflammation, the NRF2 pathway plays important roles in the pathogenesis of common inflammatory diseases and cancer in many tissues and organs, including the skin. The electrophile and NRF2 activator dimethyl fumarate (DMF) is an established and efficient drug for patients suffering from the common inflammatory skin disease psoriasis and the neuro-inflammatory disease multiple sclerosis (MS). In this review, we discuss possible molecular mechanisms underlying the therapeutic activity of DMF and other NRF2 activators. Recent evidence suggests that electrophiles not only activate NRF2, but also target other inflammation-associated pathways including the transcription factor NF-κB and the multi-protein complexes termed inflammasomes. Inflammasomes are central regulators of inflammation and are involved in many inflammatory conditions. Most importantly, the NRF2 and inflammasome pathways are connected at different levels, mainly antagonistically. Full article
Show Figures

Figure 1

30 pages, 1330 KiB  
Review
The Role of Reactive Oxygen Species in Arsenic Toxicity
by Yuxin Hu, Jin Li, Bin Lou, Ruirui Wu, Gang Wang, Chunwei Lu, Huihui Wang, Jingbo Pi and Yuanyuan Xu
Biomolecules 2020, 10(2), 240; https://doi.org/10.3390/biom10020240 - 05 Feb 2020
Cited by 198 | Viewed by 9932
Abstract
Arsenic poisoning is a global health problem. Chronic exposure to arsenic has been associated with the development of a wide range of diseases and health problems in humans. Arsenic exposure induces the generation of intracellular reactive oxygen species (ROS), which mediate multiple changes [...] Read more.
Arsenic poisoning is a global health problem. Chronic exposure to arsenic has been associated with the development of a wide range of diseases and health problems in humans. Arsenic exposure induces the generation of intracellular reactive oxygen species (ROS), which mediate multiple changes to cell behavior by altering signaling pathways and epigenetic modifications, or cause direct oxidative damage to molecules. Antioxidants with the potential to reduce ROS levels have been shown to ameliorate arsenic-induced lesions. However, emerging evidence suggests that constructive activation of antioxidative pathways and decreased ROS levels contribute to chronic arsenic toxicity in some cases. This review details the pathways involved in arsenic-induced redox imbalance, as well as current studies on prophylaxis and treatment strategies using antioxidants. Full article
Show Figures

Figure 1

Back to TopTop