8th BenBedPhar Scientific meeting

Thirty years since the discovery of NRF2

Location: Lisbon, Portugal

Participants: members of the Management Committee and Work Groups of the COST Action CA20121

Contact person

Dr Sandra Tenreiro
Email: stenreiro@nms.unl.pt

Dr Fernando Antunes
Email: fantunes@fc.ul.pt

Overview.

We are excited to announce the 8th BenBedPhar Meeting, themed “Thirty Years since the Discovery of NRF2.” This scientific gathering serves as a crucial platform for experts in the field to convene and discuss advancements in understanding the NRF2 pathway’s role in health and disease.

Celebrating thirty years of NRF2 research, the meeting will present and discuss a wide spectrum of recent developments. Topics will include NRF2’s role in brain diseases, oxidative stress, inflammation, and metabolism, as well as advances in new therapeutic approaches, drug discovery, and the development of innovative research tools.

With the ultimate goal of facilitating critical connections necessary for advancing the NRF2 field, BenBedPhar continues its mission to provide a dynamic space for researchers to collaborate, share findings, and foster connections across various life science disciplines. We remain steadfast in our commitment to promoting young researchers and encourage them to present their work.

Organizers.

Sandra Teneiro

Sandra Tenreiro

Fernando Antunes

Fernando Antunes

Preliminary program:

Thursday, October 10, 2024

O.01 | Unexpected cytotoxicity of Geldanamycin-based Heat Shock Protein  90 (HSP90) inhibitors in the context of NAD(P)H:Quinone Oxidoreductase 1  (NQO1) deficiency

Adele Valentova, Amy Bankier, Isavella Petrou, Oliver Read, Albena T. Dinkova-Kostova

Division of Cellular and Systems Medicine, University of Dundee School of Medicine, Dundee, United Kingdom
e-mail: a.dinkovakostova@dundee.ac.uk

Recent studies have demonstrated that geldanamycin-derived inhibitors of heat shock protein 90 (HSP90) are synthetic lethal with Nrf2 activation due to the enzymatic conversion of their quinone forms to more potent hydroquinones, which is catalyzed by the classical Nrf2 target NAD(P)H:quinone oxidoreductase 1 (NQO1). This study focused on exploiting these inhibitors as a potential synthetic lethal treatment strategy for glioblastoma multiforme (GBM), a highly aggressive brain tumor with limited treatment options and a poor prognosis. We aimed to test the hypothesis that geldanamycin-derived HSP90 inhibitors, such as 17-N
allylamino-17-demethoxygeldanamycin (17-AAG) and 17-dimethylaminoethylamino-17- demethoxygeldanamycin (17-DMAG), combined with Nrf2 activation and high levels of NQO1, increase sensitivity to cell death. We found that pharmacological Nrf2 activation, and consequently higher levels of NQO1, increased the cytotoxicity of 17-AAG and 17-DMAG, as expected. Importantly, this was despite the activation of the cytoprotective heat shock response, a common limitation of HSP90 inhibition. However, contrary to expectations, the cytotoxicity of 17-AAG and 17-DMAG was high in GBM cells deficient of NQO1. These results demonstrate the protective role of NQO1 against quinone-induced toxicity and further highlight the complexity of factors that influence the sensitivity to HSP90 inhibitors.
Funding: Medical Research Council UK (MR/W023806/1).

Albena T. Dinkova-Kostovais a Professor of Chemical Biology at the University of Dundee School of Medicine (UK). She graduated in Biochemistry and Microbiology from Sofia University (Bulgaria) and obtained her PhD degree in Biochemistry and Biophysics from Washington State University (USA). She subsequently trained in Pharmacology at Johns Hopkins University (USA), where she continues to hold an Adjunct Professor position. She joined the University of Dundee in 2007 as a Research Councils UK Academic Fellow and a research group leader. Her group collaborates with basic scientists and clinicians, and with the pharmaceutical industry. In her research, at the interface of Chemical Biology and Medicine, she aims to understand how cells and organisms respond to oxidative, inflammatory, and metabolic stress, and to develop strategies for protection against chronic disease. She was named among the top influential academics in Clarivate’s Highly Cited Researchers 2019, 2020, 2021, 2022 and 2023 lists.

O.02 | Endothelial miR-34a deletion guards NRF2 KO mice against aneurysm  development

Aleksandra Kopacz1, Damian Kloska1, Anna Bar2, Marta Targosz-Korecka3, Dominik Cysewski4, Agnieszka Mozdzierz1, Stefan Chlopicki2, Alicja Jozkowicz1, Anna Grochot Przeczek1

1 Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
2 Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland. 3 Department of Physics of Nanostructures and Nanotechnology, Institute of Physics, Jagiellonian University, Krakow, Poland.
4 Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.

e-mail: anna.grochot-przeczek@uj.edu.pl

An abdominal aortic aneurysm (AAA) is a life-threatening, age-associated dilatation of the abdominal aorta. Endothelial cell (EC) dysfunction is proposed to play a role in AAA formation. NRF2 and miR-34a are crucial regulators of ageing and EC biology. Thus, we hypothesized that they might have a decisive influence on the physiology of blood vessels and AAA development. NRF2 transcriptional deficiency in mice (NRF2 tKO) caused ultrastructural changes in the aorta and led to its premature ageing. Formation of AAA was favored in NRF2 tKO animals compared to wild-type counterparts (NRF2 WT). The level of miR-34a increased in the EC layer of NRF2 tKO aorta, and in response to treatment with angiotensin II (Ang II), a hypertensive peptide, in serum and ECs. We efficiently rescued EC NRF2-dependent premature ageing and AAA formation in NRF2 tKO mice using the EC specific knockout of miRNA-34a, which implies the significance of NRF2, miR-34a and intimal layer in the susceptibility to AAA. However, contrary to previously postulated mechanisms, in our hands the maintenance of specialized functions of ECs was not the primary determinant of the aneurysm formation. We propose instead that EC proliferation protects against AAA and can confer aneurysm stability.
Funding: National Science Centre grants 2016/22/E/NZ3/00405 and 2021/43/B/NZ4/02130.

Anna Grochot-Przeczek is an associate professor in the Department of  Medical Biotechnology, Faculty of Biochemistry, Biophysics, and Biotechnology,  Jagiellonian University in Krakow, Poland. She studies the molecular  mechanisms that regulate the function of endothelial cells and blood vessels  with a focus on the NRF2/KEAP1 pathway, ageing, and protein S-nitrosation.  Currently, she investigates the importance of NRF2/KEAP1 imbalance and loss  of proteostasis in blood vessel function.

 

O.03 | A preclinical approach to explore the NRF2 pathway as a therapeutic  strategy in age-related macular degeneration 

Ana S. Falcão, Margarida Pedro, Luisa de Lemos, Shuvajit Rakshit, Pedro Antas, Sandra Tenreiro and Miguel C. Seabra

iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa; 1169-056 Lisboa, Portugal.

e-mail: miguel.seabra@nms.unl.pt 

Age-related macular degeneration (AMD) is the most common blinding disease in the  western world and is currently incurable. Although the exact causes of AMD are not clear, the  primary origin of pathology appears to be in the retinal pigment epithelium (RPE). These cells  are responsible for the daily digestion of photoreceptor outer segments (POS), which  imposes a heavy continuous burden on the lysosomal network. We have developed an in  vitro model that recapitulates some AMD features, where feeding human RPE monolayers  with porcine POS leads to accumulation of autofluorescent granules (AFG) similar to  lipofuscin in vivo. Evidence suggests that the undigested autofluorescent material is a source  of oxidative stress impairing RPE health. Our hypothesis is that induction of an anti-oxidative  stress response mediated by NRF2 could represent a therapeutic strategy in  early/intermediate AMD. Focusing on repurposing clinically approved drugs that are described to be pharmacological modulators of NRF2, we are testing these compounds in  our RPE AMD in vitro model and evaluating their ability to decrease AFG formation. To further  dissect the role of NRF2 in this model, we are using gene editing tools to develop a stable  NRF2 KO line using CRISPR/Cas9 technology in RPE cells, as well as an adeno-associated  virus (AAV)-mediated NRF2 gene augmentation. We are also optimizing an inducible mice  model of AMD, characterized by a single intraperitoneal administration of NaIO3, which will  be used to validate the in vitro data. Results on these AMD models will be presented and  discussed to validate Nrf2 as a new therapeutic target not only for AMD, but also for other  age-related diseases, contributing to healthier aging. 

Funding: Project funded by “La Caixa Foundation” (NASCENT HR22-00569). R&D unit  [iNOVA4Health] (UIDB/04462/2020 and UIDP/04462/2020) and LS4FUTURE Associated  Laboratory (LA/P/0087/2020) funded by FCT.

Miguel Seabra, MD, is a leading researcher in the field of cellular and molecular  medicine. MS holds a PhD from University of Texas Southwestern Medical  Center (1992), where he was an Assistant Professor, before moving to the  Faculty of Medicine, Imperial College London where he became Full Professor  and head of Section until 2007. MS is now Full Professor at the Medical School,  UNL, Portugal, where he coordinates research in Cellular and Molecular  Medicine and holds several chairs. More recently, he became Head of Global  Eye Initiative at Fundação Champalimaud. MS was president of Fundação para  a Ciência e a Tecnologia (FCT) from 2012 to 2015, the portuguese public  funding agency for science and research in Portugal.

O.04 | Quinolyl nitrone derivative protect from auditory cell oxidative  damage and noise-induced hearing loss 

Silvia Murillo-Cuesta1,2,3, Julio Contreras1,2,4, Mourad Chioua5, Jose Manuel Zubeldia2,6, Jose  Luis Marco-Contelles2,5, Isabel Varela-Nieto1,2,3 

1Institute for Biomedical Research “Alberto Sols”, Spanish National Research Council-Autonomous University  of Madrid (CSIC-UAM), 28029 Madrid, Spain. 
2 Centre for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III (ISCIII),  28029 Madrid, Spain. 
3 Hospital La Paz Institute for Health Research (IdiPAZ), 28029 Madrid, Spain. 
4 Anatomy and Embryology Department, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid,  Spain. 
5 Laboratory of Medicinal Chemistry, Institute of Organic Chemistry (CSIC), Juan de la Cierva 3, Madrid 29006,  Spain. 
6 Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain. 

e-mail:  i.varela.nieto@csic.es

Noise-induced hearing loss (NIHL) is one of the most common sensorineural hearing  deficits worldwide, being excessive oxidative stress one of the main mechanisms underlying  it. Protecting the cochlea from oxidative stress is thus an approach for the prevention and  repair of NIHL. A variety of antioxidant molecules, including N-acetyl-L-cysteine, acetyl-L carnitine and nitrone-based antioxidants (HPN-07 and 4-OHPBN), have been tested. NRF2  is a transcriptional activator of the defense mechanisms against oxidative stress widely  expressed in the cochlea, including inner and outer hair cells, supporting cells and spiral  ganglion neurons. Thus, NRF2 has become a potential new strategic target for the  development of hearing protection drugs. Here we evaluate the safety and efficacy of a novel  quinolyl nitrone (QN23) in HEI-OC1 cells and in a NIHL mouse model. Our results showed  that pretreatment and co-administration with QN23 of HEI-OC1 cell cultures improved cell  survival after H202 oxidative stress challenge. Similarly, systemic administration of QN23 to  mice was well-tolerated and reduced acute auditory threshold shifts one day after noise  exposure in a dose-dependent manner. Noise challenge modified cochlear gene expression of Nrf2, Ho1, Nqo1, Nox3, Nox4, Nlrp3 and Il1b, whilst treatment with QN23 normalized it. 
These results indicate that QN23 protects hearing from noise exposure by modulating  oxidative stress. 

Funding: MINECO/FEDER PID2020-115274RB-I00/AEI/10.13039/501100011033 to IV-N and SM-C;  0551-PSL-6-E NITROPROHEAR grants to IV-N and RED2022-134511-T RedInflama to JM-C. 

Prof. Isabel Varela-Nieto graduated and earned her doctorate in Chemistry, Biochemistry Section, at the University Complutense of Madrid (Spain). She has been a visiting guest scientist at the Medical Schools of Uppsala (FEBS Fellow, Sweden) and San Diego (MEC Sabbatical, USA). She is Professor at the CSIC and group leader at the CIBER of rare diseases (CIBERER, ISCIII) in Madrid. From the early 1990s she has been studying hearing neurobiology, myelinopathies and IGF-1 actions. She is currently the president of the Spanish Biochemistry and Molecular Biology Society (SEBBM) and a member of the Spanish ministerial commission for ISC. As SEBBM president represents Spain in FEBS and IUBMB.

O.05 | Bile acids as immunomodulators and NRF2 activators

Srđan Bjedov1, Goran Stegnjaić2, Suzana Stanisavljević2, Milica Lazarević2, Ivan Pilipović2, Marija Sakač1, Đorđe Miljković2

1 Department of Chemistry, Biochemistry, and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, 21000, Novi Sad, Serbia.
2 Department of Immunology, Institute for Biological Research “Siniša Stanković” – National Institute of the Republic of Serbia, University of Belgrade, Despota Stefana 142, 11000 Belgrade, Serbia.

e-mail: georgije_zw@yahoo.com

Recent studies have shown that bile acids have a profound effect on intestinal T cells, T helper (Th)17 and T regulatory (Treg) cells in particular. Th17 and Treg are essential for immune homeostasis in the gut, as their proper functioning is required for efficient elimination of pathogenic microorganisms, as well as for prevention of excessive inflammation. Moreover, intestinal Th17 and Treg have a decisive role in immune homeostasis at the systemic level, and their dysfunction has been implicated in the pathogenesis of immune
mediated diseases, including autoimmune and neurodegenerative diseases, to name but a few. We have recently designed a bile acid derivative SB140 with the increased electrophilic functionality in order to obtain an immunomodulatory compound with enhanced ability to activate NRF2. Indeed, SB140 was shown to exert potent immunomodulatory effects on encephalitogenic T cells and to increase expression of NRF2-regulated genes (https://doi.org/10.3390/ijms25137136). A possibility to use bile acids and their derivatives for the treatment of immune-mediated disorders will be discussed, with the focus on their actions performed through NRF2activation.

Funding: This research was funded by the Ministry of Science, Technological Development, and Innovation, Republic of Serbia (Contract No. 451-03-66/2024-03/200007, 451-03- 66/2024-03/ 200125 & 451-03-65/2024-03/200125).

Đorđe Miljković is a research professor (from 2008) at the Institute for Biological Research „Siniša Stanković“, University of Belgrade, where he is the head of the Department of Immunology (appointed in 2015) and leader of the Group for Neuroimmunology (from 2010). He obtained his PhD in immunology at the University of Belgrade in 2002. His main research interest is in autoimmunity, multiple sclerosis in particular. His recent and ongoing projects:
• Cellular and molecular mechanisms of recovery of rats from experimental autoimmune encephalomyelitis
• Characterization of cell death mechanisms in the central nervous system of rats suffering from experimental autoimmune encephalomyelitis
• Human gut microbiota transfer for novel insights into central nervous system autoimmunity pathogenesis
• The role of gut microbiota and gut immune cells in the CNS-directed autoimmunity induced in rats without the use of the complete Freund’s
adjuvant
• Modulation of gut ILC3 by a FFAR2 agonist for the treatment of autoimmune diseases

O.06 | Interplay between NRF2, AQP3 and AQP5 in breast cancer: A pathway to therapy resistance?

Monika Mlinarić1, Ivan Lučić1, Ilija Guteša2, Sanda Bubanović2, Lidija Milković1, Ana Čipak Gašparović1

1 Ruđer Bošković Institute, Zagreb, Croatia.
2 Univesity Hospital for Tumors, University Hospital Centre “Sestre milosrdnice”, Zagreb, Croatia.

e-mail: acipak@irb.hr

NRF2, an antioxidant transcription factor, plays a dual role in tumorigenesis. In normal cells, NRF2 enhances resistance to oxidative-stress-based therapies, thus promoting tumorigenesis. However, in cancer cells, NRF2 provides protection from oxidative-stress based therapy and is therefore pro-tumorigenic factor. Peroxiporins, members of aquaporin family, facilitate transport of hydrogen peroxide along with water and glycerol. By modulating H2O2 levels, peroxiporins modulate signaling pathways involved in tumor development and progression This study investigates the expression levels of NRF2, AQP3, and AQP5 in breast cancer tissues before and after therapy. In addition, we studied these relations using the breast cancer cell lines MCF7, SkBr3 and SUM159PT, and non-tumorigenic cell line MCF10A. Our results suggest that changes in the expression profile of one aquaporin can affect the expression of the other. In addition, NRF2 activation alters the expression pattern of AQP3. These results indicate a potential connection between different aquaporins, suggesting they should be studied together. Furthermore, NRF2 activation can influence the expression pattern of AQP3. These interesting findings suggest that aquaporins could be a factor triggered by cancer therapy and related to NRF2 activity. This represents a novel pathway that may contribute to breast cancer resistance.

Funding:
Croatian Science Foundation, Grant Number IP-2020-02-3617.

Ana Čipak Gašparović is a Senior Research Associate and Head of the Laboratory for Membrane Transport and Signaling, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia. Her research focuses on the role of oxidative stress and antioxidative response in the resistance to cancer treatment. Recently, her research included aquaporins in breast and colon cancer. Special emphasis is given to peroxiporins, specific aquaporins which, in addition to water and glycerol, channel hydrogen peroxide, and as a consequence contribute to oxidative and antioxidative response of the cell. She is interested in the regulation of NRF2 pathway in response to peroxiporins, and their influence on the development of therapy resistance.

O.07 | The role of NRF2 in the adverse effects of anticancer therapies: focus on cardio-oncology

Vera Marisa Costa1,2

1 Associate Laboratory i4HB – Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal.
2 UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.

e-mail: veramcosta@ff.up.pt 

Cancer treatment has improved significantly, leading to higher survivorship but also severe adverse effects. Cardiotoxicity is a leading cause of death among cancer survivors, with the probability of dying from cardiovascular disease increasing over time and sometimes even surpassing cancer mortality. Doxorubicin (DOX), a topoisomerase II inhibitor used in several cancers, causes both short- and long-term cardiotoxicity. In vivo, the roles of inflammation and antioxidant defenses on DOX’s cardiotoxicity were evaluated in adult and elderly CD-1 male mice after a 3-week exposure to DOX (cumulative dose 9 mg/kg). In adult mice, 1-week after DOX (short-term assessment), cardiac markers related to inflammation and redox homeostasis, such as TNFR2, glutathione peroxidase 1, catalase, and iNOS, increased. Five months after DOX administration (long-term effects), NRF2 and SOD2 expression increased, as well as myeloperoxidase, IL-33, and TNF-α. Elderly mice showed short-term (1 week post-DOX) cardiac histological damage and fibrosis, which persisting up to 2 months after DOX. One week after DOX, NF-κB p65 immunopositive cells, together with the expression of iNOS and IL-33, increased. Two months post-DOX, there was an increase in glutathione peroxidase 1 and Bax along with decreased NRF2, NF-κB p65, and myeloperoxidase. Although NRF2appears to be modulated by DOX, in cultured cardiac cells (AC16 cells), NRF2modulators did not prevent or increase DOX-induced cytotoxicity. Overall, our data indicate that oxidative-stress-linked pathways, namely the activation of NRF2, need to be addressed together with other pathways, such as inflammation, to find new therapeutic strategies to decrease the cardiotoxicity of cancer treatments.

Funding:
National funds from FCT – Fundação para a Ciência e a Tecnologia, I.P., supported this work in the scope of the projects UIDP/04378/2020 and UIDB/04378/2020 of the Research Unit on Applied Molecular Biosciences – UCIBIO, and of the project LA/P/0140/2020 of the Associate Laboratory Institute for Health and Bioeconomy – i4HB.

Ana Čipak Gašparović is a Senior Research Associate and Head of the Laboratory for Membrane Transport and Signaling, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia. Her research focuses on the role of oxidative stress and antioxidative response in the resistance to cancer treatment. Recently, her research included aquaporins in breast and colon cancer. Special emphasis is given to peroxiporins, specific aquaporins which, in addition to water and glycerol, channel hydrogen peroxide, and as a consequence contribute to oxidative and antioxidative response of the cell. She is interested in the regulation of NRF2 pathway in response to peroxiporins, and their influence on the development of therapy resistance.

O.08 | NRF2/HO-1 pathway regulates the anti-oxidant and anti-inflammatory response in UVB-exposed primary human keratinocytes

Agnes S. Klar1,2,3, Katarzyna Michalak-Micka1,2,3, Monika Nänni1,2,3, Thomas Biedermann1,2,3, Ueli Moehrlen2,3,4

1 Tissue Biology Research Unit, University Children’s Hospital Zurich, Zurich, Switzerland. 2 University of Zurich, Zurich, Switzerland.
3 Children’s Research Centre, University Children’s Hospital Zurich, Zurich, Switzerland. 4 Department of Surgery, University Children’s Hospital Zurich, Zurich, Switzerland.

e-mail: agnes.klar@kispi.uzh.ch

The NRF2/HO-1 pathway plays a crucial role in the anti-oxidant and inti-inflammatory responses in human skin, protecting from UVB-induced cell injury. Our results showed that both NRF2 and HO-1 expression was immediately and transiently up-regulated in primary human keratinocytes following a single dose of 250mJ/cm2 UVB. In addition, we observed a time-dependent upregulation of the NOTCH1 downstream target genes, including HES1, IRF6, p21, and ΔNp63, indicating that NOTCH1 is involved the initial defensive mechanisms against ROS following UVB-challenge. Further, we investigated the crosstalk mechanism between NRF2and NFκB in UVB-challenged keratinocytes, and detected the phosphorylation/activation of p65, one of the main NFκB subunits, up to 3h upon UVB irradiation following its downregulation after 8h. Further, we investigated NRF2signaling in a human pigmented dermo-epidermal skin substitutes (pigmDESS) after transplantation onto immunodeficient rats. UVB exposure of the epidermis activated in this model NFκB expression leading to elevated IL-1beta and IL-6 transcripts in epidermal keratinocytes of pigmDESS. Additionally, we detected in this model an immediate activation of the NRF2pathway, triggering upregulation of Notch1, and its downstream effectors, activating directly the initial defensive mechanisms against ROS following UVB-challenge. Our study provides evidence that NRF2 enhances the expression of the downstream effectors of Notch1 and activates NFκB. These findings highlight the importance of anti-ROS machinery upon UVB challenge and unravel the interplay between the distinct mechanisms within the human skin.

Funding:
This study was supported by research funding from the Swiss National Science Foundation (SNSF).

Agnes S. Klar graduated from the University of Konstanz, Germany with a master degree in Biological Sciences in 2009, and received her PhD from the University of Zurich in 2014. She leads her own group at the Tissue Biology Research Unit at the University Children’s Hospital Zurich since 2019. PI is a specialist in the field of regenerative medicine focusing on skin tissue engineering, wound healing, and skin diseases. In particular, her research interests are characterization and application of human skin cell-based analogs. In particular, her innovations contributed significantly to establish denovoSkinTM, a tissue-engineered product containing autologous keratinocytes and fibroblasts. This analog has recently successfully completed phase II clinical trials for burn injuries. Moreover, her research group conducts also studies combining aspects related to skin healing and scarring, during skin regeneration
following graft transplantation. Her scientific track record includes 48 peer-reviewed scientific publications and the current Hirsch index is 21 (̴1155 citations).

O.09 | Anti-inflammatory and cytoprotective actions of HYCOs, NRF2 activators that simultaneously release carbon monoxide

Roberta Foresti, Roberto Motterlini

Faculty of Health, University Paris-Est Créteil, INSERM U955, Créteil, France.

e-mail: roberta.foresti@inserm.fr

Heme oxygenase-1 (HO-1) is one of several targets proteins that are dependent on NRF2 activation. HO-1 degrades heme to carbon monoxide (CO) and biliverdin/bilirubin, which possess anti-inflammatory and antioxidant properties, respectively. To exploit the positive pharmacological effects of NRF2 activation and CO, we recently developed new hybrid molecules (HYCOs) consisting of CO-releasing molecules conjugated to fumaric esters known to activate NRF2/HO-1. Fourteen compounds were tested in human monocytes and keratinocytes in vitro as well as in vivo models of inflammation. The effects of HYCOs were compared to: a) dimethyl fumarate (DMF), a known fumaric ester used in the clinic and a CO-releasing molecule alone. Selected HYCOs efficiently donated CO, up-regulated N
NRF2rf2/HO-1 and elicited a strong reduction in anti-inflammatory markers in monocytes stimulated by LPS. This effect was stronger than that observed with DMF or CO-RM alone, indicating the enhanced potency of HYCOs compared to the separate entities. In vivo, HYCOs given orally to mice exerted beneficial effects by accelerating skin wound closure and reduced psoriasis-mediated inflammation. In new unpublished data we also show that HYCOs protect a human cardiac cell line from hypoxia-reoxygenation damage, as demonstrated by maintenance of cell viability, decrease in lactate dehydrogenase release and preservation of cellular myoglobin content. Importantly, HYCOs were given at the step of reoxygenation, recapitulating more closely a potential clinical setting. These results support the idea that HYCOs possessing a dual mode of action could be applied to combat inflammation and oxidative stress. 

Funding: Société d’Accélération de Transfert Technologique (SATT)-Ile de France Innovation, Fédération Française de Cardiologie, Fondation pour la Recherche Médicale.

Roberta Foresti is Professor of Biochemistry at the University Paris-Est Créteil and Responsible for International relations at the Mondor Institute of Biomedical research (IMRB). Dr Foresti has been interested for a long time on heme oxygenase-1 (HO-1), a gene regulated by the NRF2 system. In addition to basic research that explores the significance of HO-1 and its products carbon monoxide (CO) and bilirubin in biology and disease, she also works on drug discovery approaches that targets HO-1 and NRF2 in inflammatory and stress conditions. Recently, she has developed an interest on NRF2 and CO in obesity and metabolism.

O.10 | Possible role of P2X7 on NLRP3 inflammasome regulation by NRF2/DJ-1 pathway stimulation in diabetic nephropathy

María José Caballero-Herrero1, Laura Hurtado1, Cristina Molina1, Celisa Arias-Sanchez2, Esther Jumilla1, Pablo Pelegrin2, Julieta Schachter1, Santiago Cuevas1

1 BioMedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain.
2Immunology department, University of Murcia, Murcia, Spain.

e-mail: santicuevas@gmail.com

The inflammasome is an important regulator of the inflammatory response in renal diseases. The renal DJ-1 protein exhibits anti-inflammatory properties and is involved in the regulation of NRF2. To explore novel pharmacological applications, we designed ND-13, a peptide consisting of 13 highly conserved amino acids from the DJ-1 sequence. Peripheral blood mononuclear cells (PBMC) were isolated from diabetic nephropathy patients and controls. Mouse bone marrow macrophages (BMDM) were treated with ND-13. Diabetes was induced in C57Bl/6 mice by injection of streptozotocin (STZ) and treated with ND-13 or MCC950, a specific NLRP3 inflammasome inhibitor. PBMC from patients with diabetic nephropathy treated with LPS/ATP showed a tendency to increase IL-1β release compared to controls, suggesting a potential role of NLRP3 inflammasome in diabetic nephropathy pathogenesis. IL-1β levels in BMDM medium increased after NLRP3 inflammasome stimulation with LPS+ATP, and were significantly decreased after ND-13 pretreatment. STZ induced diabetes in mice significantly increased the mRNA expression of COL-I, COL-II, TGF-β, IL-6, TNF-α and P2X7 in the renal cortex, which was partially prevented by ND-13 and MCC950 pretreatment. IL-1β release increased in peritoneal macrophages, suggesting that the inflammasome is activated in diabetes, and ND-13 treatment normalised its activity. P2X7 mRNA expression was also increased in diabetic peritoneal macrophages.
Conclusions: ND-13 may attenuate the deleterious effects associated with inflammasome activation in renal disease. Furthermore, our data suggest that P2X7 may be involved in the molecular mechanism by which ND-13 exerts such a protective role by the attenuation of inflammasome activation in diabetic nephropathy.

Funding:
SC was funded by Fundación Séneca 21921/PI/22, Instituto de Salud Carlos III PI22/00129 co-funded by the European Union. Funding sources provided financial support but were not involved in study design, collection, analysis and interpretation of data.

Santiago Cuevas: 22 years of experience in the academy and industry in basic, translational and clinical research studying the pathways involved in the regulation of oxidative stress and inflammation in the pathogenesis of hypertension and renal diseases. Ten years of research experience in the United States on the field. At the present, I am a Principal Investigator Miguel Servet in the Institute of Biomedical Research in Murcia (IMIB). I am a team leader in the Unit of Molecular Inflammation at the IMIB, where we study the molecular mechanism involved in inflammasome regulation and its role in the pathogenesis of several renal and cardiovascular diseases, and determine the protective effects of new anti-inflammatory pharmacological approaches in diabetic nephropathy.

O.11 | Lupeol modulates redox homeostasis and mitigates oxysterol induced dysfunction in monocyte-derived dendritic cells via NRF2 pathway activation

Sarmistha Saha1, Elisabetta Profumo2, Luciano Saso3, Brigitta Buttari2

1 Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, Uttar Pradesh, India. sarmistha.saha@gla.ac.in
2 Department of Cardiovascular and Endocrine-metabolic Diseases, and Aging, Istituto Superiore di Sanità, Rome, Italy. elisabetta.profumo@iss.it
3 Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, Rome, Italy. luciano.saso@uniroma1.it

e-mail: brigitta.buttari@iss.it

The redox balance is crucial for dendritic cells (DCs) to function properly and protect the body from infections and even cancer. If this balance is altered, DCs may undergo morphological and functional changes, which can lead to chronic inflammation or immune deficiency diseases. Oxysterols, oxidized cholesterol derivatives, cause severe dysfunctions in cell organelles, especially mitochondria, thus influencing DC functions, from DC maturation to migration in age-related diseases such as cardiovascular and neurodegenerative diseases. Here, we investigated whether the natural compound lupeol, a pentacyclic triterpene, induces phenotypical and functional changes in human immature monocyte
derived DCs (moDCs) and counteracts the proinflammatory signaling triggered by the oxysterol 7-ketocholesterol (7KC). Flow cytometric and immunochemical analysis showed that the treatment with lupeol of moDCs for 20 h specifically induced a typical tolerogenic phenotype of reduced costimulatory molecules and low production of proinflammatory cytokines. Pretreatment of moDCs with lupeol prevented the release of IL-12p40, IL-1β, and the upregulation of CD86, CD40, HLA-DR and CCR7 expression triggered by 7KC. Lupeol exerted its influence on redox homeostasis by regulating the generation of reactive oxygen species and the functionality of the antioxidant system, by increasing the expression levels of NRF2, HO-1, and NQO1. The NRF2 antagonist ML385 treatment (5 µM for 16 h) abrogated the protective effects of lupeol on 7KC-treated moDCs. The present results indicate the potential of lupeol, which suppresses the redox alterations caused by oxysterols on DCs by activating the NRF2 pathway as an adjunctive drug in diseases that involve DC dysfunction.

Funding:
This work was supported by the Istituto Superiore di Sanità (ISS- Italian National Institute of Health), Italy C.R. 2024 to E.P. and B.B.

Brigitta Buttari is a senior researcher at Istituto Superiore di Sanità (ISS- Italian National Institute of Health) in Rome, with extensive experience in molecular and cellular biology, particularly in the immunological and cardiovascular fields. She has led significant research projects, including studies on the NRF2 pathway in noncommunicable diseases and the anti-inflammatory effects of natural compounds. Dr. Buttari has published over 90 articles in international journals, earning an H-index of 31. Her collaborative work spans multiple international projects, and she is recognized for her leadership in scientific research and contributions to academia.

O.12 | Oxidative lipid alterations and NRF2: Implications for synaptic preservation in Alzheimer’s disease

Daniel Carnicero-Senabre¹, Mariana A Barata², Cláudia Guimas Almeida², Antonio Cuadrado¹ and Ana I. Rojo¹

1 Department of Biochemistry and BIomedical Research Institute “Sols-Monreale”, Faculty of Medicine, Autonomous University of Madrid; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED); Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain. 2iNOVA4Health, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal.

e-mail: airojo@iib.uam.es

Neuronal circuits in Alzheimer’s disease (AD) are compromised by progressive synapse loss, leading to neuronal death. Understanding the molecular mechanisms preserving synaptic homeostasis could aid in developing drugs to halt neurodegeneration. This study focuses on how oxidative lipid alterations, essential for synaptic vesicle cycles, neurotransmitter release, and signaling pathways, impact synapse homeostasis. NRF2, a key mitigator of lipid peroxidation and modulator of lipid metabolic pathways, suggests that increasing NRF2 function may alleviate conditions like AD. We assessed 544 lipid species in NRF2-null and wild-type samples via untargeted lipidomics, finding that NRF2 deficiency disrupts several lipid species, including six ether-linked phospholipids associated with lipid peroxidation (LPO). To understand NRF2 deficiency’s impact on synapses, we analyzed colocalization of excitatory (vGLUT1 and PSD95) and inhibitory (vGAT and GEPHYRIN) synapse markers in brain slices and neuronal cultures. Our findings show that NRF2 deficiency, treatment with 4-Hydroxynonenal (4-HNE, an LPO-derived aldehyde), or sn-1-O hexadecylglycerol (HG, an ether-linked lipid precursor) alters synaptic molecular composition. Importantly, pharmacological activation of NRF2 with 6-(Methylsulfinyl) hexyl isothiocyanate (6-MSITC) prevented synaptic contact loss induced by HG. In summary, NRF2 is an essential regulator of synaptic homeostasis, indicating its potential as a therapeutic target for neurodegenerative diseases such as AD, which are characterized by progressive synaptic loss.

Funding: Spanish Ministry of Economy and Competitiveness (MINECO) (grants PID-2021- 122766OB-100 and PDC2022-133765-I00), CIBERned (ISCIII), Autonomous Community of Madrid (grant P2022/BMD-7230). This study is based upon work from COST Action CA20121, supported by COST (European Cooperation in Science and Technology) (www.cost.eu) (https://benbedphar.org/about-benbedphar/).

Ana I Rojo studied Biochemistry and Molecular Biology at the Autonomous University of Madrid (2001 and 2002), holds a PhD in Biochemistry (graduated in 2006), and since 2017 is professor in Biochemistry at the Autonomous University of Madrid (Faculty of Medicine). As professor, she has participated in multiple teaching activities for the degrees of Biochemistry, Medicine, and Nursing, with special focus on research training. She has been holder of different competitive fellowships and contracts. Her professional career is focused on the study of the molecular basis of neurodegenerative diseases and in the search for novel brain protective therapies with a special focus on redox biology and NRF2 transcription factor. Nowadays, she is exploring the role of NRF2 in the pathogenesis of Alzheimer’s disease and lateral amyotrophic sclerosis as principal investigator. She has published over 50 primary and review articles and participated in more than 30 congress.

Friday, October 11, 2024

O.13 | NRF2 modulators landscape: Patent challenges and strategies for improving accessibility

Alexandra-Klimovich Mickael1, Mariusz Sacharczuk1, Michel Edwar Mickael2

1 PM Research Center, Väpnaregatan 22, 58649 Linköping, Sweden.
2Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Ul. Postepu 36A, Jastrzębiec, 05-552, Magdalenka, Poland.

e-mail: michel.mickael77@gmail.com

Nrf2 is a pivotal transcription factor that regulates numerous genes and pathways, significantly influencing various diseases. The increasing acknowledgment of NRF2’ role as a critical therapeutic target is marred by the patent strategies of large pharmaceutical companies, which often lead to inflated drug prices through monopoly control, patent evergreening, and the establishment of patent thickets. These tactics result in drug costs far exceeding their production expenses, thereby restricting access. Currently available NRF2 modulators, such as Omaveloxolone (RTA 408), Dimethyl fumarate (Tecfidera), and Bardoxolone ethyl (CDDO-Me), hold promise for treating several conditions. However, the complex acquisition landscape complicates accessibility. For example, Biogen’s acquisition of Reata Pharmaceuticals for $7.3 billion illustrates the competitive and costly nature of pharmaceutical acquisitions. While Omaveloxolone has received FDA and EU approval for Friedreich’s ataxia, and Phase II data suggests that Larimar Therapeutics’ nomlabofusp could challenge Biogen’s Skyclarys, the overarching patent policies continue to impede fair access. To address these challenges, several solutions can be considered: reforming patent laws to limit excessive extensions and thickets, enhancing transparency in drug pricing, and incentivizing competition through regulatory measures. Additionally, promoting policies that support generic and biosimilar entry into the market could mitigate monopolistic practices. Such reforms are essential to balance innovation with equitable access, ensuring that the benefits of NRF2 modulators are accessible to patients in both high- and low-income regions.

Michel-Edwar Mickael is an associate professor at the Institute of Genetics and Animal Biotechnology near Warsaw, Poland. His lab is interested in the Th17/Treg axis and its implications in autoimmune diseases. Michel learned several experimental and computational techniques while doing Ph.D. and postdoc training at Durham University, Karolinska Institutet, Victor Babes Institute, UKE (Germany), and UAB (Alabama).

O.14 | NetRF2: A novel pipeline for network-based analysis of NRF2, and its targets, interactions, variants and expressions

Muazzez Celebi Cinar1, Aida Rezaei1, and Ozlen Konu1

1 Bilkent University, Department of Molecular Biology and Genetics, 06800 Ankara Turkey.

e-mail: konu@fen.bilkent.edu.tr

NRF2, encoded by NFE2L2 gene in humans, plays a centralized role in redox biology as well as non-redox functions. Although being widely investigated in a variety of fields that range from targeting drugs to biological pathways, all of the valuable information discovered has been stored within different databases. Here, we introduce a scalable pipeline to gather NRF2-related data and a user-friendly Shiny web application, called NetRF2, to enable users to obtain NRF2 relevant data in the form of reorganized tables and mono- and/or multi-partite networks. This web application provides researchers without strong programming backgrounds access to NRF2 variants in humans and model organisms, phenotypes associated with NRF2 variants, NRF2-targeting drugs, NRF2 targets, interactions, and pathways all in one user-friendly web interface. Beyond the gene-disease relation, NetRF2 gives causal variants for phenotypes gathering detailed data using Ensembl and GWAS databases. Moreover, with the integration of Human Protein Atlas (HPA) data, the user can obtain expression profiles of human genes under filtered conditions in both normal and cancer tissues while tissue-specifically observing NRF2 targets and interactions. Gathering NRF2-related data in one place, this study also has the potential to strengthen networking among multidisciplinary teams. Having the modularity for integration of additional types of data and resources, the NetRF2 pipeline can be adapted for other genes in the future.

Funding: The European Horizon’s research and innovation program HORIZON-HLTH-2022- STAYHLTH-02 under agreement No 101095679 and COST Action CA20121. Funded by the European Union. Views and opinions expressed are however those of the author(s) only and do not necessarily reflect those of the European Union. Neither the European Union nor the granting authority can be held responsible for them.

Dr. Özlen Konu graduated from METU in 1987 with a B.S. degree in Biological Sciences and pursued her graduate studies in the Biology Department at Texas Tech University, and received her M.S. and Ph.D. degrees in 1992 and 1999, respectively. She was a postdoctoral research fellow at the University of Tennessee at Memphis during 2000-2002. Since September 2002, she is a faculty member at the Dept. Molecular Biology and Genetics, Bilkent University in Turkey. Dr. Konu’s research interests include web tool development for comparative transcriptomics analyses of zebrafish and mammals in cancer cell signaling as well as development of methodologies for screens in zebrafish glioma and liver steatosis models.

O.15 | Carbon nano-onions for targeted drug delivery

Silvia Giordani1

1 School of Chemical Sciences, Dublin City University, Dublin, Ireland.

e-mail: silvia.giordani@dcu.ie

In this presentation, carbon nano-onions (CNOs) will be discussed as a potential vesicle for nanocarrier-type drug delivery systems.1 CNOs, or multi-layer fullerenes, consist of multiple concentric layers of sp2 hybridized carbon and are emerging as platforms for biomedical applications because of their ability to be internalized by cells and low toxicity.2 In my research group we have developed methodology for the synthesis of pure, monodispersed CNOs and various chemical functionalization strategies for the introduction of different functionalities (receptor targeting unit and imaging unit) onto the surface of the CNOs. The modified CNOs display high brightness and photostability in aqueous solutions and are selectively taken up by different cancer cell lines without significant cytotoxicity. Supramolecular functionalization with biocompatible polymers is an effective strategy to develop engineered drug carriers for targeted delivery applications. We reported the use of a hyaluronic acid-phospholipid (HA-DMPE) conjugate to target CD44 overexpressing cancer cells, while enhancing solubility of the nanoconstruct. Non-covalently functionalized CNOs with HA-DMPE show excellent in vitro cell viability in human breast carcinoma cells overexpressing CD44 and are uptaken to a greater extent compared to human ovarian carcinoma cells with an undetectable amount of CD44. In addition, they possess high in vivo biocompatibility in zebrafish during the different stages of development suggesting a high degree of biosafety of this class of nanomaterials.3 Our results encouraged us to further develop them as targeted diagnostics or therapeutics nanocarriers. We successfully loaded the CNO-based nanocarrier with chemotherapeutic prodrugs derived from gemcitabine, and showed remarkable efficacy in killing pancreatic adenocarcinoma cells.

Funding: Irish Research Council and Science Foundation Ireland.

Silvia Giordani joined the School of Chemical Sciences at Dublin City University as Professor Chair of Nanomaterials in 2018. Previously she received her PhD in Chemistry from the University of Miami, USA and carried out postdoctoral research at Trinity College Dublin (TCD) and at the University of Trieste, Italy. In 2007 she received the prestigious President of Ireland Young Researcher Award and was a Research Assistant Professor at TCD from 2007 to 2013. In 2013 she founded and directed the new “Nano Carbon Materials” research lab at the Istituto Italiano di Tecnologia (IIT) and in December 2016 she was appointed Associate Professor in Organic Chemistry at the University of Turin, Italy. Her main research interests are in the design, synthesis, and characterization of a wide range of nanomaterials for applications in smart and responsive bio-related nanotechnologies. She is the author/co-author of more than 160 manuscripts, reviews and book chapters. She is the recipient of many international prizes and honours including the L’Oreal UNESCO for Women in Science fellowship, the William Evans visiting fellowship from the University of Otago (New Zealand) and was a Visiting Scientist to the Bio Nano Institute at Toyo University (Japan).

O.16 | How to study NRF2 isoforms?

Alicja Dziadosz1&, Sara Mikac1&, Artur Piróg1, Zuzanna Urban-Wójciuk, Monikaben Padariya, Umesh Kalathiya, Sachin Kote and Alicja Sznarkowska1

1 University of Gdansk, International Centre for Cancer Vaccine Science, Kladki 24, 80-822, Gdansk, Poland. & equal contribution.

e-mail: alicja.sznarkowska@ug.edu.pl

NRF2 gene (NFE2L2) gives rise to several different transcripts due to alternative splicing and existence of two promoters. Transcript 2, originating from second promoter, is the second highest expressed transcript, after the transcript 1, encoding the full-length NRF2protein of 605 amino acids. Transcript 2 gives rise to NRF2 isoform 2, which is 16 amino acids shorter at the N-terminus than the full-length form. It is challenging to differentiate between these isoforms in SDS-PAGE due to little mass difference and problematic detection of NRF2 with antibodies, which will be discussed together with mass-spectrometry based ways of NRF2 identification.

Funding: This work is supported by Polish National Science Centre SONATA nr 2021/43/D/NZ1/02059 and International Centre for Cancer Vaccine Science, University of Gdansk (Fundacja na rzecz Nauki Polskiej: MAB/3/2017).

Alicja Sznarkowska: I am a PI at the international Centre for Cancer Vaccine Science and my scientific interests are vast, ranging from the origin of immune system, through the source of antigens for Major Histocompatibility Class I pathway, to the studies of protein isoforms. I am interested in how information encoded in a gene is transcribed and translated to have different meaning. For that, I study alternative transcription and translation events and functions of their products with the example of NRF2.

O.17 | NRF2 and BCL2 family proteins

Monika A Jakubowska1

1 Malopolska Centre of Biotechnology, Jagiellonian University in Krakow, ul. Gronostajowa 7a, 30-387, Krakow, Poland.

e-mail: monika.jakubowska@uj.edu.pl

Current medical predictions report pancreatic cancer to become the second cause of cancer-related deaths by 2030, mainly because of a lack of effective treatment. Although the strategies of (in)direct targeting a genetic driver of pancreatic cancer development, a mutant KRAS gene, have proven futile, other therapeutic approaches of multi-drug intervention against this form of cancer have entered the clinic. However, these strategies use combinations of cytostatic drugs that mainly target cancerous pancreatic epithelia. Targeting cancer-associated fibroblasts, which in solid pancreatic tumors extensively produce acellular stroma of a hydrogel-like matrix and protein fibers, remains an unmet clinical need. So far, studies of other diseases characterized by strong fibrosis, such as scleroderma, have shown that fibroblasts may evade pro-apoptotic stress signals by changing the balance of BCL-2 family proteins. Under normal conditions, these proteins sensitize the cell to death signals, inducing apoptosis, but in apoptosis-resistant cells, changes in levels of BCL-2 family proteins make the cell resistant to stress. Surprisingly, a similar pattern of the BCL-2 expression characterizes deadly blood cancers: leukemias, and lymphomas. In 2016 small molecule drug, a BCL-2 inhibitor Venetoclax, was approved for clinical use against blood cancer. This has inspired our group to test BCL-2 inhibitors in a therapeutic combination with a cytostatic drug, using pre-clinical models of pancreatic cancer that differ in the content of the fibrotic matrix in a tumor. In this presentation, I am showcasing our unpublished results, and discussing the most recent literature data about the link between NRF-2 and BCL-2 proteins/small molecule inhibitors.

Funding:
the Foundation for Polish Science (Homing 3/2017), the National Science Centre Poland (Sonata-Bis 12: 2022/46/E/NZ4/00441).

Since 2018 Monika Jakubowska has been an Assistant Professor in Cell Physiology/Cell Signaling in the group Molecular Mechanisms of Disease, and a Research Director of the BSL-3+ Animal Unit, both at the Malopolska Centre of Biotechnology, the Jagiellonian University in Krakow, Poland. Monika and her team investigate the role of redox imbalance in pancreatic (patho)physiology, e.g., pancreatic inflammation and cancer. Other Monika’s interests center on transition metal ions, mainly copper and iron, in the (patho)physiology of the gastrointestinal tract and animal models of cancer, sterile inflammation, and infectious diseases.

O.18 | Novel small molecules and peptidomimetics inhibitors of NRF2– KEAP1 determined via structure-based virtual screening

Kemal Yelekci, Damla Dere

Kadir Has University, Istanbul, Turkey.

e-mail: monika.jakubowska@uj.edu.pl

Kelch-like ECH-associated protein 1 (Keap1) is an important drug target for neurodegenerative diseases, chronic obstructive pulmonary disease, various inflammatory conditions, and cancer. In recent years, various inhibitors with diverse chemical structures have been reported to have high potency for the Keap1- NRF2 target; however, a few molecules have managed to translate into the clinical phase as drug candidates. This study presents a structure-based virtual screening (SBVS) method to discover novel peptidomimetics and small molecules for Keap1 and nuclear factor erythroid 2-related factor 2 (NRF2). The x-ray crystal structure of the human Keap1 Kelch domain (PDB: 4XMB) was downloaded and prepared utilizing Biovia DS 4.5 Studio protocols for virtual screening. Sixty five thousand small molecules were selected from the ZINC15 small molecule libraries, and peptidomimetic candidates having three peptide structures were prepared from commercially available non-natural amino acids. These modified peptide-based candidates are potential lead structures for NRF2 targets and excellent leads for further optimization. Molecules with higher binding energies were selected against the Keap1 target above the threshold values. Poses of the potent inhibitors in the binding cavity were visualized. The molecular dynamics behaviors of these final compounds were investigated by performing an additional MD simulation study. Overall, the combining of several computational approaches unveiled the critical structural features essential for the inhibitory activity that will shed light on the future development of novel potent drugs against the Keap1- NRF2 target.

Kemal Yelekci: Graduated from Middle East Technical University, Chemistry Department. He was granted the Fulbright scholarship to pursue his doctoral degree in the USA. He received his Ph. D. degree from Ohio University in synthetic organic chemistry. He worked at Northwestern University (USA) as a postdoc in medicinal chemistry and drug design and synthesis. After returning to Turkiye, he was promoted to Associate professor (1989) and Full Professor (1996) at Marmara University. He joined Kadir Has University in 2001 to present. His main interests are research and development involving studies of drug design and drug action, receptor-ligand interaction and transporter protein mechanisms, in silico screening, calculation of free energy profile, elucidation of the flexibility and dynamical behavior of the protein-substrate complex.

O.19 | Proteome and redox instability as major drivers and hallmarks of aging

Despoina D. Gianniou, Sentiljana Gumeni, Ioannis P. Trougakos

Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece.

e-mail: itrougakos@biol.uoa.gr;
Web:
http://scholar.uoa.gr/itrougakos/home

Proteome stability (also referred to as proteostasis) is critical for proper cellular functionality and consequently organismal health, and it is ensured by an extensive compartment-specific network of machineries known as the proteostasis network (PN). Key components of the PN are the two main proteolytic machineries, namely the autophagy lysosome- (ALP) and the ubiquitin proteasome- (UPP) pathways, along with several stress
responsive cellular sensors including (among others) heat shock factor 1 (Hsf1) and the nuclear factor erythroid 2-related factor 2 (Nrf2) pathways. Under conditions of proteome instability and/or increased oxidative load a fully functional (i.e., young) biological system engages the PN which addresses the triage decision of protein fold, hold, or degrade, and restores normal cell oxidative load. Nonetheless, all PN modules gradually become dysfunctional during the highly unnatural process of aging; this output results in, among others, sustained accumulation of free radicals, beyond a physiological level, which promotes oxidative damage and stochastic modification of all cellular biomolecules. These processes will be discussed along with plausible interventions that can likely increase healthy aging.

Ioannis Trougakos obtained his Ph.D. in Cellular-Developmental Biology from the National and Kapodistrian University of Athens (NKUA), Greece. He has worked as Research Scientist at EMBL, Germany, CBM “Severo Ochoa”, Spain and at NHRF, Athens, Greece; he was also research visitor at EMBL and at the Netherlands Cancer Institute. Prof. Trougakos was elected Research Lecturer at NHRF and currently serves as Professor and Director of the “Cell Biology” lab at the Faculty of Biology, NKUA. He is also appointed Adjunct Professor of “Systems Biology of Ageing and Cancer” at the European University of Cyprus. He is the Head of the “Ageing and Age-Related Diseases” group (http://scholar.uoa.gr/itrougakos) at NKUA. Prof. Trougakos has published articles in high-ranking journals, chapters in international books; he is also co inventor in several patents. His group is funded by private (GR, EU, USA) and public (GR, EU) entities; also, the group participates in contractual activities with the Industry.

O.20 | Hypothermia’s molecular mechanisms and the role of the antioxidative system

K.-L. Grassman1, H. Vellama1, F.M. Sirkel1, T. Jagomäe1, Rando Porosk1, L. Tarve1, T. Visnapuu1, R. Reimets1, C.A. Hundahl1, E. Vasar1, H. Luuk1, K.-L. Eskla1

1 University of Tartu, Institute of Biomedicine and Translational Medicine, Tartu, Estonia.

e-mail: kattriliis@gmail.com

Ischemia reperfusion injury poses a significant challenge in various medical contexts, necessitating the development of effective treatments. Hypothermia has shown potential as a strategy to reduce cellular damage caused by oxygen deprivation. This study explores the molecular mechanisms behind the protective effects of hypothermia using an in vitro model. We found that hypothermia exerts a dual effect on cellular metabolism, simultaneously reducing metabolic activity and enhancing stress tolerance. Specifically, maintaining a mild hypothermic state at 32°C appears to be the optimal temperature for achieving this balance. Our findings indicate that hypothermia alleviates hypoxia-induced metabolic disruptions, such as changes in lactate:pyruvate ratios and gene expressions linked to reductive stress. Remarkably, we discovered that hypothermia influences gene expression more extensively than hypoxia, with over 3000 genes responding to cooling. Interestingly, the hypothermia
induced activation of hypoxia response element (HRE) transcriptional activity is mediated by HIF1B, rather than HIF1A or HIF2A. At 32°C, hypothermia activates the Nrf2 pathway, enhancing the expression of thioredoxin and glutathione-related genes, and increasing glutathione levels. Additionally, pre-conditioning with hypothermia boosts cellular resilience to oxidative stress. These insights into the molecular mechanisms of hypothermia’s potentially therapeutic effects can be applied to in vivo models for further evaluation. Understanding the interactions between hypothermia, metabolic regulation, and stress response pathways could lead to innovative therapeutic strategies for mitigating ischemia reperfusion injury.

Funding: This research was supported by Estonian Research Council PUT1077, PUT120, PRG685, IUT20-42, PSG959; ERDF2014-2020.4.01.15-0012 and Baltic Research Programme of the EA EEZ/BPP/VIAA/2021/8.

Kattri-Liis Eskla In 2019, she earned her PhD in neuroscience from the University of Tartu (Estonia), discovering hypothermia’s profound impact on the antioxidant system. Her journey took her to Emory University School of Medicine (USA), where she explored novel treatments for cardiovascular diseases involving hydrogen sulfide and therapeutic lymphangiogenesis. Transitioning to the University of Birmingham (UK) in 2021, she embarked on collaborative research into hypoxic biology. Currently, her work challenges conventional notions about hypothermia, suggesting that its therapeutic effects are not solely due to metabolic depression. She is dedicated to unraveling the mechanisms behind hypothermia and reductive stress, aiming to transform the way we approach treating hypoxic/ischemic conditions (stroke, heart attack, organ transplantation, HIE) and disorders associated with electron transport chain dysfunction and reactive oxygen species formation.

O.21 | Dissecting NRF2 activation by physiological levels of hydrogen peroxide

Dina Dikovskaya1,2, Boushra Bathish 1, Albena T. Dinkova-Kostova1

1 School of Medicine, University of Dundee, UK.
2 Peninsula Medical School, University of Plymouth, UK.

e-mail: ddikovskaya@dundee.ac.uk

NRF2 mediates the stress response to pathological levels of reactive oxygen species (ROS) and xenobiotics via transient activation of ~200 cytoprotective genes. In unstressed conditions, Keap1 binds to NRF2 and recruits it to Cullin3-RING E3-ubiquitin ligase that targets NRF2 for degradation. Modifications of Keap1 cysteine residues by ROS or electrophiles interfere with Keap1-dependent degradation of NRF2, resulting in its accumulation and nuclear translocation. Using FLIM-FRET-based assays, we have previously shown that the NRF2-Keap1 complex adopt a more compact conformation in response to electrophiles. However, how physiological oxidants affect the NRF2-Keap1 complex is unknown. To gain insight into NRF2induction by physiological ROS, we monitored temporal changes in NRF2, NRF2-Keap1 complex and Keap1 oxidation upon exposure to a sub-lethal level of hydrogen peroxide (H2O2), a major physiological ROS and a signalling molecule, in live primary human fibroblasts. 75µM H2O2 caused transient oxidation of Keap1 that was not strongly influenced by the depletion of Peroxiredoxins 1 and/or 2. H2O2 induced changes in NRF2-Keap1 complex similar to those caused by electrophilic Nrf2 inducer Sulforaphane but independent of C151 in Keap1. Comparing the above kinetics revealed that while Keap1 oxidation was resolved before decrease in H2O2, the changes in NRF2-Keap1 complex persisted, even after H2O2 levels were reduced by N-acetyl cysteine (NAC). A transient NRF2 upregulation by H2O2 trailed that of Keap1 oxidation, and NAC accelerated the return of NRF2 to the basal level. Our data suggests that NRF2 degradation after its induction by physiological ROS can restart without restoration of NRF2-Keap1 complex conformation.

Funding:
BBSRC, Tenovus Scotland.

Dina Dikovskaya: I graduated from Tomsk State University in Russia and did my PhD in Karlsruhe, Germany. My first postdoctoral project at the University of Dundee, UK, investigated mechanisms of chromosomal instability and cell division in colorectal cancers. Cell division (or its absence) was also a focus of my next postdoc at the CRUK Beatson Laboratories in Glasgow, UK, where I studied cellular senescence. Wanting to know more about regulation of oxidative stress in senescence, I joined Albena Dinkova-Kostova lab at the University of Dundee, where I developed several methods to monitor oxidative stress response in live cells. During my next postdoctoral work that dealt with LRRK2 in mouse models of Crohn’s Disease in MRC-PPU, University of Dundee, I won a BSI grant to measure Nrf2 activation in Crohn’s Disease. I am now moving to University of Plymouth to take a lecturer position and start my own lab to study Nrf2.

O.22 | Laminin-α2 chain deficiency leads to multi-organ oxidative stress

Susana G Martins1,2,*, Mafalda Pita1,2,*, Sharadha Dayalan Naidu3, Albena T Dinkova Kostova3, Sólveig Thorsteinsdóttir1,2,# & Ana Rita Carlos1,2,#

1 Centre for Ecology, Evolution and Environmental Changes (cE3c) & CHANGE, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal.
2 Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal.
3 Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, U.K.
*co-first authors; # co-senior authors.

e-mail: sgmartins@ciencias.ulisboa.pt

LAMA2-congenital muscular dystrophy (LAMA2-CMD) is caused by mutations in LAMA2, encoding the laminin-α2 chain, a structural component of the extracellular matrix (ECM). Most studies have focused on the analysis of LAMA2 role in the muscle, and, accordingly, we have recently shown that LAMA2-CMD onset in skeletal muscle occurs in utero accompanied by profound transcriptional changes and imbalanced redox metabolism. However, LAMA2 is also expressed in other less studied organs including the heart, kidney and liver. Although the composition of the ECM may share strong similarities in different organs, the pathways involved in the ECM-cell communication may differ tremendously. Thus, this work aimed at analyzing the impact of the Lama2-deficiency in the heart, kidney and liver. For that, we collected liver, kidney and heart samples from dyW fetuses, a LAMA2- CMD mouse model, during the onset of the disease and analyzed different pathways. This analysis revealed that Lama2-deficiency leads to alterations in the antioxidant response in all three organs, including altered levels of NRF2 and its target genes, such as HO-1. Additionally, there were also observed changes in mitochondria functioning, glutathione peroxidases (GPX) and p38 pathway. To better understand if this phenotype is cell autonomous, we isolated cells from the different organs, cultured them in vitro and analyzed some oxidative stress markers. Our results suggest that some alterations are cell autonomous, while most seem to be non-cell autonomous. Overall, this study provides a novel and unique analysis of the impact of Lama2-deficiency, which leads to altered redox balance, beyond the well-characterized muscle phenotype.

Funding: Fundação para a Ciência e Tecnologia (FCT, Portugal; 2022.10813.BD; CEECIND/01589/2017; doi:10.54499/PTDC/BTM-ORG/1383/2020; doi:10.54499/UIDB/00329/2020); COST Action CA20121 (BenBedPhar) (E-COST-GRANT-CA20121-a076558c), Association Française contre les Myopathies (AFM) Téléthon (contract no. 23049); Henrique Meirelles who chose to support the MATRIHEALTH Project (CC1036); Microscopy Facility of FCUL (PPBI-POCI-01-0145-FEDER-022122); CONGENTO LISBOA-01-0145-FEDER
022170 (FCT, Lisboa2020, Por2020, ERDF).

Susana G Martins is a PhD Student at the laboratories of Molecular Mechanisms of Disease (MMD) and Extracellular Matrix in Development and Disease (E MDD) at cE3c (Faculty of Sciences, Lisbon University) under the supervision of Dr. Ana Rita Carlos and Dr. Sólveig Thorsteinsdóttir. Her research is focused on understanding how Lama2-deficiency affects different organs and unravel possible therapeutic target. This work already led to the publication of two reviews and one research article (Martins et al. (2024), under revision). In 2022, she was one of the winners of the Participatory Budget of cE3c and, last year, she received a grant for a Short-Term Scientific Mission within BenBedPhar Cost Action.

O.23 | 30 Years of NRF2: Charting sustainable pathways for the future of the BenBedPhar Action

Theo Zacharis1

1 Greek Scientists Society.

e-mail: theo@greek-scientists-society.org

As we mark the 30th anniversary of the discovery of NRF2, a key regulator of cellular defence mechanisms, we stand at a critical juncture to leverage this knowledge for sustainable advancements in biomedicine. This presentation will explore the dissemination and exploitation avenues for the NRF2 research and its implications for the BenBedPhar Action, an initiative dedicated to enhancing therapeutic strategies and pharmaceutical innovations. The session will explore the integration of NRF2 pathways in developing robust and environmentally sustainable pharmaceutical practices. We will discuss how the BenBedPhar Action can harness the potential of NRF2 to foster community well-being through targeted biotechnological interventions, aiming for a balance between scientific progress and ecological responsibility.
Key topics will include:
• The evolution of NRF2 research over the past three decades
• Current and future applications of NRF2 in pharmaceutical sciences • Strategic planning for sustainability within the BenBedPhar framework Through this exploration, we aim to outline a visionary roadmap for the BenBedPhar Community, ensuring that future biotechnological en

Theo Zacharis is an Innovation & Strategy Advisor with a B.Sc. in Business Administration from the Empire State University and an MBA from ALBA Athens Graduate Business School. He began his career as Marketing Manager at adidas Hellas before founding Strategic Foresight Hellas in 1997. In 2018, he launched bioGLOT Ventures in Cambridge, UK, an Innovation & Strategy Advisory firm. Theo is also the founder and leader of the Greek Scientists Society (GSS), the largest global network of Scientists and Technologists with over 33,000 members. His expertise spans organisational consultancy, IT solutions, and managing complex projects, including national and European co-financed initiatives. He is deeply committed to fostering scientific collaboration and innovation. His work includes advising start-ups on business planning, technology transfer, and investment strategies, with a particular focus on fostering innovation ecosystems and supporting early-stage ventures. He is an expert in Dissemination and Exploitation.

O.24 | Pharmacologic and genetic activation of NRF2 confers anti-fibrotic effects

Sharadha Dayalan Naidu1, Ralitsa R. Madsen2, Iain M. Phair1, Boushra Al-Bathish1, Abel D. Ang1, Maureen Higgins1, Pingting Bian1, Dorothy Kisielewski1, Terry W. Moore3, W. Christian Wigley 4, John D. Hayes1, Albena T. Dinkova-Kostova1,5

1 Division of Cellular & Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, James Arrott Drive, Dundee, Scotland, UK.
2 Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK.
3 Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL, USA 4 Reata Pharmaceuticals, Irving, TX, USA.
5 Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.

e-mail: s.z.dayalannaidu@dundee.ac.uk

Chronic liver injury, often resulting in oxidative stress burden, causes liver fibrosis, which is characterized by excessive scarring. Unresolved hepatic fibrosis leads to cirrhosis, and ultimately liver failure which requires liver transplantation. To date, there are no clinically approved drugs to treat liver fibrosis. Quiescent hepatic stellate cells (HSCs) when transdifferentiated into proliferative myofibroblasts by highly potent cytokines such as transforming growth factor beta 1 (Tgfβ1) are the main cells responsible for scar formation in the liver. We hypothesized that NRF2 activation in HSCs can circumvent conditions of oxidative stress, thereby inhibiting fibrogenesis. First, we used a genetic approach to manipulate the levels of NRF2. In unstimulated HSCs, NRF2 knockdown (NRF2-KD) increased the expression of pro-fibrotic markers, and to a further extent upon stimulation with Tgfβ1 compared to the wild-type cells. Conversely, compared to the wild-type, in Keap1-KD cells (with high NRF2 levels), the expression of pro-fibrotic markers was suppressed. To activate NRF2 pharmacologically in the HSCs, we used two classes of NRF2 activators, an electrophilic cyanoenone triterpenoid (TP), and a non-electrophilic Keap1- NRF2 protein protein interaction inhibitor (PPII). Treatment with both types of NRF2 activators reduced the Tgfβ1-induced expression of pro-fibrotic markers. Additionally, a global overview using high
resolution proteomics further revealed that pharmacological NRF2 activation orchestrates an anti-fibrotic program. In conclusion, our results suggest that NRF2 activation induces an anti fibrotic program. Considering that one of the TPs has been approved for clinical use, our findings support the development of such compounds as potential therapeutic agents to prevent and treat liver fibrosis.

Funding: Reata Pharmaceuticals, Tenovus Scotland.

Sharadha Dayalan Naidu graduated with a Ph.D. in Medicine from the University of Dundee in 2016. She joined the Cullman Chemoprotection Centre at Johns Hopkins University in 2017 as a postdoctoral research scholar in the Department of Pharmacology and Molecular Sciences, focusing on identifying the mechanisms of action of novel compounds. Returning to Scotland in 2019, she continued her postdoctoral research in Professor Albena T. Dinkova
Kostova’s laboratory. Her current research explores the benefits of various classes of Nrf2 activators in treating liver fibrosis. She has published over 25 original and review articles, primarily on Nrf2-Keap1 biology. Beyond her research, Sharadha is committed to mentoring the next generation of scientists and plays an active role in supporting their academic and professional development.

O.25 | Bdnf-NRF2 crosstalk in depression disorder

Marlene Santos1,4, Renato Caldevilla1,2, Stephanie Morais2, Serafim Carvalho3, Rui Medeiros4,5, Maria Fátima Barroso2*

1 REQUIMTE/LAQV, Escola Superior de Saúde, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida, 400, 4200-072, Porto, Portugal.
2 REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico, Rua Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal.
3 Hospital de Magalhães Lemos, 4149-003 Porto, Portugal.
4 CI-IPOP, Centro de Investigação do Instituto Português de Oncologia do Porto, Rua Dr. António Bernardino de Almeida 865, 4200-072, Porto, Portugal.
5 Liga Portuguesa Contra o Cancro, Departamento de Investigação, Estrada Interior da Circunvalação 6657, 4200- 172, Porto, Portugal.

e-mail: mes@ess.ipp.pt

The World Health Organization estimates that major depressive disorder (MDD) affects over 264 million individuals globally, posing a significant public health challenge. Treatment resistant depression (TRD) represents a severe form of MDD with poor treatment outcomes. Genetic variations are known to impact MDD treatment responses, yet genome-wide association studies have struggled to identify consistent marker alleles. Previous research has linked the Brain Derived Neurotrophic Factor (BDNF) genetic polymorphism with TRD. BDNF is essential for neuronal survival and neuroplasticity, processes influenced by antidepressant treatment, and regulated by transcription factors like Nuclear factor erythroid 2-related factor 2 (NRF2). NRF2 regulates antioxidant and anti-inflammatory responses and plays a crucial role in depression pathogenesis. NRF2 knockout mice exhibit reduced BDNF levels and depression-like behaviors, indicating that NRF2activation enhances BDNF expression and antidepressant efficacy. The BDNF rs6265 (Val66Met) polymorphism is associated with variations in antidepressant response rates. Research suggests that the interaction between BDNF and NRF2 pathways could enhance antidepressant effectiveness. NRF2 activation, such as through the compound sulforaphane, has demonstrated rapid antidepressant effects by increasing BDNF expression. Lower levels of NRF2 and BDNF are observed in stress-induced depression models, and ketamine treatment influences NRF2-related genes. Simultaneously, there is a growing need for efficient genotyping methods, and genosensors offer a promising solution. This presentation will address the interplay between BDNF and NRF2 in depression, explore its relationship in antidepressant response, and present a putative genosensor for BDNF rs6265 (Val66Met) polymorphism identification, improving antidepressant treatment outcome.

Funding: This work received financial support from FCT/MCTES (UIDP/50006/2020 DOI 10.54499/UIDP/50006/2020) through national funds and the Ibero-American Program on Science and Technology CYTED (GENOPSYSEN, P222RT011).

Marlene Santos (MS) (Orcid: 0000-0001-5020-5942) holds a PhD in Biomedicine, an MSc in Molecular Genetics, and a Degree in Pharmacy. She is a Coordinator Professor at the Escola Superior de Saúde (E2S), Polytechnic Institute of Porto, where she serves as the Director of the Master’s Degree in Pharmacy program. She is an integrated Investigator at LAQV – REQUIMTE and leads Working Group 1 on “Patient stratification tools for optimal treatment with therapeutic antibodies” under the COST Action ENOTTA. Her research interests include the study of biomarkers for psychiatric and neurological diseases and treatment response outcomes, with her primary publications in the fields of Pharmacogenomics and Neuropsychopharmacology. She also serves as Academic Editor for “Depression and Anxiety” journal.

O.26 | Isothiocyanates for protection against neurodegenerative diseases

Antonio Cuadrado1, Ana I. Rojo1, Eduardo Cazalla1, Isao Okunishi2, Manuel Debasa1, and Angel J. García-Yagüe1

1 Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM). Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM). Instituto de Investigación Sanitaria La Paz (IdiPaz). Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
2 Kinjirushi Co, Ltd. 2-61Yahata-Hontori, Nakagawa-ku, Nagoya-shi, Aichi.454-8526, Japan.

e-mail: antonio.cuadrado@uam.es

Tauopathies are over 25 different neurodegenerative diseases that present brain accumulation of microtubule-associated protein TAU in hyperphosphorylated, fibrillar aggregates and that participate in damage of subcortical and cortical structures. Abnormally hyperphosphorylated TAU is accumulated as intraneuronal tangles of paired helical filaments (PHF), twisted ribbons, and or straight filaments and disturb axonal transport, leading to neural damage, formation of reactive oxygen species, and uncontrolled neuroinflammation. We have developed a mouse model of progressive supranuclear palsy (PSP), a rare tauopathy that comprises dementia and atypical parkinsonism, and is amenable to orphan designation. Several studies have reported that isothiocyanates such as Sulforaphane (SFN) or Hexaraphane (6-MSITC) exert neuroprotective function in the brain of neurodegenerative animal models. This work intends to study whether the 6-MSITC might regulate the levels of phosphorylated TAU and identify its mechanism of action. Nerve hippocampus cell lines and primary neurons revealed that 6-MSITC decreased the levels of phosphorylated TAU (epitopes AT8 and PHF1). Surprisingly, 6-MSITC regulates phospho-TAU levels through a mechanism that is NRF2-independent. On the other hand, we found that 6-MSITC alters the balance between TAU-kinases and phosphatases to favor phosphorylation of this protein. Hexaraphane is a promising pharmacological candidate to regulate TAU hyperphosphorylation in neurodegenerative diseases.

Funding: Spanish Ministry of Economy and Competitiveness (MINECO) (grants PID2019- 110061RB-I00, PID-2021-122766OB-100 and PDC2021-121421-I00, PDC2022-133765-I00, and CIBERned (ISCIII), Autonomous Community of Madrid (grant P2022/BMD-7230). Colaborative agreement with Kinhirushi Co, Ltd. This study is based upon work from COST Action CA20121, supported by COST (European Cooperation in Science and Technology) (www.cost.eu).

Antonio Cuadrado is a full professor of Biochemistry and Molecular Biology at the Department of Biochemistry, Medical School, Autonomous University of Madrid. He obtained his PhD degree in Biology in 1985 and enjoyed several postdoctoral stays in the National Cancer Institute-NIH with the help of Fulbright and Fogarty fellowships. He established his independent laboratory as Professor of Biochemistry in 1997 with a main interest on the study of molecular mechanisms involved in initiation and progression of chronic diseases. For the past years his main lane of research has been the validation of transcription factor NRF2, master regulator of cellular homeostasis as a new therapeutic target in NASH. His current interest is the development of new NRF2-modulating drugs. Dr. Cuadrado has published over 160 primary and review articles, of which more than 80 are related to the role of NRF2 in physiological and pathological responses to disease.

O.27 | Targeting Alzheimer´s disease hallmarks with the NRF2 activator Isoeugenol

Ana Silva1, Sónia Silva2,3, Jéssica Macedo2, Patrícia Moreira1, Diana Baptista2, Joana Bicker2,4, Ana Fortuna2,4, Joana Liberal5, Beatriz Rodrigues2, Rosa Resende1, Armanda E. Santos1,2, Bruno Miguel Neves6, Cláudia Pereira1,7 and Maria Teresa Cruz1,2 1

Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3000-548 Coimbra, Portugal; 2 Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; 3 Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra Institute of Clinical and Biomedical Research, University of Coimbra, 3000-548 Coimbra, Portugal.
4 CIBIT/ICNAS – Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; 5 Polytechnic Institute of Coimbra, Coimbra Health School, 3046-854 Coimbra, Portugal; 6 Department of Medical Sciences and Institute of Biomedicine – iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal;7 Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.

e-mail: trosete@ff.uc.pt

Alzheimer´s disease (AD) is a neurodegenerative disorder and the most common cause of dementia with no effective cure or treatment. Therefore, the identification of disease-modifying therapeutics is crucial. The transcription factor nuclear factor erythroid 2–related factor 2 (NRF2) is a master controller of homeostatic functions that control redox and energy metabolism, neuroinflammation, and proteostasis, and its overall activity is compromised in AD. Most pharmacological NRF2 activators are small electrophilic molecules, a characteristic of low
molecular-weight skin allergens such as Dimethyl fumarate and Isoeugenol. Hence, we conducted a ground-breaking study to investigate Isoeugenol potential in activating NRF2 and revert some AD hallmarks. This study was performed in microglia cells exposed to LPS and neuronal cells overexpressing human APP with Swedish mutation, and in the AD double transgenic mice, APP/PS1, intranasally administered with Isoeugenol, at 11 months old. The results showed that Isoeugenol 1) activated Nrf2; 2) displayed antioxidant and anti-inflammatory effects; 3) exhibited good pharmacokinetic and pharmacodynamic profiles; 4) reduced the levels of plasma Aβ peptides; 5) reduced triglyceride and LDL cholesterol levels in treated mice, and 6) improved the memory deficits observed in old mice, reinforcing its potential to alleviate AD hallmarks. This study positions Isoeugenol as a promising pleiotropic therapeutic molecule, bringing hope for the implementation of a new therapeutic approach for AD.

Funding:
COMPETE 2020 – Operational Programme for Competitiveness and Internationalisation and Portuguese national funds via FCT – Fundação para a Ciência e a Tecnologia, under projects POCI-01-0145-FEDER-029369 and UIDB/04539/2020, UIDP/04539/2020 and LA/P/0058/2020 and also supported by the COST action BenBedPhar: and HORIZON-RIA – HORIZON Research and Innovation Actions (Grant agreement ID: 101080329).

Maria Teresa Cruz (MT Cruz) holds a degree in Pharmaceutical Sciences (1991) from the University of Lisbon, a master’s in Drug Technologies (1996), and a PhD in Pharmacology (2004) from the University of Coimbra. She is Associate Professor with Habilitation at the Faculty of Pharmacy and Group Leader at the Centre for Innovative Biomedicine and Biotechnology. She is a European Certified Pharmacologist and Toxicologist, and has actively participated in 49 national and international funded projects, leading 21 of them, including those funded by the European Union 7th Framework Programme and John Hopkins, USA. She has contributed to 7 international patents, filed 4 provisional patent requests, authored 6 book chapters, and published 207 articles in SCI indexed journals. Listed among the “World’s Top 2% Scientists” for citations in 2022, MT Cruz has an h-index of 39 with 7867 citations (Web of Science). She has supervised or co-supervised 20 doctoral (9 completed), 28 master (22 completed), and 3 post-doctoral students.

O.28 | A novel protein-protein NRF2 inducer with anti-neuroinflammatory effects

Manuela G. Lopez1, Eric del Sastre1, Lucía Viqueira1, Cristina Fernández-Mendívil1, Elsa Cortes1, Silvia Santamaría1, Antonio Cuadrado3, Rafael León2, and María Isabel Rodríguez Franco2

1 Departamento de Farmacología y Terapéutica, Facultad de Medicina. Instituto Teofilo Hernando. Universidad Autónoma de Madrid, Spain.
2Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28006 Madrid, Spain. 3Departmento de Bioquímica, Facultad de Medicina, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ UAM-CSIC, Universidad Autónoma de Madrid, Madrid, Spain.

e-mail: manuela.garcia@uam.es

NRF2 is a transcription factor that is being proposed as a target for neurodegenerative diseases as it regulates a wide array of genes implicated in controlling cellular redox status and inflammation. Electrophilic NRF2 inducers are highly reactive molecules that can cause off-target effects. Therefore, NRF2-KEAP1 protein-protein inhibitors (PPI) could be a safer alternative. In the course of a drug discovery program, we have identified ND523 as PPI lead compound. Here we study its mechanism of action and its anti-neuroinflammatory effects. The NRF2 inducing mechanism has been evaluated in AREc32 cells, by superficial plasmon resonance, GSH conjugation and lC-IT-MS analysis, and by using WT, KEAP1 KO and NRF2-
KO MEFs. The potential anti-neuroinflammatory effect has been evaluated using LPS in vitro (BV2 cells and primary glial cultures) and in vivo. Sickness behaviour in vivo and cytokines levels measured by qPCR and multiplex ELISA were evaluated. ND523 is a non-electrophilic compound that induces NRF2 by inhibiting NRF2/KEAP1 interaction. It showed a good anti neuroinflammatory profile in a microglial murine cell line and in rat primary glial cell cultures; these effects were lost in NRF2 KO cells. The anti-neuroinflammatory effects were corroborated in vivo as shown by a reduction of cytokines 4 and 24 h post LPS-injection, reduction in brain microgliosis and improvements in locomotion and cognitive decline. ND523 is a first in class protein-protein inhibitor of NRF2/KEAP1 with potential therapeutic interest in neurodegenerative diseases with neuroinflammation.

Funding: Spanish Ministry of Economy and Competence Refs. PDC2022-133809-I00 and PID2021- 125986OB-I00 to MGL; PID2021-122650OB-I00 to MIRF and PID2021-123481OB-I00 to RL; and General Council for Research and Innovation of the Community of Madrid Refs. B2017/BMD-3827 and P2022/BMD-7230 to MGL.
Disclosure statement: Cristina Fernández-Mendívil and Eric del Sastre López are former employees of Instituto Teófilo Hernando and Universidad Autónoma de Madrid.

Manuela G. Lopez is MD PhD and full professor of Pharmacology at the Department of Pharmacology in the School of Medicine, Universidad Autónoma de Madrid (UAM), Spain. Currently, she heads the Institute Teofilo Hernando for drug discovery (http://www.ifth.es/) that belongs to UAM. Her group, “NeuroprotectionLab” (http://neurodiscovery-ndd.com/gt1), has particular interest in the identification of new potential therapeutic targets to develop innovative and disease modifying therapies for neurodegenerative diseases, with special focus in modulating neuroinflammation (microglia-astrocyte interaction), oxidative stress and autophagy. Within the field of NRF2, she has contributed to the understanding of NRF2 in pain, depression, stroke and neurodegenerative diseases, together with the development of different NRF2 multitarget drugs in collaboration with medicinal chemists. Currently, she is coordinating a drug development project to identify Keap1-NRF2 inhibitors with potential use in Alzheimer’s disease.

O.29 | Differential effects of diphenyl diselenide (PhSe)2 on mitochondria related pathways depending on the cellular energy status in bovine vascular endothelial cells

Letícia Selinger Galant1,2, Laura Doblado2, Rafael Radi3, João Batista Teixeira da Rocha4, Andreza Fabro de Bem 1,5*, Maria Monsalve2

1 Biochemistry PhD Program, Federal University of Santa Catarina, Florianopolis, SC, Brazil. 2Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM). Arturo Duperier 4. 28029, Madrid, Spain. 3 Center for Free Radical and Biomedical Research (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
4 Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Brazil. 5 Departament of Physiological Science, Institute for Biological Sciences; University of Brasília, Brasília, Brazil.

e-mail: mpmonsalve@iib.uam.es

Cellular energy metabolism varies depending on tissue and cell type, as well as the availability of energy substrates and energy demands. We recently investigated the variations in cellular metabolism and antioxidant responses in primary bovine vascular endothelial cells (BAECs) under different energetic substrate conditions in vitro, specifically glucose or galactose. In this context, pharmacological agents may exert different effects on cells depending on their energy metabolism status. In this study, we aimed to characterize the effects of (PhSe)2, a redox
active molecule known for its prominent cardiovascular effects, on redox-bioenergetic cellular pathways under glycolytic or oxidative conditions in BAECs. Under glucose conditions, (PhSe)2 positively impacted mitochondrial oxidative capacity, as assessed by respirometry, and was associated with changes in mitochondrial cellular dynamics. However, these changes were not observed in cells cultured with galactose. Although (PhSe)2 induced the nuclear translocation of NRF2 in both glucose and galactose media, NRF2 remained in the nuclei of cells cultured in galactose for a longer duration. Additionally, activation of FOXO3a was only detected in galactose media. Notably, (PhSe)2 strongly induced the expression of genes controlling mitochondrial antioxidant capacity and glutathione synthesis and recycling in glucose media, whereas its effects in galactose media were primarily focused on glutathione homeostasis. In conclusion, our findings underscore the critical influence of cellular metabolic status on the antioxidant capacity of redox
active molecules such as (PhSe)2.

Funding: This research was funded by MCIN/AEI, grant numbers RTI2018-093864-B-I00 and PID2021- 122765OB-I00, the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement 721236-TREATMENT and the Brazilian institutions: Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Fundação de Apoio à Pesquisa do Distrito Federal (FAPDF grants 00193-00000884/2021-89 and 00193-00002348/2022-07) and Instituto Nacional de Ciência e Tecnologia e Neuro-ImunoModulação (INCT-NIM grant 485489/2014-1).

María Monsalve holds a doctorate in Biochemistry and Molecular Bology from the Autonomous University of Madrid and is CSIC Scientific Researcher at the Sols Morreale Biomedical Research Institute (CSIC-UAM), where she leads the Mitochondrial Function in Health and Disease group whose main focus is to understand the molecular bases involved in the regulation of mitochondrial function, to understand how its alteration leads to the development of diseases, and to seek the application of this knowledge to the development of diagnostic tools and new therapeutic approaches. Dr. María Monsalve has represented Spain in three COST (European Cooperation in Science & Technology) Actions, has been a member of the European Society SFRR-E Council, has been responsible for the IIBm Seminar’s Commission and is currently Vice
Director of the Spanish GEIRLI Group, has coordinated the Free Radicals and Oxidative Stress Group of the SEBBM Society and is currently a member of its Council (2020-2024).

O.30 | NRF2 targeting in arterial hypertension associated with sleep apnea

António B. Pimpão1, M. João Correia1, Elsa Mecha2,3, Dalila G. F. Fernandes3, Teresa Serra2,3, Antonio Melo Junior1, João B. Vicente3, Xavier Coumoul4, Emília C. Monteiro1, Maria R. Bronze2,3, Sofia A. Pereira1

1iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa (UNL), Portugal.
2iBET-Instituto de Biologia Experimental e Tecnológica, Portugal.
3Instituto de Tecnologia Química e Biológica António Xavier, UNL, Portugal.
4INSERM UMR-S 1124, 3TS, Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, Université de Paris, France.

e-mail: antonio.pimpao@nms.unl.pt

Chronic intermittent hypoxia (CIH) is responsible for the development of drug-resistant hypertension (HTN) in patients with obstructive sleep apnea (OSA). We were pioneers in linking aryl hydrocarbon receptor (AhR) activation and cysteine redox dynamics with chronic intermittent hypoxia (CIH)-induced elevated blood pressure. We also found that hydroxytyrosol (OHTyr), the most bioactive phenolic compound on extra-virgin olive oil and a known NRF2 agonist reverts the CIH-induced elevated blood pressure and decreases renal AhR activation. Herein, we aim at investigating the impact of OHTyr treatment in the AhR-HIF interplay and cysteine metabolism in CIH conditions in different organs (e.g., kidney, liver). The study has the Ethical approval by DGAV and NMS ethic committee. Briefly, male Wistar rats (n≥5/group) underwent CIH (21-5% O2, 5.6 cycles/h, 10.5h/day, during animals’ inactive period) for 21 days to establish HTN, followed by oral gavage with OHTyr (15mg/kg/day in vegetable oil) for 14 days under CIH. Animals under 35 days of normoxia (Nx) or CIH were used as controls. AHR-related signaling (e.g., CYP1A1, PON1, HIFs) and cysteine metabolic enzymes (e.g., CBS, MST) were assessed by Western Blot and quantification of cysteine related thiols (e.g., GSH, Cys) conducted by HPLC-FD. CIH exposure altered the expression of AHR-related proteins. OHTyr reversed renal CIH-increased CYP1A1 levels and normalized the levels of oxidized cysteine and CIH-altered cysteine metabolic enzymes. Altogether, our findings suggest that OHTyr impact in CIH-induced AhR activation is tissue dependent, besides acting in cysteine metabolism/H2S production. We also identify AHR pharmacological pathways, putatively relevant to OSA-HTN.

Funding: This work was supported by Fundação para Ciência e Tecnologia [PTDC/MED TOX/30418/2017] and iNOVA4Health [IUDB/004462/2020/TL2/P2]. A.B.P. and M.J.C. are supported by FCT grants [2022.11188.BD and SFRH/BD/131331/2017, respectively].

António B. Pimpão is graduated in Human Biology from the University of Évora and has a master’s degree in Molecular Genetics and Biomedicine from the Faculty of Science and Technology – NOVA University of Lisbon. He is actually a Ph.D. student at the Renal, Cardiovascular and Metabolic Pharmacology Lab, led by professor Sofia Pereira at NOVA Medical School, where he is exploring the aryl hydrocarbon receptor (AHR) and its related signaling in obstructive sleep apnea-related arterial hypertension, as well as its pharmacological modulation as a putative therapeutic approach. Besides, he’s research interests focus mainly in renal-cardiovascular diseases and in identifying novel approaches to treat them.

O.31 | NRF2 regulates a neuronal fate by modification of mitochondrial metabolism, Ca2+ handling and permeability transition

Andrey Y. Abramov1

1 Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK.

e-mail: a.abramov@ucl.ac.uk

Mitochondria are organelles which are responsible for multiple functions in the cells including energy production, reactive oxygen production, calcium signaling and regulation of cell death/survival. Fast and transient permeability of mitochondria (mitochondrial permeability transition, mPT) is shown to be an initial step in the mechanism of apoptotic and necrotic cell death which acts as a regulator of tissue regeneration but for postmitotic neurons as it leads to irreparable loss of cells and function. Two major players of mitochondrial function – Ca2+ and reactive oxygen species (ROS) are triggers for the opening the mitochondrial permeability transition pore (mPTP) –and cascade mechanism of cell death. Activation of Nrf2 in primary neurons and astrocytes with omaveloxolone led to stimulation of the mitochondrial energy metabolism, increase in mitochondrial membrane potential that increase ROS production in mitochondria. However, activation of Nrf2 in co-culture of neurons and astrocytes modify mitochondrial Ca2+ handling resulting in faster calcium efflux out of mitochondria. This effect led to increase mitochondrial Ca2+ capacity in neurons and astrocytes that protects these cells against calcium induced opening of mitochondrial permeability transition pore and cell death. This effect may explain neuroprotective effect of Nrf2 activation in familial forms of neurodegenerative diseases including Parkinson’s disease and Alzheimer’s disease and ischemia/reperfusion injury.

Andrey Y. Abramov is a Professor at the Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology. He studies the role of mitochondria, calcium signalling and redox biology in physiology of the Central nervous system and in the mechanism of the pathology of neurodegenerative disorders. In the last decade in collaboration with Professor Dinkova-Kostova we identified novel and underestimated role of Nrf2 in mitochondrial bioenergetics.

O.32 | Brain metabolic alterations promote faulty proteostasis and redox imbalance in Down syndrome neuropathology

Fabio Di Domenico1, Francesca Prestia1, Chiara Lanzillotta1, Antonella Tramutola1, Eugenio Barone1, Marzia Perluigi1

1 Departement of Biochemical Science, Sapienza University of Rome, Italy.

e-mail: fabio.didomenico@uniroma1.it

A maladaptive response, such as chronic unfolded protein response (UPR) activation, provides a link between the accumulation of misfolded proteins, increased oxidative damage and neurotoxicity. We recently reported that the dysregulation of the PERK branch of the UPR is associated with altered Nrf2 response contributing to the progression of Alzheimer-like signatures in Down syndrome (DS) brain. In addition, DS brain demonstrate that metabolic defecta occurs in parallel to faulty proteostasis and redox imbalance and are associated and cognitive decline. Our study aimed to understand in vitro and in vivo the pronicity of DS phenotype in developing defects of proteostasis and of Nrf2 response under aberrant metabolic stimuli. Our data on primary cortical neurons and astrocytes isolated from euploid and DS mice demonstrated that IPA-induced metabolic stress led to the de-regulation of the PERK/eIF2a axis, and of Nrf2-related antioxidant responses. However, by pharmacologically targeting the UPR we were able to ameliorate metabolic defects, improve proteostasis and reduce oxidative damage. Similarly, the IPA-induced accumulation of tau and Aβ, observed in DS LCLs, was cleared after the administration of UPR targeting agents rescuing proteostasis and Nrf2 induction. Further, in DS mice we observed that the early administration of high fat diet was able to promote aberrant proteostasis and redox imbalance in a gender and brain region-specific manner. Our results suggest that metabolic defects occurring in DS contribute to the failure of the proteostasis network and to the de-regulation of Nrf2 signal, thus promoting major critical events that leads to neurodegeneration and cognitive decline.

Funding: Jerome Lejeune Foundation grant #2280_2023B “Nutrient-dependent alteration of the Unfolded Protein Response in Down syndrome brain: tackling the diet/cognition axis”.

Fabio Di Domenico is Full professor of Biochemistry at Sapienza University of Rome. Under the supervision of Prof. Butterfield, at University of Kentucky, he has been involved in the application of redox proteomics studies to unravel the neurodegenerative mechanisms associated with increased oxidative damage in Alzheimer Disease. His research is currently focused in understanding the mechanisms that redox imbalance and defective proteostasis in the development of Alzheimer-like dementia. Collected data from his laboratory postulate that aberrant proteostasis, observed in both Alzheimer and Down syndrome patients, is strictly associated with the increase of oxidative damage as result of compromised antioxidant response and faulty protein degradative systems. Recently, his studies revealed that the chronic induction of the unfolded protein response and its aberrant relationship with Nrf2 response hold a prominent role in the development of dementia in the brain from Alzheimer and Down Syndrome patients.

O.33 | Identification of prognostic DNA methylation changes in NRF2 metabolic dysfunction associated steatotic liver disease

Evelien Van Dijck1,2, Wim Van Hul1, Wim Vanden Berghe2

1 University of Antwerp – Center of Medical Genetics, Belgium.
2 University of Antwerp – Cell Death Signaling lab, Belgium.

e-mail: wim.vandenberghe@uantwerpen.be

Metabolic dysfunction associated steatotic liver disease (MASLD) is a heterogeneous and progressive liver disease affecting up to 1/3th of the population. It’s etiology is complex and involves (interplay among) genetic, metabolic and environmental factors. The surge in MASLD incidence coincides with changing dietary habits, shifting research towards epigenetics, which links disease to environmental and lifestyle cues. Currently, histological assessment of liver biopsy is the golden standard in diagnostics, but it relies on semiquantitative assessment. To better understand the pathophysiological processes implicated in MASLD and identify new diagnostic biomarkers leveraging added value for clinical diagnosis, we examined liver biopsy DNA methylation changes in MASLD stages.Our pilot study cohort consists of 22 patients with Western-Diet associated obesity, with(out) associated MASLD. The cohort is subdivided into four incremental categories from no MASLD to severe steatohepatitis with fibrosis. Analysis of incremental MASLD-stage identified 79842 differentially methylated probes (DMPs). Pathway analysis revealed enrichment in 118 processes, mostly related to metabolic pathways, inflammation and oxidative stress, coinciding with progressive DNA hypermethylation and gene silencing of the liver-specific nuclear receptor PPARα, a key regulator of lipid metabolism. Remarkably, genetic and lipid diet-induced loss of PPARα function triggers compensatory activation of multiple lipid sensing transcription factors and epigenetic writer eraser-reader proteins, which promotes the epigenetic transition from lipid metabolic stress towards ferroptosis and pyroptosis lipid hepatoxicity pathways associated with advanced MASLD.Due to the limitations associated with histological MASLD-scoring, epigenetic age acceleration (EAA) (residual from the regression of chronological age with epigenetic age) was calculated using Horvath’s algorithm as a read-out for general liver damage. EAA correlates with histological liver-damage associated features, and MASLD stage explains 58% of EAA variance in a linear regression model. DMP analysis of EAA shows enrichment of 134 pathways, displaying an 80% overlap with the MASLD identified pathways. Several uniquely identified pathways have well-known associations with MASLD including Nrf2, Wnt, cAMP, adipocytokine and TNF signaling pathways, and pathways related to lipid metabolism, indicating an added value for EAA analysis. Since oxidative stress is a causative factor of MASLD, resulting in liver inflammation and fibrosis, we also evaluated possible associations with anti-inflammatory and antioxidant properties of the paraoxonase (PON) protein family (PON1, PON2 and PON3) through SNPs, activity and DNA methylation analysis. PONs protect against oxidation by hydrolyzing (lipid) peroxides and lactones. However, lactonase and arylesterase activities or SNPs were not correlated to MASLD or EAA. In contrast, methylation of individual PON-CpGs shows significant correlations with EAA but not with MASLD. Finally, an association with a differently methylated region in PON3 was identified for MASLD.

Prof. Wim Vanden Berghe obtained his PhD at the University of Ghent (UGent) in 1999 in the Faculty of Science Biochemistry-Biotechnology. After postdoctoral research at various research institutions (University of Montpellier-France, Stellenbosch-SouthAfrica and Oxford-UK), Wim Vanden Berghe was appointed professor of Epigenetics in 2009 at the cell death epigenetic signaling lab (CDES
IPPON) (UAntwerp/UGent, Belgium). With his current research, he is characterizing epigenetic plasticity of redox/kinase signaling pathways to modulate ferroptosis disease phenotypes with phytomedicinal or pharmacological compounds to improve treatment of cancer, CVD, obesity, NAFLD, autism spectrum disorders or neurodegeneration/development.

O.34 | Assessment of fructose consumption on blood-brain and testicular barriers, and NRF2 modulation: Evaluation of hawthorn treatment

Onur Gökhan Yildirim1, Hatice Iskender2, Eda Dokumacıoğlu2, Kübra Asena Terim Kapakin3, Ali Doğan Ömür4 and Metin Kiliçlioğlu3

1 Medicinal-Aromatic Plants Application and Research Center, Artvin Coruh University, Artvin, Turkey. 2 Department of Nutrition and Dietetics, Faculty of Healthy Sciences, Artvin Coruh University, Artvin, Turkey. 3 Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey. 4 Department of Reproduction and Artificial Insemination, Institute of Health Sciences, Atatürk University, Erzurum, Turkey.

e-mail: ogyildirim@artvin.edu.tr

The escalating consumption of high-fructose content refined foods has been closely linked to the increasing prevalence of functional disturbances within the male reproductive system. Both epidemiological and experimental studies have consistently demonstrated the pivotal role of high-fructose diets in the pathogenesis of chronic diseases. Furthermore, these findings strongly suggest that fructose may exacerbate oxidative stress, induce testicular damage, and contribute to a spectrum of fertility-related disorders. The components of the hawthorn plant (Crataegus microphylla), encompassing its fruits, flowers, and leaves, are abundant in bioactive compounds and widely distributed across our region. These botanical elements exhibit a diverse array of properties, including antimicrobial, anti-inflammatory, antioxidant, anticancer, antidiabetic, antihyperlipidemic, antihyperglycemic, anticoagulant, anti-atherosclerotic, gastroprotective, and neuroprotective effects. This study investigated the impact of Crataegus microphylla on the levels of Vimentin, NRF2, Testosterone, and Connexin 43 in both testicular and brain tissues of rats subjected to a 15-week fructose diet. The findings revealed a decrease in serum testosterone levels and an increase in Vimentin, Connexin 43, and NRF2 values within the testicular tissue of rats administered fructose. Conversely, these parameters exhibited decreased levels while testosterone levels increased in the group treated with Crataegus microphylla. In the brain tissue, an increase in vimentin and NRF2 values was noted in rats exposed to fructose, whereas these values decreased in the group treated with Crataegus. This observation suggests that Crataegus microphylla mitigates the detrimental effects induced by fructose in brain tissue. Additionally, epididymal sperm density and motility were significantly lower in the fructose group compared to the other experimental groups examined in this study. In the hawthorn group, the rate of dead spermatozoa was notably lower compared to the other experimental groups, where it was found to be higher. However, no statistically significant difference was observed between the groups in terms of abnormal spermatozoa rate.

I’m Onur Gökhan Yildirim from Türkiye, and also a pharmacologist. I’m director of the Medicinal-Aromatic Plants Application and Research Center, Artvin Coruh University, Türkiye. While we investigate solutions from natural herbal sources to cure diseases such as metabolic syndrome, diabetes and obesity, we also illuminate the disease mechanisms of these diseases.

Saturday, October 12, 2024

Cost.

COST (European Cooperation in Science and Technology) is a funding agency for research and innovation networks. Our Actions help connect research initiatives across Europe and enable scientists to grow their ideas by sharing them with their peers. This boosts their research, career and innovation.

BenBedPhar CA20121.

Action details.

Grant Holder: Universidad Autónoma de Madrid
Start of Action: 19 October, 2021
End of Action: 18 October, 2025
Entry into force: DATE HERE
CSO approval date: 25 May 2021
Action email: info@benbedphar.org