1
|
Marques ES, Severance EG, Arsenault P, Zahn SM, Timme-Laragy AR. Activation of Nrf2 at Critical Windows of Development Alters Tissue-Specific Protein S-Glutathionylation in the Zebrafish ( Danio rerio) Embryo. Antioxidants (Basel) 2024; 13:1006. [PMID: 39199250 PMCID: PMC11352166 DOI: 10.3390/antiox13081006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Activation of Nrf2-the master regulator of antioxidative response-at different stages of embryonic development has been shown to result in changes in gene expression, but the tissue-specific and downstream effects of Nrf2 activation during development remain unclear. This work seeks to elucidate the tissue-specific Nrf2 cellular localization and the downstream changes in protein S-glutathionylation during critical windows of zebrafish (Danio rerio) development. Wild-type and mutant zebrafish embryos with a loss-of-function mutation in Nrf2a were treated with two canonical activators, sulforaphane (SFN; 40 µM) or tert-butylhydroquinone (tBHQ; 1 µM), for 6 h at either pharyngula, hatching, or the protruding-mouth stage. Nrf2a protein and S-glutathionylation were visualized in situ using immunohistochemistry. At the hatching stage, Nrf2a protein levels were decreased with SFN, but not tBHQ, exposure. Exposure to both activators, however, decreased downstream S-glutathionylation. Stage- and tissue-specific differences in Nrf2a protein and S-glutathionylation were identified in the pancreatic islet and liver. Protein S-glutathionylation in Nrf2a mutant fish was increased in the liver by both activators, but not the islets, indicating a tissue-specific and Nrf2a-dependent dysregulation. This work demonstrates that critical windows of exposure and Nrf2a activity may influence redox homeostasis and highlights the importance of considering tissue-specific outcomes and sensitivity in developmental redox biology.
Collapse
Affiliation(s)
| | | | | | | | - Alicia R. Timme-Laragy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA (E.G.S.)
| |
Collapse
|
2
|
Bartolini D, Stabile AM, Migni A, Gurrado F, Lioci G, De Franco F, Mandarano M, Svegliati-Baroni G, Di Cristina M, Bellezza G, Rende M, Galli F. Subcellular distribution and Nrf2/Keap1-interacting properties of Glutathione S-transferase P in hepatocellular carcinoma. Arch Biochem Biophys 2024; 757:110043. [PMID: 38789086 DOI: 10.1016/j.abb.2024.110043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
The oncogene and drug metabolism enzyme glutathione S-transferase P (GSTP) is also a GSH-dependent chaperone of signal transduction and transcriptional proteins with key role in liver carcinogenesis. In this study, we explored this role of GSTP in hepatocellular carcinoma (HCC) investigating the possible interaction of this protein with one of its transcription factor and metronome of the cancer cell redox, namely the nuclear factor erythroid 2-related factor 2 (Nrf2). Expression, cellular distribution, and function as glutathionylation factor of GSTP1-1 isoform were investigated in the mouse model of N-nitrosodiethylamine (DEN)-induced HCC and in vitro in human HCC cell lines. The physical and functional interaction of GSTP protein with Nrf2 and Keap1 were investigated by immunoprecipitation and gene manipulation experiments. GSTP protein increased its liver expression, enzymatic activity and nuclear levels during DEN-induced tumor development in mice; protein glutathionylation (PSSG) was increased in the tumor masses. Higher levels and a preferential nuclear localization of GSTP protein were also observed in HepG2 and Huh-7 hepatocarcinoma cells compared to HepaRG non-cancerous cells, along with increased basal and Ebselen-stimulated levels of free GSH and PSSG. GSTP activity inhibition with the GSH analogue EZT induced apoptotic cell death in HCC cells. Hepatic Nrf2 and c-Jun, two transcription factors involved in GSTP expression and GSH biosynthesis, were induced in DEN-HCC compared to control animals; the Nrf2 inhibitory proteins Keap1 and β-TrCP also increased and oligomeric forms of GSTP co-immunoprecipitated with both Nrf2 and Keap1. Nrf2 nuclear translocation and β-TrCP expression also increased in HCC cells, and GSTP transfection in HepaRG cells induced Nrf2 activation. In conclusion, GSTP expression and subcellular distribution are modified in HCC cells and apparently contribute to the GSH-dependent reprogramming of the cellular redox in this type of cancer directly influencing the transcriptional system Nrf2/Keap1.
Collapse
Affiliation(s)
- Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, 06126, Perugia, Italy.
| | - Anna Maria Stabile
- Department of Medicine and Surgery, University of Perugia, 06132, Perugia, Italy.
| | - Anna Migni
- Department of Pharmaceutical Sciences, University of Perugia, 06126, Perugia, Italy.
| | - Fabio Gurrado
- Department of Gastroenterology, Marche Polytechnic University, Ancona, Italy.
| | - Gessica Lioci
- Department of Gastroenterology, Marche Polytechnic University, Ancona, Italy.
| | | | - Martina Mandarano
- Department of Medicine and Surgery, University of Perugia, 06132, Perugia, Italy
| | - Gianluca Svegliati-Baroni
- Department of Gastroenterology, Marche Polytechnic University, Ancona, Italy; Obesity Center, Marche Polytechnic University, Ancona, Italy and Liver Injury and Transplant Unit, Ancona, Italy.
| | - Manlio Di Cristina
- Department of Medicine and Surgery, University of Perugia, 06132, Perugia, Italy.
| | - Guido Bellezza
- Department of Medicine and Surgery, University of Perugia, 06132, Perugia, Italy.
| | - Mario Rende
- Department of Medicine and Surgery, University of Perugia, 06132, Perugia, Italy.
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, 06126, Perugia, Italy.
| |
Collapse
|
3
|
Zhang CY, Yang M. Anti-oxidative stress treatment and current clinical trials. World J Hepatol 2024; 16:294-299. [PMID: 38495278 PMCID: PMC10941751 DOI: 10.4254/wjh.v16.i2.294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/08/2024] [Accepted: 02/05/2024] [Indexed: 02/27/2024] Open
Abstract
Oxidative stress disturbs the balance between the production of reactive oxygen species (ROS) and the detoxification biological process. It plays an important role in the development and progression of many chronic diseases. Upon exposure to oxidative stress or the inducers of ROS, the cellular nucleus undergoes some biological processes via different signaling pathways, such as stress adaption through the forkhead box O signaling pathway, inflammatory response through the IκB kinase/nuclear factor-κB signaling pathway, hypoxic response via the hypoxia-inducible factor/prolyl hydroxylase domain proteins pathway, DNA repair or apoptosis through the p53 signaling pathway, and antioxidant response through the Kelch-like ECH-associated protein 1/nuclear factor E2-related factor 2 signaling pathway. These processes are involved in many diseases. Therefore, oxidative stress has gained more attraction as a targeting process for disease treatment. Meanwhile, anti-oxidative stress agents have been widely explored in pre-clinical trials. However, only limited clinical trials are performed to evaluate the efficacy of anti-oxidative stress agents or antioxidants in diseases. In this letter, we further discuss the current clinical trials related to anti-oxidative stress treatment in different diseases. More pre-clinical studies and clinical trials are expected to use anti-oxidative stress strategies as disease treatment or dietary supplementation to improve disease treatment outcomes.
Collapse
Affiliation(s)
- Chun-Ye Zhang
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, United States
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States.
| |
Collapse
|
4
|
Petrovic M, Simic T, Djukic T, Radic T, Savic-Radojevic A, Zekovic M, Durutovic O, Janicic A, Milojevic B, Kajmakovic B, Zivkovic M, Bojanic N, Bumbasirevic U, Coric V. The Polymorphisms in GSTO Genes ( GSTO1 rs4925, GSTO2 rs156697, and GSTO2 rs2297235) Affect the Risk for Testicular Germ Cell Tumor Development: A Pilot Study. Life (Basel) 2023; 13:1269. [PMID: 37374052 DOI: 10.3390/life13061269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/24/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Members of the omega class of glutathione transferases (GSTs), GSTO1, and GSTO2, catalyze a range of reduction reactions as a part of the antioxidant defense system. Polymorphisms of genes encoding antioxidant proteins and the resultant altered redox profile have already been associated with the increased risk for testicular germ cell cancer (GCT) development. The aim of this pilot study was to assess the individual, combined, haplotype, and cumulative effect of GSTO1rs4925, GSTO2rs156697, and GSTO2rs2297235 polymorphisms with the risk for testicular GCT development, in 88 patients and 96 matched controls, through logistic regression models. We found that carriers of the GSTO1*C/A*C/C genotype exhibited an increased risk for testicular GCT development. Significant association with increased risk of testicular GCT was observed in carriers of GSTO2rs2297235*A/G*G/G genotype, and in carriers of combined GSTO2rs156697*A/G*G/G and GSTO2rs2297235*A/G*G/G genotypes. Haplotype H7 (GSTO1rs4925*C/GSTO2rs2297235*G/GSTO2rs156697*G) exhibited higher risk of testicular GCT, however, without significant association (p > 0.05). Finally, 51% of testicular GCT patients were the carriers of all three risk-associated genotypes, with 2.5-fold increased cumulative risk. In conclusion, the results of this pilot study suggest that GSTO polymorphisms might affect the protective antioxidant activity of GSTO isoenzymes, therefore predisposing susceptible individuals toward higher risk for testicular GCT development.
Collapse
Affiliation(s)
- Milos Petrovic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Tatjana Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Department of Medical Sciences, Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| | - Tatjana Djukic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Tanja Radic
- Institute of Mental Health, 11000 Belgrade, Serbia
| | - Ana Savic-Radojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milica Zekovic
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Otas Durutovic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandar Janicic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Bogomir Milojevic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Boris Kajmakovic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Marko Zivkovic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Nebojsa Bojanic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Uros Bumbasirevic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Vesna Coric
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
5
|
Orlewska K, Klusek J, Głuszek S, Klusek J, Witczak B, Wawszczak M, Madej Ł, Marzec MT, Orlewska E. Glutathione S-Transferase P1 Genetic Variant's Influence on the HbA1c Level in Type Two Diabetic Patients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1520. [PMID: 36674274 PMCID: PMC9859603 DOI: 10.3390/ijerph20021520] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
GST (glutathione S-transferases) are capable of influencing glucose homeostasis, probably through regulation of the response to oxidant stress. The aim of our study was to investigate the relationship between GSTP1 gene polymorphism and glycated hemoglobin (HbA1c) levels in type two diabetic (T2D) patients. A total of 307 T2D patients were included. Analysis of the GSTP1 gene polymorphism (rs1695) was conducted using the TaqMan qPCR method endpoint genotyping. HbA1c was determined using a COBAS 6000 autoanalyzer. A univariable linear regression and multivariable linear regression model were used to investigate the association between mean HbA1c level and GSTP1 gene polymorphism, age at T2D diagnosis, T2D duration, therapy with insulin, gender, BMI, smoking status. GSTP1 Val/Val genotype, age at T2D diagnosis, T2D duration and therapy with insulin were statistically significant contributors to HbA1c levels (p < 0.05). Multivariable regression analysis revealed that GSTP1 (Val/Val vs. Ile/Ile) was associated with higher HbA1c even after adjustment for variables that showed a statistically significant relationship with HbA1c in univariable analyses (p = 0.024). The results suggest that GSTP polymorphism may be one of the risk factors for higher HbA1c in T2D patients. Our study is limited by the relatively small sample size, cross-sectional design, and lack of inclusion of other oxidative stress-related genetic variants.
Collapse
Affiliation(s)
| | - Justyna Klusek
- Collegium Medicum, Jan Kochanowski University, 25-516 Kielce, Poland
| | - Stanisław Głuszek
- Collegium Medicum, Jan Kochanowski University, 25-516 Kielce, Poland
- Department of General, Oncological and Endocrinological Surgery, Voivodeship Hospital, 25-736 Kielce, Poland
| | - Jolanta Klusek
- Institute of Biology, Jan Kochanowski University, 25-406 Kielce, Poland
| | - Bartosz Witczak
- Collegium Medicum, Jan Kochanowski University, 25-516 Kielce, Poland
| | - Monika Wawszczak
- Collegium Medicum, Jan Kochanowski University, 25-516 Kielce, Poland
| | - Łukasz Madej
- Collegium Medicum, Jan Kochanowski University, 25-516 Kielce, Poland
| | - Michał Tomasz Marzec
- Collegium Medicum, Jan Kochanowski University, 25-516 Kielce, Poland
- Department of Biomedical Sciences, University of Copenhagen, 1017 Copenhagen, Denmark
| | - Ewa Orlewska
- Collegium Medicum, Jan Kochanowski University, 25-516 Kielce, Poland
| |
Collapse
|
6
|
Marques ES, Severance EG, Min B, Arsenault P, Conlin SM, Timme-Laragy AR. Developmental impacts of Nrf2 activation by dimethyl fumarate (DMF) in the developing zebrafish (Danio rerio) embryo. Free Radic Biol Med 2023; 194:284-297. [PMID: 36528121 PMCID: PMC9906634 DOI: 10.1016/j.freeradbiomed.2022.12.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Dimethyl fumarate (DMF) is pharmaceutical activator of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates of many cellular antioxidant response pathways, and has been used to treat inflammatory diseases such as multiple sclerosis. However, DMF has been shown to produce adverse effects on offspring in animal studies and as such is not recommended for use during pregnancy. The goal of this work is to better understand how these adverse effects are initiated and the role of DMF-induced Nrf2 activation during three critical windows of development in embryonic zebrafish (Danio rerio): pharyngula, hatching, and protruding-mouth stages. To evaluate Nrf2 activation, wildtype zebrafish, and mutant zebrafish (nrf2afh318/fh318) embryos with a loss of function mutation in Nrf2a, the co-ortholog to human Nrf2, were treated for 6 h with DMF (0-20 μM) beginning at the pharyngula, hatching, or protruding-mouth stage and assessed for survival and morphology. Nrf2a mutant fish had an increase in survival, however, morphology studies demonstrated Nrf2a mutant fish had more severe deformities occurring with exposures during the hatching stage. To verify Nrf2 cellular localization and downstream impacts on protein-S-glutathionylation in situ, a concentration below the LOAEL was chosen (7 μM) for immunohistochemistry and S-glutathionylation. Embryos were imaged via epifluorescence microscopy studies, the Nrf2a protein in the body tissue was decreased with DMF only when exposed at the hatching stage, while total protein S-glutathionylation was modulated by Nrf2a activity and DMF during the pharyngula and protruding-mouth stage. The pancreatic islet and liver were further analyzed via confocal microscopy. Pancreatic islets and liver also had tissue specific differences with Nrf2a protein expression and protein S-glutathionylation. This work demonstrates how critical windows of exposure and Nrf2a activity may influence toxicity of DMF and highlights tissue-specific changes in Nrf2a protein levels and S-glutathionylation in pancreatic islet and liver during embryonic development.
Collapse
Affiliation(s)
- Emily S Marques
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Emily G Severance
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Bellis Min
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Paige Arsenault
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Sarah M Conlin
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| |
Collapse
|
7
|
Suppression of Selective Voltage-Gated Calcium Channels Alleviates Neuronal Degeneration and Dysfunction through Glutathione S-Transferase-Mediated Oxidative Stress Resistance in a Caenorhabditis elegans Model of Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8287633. [PMID: 36600949 PMCID: PMC9806690 DOI: 10.1155/2022/8287633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/18/2022] [Accepted: 10/31/2022] [Indexed: 12/05/2022]
Abstract
Calcium homeostasis plays a vital role in protecting against Alzheimer's disease (AD). In this study, amyloid-β (Aβ)-induced C. elegans models of AD were used to elucidate the mechanisms underlying calcium homeostasis in AD. Calcium acetate increased the intracellular calcium content, exacerbated Aβ 1-42 aggregation, which is closely associated with oxidative stress, aggravated neuronal degeneration and dysfunction, and shortened the lifespan of the C. elegans models. Ethylene glycol tetraacetic acid (EGTA) and nimodipine were used to decrease the intracellular calcium content. Both EGTA and nimodipine showed remarkable inhibitory effects on Aβ 1-42 aggregations by increasing oxidative stress resistance. Moreover, both compounds significantly delayed the onset of Aβ-induced paralysis, rescued memory deficits, ameliorated behavioral dysfunction, decreased the vulnerability of two major (GABAergic and dopaminergic) neurons and synapses, and extended the lifespan of the C. elegans AD models. Furthermore, RNA sequencing of nimodipine-treated worms revealed numerous downstream differentially expressed genes related to calcium signaling. Nimodipine-induced inhibition of selective voltage-gated calcium channels was shown to activate other calcium channels of the plasma membrane (clhm-1) and endoplasmic reticulum (unc-68), in addition to sodium-calcium exchanger channels (ncx-1). These channels collaborated to activate downstream events to resist oxidative stress through glutathione S-transferase activity mediated by HPGD and skn-1, as verified by RNA interference. These results may be applied for the treatment of Alzheimer's disease.
Collapse
|
8
|
Bartolini D, Arato I, Mancuso F, Giustarini D, Bellucci C, Vacca C, Aglietti MC, Stabile AM, Rossi R, Cruciani G, Rende M, Calafiore R, Luca G, Galli F. Melatonin modulates Nrf2 activity to protect porcine pre-pubertal Sertoli cells from the abnormal H 2 O 2 generation and reductive stress effects of cadmium. J Pineal Res 2022; 73:e12806. [PMID: 35524288 PMCID: PMC9539639 DOI: 10.1111/jpi.12806] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/27/2022]
Abstract
Melatonin (MLT) is a cytoprotective agent holding potential to prevent cadmium (Cd) toxicity and its impact in testicular function and fertility. In this study, we explored such potential in porcine pre-pubertal Sertoli cells (SCs). Cd toxicity resulted in impaired SC viability and function, abnormal cellular H2 O2 generation and efflux, and induction of reductive stress by the upregulation of Nrf2 expression and activity, cystine uptake and glutathione biosynthesis, glutathione-S-transferase P (GSTP) expression, and protein glutathionylation inhibition. Cd toxicity also stimulated the activity of cellular kinases (MAPK-ERK1/2 and Akt) and NFkB transcription factor, and cJun expression was increased. MLT produced a potent cytoprotective effect when co-administered with Cd to SCs; its efficacy and the molecular mechanism behind its cytoprotective function varied according to Cd concentrations. However, a significant restoration of cell viability and function, and of H2 O2 levels, was observed both at 5 and 10 μM Cd. Mechanistically, these effects of MLT were associated with a significant reduction of the Cd-induced activation of Nrf2 and GSTP expression at all Cd concentrations. CAT and MAPK-ERK1/2 activity upregulation was associated with these effects at 5 μM Cd, whereas glutathione biosynthesis and efflux were involved at 10 μM Cd together with an increased expression of the cystine transporter xCT, of cJun and Akt and NFkB activity. MLT protects SCs from Cd toxicity reducing its H2 O2 generation and reductive stress effects. A reduced activity of Nrf2 and the modulation of other molecular players of MLT signaling, provide a mechanistic rational for the cytoprotective effect of this molecule in SCs.
Collapse
Affiliation(s)
- Desirée Bartolini
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic AnatomyUniversity of PerugiaPerugiaItaly
| | - Iva Arato
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | | | - Daniela Giustarini
- Department of Biotechnology, Chemistry and PharmacyUniversity of SienaSienaItaly
| | - Catia Bellucci
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Carmine Vacca
- Department of Chemistry, Biology and BiotechnologyUniversity of PerugiaPerugiaItaly
| | | | - Anna Maria Stabile
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic AnatomyUniversity of PerugiaPerugiaItaly
| | - Ranieri Rossi
- Department of Biotechnology, Chemistry and PharmacyUniversity of SienaSienaItaly
| | - Gabriele Cruciani
- Department of Chemistry, Biology and BiotechnologyUniversity of PerugiaPerugiaItaly
| | - Mario Rende
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic AnatomyUniversity of PerugiaPerugiaItaly
| | - Riccardo Calafiore
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
- Department of Medicine and Surgery, International Biotechnological Center for Endocrine, Metabolic and Embryo‐Reproductive Translational Research (CIRTEMER)University of PerugiaPerugiaItaly
| | - Giovanni Luca
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
- Division of Medical Andrology and Endocrinology of ReproductionSaint Mary HospitalTerniItaly
- Department of Medicine and Surgery, International Biotechnological Center for Endocrine, Metabolic and Embryo‐Reproductive Translational Research (CIRTEMER)University of PerugiaPerugiaItaly
| | - Francesco Galli
- Department of Pharmaceutical SciencesUniversity of PerugiaPerugiaItaly
| |
Collapse
|
9
|
How Aging and Oxidative Stress Influence the Cytopathic and Inflammatory Effects of SARS-CoV-2 Infection: The Role of Cellular Glutathione and Cysteine Metabolism. Antioxidants (Basel) 2022; 11:antiox11071366. [PMID: 35883857 PMCID: PMC9311797 DOI: 10.3390/antiox11071366] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 12/26/2022] Open
Abstract
SARS-CoV-2 infection can cause a severe respiratory distress syndrome with inflammatory and thrombotic complications, the severity of which increases with patients’ age and presence of comorbidity. The reasons for an age-dependent increase in the risk of severe COVID-19 could be many. These include defects in the homeostatic processes that control the cellular redox and its pivotal role in sustaining the immuno-inflammatory response to the host and the protection against oxidative stress and tissue degeneration. Pathogens may take advantage of such age-dependent abnormalities. Alterations of the thiol redox balance in the lung tissue and lining fluids may influence the risk of infection, and the host capability to respond to pathogens and to avoid severe complications. SARS-CoV-2, likewise other viruses, such as HIV, influenza, and HSV, benefits in its replication cycle of pro-oxidant conditions that the same viral infection seems to induce in the host cell with mechanisms that remain poorly understood. We recently demonstrated that the pro-oxidant effects of SARS-CoV-2 infection are associated with changes in the cellular metabolism and transmembrane fluxes of Cys and GSH. These appear to be the consequence of an increased use of Cys in viral protein synthesis and to ER stress pathway activation that interfere with transcription factors, as Nrf2 and NFkB, important to coordinate the metabolism of GSH with other aspects of the stress response and with the pro-inflammatory effects of this virus in the host cell. This narrative review article describes these cellular and molecular aspects of SARS-CoV-2 infection, and the role that antivirals and cytoprotective agents such as N-acetyl cysteine may have to limit the cytopathic effects of this virus and to recover tissue homeostasis after infection.
Collapse
|
10
|
Antioxidant Genetic Profile Modifies Probability of Developing Neurological Sequelae in Long-COVID. Antioxidants (Basel) 2022; 11:antiox11050954. [PMID: 35624818 PMCID: PMC9138155 DOI: 10.3390/antiox11050954] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/06/2022] [Accepted: 05/08/2022] [Indexed: 02/06/2023] Open
Abstract
Understanding the sequelae of COVID-19 is of utmost importance. Neuroinflammation and disturbed redox homeostasis are suggested as prevailing underlying mechanisms in neurological sequelae propagation in long-COVID. We aimed to investigate whether variations in antioxidant genetic profile might be associated with neurological sequelae in long-COVID. Neurological examination and antioxidant genetic profile (SOD2, GPXs and GSTs) determination, as well as, genotype analysis of Nrf2 and ACE2, were conducted on 167 COVID-19 patients. Polymorphisms were determined by the appropriate PCR methods. Only polymorphisms in GSTP1AB and GSTO1 were independently associated with long-COVID manifestations. Indeed, individuals carrying GSTP1 Val or GSTO1 Asp allele exhibited lower odds of long-COVID myalgia development, both independently and in combination. Furthermore, the combined presence of GSTP1 Ile and GSTO1 Ala alleles exhibited cumulative risk regarding long-COVID myalgia in carriers of the combined GPX1 LeuLeu/GPX3 CC genotype. Moreover, individuals carrying combined GSTM1-null/GPX1LeuLeu genotype were more prone to developing long-COVID “brain fog”, while this probability further enlarged if the Nrf2 A allele was also present. The fact that certain genetic variants of antioxidant enzymes, independently or in combination, affect the probability of long-COVID manifestations, further emphasizes the involvement of genetic susceptibility when SARS-CoV-2 infection is initiated in the host cells, and also months after.
Collapse
|
11
|
Dobiasová S, Szemerédi N, Kučerová D, Koucká K, Václavíková R, Gbelcová H, Ruml T, Domínguez-Álvarez E, Spengler G, Viktorová J. Ketone-selenoesters as potential anticancer and multidrug resistance modulation agents in 2D and 3D ovarian and breast cancer in vitro models. Sci Rep 2022; 12:6548. [PMID: 35449387 PMCID: PMC9023544 DOI: 10.1038/s41598-022-10311-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/11/2022] [Indexed: 12/13/2022] Open
Abstract
Long-term treatment of cancer with chemotherapeutics leads to the development of resistant forms that reduce treatment options. The main associated mechanism is the overexpression of transport proteins, particularly P-glycoprotein (P-gp, ABCB1). In this study, we have tested the anticancer and multidrug resistance (MDR) modulation activity of 15 selenocompounds. Out of the tested compounds, K3, K4, and K7 achieved the highest sensitization rate in ovarian carcinoma cells (HOC/ADR) that are resistant to the action of the Adriamycin. These compounds induced oxidation stress, inhibited P-gp transport activity and altered ABC gene expression. To verify the effect of compounds, 3D cell models were used to better mimic in vivo conditions. K4 and K7 triggered the most significant ROS release. All selected selenoesters inhibited P-gp efflux in a dose-dependent manner while simultaneously altering the expression of the ABC genes, especially P-gp in paclitaxel-resistant breast carcinoma cells (MCF-7/PAX). K4, and K7 demonstrated sensitization potential in resistant ovarian spheroids. Additionally, all selected selenoesters achieved a high cytotoxic effect in 3D breast and ovarian models, which was comparable to that in 2D cultures. K7 was the only non-competitive P-gp inhibitor, and therefore appears to have considerable potential for the treatment of drug-resistant cancer.
Collapse
Affiliation(s)
- Simona Dobiasová
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, Technická 3, 166 28, Prague 6, Czechia
| | - Nikoletta Szemerédi
- Department of Medical Microbiology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, Szeged, 6725, Hungary
| | - Denisa Kučerová
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, Technická 3, 166 28, Prague 6, Czechia
| | - Kamila Koucká
- Toxicogenomics Unit, National Institute of Public Health, Šrobárova 49, 100 00, Prague, Czechia.,Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, 323 00, Pilsen, Czechia
| | - Radka Václavíková
- Toxicogenomics Unit, National Institute of Public Health, Šrobárova 49, 100 00, Prague, Czechia.,Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, 323 00, Pilsen, Czechia
| | - Helena Gbelcová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08, Bratislava, Slovakia
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, Technická 3, 166 28, Prague 6, Czechia
| | - Enrique Domínguez-Álvarez
- Instituto de Química Orgánica General (IQOG-CSIC), Consejo Superior de Investigaciones Científicas, Juan de la Cierva 3, 28006, Madrid, Spain.
| | - Gabriella Spengler
- Department of Medical Microbiology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, Szeged, 6725, Hungary.
| | - Jitka Viktorová
- Department of Biochemistry and Microbiology, Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, Technická 3, 166 28, Prague 6, Czechia.
| |
Collapse
|
12
|
Mihailović S, Džamić Z, Plješa-Ercegovac M. The role of redox homeostasis biomarkers in clear cell renal cell carcinoma development and progression. MEDICINSKI PODMLADAK 2022. [DOI: 10.5937/mp73-35557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
The clear cell renal cell carcinoma (ccRCC) is the most frequent and the most aggresive subtype of renal cell carcinoma usually detected at an already advanced stage. It might even be observed as a metabolic disease since complex molecular changes and disturbed redox homeostasis are its hallmark. As certain changes are characteristic for tumorigenesis, while some other for metastatic disease, the identification of metabolic modifications could also point out the stage of tumor progression. Hypoxia inducible factor, as a factor regulating transcription of genes encoding glycolytic enzymes, as well as controlling lipid accumulation, has a particular place in ccRCC development. Additionaly, disturbed redox homeostasis induces the Keap1/Nrf2 pathway which further modulates the synthesis of phase-II detoxifying metabolism enzymes. The upregulation of glutathione transferases, Pi class especially, inhibits kinase-dependent apoptosis that is essential in tumor progression. Furthermore, hydrogen peroxide (H2O2) acts as a signaling molecule conveying redox signals, while superoxide dismutase, as well as glutathione peroxidase are enzymes involved in its production and degradation. Hence, the activity of these enzymes impacts hydrogen peroxide levels and consequentially the ability of ccRCC cells to evade negative effect of reactive oxygen species.
Collapse
|
13
|
Zhou J, Gu L, Shi Y, Huang T, Fan X, Bi X, Lu S, Liang J, Luo L, Cao P, Yin Z. GSTpi reduces DNA damage and cell death by regulating the ubiquitination and nuclear translocation of NBS1. Cell Mol Life Sci 2021; 79:54. [PMID: 34936032 PMCID: PMC11072236 DOI: 10.1007/s00018-021-04057-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 01/22/2023]
Abstract
Glutathione S-transferase pi (GSTpi) is an important phase II detoxifying enzyme that participates in various physiological processes, such as antioxidant, detoxification, and signal transduction. The high expression level of GSTpi has been reported to be related to drug-resistant and anti-inflammatory and it functioned via its non-catalytic ligandin. However, the previous protection mechanism of GSTpi in DNA damage has not been addressed so far. Nijmegen breakage syndrome 1 (NBS1) is one of the most important sensor proteins to detect damaged DNA. Here, we investigated the interaction between GSTpi and NBS1 in HEK-293 T cells and human breast adenocarcinoma cells during DNA damage. Our results showed that overexpression of GSTpi in cells by transfecting DNA vector decreased the DNA damage level after methyl methanesulfonate (MMS) or adriamycin (ADR) treatment. We found that cytosolic GSTpi could increase NBS1 ubiquitin-mediated degradation in unstimulated cells, which suggested that GSTpi could maintain the basal level of NBS1 during normal conditions. In response to DNA damage, GSTpi can be phosphorylated in Ser184 and inhibit the ubiquitination degradation of NBS1 mediated by Skp2 to recover NBS1 protein level. Phosphorylated GSTpi can further enhance NBS1 nuclear translocation to activate the ATM-Chk2-p53 signaling pathway. Finally, GSTpi blocked the cell cycle in the G2/M phase to allow more time for DNA damage repair. Thus, our finding revealed the novel mechanism of GSTpi via its Ser184 phosphorylation to protect cells from cell death during DNA damage and it enriches the function of GSTpi in drug resistance.
Collapse
Affiliation(s)
- Jinyi Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, People's Republic of China
| | - Lili Gu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, People's Republic of China
| | - Yingying Shi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, People's Republic of China
| | - Ting Huang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, People's Republic of China
| | - Xirui Fan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, People's Republic of China
| | - Xiaowen Bi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, People's Republic of China
| | - Shuai Lu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, People's Republic of China
| | - Juanjuan Liang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, People's Republic of China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, People's Republic of China.
| | - Peng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210046, People's Republic of China.
| |
Collapse
|
14
|
Coric V, Milosevic I, Djukic T, Bukumiric Z, Savic-Radojevic A, Matic M, Jerotic D, Todorovic N, Asanin M, Ercegovac M, Ranin J, Stevanovic G, Pljesa-Ercegovac M, Simic T. GSTP1 and GSTM3 Variant Alleles Affect Susceptibility and Severity of COVID-19. Front Mol Biosci 2021; 8:747493. [PMID: 34988113 PMCID: PMC8721193 DOI: 10.3389/fmolb.2021.747493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022] Open
Abstract
Based on the premise that oxidative stress plays an important role in severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection, we speculated that variations in the antioxidant activities of different members of the glutathione S-transferase family of enzymes might modulate individual susceptibility towards development of clinical manifestations in COVID-19. The distribution of polymorphisms in cytosolic glutathione S-transferases GSTA1, GSTM1, GSTM3, GSTP1 (rs1695 and rs1138272), and GSTT1 were assessed in 207 COVID-19 patients and 252 matched healthy individuals, emphasizing their individual and cumulative effect in disease development and severity. GST polymorphisms were determined by appropriate PCR methods. Among six GST polymorphisms analyzed in this study, GSTP1 rs1695 and GSTM3 were found to be associated with COVID-19. Indeed, the data obtained showed that individuals carrying variant GSTP1-Val allele exhibit lower odds of COVID-19 development (p = 0.002), contrary to carriers of variant GSTM3-CC genotype which have higher odds for COVID-19 (p = 0.024). Moreover, combined GSTP1 (rs1138272 and rs1695) and GSTM3 genotype exhibited cumulative risk regarding both COVID-19 occurrence and COVID-19 severity (p = 0.001 and p = 0.025, respectively). Further studies are needed to clarify the exact roles of specific glutathione S-transferases once the SARS-CoV-2 infection is initiated in the host cell.
Collapse
Affiliation(s)
- Vesna Coric
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Ivana Milosevic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Infectious and Tropical Diseases, Clinical Centre of Serbia, Belgrade, Serbia
| | - Tatjana Djukic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Zoran Bukumiric
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical Statistics and Informatics, Belgrade, Serbia
| | - Ana Savic-Radojevic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Marija Matic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Djurdja Jerotic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Nevena Todorovic
- Clinic of Infectious and Tropical Diseases, Clinical Centre of Serbia, Belgrade, Serbia
| | - Milika Asanin
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Neurology, Clinical Centre of Serbia, Belgrade, Serbia
| | - Marko Ercegovac
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Cardiology, Clinical Centre of Serbia, Belgrade, Serbia
| | - Jovan Ranin
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Infectious and Tropical Diseases, Clinical Centre of Serbia, Belgrade, Serbia
| | - Goran Stevanovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Infectious and Tropical Diseases, Clinical Centre of Serbia, Belgrade, Serbia
| | - Marija Pljesa-Ercegovac
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
| | - Tatjana Simic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| |
Collapse
|
15
|
Castro-Gil MP, Torres-Mena JE, Salgado RM, Muñoz-Montero SA, Martínez-Garcés JM, López-Torres CD, Mendoza-Vargas A, Gabiño-López NB, Villa-Treviño S, Del Pozo-Yauner L, Arellanes-Robledo J, Krötzsch E, Pérez-Carreón JI. The transcriptome of early GGT/KRT19-positive hepatocellular carcinoma reveals a downregulated gene expression profile associated with fatty acid metabolism. Genomics 2021; 114:72-83. [PMID: 34861383 DOI: 10.1016/j.ygeno.2021.11.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/05/2021] [Accepted: 11/26/2021] [Indexed: 01/14/2023]
Abstract
Hepatocellular carcinoma expressing hepatobiliary progenitor markers, is considered of poor prognosis. By using a hepatocarcinogenesis model, laser capture microdissection, and RNA-Sequencing analysis, we identified an expression profile in GGT/KRT19-positive experimental tumors; 438 differentially expressed genes were found in early and late nodules along with increased collagen deposition. Dysregulated genes were involved in Fatty Acid Metabolism, RXR function, and Hepatic Stellate Cells Activation. Downregulation of Slc27a5, Acsl1, and Cyp2e1, demonstrated that Retinoid X Receptor α (RXRα) function is compromised in GGT/KRT19-positive nodules. Since RXRα controls NRF2 pathway activation, we determined the expression of NRF2 targeted genes; Akr1b8, Akr7a3, Gstp1, Abcc3, Ptgr1, and Txnrd1 were upregulated, indicating NRF2 pathway activation. A comparative analysis in human HCC showed that SLC27A5, ACSL1, CYP2E1, and RXRα gene expression is mutually exclusive with KRT19 gene expression. Our results indicate that the downregulation of Slc27a5, Acsl1, Rxrα, and Cyp2e1 genes is an early event within GGT/KRT19-positive HCC.
Collapse
Affiliation(s)
| | | | - Rosa M Salgado
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", CDMX, Mexico
| | - Said A Muñoz-Montero
- Department of Computational Genomics, National Institute of Genomic Medicine, CDMX, Mexico
| | | | | | | | | | - Saúl Villa-Treviño
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute, CDMX, Mexico
| | - Luis Del Pozo-Yauner
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Jaime Arellanes-Robledo
- Laboratory of Liver Diseases, National Institute of Genomic Medicine, CDMX, Mexico; Directorate of Cátedras, National Council of Science and Technology, CDMX, Mexico
| | - Edgar Krötzsch
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", CDMX, Mexico
| | | |
Collapse
|
16
|
Zhou X, Shin S, He C, Zhang Q, Rasband MN, Ren J, Dai C, Zorrilla-Veloz RI, Shingu T, Yuan L, Wang Y, Chen Y, Lan F, Hu J. Qki regulates myelinogenesis through Srebp2-dependent cholesterol biosynthesis. eLife 2021; 10:60467. [PMID: 33942715 PMCID: PMC8139834 DOI: 10.7554/elife.60467] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 05/01/2021] [Indexed: 01/14/2023] Open
Abstract
Myelination depends on timely, precise control of oligodendrocyte differentiation and myelinogenesis. Cholesterol is the most abundant component of myelin and essential for myelin membrane assembly in the central nervous system. However, the underlying mechanisms of precise control of cholesterol biosynthesis in oligodendrocytes remain elusive. In the present study, we found that Qki depletion in neural stem cells or oligodendrocyte precursor cells in neonatal mice resulted in impaired cholesterol biosynthesis and defective myelinogenesis without compromising their differentiation into Aspa+Gstpi+ myelinating oligodendrocytes. Mechanistically, Qki-5 functions as a co-activator of Srebp2 to control transcription of the genes involved in cholesterol biosynthesis in oligodendrocytes. Consequently, Qki depletion led to substantially reduced concentration of cholesterol in mouse brain, impairing proper myelin assembly. Our study demonstrated that Qki-Srebp2-controlled cholesterol biosynthesis is indispensable for myelinogenesis and highlights a novel function of Qki as a transcriptional co-activator beyond its canonical function as an RNA-binding protein.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Seula Shin
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Cancer Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, United States
| | - Chenxi He
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiang Zhang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Jiangong Ren
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Congxin Dai
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Rocío I Zorrilla-Veloz
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Cancer Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, United States
| | - Takashi Shingu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Liang Yuan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Graduate School of Biomedical Sciences, Tufts University, Boston, United States
| | - Yunfei Wang
- Clinical Science Division, H. Lee Moffitt Cancer Center & Research Institute, Tampa, United States
| | - Yiwen Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Fei Lan
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States.,Cancer Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, United States.,Neuroscience Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, United States
| |
Collapse
|
17
|
The Association of Polymorphisms in Nrf2 and Genes Involved in Redox Homeostasis in the Development and Progression of Clear Cell Renal Cell Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6617969. [PMID: 33953831 PMCID: PMC8068539 DOI: 10.1155/2021/6617969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/28/2020] [Accepted: 04/03/2021] [Indexed: 01/07/2023]
Abstract
Deleterious effects of SNPs found in genes encoding transcriptional factors, as well as antioxidant and detoxification enzymes, are disputable; however, their functional significance seems to modify the risk for clear cell renal cell carcinoma (ccRCC) development and progression. We investigated the effect of specific Nrf2, SOD2, GPX1 gene variants and GSTP1ABCD haplotype on ccRCC risk and prognosis and evaluated the association between GSTP1 and regulatory (JNK1/2) and executor (caspase-3) apoptotic molecule expression in ccRCC tissue samples and the presence of GSTP1 : JNK1/2 protein : protein interactions. Genotyping was performed in 223 ccRCC patients and 336 matched controls by PCR-CTTP and qPCR. Protein expression was analyzed using immunoblot, while the existence of GSTP1 : JNK1 protein : protein interactions was investigated by immunoprecipitation experiments. An increased risk of ccRCC development was found among carriers of variant genotypes of both SOD2 rs4880 and GSTP1 rs1695 polymorphisms. Nrf2 rs6721961 genetic polymorphism in combination with both rs4880 and rs1695 showed higher ccRCC risk as well. Haplotype analysis revealed significant risk of ccRCC development in carriers of the GSTP1C haplotype. Furthermore, GSTP1 variant forms seem to affect the overall survival in ccRCC patients, and the proposed molecular mechanism underlying the GSTP1 prognostic role might be the presence of GSTP1 : JNK1/2 protein : protein interactions.
Collapse
|
18
|
Amara I, Ontario ML, Scuto M, Lo Dico GM, Sciuto S, Greco V, Abid-Essefi S, Signorile A, Salinaro AT, Calabrese V. Moringa oleifera Protects SH-SY5YCells from DEHP-Induced Endoplasmic Reticulum Stress and Apoptosis. Antioxidants (Basel) 2021; 10:532. [PMID: 33805396 PMCID: PMC8065568 DOI: 10.3390/antiox10040532] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/23/2021] [Accepted: 03/19/2021] [Indexed: 12/29/2022] Open
Abstract
Moringa oleifera (MO) is a medicinal plant that has been shown to possess antioxidant, anticarcinogenic and antibiotic activities. In a rat model, MO extract (MOe) has been shown to have a protective effect against brain damage and memory decline. As an extending study, here, we have examined the protective effect of MOe against oxidative stress and apoptosis caused in human neuroblastome (SH-SY5Y) cells by di-(2-ethylhexyl) phthalate (DEHP), a plasticizer known to induce neurotoxicity. Our data show that MOe prevents oxidative damage by lowering reactive oxygen species (ROS) formation, restoring mitochondrial respiratory chain complex activities, and, in addition, by modulating the expression of vitagenes, i.e., antioxidant proteins Nrf2 and HO-1. Moreover, MOe prevented neuronal damage by partly inhibiting endoplasmic reticulum (ER) stress response, as indicated by decreased expression of CCAAT-enhancer-binding protein homologous protein (CHOP) and Glucose-regulated protein 78 (GRP78) proteins. MOe also protected SH-SY5Y cells from DEHP-induced apoptosis, preserving mitochondrial membrane permeability and caspase-3 activation. Our findings provide insight into understanding of molecular mechanisms involved in neuroprotective effects by MOe against DEHP damage.
Collapse
Affiliation(s)
- Ines Amara
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (I.A.); (M.L.O.); (M.S.); (G.M.L.D.); (S.S.); (V.G.); (V.C.)
- Laboratory for Research on Biologically Compatible Compounds, Faculty of Dental Medicine, University of Monastir, Rue Avicenne, Monastir 5019, Tunisia;
| | - Maria Laura Ontario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (I.A.); (M.L.O.); (M.S.); (G.M.L.D.); (S.S.); (V.G.); (V.C.)
| | - Maria Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (I.A.); (M.L.O.); (M.S.); (G.M.L.D.); (S.S.); (V.G.); (V.C.)
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Gianluigi Maria Lo Dico
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (I.A.); (M.L.O.); (M.S.); (G.M.L.D.); (S.S.); (V.G.); (V.C.)
| | - Sebastiano Sciuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (I.A.); (M.L.O.); (M.S.); (G.M.L.D.); (S.S.); (V.G.); (V.C.)
| | - Valentina Greco
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (I.A.); (M.L.O.); (M.S.); (G.M.L.D.); (S.S.); (V.G.); (V.C.)
| | - Salwa Abid-Essefi
- Laboratory for Research on Biologically Compatible Compounds, Faculty of Dental Medicine, University of Monastir, Rue Avicenne, Monastir 5019, Tunisia;
| | - Anna Signorile
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Piazza G. Cesare, 11, 70124 Bari, Italy
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (I.A.); (M.L.O.); (M.S.); (G.M.L.D.); (S.S.); (V.G.); (V.C.)
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (I.A.); (M.L.O.); (M.S.); (G.M.L.D.); (S.S.); (V.G.); (V.C.)
| |
Collapse
|
19
|
Kim HJ, Lee JH, Lee KB, Shin JW, Kwon MA, Lee S, Jeong EM, Cho SY, Kim IG. Transglutaminase 2 crosslinks the glutathione S-transferase tag, impeding protein-protein interactions of the fused protein. Exp Mol Med 2021; 53:115-124. [PMID: 33441971 PMCID: PMC8080825 DOI: 10.1038/s12276-020-00549-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 01/29/2023] Open
Abstract
Glutathione S-transferase (GST) from Schistosoma japonicum has been widely used as a tag for affinity purification and pulldown of fusion proteins to detect protein-protein interactions. However, the reliability of this technique is undermined by the formation of GST-fused protein aggregates after incubation with cell lysates. It remains unknown why this aggregation occurs. Here, we demonstrate that the GST tag is a substrate of transglutaminase 2 (TG2), which is a calcium-dependent enzyme that polyaminates or crosslinks substrate proteins. Mutation analysis identified four glutamine residues in the GST tag as polyamination sites. TG2-mediated modification of the GST tag caused aggregate formation but did not affect its glutathione binding affinity. When incubated with cell lysates, GST tag aggregation was dependent on cellular TG2 expression levels. A GST mutant in which four glutamine residues were replaced with asparagine (GST4QN) exhibited a glutathione binding affinity similar to that of wild-type GST and could be purified by glutathione affinity chromatography. Moreover, the use of GST4QN as a tag reduced fused p53 aggregation and enhanced the induction of p21 transcription and apoptosis in cells treated with 5-fluorouracil (5-FU). These results indicated that TG2 interferes with the protein-protein interactions of GST-fused proteins by crosslinking the GST tag; therefore, a GST4QN tag could improve the reproducibility and reliability of GST pulldown experiments.
Collapse
Affiliation(s)
- Hyo-Jun Kim
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Jin-Haeng Lee
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Ki Baek Lee
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Woong Shin
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Mee-ae Kwon
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Soojin Lee
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Eui Man Jeong
- grid.411277.60000 0001 0725 5207Department of Pharmacy, College of Pharmacy, Jeju National University, Jeju Special Self-Governing Province, Korea
| | - Sung-Yup Cho
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea ,grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - In-Gyu Kim
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea ,grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea ,grid.31501.360000 0004 0470 5905Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
20
|
Abstract
Drug metabolizing enzymes catalyze the biotransformation of many of drugs and chemicals. The drug metabolizing enzymes are distributed among several evolutionary families and catalyze a range of detoxication reactions, including oxidation/reduction, conjugative, and hydrolytic reactions that serve to detoxify potentially toxic compounds. This detoxication function requires that drug metabolizing enzymes exhibit substrate promiscuity. In addition to their catalytic functions, many drug metabolizing enzymes possess functions unrelated to or in addition to catalysis. Such proteins are termed 'moonlighting proteins' and are defined as proteins with multiple biochemical or biophysical functions that reside in a single protein. This review discusses the diverse moonlighting functions of drug metabolizing enzymes and the roles they play in physiological functions relating to reproduction, vision, cell signaling, cancer, and transport. Further research will likely reveal new examples of moonlighting functions of drug metabolizing enzymes.
Collapse
Affiliation(s)
- Philip G Board
- John Curtin School of Medical Research, ANU College of Health and Medicine, The Australian National University, Canberra, ACT, Australia
| | - M W Anders
- Department of Pharmacology and Physiology, University of Rochester Medical Center, New York, NY, USA
| |
Collapse
|
21
|
Yamashita Y, Sakakibara H, Toda T, Ashida H. Insights into the potential benefits of black soybean ( Glycine max L.) polyphenols in lifestyle diseases. Food Funct 2020; 11:7321-7339. [PMID: 32852022 DOI: 10.1039/d0fo01092h] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Black soybean (Glycine max L.), a cultivar containing abundant polyphenols in its seed coat such as anthocyanins and flavan-3-ols, has been reported to possess various health benefits toward lifestyle diseases. In this review article, the safety evaluation of polyphenol-rich black soybean seed coat extract (BE) and absorption of BE polyphenols are summarized. Additionally, we describe the antioxidant activity of BE polyphenols and their ability to induce antioxidant enzymes. The health benefits of BE and its polyphenols, such as anti-obesity and anti-hyperglycemic activities through the activation of AMP-activated protein kinase and translocation of glucose transporter 4, respectively, are also discussed. Furthermore, we found that black soybean polyphenols were involved in the improvement of vascular function. These emerging data require further investigation in scientific studies and human trials to evaluate the prevention of lifestyle diseases using black soybean polyphenols.
Collapse
Affiliation(s)
- Yoko Yamashita
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan.
| | | | - Toshiya Toda
- Department of Innovative Food Sciences, School of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya 663-8558, Japan
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan.
| |
Collapse
|
22
|
Pljesa-Ercegovac M, Savic-Radojevic A, Coric V, Radic T, Simic T. Glutathione transferase genotypes may serve as determinants of risk and prognosis in renal cell carcinoma. Biofactors 2020; 46:229-238. [PMID: 31483924 DOI: 10.1002/biof.1560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/18/2019] [Indexed: 12/25/2022]
Abstract
Renal cell carcinoma (RCC) represents a group of histologically similar neoplasms with significant intratumor and intertumor genetic heterogeneity. Recognized risk factors for RCC development include smoking, hypertension, obesity, as well as von Hippel-Lindau (VHL) disease. Inactivation of VHL, deregulated nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway, and altered redox homeostasis, together with changes in glutathione transferase (GST) profile, are considered as important contributing factors in RCC development and progression. Although the available results of both gene-gene and gene-environment analysis are quite heterogeneous, they clearly indicate that certain GST genotypes may play a role as risk modifiers, either individually or in combination with other Phase I or Phase II gene polymorphisms, as well as in subjects exposed to relevant substrates. Seemingly, GST genotyping could identify individuals with impaired detoxification in renal parenchyma that are at higher risk of developing RCC. In addition to well established roles of GSTs in conjugation and biotransformation of xenobiotics, GSTs have emerged as significant regulators of pathways determining cell proliferation and survival. Indeed, there are evidence in favor of GST significance, not only in terms of risk for RCC development, but also with respect to progression and prognosis. So far, GSTM1-active genotype was confirmed to be an independent predictor of higher risk of overall mortality. Therefore, it is reasonable to assume that certain GST variants may assist in individual RCC risk assessment, as well as postoperative prognosis. Even more, GST profiling might contribute to development of personalized targeted therapy in RCC patients.
Collapse
Affiliation(s)
- Marija Pljesa-Ercegovac
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ana Savic-Radojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vesna Coric
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tanja Radic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tatjana Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
23
|
Fuda H, Miyanaga S, Furukawa T, Umetsu S, Joko S, Roan Y, Suzuki H, Hui SP, Watanabe M, Chiba H. Flazin as a Promising Nrf2 Pathway Activator. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:12844-12853. [PMID: 31668063 DOI: 10.1021/acs.jafc.9b04600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Flazin is a β-carboline-derived alkaloid found in Japanese fermented foods. Here, the potential of flazin as an antioxidant food was studied with particular reference to its effect on the Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2) system in human hepatocytes (C3A). Flazin and flazin analogues including the decarboxylated derivative perlolyrine were chemically synthesized and compared with each other and with chlorogenic acid and curcumin. Among these compounds, flazin showed the lowest cytotoxicity (IC50 < 500 μM) and the highest capacity to activate the Keap1-Nrf2 system. It provided the largest (>3-fold of the control) cytoprotection ability against a pro-oxidant, although its radical absorbance capacity was relatively low. Flazin increased the expressions of Nrf2-dependent phase II enzyme genes and their products (NQO1, GSTP, and GSH proteins). The strong cytoprotection ability of flazin associated with low log P (0-3) is shared by sulforaphane and 3,5-dihydroxy-4-methoxybenzyl alcohol, suggesting the potential value of flazin and flazin-rich foods for the prevention of oxidation-related health disorders.
Collapse
Affiliation(s)
- Hirotoshi Fuda
- Faculty of Health Sciences , Hokkaido University , Sapporo 060-0812 , Japan
| | - Satoshi Miyanaga
- Faculty of Health Sciences , Hokkaido University , Sapporo 060-0812 , Japan
| | - Takayuki Furukawa
- Faculty of Health Sciences , Hokkaido University , Sapporo 060-0812 , Japan
| | - Satomi Umetsu
- Faculty of Health Sciences , Hokkaido University , Sapporo 060-0812 , Japan
| | - Sae Joko
- Faculty of Health Sciences , Hokkaido University , Sapporo 060-0812 , Japan
| | - Yuning Roan
- Faculty of Health Sciences , Hokkaido University , Sapporo 060-0812 , Japan
| | - Hirotaka Suzuki
- Faculty of Health Sciences , Hokkaido University , Sapporo 060-0812 , Japan
| | - Shu-Ping Hui
- Faculty of Health Sciences , Hokkaido University , Sapporo 060-0812 , Japan
| | - Mitsugu Watanabe
- Watanabe Oyster Laboratory Co. Ltd. , 490-3, Shimoongata-cho , Hachioji , Tokyo 190-0154 , Japan
| | - Hitoshi Chiba
- Department of Nutrition , Sapporo University of Health Sciences , Nakanuma Nishi-4-2-1-15 , Higashi-ku, Sapporo 007-0894 , Japan
| |
Collapse
|
24
|
Giuliani ME, Sparaventi E, Lanzoni I, Pittura L, Regoli F, Gorbi S. Precision-Cut Tissue Slices (PCTS) from the digestive gland of the Mediterranean mussel Mytilus galloprovincialis: An ex vivo approach for molecular and cellular responses in marine invertebrates. Toxicol In Vitro 2019; 61:104603. [PMID: 31330176 DOI: 10.1016/j.tiv.2019.104603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/05/2019] [Accepted: 07/17/2019] [Indexed: 11/25/2022]
Abstract
The precision-cut tissue slices (PCTS) represent a largely used biological model in mammalian research. This ex vivo approach offers the main advantages of in vitro systems, while maintaining the natural architecture of the tissue. The use of PCTS in toxicological research has been proposed for investigating the cellular effects of xenobiotics or bioactive compounds mostly in mammalian models. Their application is increasing also in marine organisms, but still limited to fish. This work validates the use of PCTS in an invertebrate species, the Mediterranean mussel Mytilus galloprovincialis. Intact tissue slices of different thicknesses (300, 350 and 400 μm) were successfully obtained from the digestive gland. The slices maintained the histological integrity and the viability after 6 h and 24 h incubation in culture medium, with some differences depending on the thickness. The enzymatic activities and mRNA levels of catalase and glutathione S-transferase, chosen as model biological endpoints, were measured until 24 h incubation, revealing the functionality of such systems. This work demonstrates the suitability of mussel PCTS for investigating molecular and cellular responses in ecotoxicological research.
Collapse
Affiliation(s)
- Maria Elisa Giuliani
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Erica Sparaventi
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Ilaria Lanzoni
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Lucia Pittura
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Francesco Regoli
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Stefania Gorbi
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy.
| |
Collapse
|
25
|
López-Rodríguez JC, Manosalva J, Cabrera-García JD, Escribese MM, Villalba M, Barber D, Martínez-Ruiz A, Batanero E. Human glutathione-S-transferase pi potentiates the cysteine-protease activity of the Der p 1 allergen from house dust mite through a cysteine redox mechanism. Redox Biol 2019; 26:101256. [PMID: 31229842 PMCID: PMC6597738 DOI: 10.1016/j.redox.2019.101256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/30/2019] [Accepted: 06/10/2019] [Indexed: 11/30/2022] Open
Abstract
Environmental proteases have been widely associated to the pathogenesis of allergic disorders. Der p 1, a cysteine-protease from house dust mite (HDM) Dermatophagoides pteronyssinus, constitutes one of the most clinically relevant indoor aeroallergens worldwide. Der p 1 protease activity depends on the redox status of its catalytic cysteine residue, which has to be in the reduced state to be active. So far, it is unknown whether Der p 1-protease activity could be regulated by host redox microenvironment once it reaches the lung epithelial lining fluid in addition to endogenous mite components. In this sense, Glutathione-S-transferase pi (GSTpi), an enzyme traditionally linked to phase II detoxification, is highly expressed in human lung epithelial cells, which represent the first line of defence against aeroallergens. Moreover, GSTpi is a generalist catalyst of protein S-glutathionylation reactions, and some polymorphic variants of this enzyme has been associated to the development of allergic asthma. Here, we showed that human GSTpi increased the cysteine-protease activity of Der p 1, while GSTmu (the isoenzyme produced by the mite) did not alter it. GSTpi induces the reduction of Cys residues in Der p 1, probably by rearranging its disulphide bridges. Furthermore, GSTpi was detected in the apical medium collected from human bronchial epithelial cell cultures, and more interesting, it increased cysteine-protease activity of Der p 1. Our findings support the role of human GSTpi from airways in modulating of Der p 1 cysteine-protease activity, which may have important clinical implications for immune response to this aeroallergen in genetically susceptible individuals.
Collapse
Affiliation(s)
- Juan Carlos López-Rodríguez
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - Juliana Manosalva
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - J Daniel Cabrera-García
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - María M Escribese
- Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo CEU, Madrid, Spain
| | - Mayte Villalba
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - Domingo Barber
- Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo CEU, Madrid, Spain
| | - Antonio Martínez-Ruiz
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain.
| | - Eva Batanero
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain.
| |
Collapse
|
26
|
Bartolini D, Wang Y, Zhang J, Giustarini D, Rossi R, Wang GY, Torquato P, Townsend DM, Tew KD, Galli F. A seleno-hormetine protects bone marrow hematopoietic cells against ionizing radiation-induced toxicities. PLoS One 2019; 14:e0205626. [PMID: 31034521 PMCID: PMC6488049 DOI: 10.1371/journal.pone.0205626] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 03/28/2019] [Indexed: 01/05/2023] Open
Abstract
2,2'-diselenyldibenzoic acid (DSBA) is a chemical probe produced to explore the pharmacological properties of diphenyldiselenide-derived agents with seleno-hormetic activity undergoing preclinical development. The present study was designed to verify in vivo the drug's properties and to determine mechanistically how these may mediate the protection of tissues against stress conditions, exemplified by ionizing radiation induced damage in mouse bone marrow. In murine bone marrow hematopoietic cells, the drug initiated the activation of the Nrf2 transcription factor resulting in enhanced expression of downstream stress response genes. This type of response was confirmed in human liver cells and included enhanced expression of glutathione S-transferases (GST), important in the metabolism and pharmacological function of seleno-compounds. In C57 BL/6 mice, DSBA prevented the suppression of bone marrow hematopoietic cells caused by ionizing radiation exposure. Such in vivo prevention effects were associated with Nrf2 pathway activation in both bone marrow cells and liver tissue. These findings demonstrated for the first time the pharmacological properties of DSBA in vivo, suggesting a practical application for this type of Se-hormetic molecules as a radioprotective and/or prevention agents in cancer treatments.
Collapse
Affiliation(s)
- Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
- * E-mail:
| | - Yanzhong Wang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States of America
| | - Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States of America
| | - Daniela Giustarini
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Ranieri Rossi
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Gavin Y. Wang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States of America
| | - Pierangelo Torquato
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Danyelle M. Townsend
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States of America
| | - Kenneth D. Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States of America
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
27
|
Shao D, Wu Z, Bai S, Fu G, Zou Z. The function of miRNA‑153 against isoflurane‑induced neurotoxicity via Nrf2/ARE cytoprotection. Mol Med Rep 2019; 19:4001-4010. [PMID: 30896808 PMCID: PMC6471563 DOI: 10.3892/mmr.2019.10056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 08/17/2018] [Indexed: 12/28/2022] Open
Abstract
The present study aimed to investigate the function of micro (mi)RNA‑153 against isoflurane‑induced neurotoxicity and its mechanism. In isoflurane‑induced mice, miRNA‑153 expression was downregulated compared with in the control group. Downregulation of miRNA‑153 induced neurocyte apoptosis, reduced cell growth and promoted oxidative stress in an in vitro model. Overexpression of miRNA‑153 reduced oxidative stress, promoted cell growth and inhibited neurocyte apoptosis within an in vitro model. Downregulation of miRNA‑153 suppressed nuclear erythroid‑2 related factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathway, which was induced via the overexpression of miRNA‑153 in vitro. The Nrf2 agonist, dimethyl fumarate (2.5 µM), induced the Nrf2/ARE signaling pathway and reduced oxidative stress to induce neurocyte apoptosis in vitro following treatment with anti‑miRNA‑153. The results of the present study suggested the function of miRNA‑153 against neurotoxicity via Nrf2/ARE‑mediated cytoprotection.
Collapse
Affiliation(s)
- Dong Shao
- Department of Anesthesiology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Zhouquan Wu
- Department of Anesthesiology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Shuying Bai
- Department of Anesthesiology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Guowei Fu
- Department of Anesthesiology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Zhiqing Zou
- Department of Anesthesiology, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
28
|
Scirè A, Cianfruglia L, Minnelli C, Bartolini D, Torquato P, Principato G, Galli F, Armeni T. Glutathione compartmentalization and its role in glutathionylation and other regulatory processes of cellular pathways. Biofactors 2019; 45:152-168. [PMID: 30561781 DOI: 10.1002/biof.1476] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/20/2022]
Abstract
Glutathione is considered the major non-protein low molecular weight modulator of redox processes and the most important thiol reducing agent of the cell. The biosynthesis of glutathione occurs in the cytosol from its constituent amino acids, but this tripeptide is also present in the most important cellular districts, such as mitochondria, nucleus, and endoplasmic reticulum, thus playing a central role in several metabolic pathways and cytoprotection mechanisms. Indeed, glutathione is involved in the modulation of various cellular processes and, not by chance, it is a ubiquitous determinant for redox signaling, xenobiotic detoxification, and regulation of cell cycle and death programs. The balance between its concentration and redox state is due to a complex series of interactions between biosynthesis, utilization, degradation, and transport. All these factors are of great importance to understand the significance of cellular redox balance and its relationship with physiological responses and pathological conditions. The purpose of this review is to give an overview on glutathione cellular compartmentalization. Information on its subcellular distribution provides a deeper understanding of glutathione-dependent processes and reflects the importance of compartmentalization in the regulation of specific cellular pathways. © 2018 BioFactors, 45(2):152-168, 2019.
Collapse
Affiliation(s)
- Andrea Scirè
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Laura Cianfruglia
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Cristina Minnelli
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Desirée Bartolini
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Pierangelo Torquato
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Giovanni Principato
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Galli
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Tatiana Armeni
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
29
|
Bartolini D, Torquato P, Piroddi M, Galli F. Targeting glutathione S-transferase P and its interactome with selenium compounds in cancer therapy. Biochim Biophys Acta Gen Subj 2019; 1863:130-143. [DOI: 10.1016/j.bbagen.2018.09.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 12/14/2022]
|
30
|
García S, Alberich S, MacDowell KS, Martínez-Cengotitabengoa M, López P, Zorrilla I, Leza JC, González-Pinto A. Association Between Medication Adherence and Oxidative Stress in Patients With First-Episode Mania. Front Psychiatry 2019; 10:162. [PMID: 30971964 PMCID: PMC6445053 DOI: 10.3389/fpsyt.2019.00162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 03/05/2019] [Indexed: 11/13/2022] Open
Abstract
Poor adherence is a major problem in patients with manic episodes that impairs functionality and has unknown effects on oxidative stress. The objective of this study was to analyze the relationship between adherence to medication, severity of symptoms and oxidative stress in a sample of patients with a first episode of mania. A longitudinal, 6-month study was performed in 60 patients, who were classified as adherent and non-adherent to medication (mainly antipsychotics). Blood levels of oxidative stress parameters and expression of the antioxidant nuclear transcription factor NRF2 in mononuclear cells of peripheral blood were assessed at baseline and at the end of follow-up. In addition, clinical symptoms and functioning were evaluated. Linear multivariate regression was used to determine the relationship between adherence, oxidative stress, and clinical symptoms. Finally, 44 patients completed follow-up. The results of this study showed that at 6-month follow-up, adherence was significantly associated with better functioning and reduced clinical symptoms. Additionally, more severe symptoms were associated with increased levels of oxidative stress and antioxidant parameters. At study completion, non-adherents exhibited greater levels of antioxidants than adherent patients. In conclusion, poor adherence to medication is associated with a poorer prognosis in the medium term and causes increased antioxidant response.
Collapse
Affiliation(s)
- Saínza García
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,School of Medicine, University of the Basque Country, Vitoria, Spain
| | - Susana Alberich
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain
| | - Karina S MacDowell
- Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid, IUIN and IIS Hospital 12 de Octubre, Madrid, Spain
| | - Mónica Martínez-Cengotitabengoa
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,School of Medicine, University of the Basque Country, Vitoria, Spain.,Psychobiology Department, National Distance Education University (UNED), Vitoria, Spain
| | - Purificación López
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,School of Medicine, University of the Basque Country, Vitoria, Spain
| | - Iñaki Zorrilla
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,School of Medicine, University of the Basque Country, Vitoria, Spain
| | - Juan Carlos Leza
- Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid, IUIN and IIS Hospital 12 de Octubre, Madrid, Spain
| | - Ana González-Pinto
- Department of Psychiatry, University Hospital of Alava-Santiago, Vitoria, Spain.,Centre for Biomedical Research Network on Mental Health (CIBERSAM), Madrid, Spain.,School of Medicine, University of the Basque Country, Vitoria, Spain
| |
Collapse
|
31
|
Higareda-Almaraz JC, Karbiener M, Giroud M, Pauler FM, Gerhalter T, Herzig S, Scheideler M. Norepinephrine triggers an immediate-early regulatory network response in primary human white adipocytes. BMC Genomics 2018; 19:794. [PMID: 30390616 PMCID: PMC6215669 DOI: 10.1186/s12864-018-5173-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/16/2018] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Norepinephrine (NE) signaling has a key role in white adipose tissue (WAT) functions, including lipolysis, free fatty acid liberation and, under certain conditions, conversion of white into brite (brown-in-white) adipocytes. However, acute effects of NE stimulation have not been described at the transcriptional network level. RESULTS We used RNA-seq to uncover a broad transcriptional response. The inference of protein-protein and protein-DNA interaction networks allowed us to identify a set of immediate-early genes (IEGs) with high betweenness, validating our approach and suggesting a hierarchical control of transcriptional regulation. In addition, we identified a transcriptional regulatory network with IEGs as master regulators, including HSF1 and NFIL3 as novel NE-induced IEG candidates. Moreover, a functional enrichment analysis and gene clustering into functional modules suggest a crosstalk between metabolic, signaling, and immune responses. CONCLUSIONS Altogether, our network biology approach explores for the first time the immediate-early systems level response of human adipocytes to acute sympathetic activation, thereby providing a first network basis of early cell fate programs and crosstalks between metabolic and transcriptional networks required for proper WAT function.
Collapse
Affiliation(s)
- Juan Carlos Higareda-Almaraz
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Metabolic Control, Medical Faculty, Technical University, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- NMR laboratory, Institute of Myology, Hopital Universitaire Pitie Salpetriere, Paris, France
| | - Michael Karbiener
- Department of Phoniatrics, ENT University Hospital, Medical University of Graz, Graz, Austria
| | - Maude Giroud
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Metabolic Control, Medical Faculty, Technical University, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Florian M. Pauler
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Present Address: Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Teresa Gerhalter
- Present Address: Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Metabolic Control, Medical Faculty, Technical University, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcel Scheideler
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Metabolic Control, Medical Faculty, Technical University, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- NMR laboratory, Institute of Myology, Hopital Universitaire Pitie Salpetriere, Paris, France
| |
Collapse
|
32
|
Dong SC, Sha HH, Xu XY, Hu TM, Lou R, Li H, Wu JZ, Dan C, Feng J. Glutathione S-transferase π: a potential role in antitumor therapy. Drug Des Devel Ther 2018; 12:3535-3547. [PMID: 30425455 PMCID: PMC6204874 DOI: 10.2147/dddt.s169833] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Glutathione S-transferase π (GSTπ) is a Phase II metabolic enzyme that is an important facilitator of cellular detoxification. Traditional dogma asserts that GSTπ functions to catalyze glutathione (GSH)-substrate conjunction to preserve the macromolecule upon exposure to oxidative stress, thus defending cells against various toxic compounds. Over the past 20 years, abnormal GSTπ expression has been linked to the occurrence of tumor resistance to chemotherapy drugs, demonstrating that this enzyme possesses functions beyond metabolism. This revelation reveals exciting possibilities in the realm of drug discovery, as GSTπ inhibitors and its prodrugs offer a feasible strategy in designing anticancer drugs with the primary purpose of reversing tumor resistance. In connection with the authors' current research, we provide a review on the biological function of GSTπ and current developments in GSTπ-targeting drugs, as well as the prospects of future strategies.
Collapse
Affiliation(s)
- Shu-Chen Dong
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Huan-Huan Sha
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Xiao-Yue Xu
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Tian-Mu Hu
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
| | - Rui Lou
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Huizi Li
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Jian-Zhong Wu
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Chen Dan
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Jifeng Feng
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| |
Collapse
|
33
|
García S, Alberich S, Martínez-Cengotitabengoa M, Arango C, Castro-Fornieles J, Parellada M, Baeza I, Moreno C, Micó JA, Berrocoso E, Graell M, Otero S, Simal T, González-Pinto A. The complex association between the antioxidant defense system and clinical status in early psychosis. PLoS One 2018; 13:e0194685. [PMID: 29698400 PMCID: PMC5919675 DOI: 10.1371/journal.pone.0194685] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 03/07/2018] [Indexed: 11/19/2022] Open
Abstract
Oxidative stress is a pathophysiological mechanism potentially involved in psychiatric disorders. The objective of this study was to assess the relationship between total antioxidant status (TAS) and the functional status of patients with a first episode of psychosis at the onset of the disease. For this purpose, a sample of 70 patients aged between 9 and 17 years with a first episode of psychosis were followed up for a period of two years. Blood samples were drawn to measure TAS levels at three time points: at baseline, at one year, and at two years. Clinical symptoms and functioning were also assessed at the same time points using various scales. Linear regression analysis was performed to investigate the relationship between TAS and clinical status at each assessment, adjusting for potential confounding factors. The distribution of clinical variables was grouped in different percentiles to assess the dose-response in the relation between clinical variables and TAS. At baseline, patient's score on Children's Global Assessment Scale (CGAS) was directly and significantly associated with TAS with a monotonic increase in percentiles, and surprising this association was reversed after one and two years of follow-up with a monotonic decrease. In summary at the onset of the illness, TAS is positively related to clinical status, whereas as the illness progresses this correlation is reversed and becomes negative. This may be the result of an adaptive response.
Collapse
Affiliation(s)
- Saínza García
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- BioAraba Research Institute, OSI Araba, Department of Psychiatry, Araba University Hospital, Vitoria, Spain
- University of the Basque Country, Vitoria, Spain
| | - Susana Alberich
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- BioAraba Research Institute, OSI Araba, Department of Psychiatry, Araba University Hospital, Vitoria, Spain
| | - Mónica Martínez-Cengotitabengoa
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- BioAraba Research Institute, OSI Araba, Department of Psychiatry, Araba University Hospital, Vitoria, Spain
- University of the Basque Country, Vitoria, Spain
- Psychobiology Department, National Distance Education University (UNED), Vitoria, Spain
| | - Celso Arango
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- Child and Adolescent Psychiatry Department, Gregorio Marañón General University Hospital, IiSGM, Madrid, Spain
- School of Medicine, Complutense University, Madrid, Spain
| | - Josefina Castro-Fornieles
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Child and Adolescent Psychiatry and Psychology, SGR489, Institute of Neuroscience, Hospital Clínic of Barcelona, Barcelona, Spain
- Department of Psychiatry and Clinical Psychobiology, University of Barcelona, Barcelona, Spain
| | - Mara Parellada
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- Child and Adolescent Psychiatry Department, Gregorio Marañón General University Hospital, IiSGM, Madrid, Spain
- School of Medicine, Complutense University, Madrid, Spain
| | - Inmaculada Baeza
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Child and Adolescent Psychiatry and Psychology, SGR489, Institute of Neuroscience, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Carmen Moreno
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- Child and Adolescent Psychiatry Department, Gregorio Marañón General University Hospital, IiSGM, Madrid, Spain
- School of Medicine, Complutense University, Madrid, Spain
| | - Juan Antonio Micó
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- Neuropsychopharmacology & Psychobiology Research Group, University of Cádiz, Cadiz, Spain
- Institute of Research and Innovation in Biomedical Sciences, INiBICA, Cádiz, Spain
| | - Esther Berrocoso
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- Neuropsychopharmacology & Psychobiology Research Group, University of Cádiz, Cadiz, Spain
- Institute of Research and Innovation in Biomedical Sciences, INiBICA, Cádiz, Spain
| | - Montserrat Graell
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- Child and Adolescent Psychiatry and Psychology Department, Niño Jesús University Children's Hospital, La Princesa Institute for Health Research, Madrid, Spain
| | - Soraya Otero
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- Department of Psychiatry, Marqués de Valdecilla University Hospital, IDIVAL, Santander, Spain
- Child and Adolescent Psychiatry Unit, Marqués de Valdecilla University Hospital, Santander, Spain
- School of Medicine, University of Cantabria, Santander, Spain
| | - Tatiana Simal
- Department of Psychiatry, Miguel Servet University Hospital, Zaragoza, Spain
| | - Ana González-Pinto
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), Madrid, Spain
- BioAraba Research Institute, OSI Araba, Department of Psychiatry, Araba University Hospital, Vitoria, Spain
- University of the Basque Country, Vitoria, Spain
- * E-mail:
| |
Collapse
|
34
|
Suojalehto H, Lindström I, Wolff H, Puustinen A. Nasal protein profiles in work-related asthma caused by different exposures. Allergy 2018; 73:653-663. [PMID: 28960398 DOI: 10.1111/all.13325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND The mechanisms of work-related asthma (WRA) are incompletely delineated. Nasal cell samples may be informative about processes in the lower airways. Our aim was to determine the nasal protein expression profiles of WRA caused by different kind of exposures. METHODS We collected nasal brush samples from 82 nonsmoking participants, including healthy controls and WRA patients exposed to (i) protein allergens, (ii) isocyanates and (iii) welding fumes the day after relevant exposure. The proteome changes in samples were analysed by two-dimensional difference gel electrophoresis, and the differentially regulated proteins found were identified by mass spectrometry. Immunological comparison was carried out using Western blot. RESULTS We detected an average of 2500 spots per protein gel. Altogether, 228 protein spots were chosen for identification, yielding 77 different proteins. Compared to the controls, exposure to protein allergens had the largest effects on the proteome. Hierarchical clustering revealed that protein allergen- and isocyanate-related asthma had similar profiles, whereas asthma related to welding fumes differed. The highly overrepresented functional categories in the asthma groups were defence response, protease inhibitor activity, inflammatory and calcium signalling, complement activation and cellular response to oxidative stress. Immunological analysis confirmed the found abundance differences in galectin 10 and protein S100-A9 between the groups. CONCLUSIONS Work-related asthma patients exposed to protein allergens and isocyanates elicit similar nasal proteome responses and the profiles of welders and healthy controls were alike. Revealed biological activities of the protein expression changes are associated with allergic inflammation and asthma.
Collapse
Affiliation(s)
- H. Suojalehto
- Occupational Medicine; Finnish Institute of Occupational Health; Helsinki Finland
| | - I. Lindström
- Occupational Medicine; Finnish Institute of Occupational Health; Helsinki Finland
| | - H. Wolff
- Work Environment Laboratories; Finnish Institute of Occupational Health; Helsinki Finland
| | - A. Puustinen
- Unit of Systems Toxicology; Finnish Institute of Occupational Health; Helsinki Finland
- Verifin; Department of Chemistry; University of Helsinki; Helsinki Finland
| |
Collapse
|
35
|
Nrf2-p62 autophagy pathway and its response to oxidative stress in hepatocellular carcinoma. Transl Res 2018; 193:54-71. [PMID: 29274776 DOI: 10.1016/j.trsl.2017.11.007] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/06/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023]
Abstract
Deregulation of autophagy is proposed to play a key pathogenic role in hepatocellular carcinoma (HCC), the most common primary malignancy of the liver and the third leading cause of cancer death. Autophagy is an evolutionarily conserved catabolic process activated to degrade and recycle cell's components. Under stress conditions, such as oxidative stress and nutrient deprivation, autophagy is an essential survival pathway that operates in harmony with other stress response pathways. These include the redox-sensitive transcription complex Nrf2-Keap1 that controls groups of genes with roles in detoxification and antioxidant processes, intermediary metabolism, and cell cycle regulation. Recently, a functional association between a dysfunctional autophagy and Nrf2 pathway activation has been identified in HCC. This appears to occur through the physical interaction of the autophagy adaptor p62 with the Nrf2 inhibitor Keap1, thus leading to increased stabilization and transcriptional activity of Nrf2, a key event in reprogramming metabolic and stress response pathways of proliferating hepatocarcinoma cells. These emerging molecular mechanisms and the therapeutic perspective of targeting Nrf2-p62 interaction in HCC are discussed in this paper along with the prognostic value of autophagy in this type of cancer.
Collapse
|
36
|
Bocedi A, Noce A, Rovella V, Marrone G, Cattani G, Iappelli M, De Paolis P, Iaria G, Sforza D, Gallù M, Tisone G, Di Daniele N, Ricci G. Erythrocyte glutathione transferase in kidney transplantation: a probe for kidney detoxification efficiency. Cell Death Dis 2018; 9:288. [PMID: 29459773 PMCID: PMC5833821 DOI: 10.1038/s41419-018-0289-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023]
Abstract
Erythrocyte glutathione transferase (e-GST) is overexpressed in case of increased blood toxicity and its level correlates with the kidney disease progression. Thus, it represents a probe of kidney efficiency against circulating toxins. We measured the activity of e-GST in patients with transplant kidney from living and cadaver donors, correlated its level to biochemical parameters of kidney function, and measured the level of oxidized albumin as a probe of oxidative stress using a new simple procedure. Interestingly, the activity of e-GST in transplant patients from cadaver donors (N = 153) is very high (11.7 U/gHb) compared to healthy subjects (N = 80) ( 5.6 U/gHb). Lower values were observed in transplant patients with kidney from living donors (N = 16) (9.8 U/gHb). Except for steroids, no correlation has been found with the immunosuppressive therapies and routine clinical and laboratory parameters. Also serum oxidized albumin, which reveals oxidative stress, is significantly higher in transplant patients from cadaver donors (53%) compared to that from living donors (36%). Overall, these data indicate that most of transplant kidneys from cadavers lost part of the detoxifying power against circulating toxins and suffer a relevant oxidative stress compared to those coming from living donors. A case report suggests that e-GST could represent a very early marker of incipient graft rejection. In conclusion, e-GST may be used to check the decline or maintenance of the kidney detoxification competence during post-transplantation course.
Collapse
Affiliation(s)
- Alessio Bocedi
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Annalisa Noce
- Department of Systems Medicine, Hypertension and Nephrology Unit, University of Rome, Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Valentina Rovella
- Department of Systems Medicine, Hypertension and Nephrology Unit, University of Rome, Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Giulia Marrone
- Department of Systems Medicine, Hypertension and Nephrology Unit, University of Rome, Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Giada Cattani
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Massimo Iappelli
- Nephrology and Transplant Unit, A.O.S. Camillo-Forlanini, Circonvallazione Gianicolense 87, 00152, Rome, Italy
| | - Paolo De Paolis
- Nephrology and Transplant Unit, A.O.S. Camillo-Forlanini, Circonvallazione Gianicolense 87, 00152, Rome, Italy
| | - Giuseppe Iaria
- Liver and kidney Transplant Centre, University of Rome, Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Daniele Sforza
- Liver and kidney Transplant Centre, University of Rome, Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Mariacarla Gallù
- Department of Systems Medicine, Hypertension and Nephrology Unit, University of Rome, Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Giuseppe Tisone
- Liver and kidney Transplant Centre, University of Rome, Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, Hypertension and Nephrology Unit, University of Rome, Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Giorgio Ricci
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy.
| |
Collapse
|
37
|
Murdolo G, Bartolini D, Tortoioli C, Piroddi M, Torquato P, Galli F. Selenium and Cancer Stem Cells. Adv Cancer Res 2017; 136:235-257. [PMID: 29054420 DOI: 10.1016/bs.acr.2017.07.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Selenium (Se) is an essential micronutrient that functions as "redox gatekeeper" and homeostasis factor of normal and cancer cells. Epidemiology and experimental studies, in the last years suggested that both inorganic and organic forms of Se may have favorable health effects. In this regard, a protective action of Se on cellular systems that may help preventing cancer cell differentiation has been demonstrated, while the hypothesis that Se compounds may cure cancer and its metastatic diffusion appears speculative and is still a matter of investigation. Indeed, the overall actions of Se compounds in carcinogenesis are controversial. The recognition that cancer is a stem cell disease instigated major paradigm shifts in our basic understanding of cancer and attracted a great deal of interest. Although current treatment approaches in cancer are grounded in the need to kill the majority of cancer cells, targeting cancer stem cells (CSCs) may hold great potential in improving cancer treatment. In this respect, Se compounds have been demonstrated modulating numerous signaling pathways involved in CSC biology and these findings are now stimulating further research on optimal Se concentrations, most effective and cancer-specific Se compounds, and inherent pathways involved in redox and metabolic regulation of CSCs. In this review, we summarize the current knowledge about the effects of Se compounds on CSCs, by focusing on redox-dependent pathways and main gene regulation checkpoints that affect self-renewal, differentiation, and migration responses in this subpopulation of cancer cells.
Collapse
Affiliation(s)
- Giuseppe Murdolo
- Section of Internal Medicine, Endocrine and Metabolic Sciences, University of Perugia, Perugia, Italy.
| | | | - Cristina Tortoioli
- Section of Internal Medicine, Endocrine and Metabolic Sciences, University of Perugia, Perugia, Italy
| | | | | | | |
Collapse
|
38
|
Egea J, Fabregat I, Frapart YM, Ghezzi P, Görlach A, Kietzmann T, Kubaichuk K, Knaus UG, Lopez MG, Olaso-Gonzalez G, Petry A, Schulz R, Vina J, Winyard P, Abbas K, Ademowo OS, Afonso CB, Andreadou I, Antelmann H, Antunes F, Aslan M, Bachschmid MM, Barbosa RM, Belousov V, Berndt C, Bernlohr D, Bertrán E, Bindoli A, Bottari SP, Brito PM, Carrara G, Casas AI, Chatzi A, Chondrogianni N, Conrad M, Cooke MS, Costa JG, Cuadrado A, My-Chan Dang P, De Smet B, Debelec-Butuner B, Dias IHK, Dunn JD, Edson AJ, El Assar M, El-Benna J, Ferdinandy P, Fernandes AS, Fladmark KE, Förstermann U, Giniatullin R, Giricz Z, Görbe A, Griffiths H, Hampl V, Hanf A, Herget J, Hernansanz-Agustín P, Hillion M, Huang J, Ilikay S, Jansen-Dürr P, Jaquet V, Joles JA, Kalyanaraman B, Kaminskyy D, Karbaschi M, Kleanthous M, Klotz LO, Korac B, Korkmaz KS, Koziel R, Kračun D, Krause KH, Křen V, Krieg T, Laranjinha J, Lazou A, Li H, Martínez-Ruiz A, Matsui R, McBean GJ, Meredith SP, Messens J, Miguel V, Mikhed Y, Milisav I, Milković L, Miranda-Vizuete A, Mojović M, Monsalve M, Mouthuy PA, Mulvey J, Münzel T, Muzykantov V, Nguyen ITN, Oelze M, Oliveira NG, Palmeira CM, Papaevgeniou N, Pavićević A, Pedre B, Peyrot F, Phylactides M, Pircalabioru GG, Pitt AR, Poulsen HE, Prieto I, Rigobello MP, Robledinos-Antón N, Rodríguez-Mañas L, Rolo AP, Rousset F, Ruskovska T, Saraiva N, Sasson S, Schröder K, Semen K, Seredenina T, Shakirzyanova A, Smith GL, Soldati T, Sousa BC, Spickett CM, Stancic A, Stasia MJ, Steinbrenner H, Stepanić V, Steven S, Tokatlidis K, Tuncay E, Turan B, Ursini F, Vacek J, Vajnerova O, Valentová K, Van Breusegem F, Varisli L, Veal EA, Yalçın AS, Yelisyeyeva O, Žarković N, Zatloukalová M, Zielonka J, Touyz RM, Papapetropoulos A, Grune T, Lamas S, Schmidt HHHW, Di Lisa F, Daiber A. European contribution to the study of ROS: A summary of the findings and prospects for the future from the COST action BM1203 (EU-ROS). Redox Biol 2017; 13:94-162. [PMID: 28577489 PMCID: PMC5458069 DOI: 10.1016/j.redox.2017.05.007] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/12/2022] Open
Abstract
The European Cooperation in Science and Technology (COST) provides an ideal framework to establish multi-disciplinary research networks. COST Action BM1203 (EU-ROS) represents a consortium of researchers from different disciplines who are dedicated to providing new insights and tools for better understanding redox biology and medicine and, in the long run, to finding new therapeutic strategies to target dysregulated redox processes in various diseases. This report highlights the major achievements of EU-ROS as well as research updates and new perspectives arising from its members. The EU-ROS consortium comprised more than 140 active members who worked together for four years on the topics briefly described below. The formation of reactive oxygen and nitrogen species (RONS) is an established hallmark of our aerobic environment and metabolism but RONS also act as messengers via redox regulation of essential cellular processes. The fact that many diseases have been found to be associated with oxidative stress established the theory of oxidative stress as a trigger of diseases that can be corrected by antioxidant therapy. However, while experimental studies support this thesis, clinical studies still generate controversial results, due to complex pathophysiology of oxidative stress in humans. For future improvement of antioxidant therapy and better understanding of redox-associated disease progression detailed knowledge on the sources and targets of RONS formation and discrimination of their detrimental or beneficial roles is required. In order to advance this important area of biology and medicine, highly synergistic approaches combining a variety of diverse and contrasting disciplines are needed.
Collapse
Affiliation(s)
- Javier Egea
- Institute Teofilo Hernando, Department of Pharmacology, School of Medicine. Univerisdad Autonoma de Madrid, Spain
| | - Isabel Fabregat
- Bellvitge Biomedical Research Institute (IDIBELL) and University of Barcelona (UB), L'Hospitalet, Barcelona, Spain
| | - Yves M Frapart
- LCBPT, UMR 8601 CNRS - Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | | | - Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Kateryna Kubaichuk
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Ulla G Knaus
- Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| | - Manuela G Lopez
- Institute Teofilo Hernando, Department of Pharmacology, School of Medicine. Univerisdad Autonoma de Madrid, Spain
| | | | - Andreas Petry
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Rainer Schulz
- Institute of Physiology, JLU Giessen, Giessen, Germany
| | - Jose Vina
- Department of Physiology, University of Valencia, Spain
| | - Paul Winyard
- University of Exeter Medical School, St Luke's Campus, Exeter EX1 2LU, UK
| | - Kahina Abbas
- LCBPT, UMR 8601 CNRS - Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Opeyemi S Ademowo
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Catarina B Afonso
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B47ET, UK
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Haike Antelmann
- Institute for Biology-Microbiology, Freie Universität Berlin, Berlin, Germany
| | - Fernando Antunes
- Departamento de Química e Bioquímica and Centro de Química e Bioquímica, Faculdade de Ciências, Portugal
| | - Mutay Aslan
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Markus M Bachschmid
- Vascular Biology Section & Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Rui M Barbosa
- Center for Neurosciences and Cell Biology, University of Coimbra and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Vsevolod Belousov
- Molecular technologies laboratory, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - David Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota - Twin Cities, USA
| | - Esther Bertrán
- Bellvitge Biomedical Research Institute (IDIBELL) and University of Barcelona (UB), L'Hospitalet, Barcelona, Spain
| | | | - Serge P Bottari
- GETI, Institute for Advanced Biosciences, INSERM U1029, CNRS UMR 5309, Grenoble-Alpes University and Radio-analysis Laboratory, CHU de Grenoble, Grenoble, France
| | - Paula M Brito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal; Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Guia Carrara
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ana I Casas
- Department of Pharmacology & Personalized Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Afroditi Chatzi
- Institute of Molecular Cell and Systems Biology, College of Medical Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow, UK
| | - Niki Chondrogianni
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, 48 Vas. Constantinou Ave., 116 35 Athens, Greece
| | - Marcus Conrad
- Helmholtz Center Munich, Institute of Developmental Genetics, Neuherberg, Germany
| | - Marcus S Cooke
- Oxidative Stress Group, Dept. Environmental & Occupational Health, Florida International University, Miami, FL 33199, USA
| | - João G Costa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal; CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Lisboa, Portugal
| | - Antonio Cuadrado
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid. Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Pham My-Chan Dang
- Université Paris Diderot, Sorbonne Paris Cité, INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, Faculté de Médecine Xavier Bichat, Paris, France
| | - Barbara De Smet
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium; Structural Biology Research Center, VIB, 1050 Brussels, Belgium; Department of Biomedical Sciences and CNR Institute of Neuroscience, University of Padova, Padova, Italy; Pharmahungary Group, Szeged, Hungary
| | - Bilge Debelec-Butuner
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Bornova, Izmir 35100, Turkey
| | - Irundika H K Dias
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Joe Dan Dunn
- Department of Biochemistry, Science II, University of Geneva, 30 quai Ernest-Ansermet, 1211 Geneva-4, Switzerland
| | - Amanda J Edson
- Department of Molecular Biology, University of Bergen, Bergen, Norway
| | - Mariam El Assar
- Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Getafe, Spain
| | - Jamel El-Benna
- Université Paris Diderot, Sorbonne Paris Cité, INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, Faculté de Médecine Xavier Bichat, Paris, France
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Medical Faculty, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Ana S Fernandes
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Lisboa, Portugal
| | - Kari E Fladmark
- Department of Molecular Biology, University of Bergen, Bergen, Norway
| | - Ulrich Förstermann
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Medical Faculty, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Medical Faculty, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Helen Griffiths
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK; Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Vaclav Hampl
- Department of Physiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alina Hanf
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Jan Herget
- Department of Physiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pablo Hernansanz-Agustín
- Servicio de Immunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas Alberto Sols, Madrid, Spain
| | - Melanie Hillion
- Institute for Biology-Microbiology, Freie Universität Berlin, Berlin, Germany
| | - Jingjing Huang
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium; Structural Biology Research Center, VIB, 1050 Brussels, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium; Brussels Center for Redox Biology, Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Serap Ilikay
- Harran University, Arts and Science Faculty, Department of Biology, Cancer Biology Lab, Osmanbey Campus, Sanliurfa, Turkey
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Vincent Jaquet
- Dept. of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - Jaap A Joles
- Department of Nephrology & Hypertension, University Medical Center Utrecht, The Netherlands
| | | | | | - Mahsa Karbaschi
- Oxidative Stress Group, Dept. Environmental & Occupational Health, Florida International University, Miami, FL 33199, USA
| | - Marina Kleanthous
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Lars-Oliver Klotz
- Institute of Nutrition, Department of Nutrigenomics, Friedrich Schiller University, Jena, Germany
| | - Bato Korac
- University of Belgrade, Institute for Biological Research "Sinisa Stankovic" and Faculty of Biology, Belgrade, Serbia
| | - Kemal Sami Korkmaz
- Department of Bioengineering, Cancer Biology Laboratory, Faculty of Engineering, Ege University, Bornova, 35100 Izmir, Turkey
| | - Rafal Koziel
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Damir Kračun
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Karl-Heinz Krause
- Dept. of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - Vladimír Křen
- Institute of Microbiology, Laboratory of Biotransformation, Czech Academy of Sciences, Videnska 1083, CZ-142 20 Prague, Czech Republic
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, UK
| | - João Laranjinha
- Center for Neurosciences and Cell Biology, University of Coimbra and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Antonio Martínez-Ruiz
- Servicio de Immunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Reiko Matsui
- Vascular Biology Section & Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Gethin J McBean
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Stuart P Meredith
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B47ET, UK
| | - Joris Messens
- Structural Biology Research Center, VIB, 1050 Brussels, Belgium; Brussels Center for Redox Biology, Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Verónica Miguel
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Yuliya Mikhed
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Irina Milisav
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology and Faculty of Health Sciences, Ljubljana, Slovenia
| | - Lidija Milković
- Ruđer Bošković Institute, Division of Molecular Medicine, Zagreb, Croatia
| | - Antonio Miranda-Vizuete
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Miloš Mojović
- University of Belgrade, Faculty of Physical Chemistry, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - María Monsalve
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Pierre-Alexis Mouthuy
- Laboratory for Oxidative Stress, Rudjer Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - John Mulvey
- Department of Medicine, University of Cambridge, UK
| | - Thomas Münzel
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Vladimir Muzykantov
- Department of Pharmacology, Center for Targeted Therapeutics & Translational Nanomedicine, ITMAT/CTSA Translational Research Center University of Pennsylvania The Perelman School of Medicine, Philadelphia, PA, USA
| | - Isabel T N Nguyen
- Department of Nephrology & Hypertension, University Medical Center Utrecht, The Netherlands
| | - Matthias Oelze
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Nuno G Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos M Palmeira
- Center for Neurosciences & Cell Biology of the University of Coimbra, Coimbra, Portugal; Department of Life Sciences of the Faculty of Sciences & Technology of the University of Coimbra, Coimbra, Portugal
| | - Nikoletta Papaevgeniou
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, 48 Vas. Constantinou Ave., 116 35 Athens, Greece
| | - Aleksandra Pavićević
- University of Belgrade, Faculty of Physical Chemistry, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - Brandán Pedre
- Structural Biology Research Center, VIB, 1050 Brussels, Belgium; Brussels Center for Redox Biology, Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Fabienne Peyrot
- LCBPT, UMR 8601 CNRS - Paris Descartes University, Sorbonne Paris Cité, Paris, France; ESPE of Paris, Paris Sorbonne University, Paris, France
| | - Marios Phylactides
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | | | - Andrew R Pitt
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B47ET, UK
| | - Henrik E Poulsen
- Laboratory of Clinical Pharmacology, Rigshospitalet, University Hospital Copenhagen, Denmark; Department of Clinical Pharmacology, Bispebjerg Frederiksberg Hospital, University Hospital Copenhagen, Denmark; Department Q7642, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ignacio Prieto
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Maria Pia Rigobello
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Natalia Robledinos-Antón
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid. Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Leocadio Rodríguez-Mañas
- Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Getafe, Spain; Servicio de Geriatría, Hospital Universitario de Getafe, Getafe, Spain
| | - Anabela P Rolo
- Center for Neurosciences & Cell Biology of the University of Coimbra, Coimbra, Portugal; Department of Life Sciences of the Faculty of Sciences & Technology of the University of Coimbra, Coimbra, Portugal
| | - Francis Rousset
- Dept. of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - Tatjana Ruskovska
- Faculty of Medical Sciences, Goce Delcev University, Stip, Republic of Macedonia
| | - Nuno Saraiva
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Lisboa, Portugal
| | - Shlomo Sasson
- Institute for Drug Research, Section of Pharmacology, Diabetes Research Unit, The Hebrew University Faculty of Medicine, Jerusalem, Israel
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany; DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Mainz, Germany
| | - Khrystyna Semen
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Tamara Seredenina
- Dept. of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - Anastasia Shakirzyanova
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - Thierry Soldati
- Department of Biochemistry, Science II, University of Geneva, 30 quai Ernest-Ansermet, 1211 Geneva-4, Switzerland
| | - Bebiana C Sousa
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B47ET, UK
| | - Corinne M Spickett
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Ana Stancic
- University of Belgrade, Institute for Biological Research "Sinisa Stankovic" and Faculty of Biology, Belgrade, Serbia
| | - Marie José Stasia
- Université Grenoble Alpes, CNRS, Grenoble INP, CHU Grenoble Alpes, TIMC-IMAG, F38000 Grenoble, France; CDiReC, Pôle Biologie, CHU de Grenoble, Grenoble, F-38043, France
| | - Holger Steinbrenner
- Institute of Nutrition, Department of Nutrigenomics, Friedrich Schiller University, Jena, Germany
| | - Višnja Stepanić
- Ruđer Bošković Institute, Division of Molecular Medicine, Zagreb, Croatia
| | - Sebastian Steven
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Kostas Tokatlidis
- Institute of Molecular Cell and Systems Biology, College of Medical Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow, UK
| | - Erkan Tuncay
- Department of Biophysics, Ankara University, Faculty of Medicine, 06100 Ankara, Turkey
| | - Belma Turan
- Department of Biophysics, Ankara University, Faculty of Medicine, 06100 Ankara, Turkey
| | - Fulvio Ursini
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Jan Vacek
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, Hnevotinska 3, Olomouc 77515, Czech Republic
| | - Olga Vajnerova
- Department of Physiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kateřina Valentová
- Institute of Microbiology, Laboratory of Biotransformation, Czech Academy of Sciences, Videnska 1083, CZ-142 20 Prague, Czech Republic
| | - Frank Van Breusegem
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Lokman Varisli
- Harran University, Arts and Science Faculty, Department of Biology, Cancer Biology Lab, Osmanbey Campus, Sanliurfa, Turkey
| | - Elizabeth A Veal
- Institute for Cell and Molecular Biosciences, and Institute for Ageing, Newcastle University, Framlington Place, Newcastle upon Tyne, UK
| | - A Suha Yalçın
- Department of Biochemistry, School of Medicine, Marmara University, İstanbul, Turkey
| | | | - Neven Žarković
- Laboratory for Oxidative Stress, Rudjer Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - Martina Zatloukalová
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, Hnevotinska 3, Olomouc 77515, Czech Republic
| | | | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Andreas Papapetropoulos
- Laboratoty of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Tilman Grune
- German Institute of Human Nutrition, Department of Toxicology, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Santiago Lamas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Harald H H W Schmidt
- Department of Pharmacology & Personalized Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Fabio Di Lisa
- Department of Biomedical Sciences and CNR Institute of Neuroscience, University of Padova, Padova, Italy.
| | - Andreas Daiber
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany; DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Mainz, Germany.
| |
Collapse
|
39
|
Domazetovic V, Fontani F, Marcucci G, Iantomasi T, Brandi ML, Vincenzini MT. Estrogen inhibits starvation-induced apoptosis in osteocytes by a redox-independent process involving association of JNK and glutathione S-transferase P1-1. FEBS Open Bio 2017; 7:705-718. [PMID: 28469982 PMCID: PMC5407897 DOI: 10.1002/2211-5463.12216] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 12/25/2022] Open
Abstract
Estrogen deficiency causes bone loss as a result of microdamage, oxidative stress, and osteocyte apoptosis. A relationship between oxidative stress‐induced apoptosis, c‐Jun N‐terminal kinase (JNK) activation, and expression of factors involved in bone remodeling has been demonstrated in osteocytes. However, the molecular regulation of these events in osteocytes treated with 17β‐estradiol (17β‐E2) remains unexplored. The MLO‐Y4 murine osteocyte‐like cell line was used as a model to study starvation‐induced apoptosis and ROS production during 17β‐E2 treatment. Expression of glutathione S‐transferase P1‐1 (GSTP1‐1), receptor activator kB ligand (RANKL), osteoprotegerin (OPG), sclerostin, and kinases activation were measured by western blot. In addition, the GSTP1‐1/JNK association was assessed by immunoprecipitation, and GSTP1‐1 involvement in the osteocyte response to 17β‐E2 was detected by specific siRNA transfection. 17β‐E2 prevents starvation‐induced apoptosis (DNA fragmentation and caspase activation), the increase in sclerostin expression and the RANKL/OPG ratio, which are all related to JNK activation due to oxidative stress in osteocytes. This occurs through GSTP1‐1 overexpression, which can inhibit JNK activation by formation of a GSTP1‐1/JNK complex. No early antioxidant action of 17β‐E2 has been found but the estrogen effect is similar to N‐acetylcysteine which, by increasing the intracellular redox state, maintains JNK bound to GSTP1‐1. Thus, the antiapoptotic and osteogenic effect of 17β‐E2 in MLO‐Y4 occurs by a redox‐independent process involving GSTP1‐1/JNK association. This study clarifies at molecular level the effect of 17β‐E2 on osteocyte activity and identifies a possible role of GSTP1‐1 and JNK activity in bone remodeling and repair mechanisms.
Collapse
Affiliation(s)
- Vladana Domazetovic
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (Biochemistry section) University of Florence Italy
| | - Filippo Fontani
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (Biochemistry section) University of Florence Italy
| | - Gemma Marcucci
- Department of Surgery and Translational Medicine (Endocrinology Section) University of Florence Italy
| | - Teresa Iantomasi
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (Biochemistry section) University of Florence Italy
| | - Maria Luisa Brandi
- Department of Surgery and Translational Medicine (Endocrinology Section) University of Florence Italy
| | - Maria Teresa Vincenzini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (Biochemistry section) University of Florence Italy
| |
Collapse
|
40
|
Extract from Periostracum cicadae Inhibits Oxidative Stress and Inflammation Induced by Ultraviolet B Irradiation on HaCaT Keratinocytes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:8325049. [PMID: 28465707 PMCID: PMC5390570 DOI: 10.1155/2017/8325049] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/21/2017] [Accepted: 03/06/2017] [Indexed: 11/18/2022]
Abstract
Periostracum cicadae is widely used for the treatment of skin diseases such as eczema, pruritus, and itching. The current study sought to evaluate the effect of P. cicadae extract on ultraviolet B (UVB) irradiation and identify the mechanisms involved. Photodamage-protective activity of P. cicadae extracts against oxidative challenge was screened using HaCaT keratinocytes. P. cicadae extracts did not affect cell viability but decreased reactive oxygen species (ROS) production. The extract attenuates the expression of interleukin-6 (IL-6), matrix metalloproteinase-2 (MMP-2), and MMP-9 in UVB-treated HaCaT cells. Also, P. cicadae abrogated UVB-induced activation of NF-κB, p53, and activator protein-1 (AP-1). The downmodulation of IL-6 by P. cicadae was inhibited by the p38 inhibitor (SB203580) or JNK inhibitor (SP600125). Moreover, the extract attenuated the expression of NF-κB and induced thrombomodulin in keratinocytes and thereby effectively downregulated inflammatory responses in the skin. The nuclear accumulation and expression of NF-E2-related factor (Nrf2) were increased by P. cicadae treatment. Furthermore, treatment with P. cicadae remarkably ameliorated the skin's structural damage induced by irradiation. This study demonstrates that P. cicadae may protect skin cells against oxidative insult by modulating ROS concentration, IL-6, MMPs generation, antioxidant enzymes activity, and cell signaling pathways.
Collapse
|
41
|
Piroddi M, Albini A, Fabiani R, Giovannelli L, Luceri C, Natella F, Rosignoli P, Rossi T, Taticchi A, Servili M, Galli F. Nutrigenomics of extra-virgin olive oil: A review. Biofactors 2017; 43:17-41. [PMID: 27580701 DOI: 10.1002/biof.1318] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 07/08/2016] [Accepted: 07/08/2016] [Indexed: 12/11/2022]
Abstract
Nutrigenomics data on the functional components of olive oil are still sparse, but rapidly increasing. Olive oil is the main source of fat and health-promoting component of the Mediterranean diet. Positive effects have been observed on genes involved in the pathobiology of most prevalent age- and lifestyle-related human conditions, such as cancer, cardiovascular disease and neurodegeneration. Other effects on health-promoting genes have been identified for bioactive components of olives and olive leafs. Omics technologies are offering unique opportunities to identify nutritional and health biomarkers associated with these gene responses, the use of which in personalized and even predictive protocols of investigation, is a main breakthrough in modern medicine and nutrition. Gene regulation properties of the functional components of olive oil, such as oleic acid, biophenols and vitamin E, point to a role for these molecules as natural homeostatic and even hormetic factors with applications as prevention agents in conditions of premature and pathologic aging. Therapeutic applications can be foreseen in conditions of chronic inflammation, and particularly in cancer, which will be discussed in detail in this review paper as major clinical target of nutritional interventions with olive oil and its functional components. © 2016 BioFactors, 43(1):17-41, 2017.
Collapse
Affiliation(s)
- Marta Piroddi
- Department of Pharmaceutical Sciences, Nutrition and Clinical Biochemistry Lab, University of Perugia, Italy
| | - Adriana Albini
- IRCCS MultiMedica, Scientific and Technology Pole, Milan, Italy
| | - Roberto Fabiani
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Italy
| | - Lisa Giovannelli
- NEUROFARBA - Section of Phamacology and Toxicology, University of Firenze, Italy
| | - Cristina Luceri
- NEUROFARBA - Section of Phamacology and Toxicology, University of Firenze, Italy
| | - Fausta Natella
- CREA-NUT, Consiglio per La Ricerca in Agricoltura E L'Analisi Dell'Economia Agraria, Food and Nutrition Research Centre, via Ardeatina 546, 00178, Roma, Italy
| | - Patrizia Rosignoli
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Italy
| | - Teresa Rossi
- Research and Statistics, Department, IRCCS "Tecnologie Avanzate E Modelli Assistenziali in Oncologia", Laboratory of Translational Research, Arcispedale S. Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - Agnese Taticchi
- Department of Agricultural Food and Environmental Sciences, University of Perugia, Italy
| | - Maurizio Servili
- Department of Agricultural Food and Environmental Sciences, University of Perugia, Italy
| | - Francesco Galli
- Department of Pharmaceutical Sciences, Nutrition and Clinical Biochemistry Lab, University of Perugia, Italy
| |
Collapse
|
42
|
Bartolini D, Sancineto L, Fabro de Bem A, Tew KD, Santi C, Radi R, Toquato P, Galli F. Selenocompounds in Cancer Therapy: An Overview. Adv Cancer Res 2017; 136:259-302. [PMID: 29054421 DOI: 10.1016/bs.acr.2017.07.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
In vitro and in vivo experimental models clearly demonstrate the efficacy of Se compounds as anticancer agents, contingent upon chemical structures and concentrations of test molecules, as well as on the experimental model under investigation that together influence cellular availability of compounds, their molecular dynamics and mechanism of action. The latter includes direct and indirect redox effects on cellular targets by the activation and altered compartmentalization of molecular oxygen, and the interaction with protein thiols and Se proteins. As such, Se compounds interfere with the redox homeostasis and signaling of cancer cells to produce anticancer effects that include alterations in key regulatory elements of energy metabolism and cell cycle checkpoints that ultimately influence differentiation, proliferation, senescence, and death pathways. Cys-containing proteins and Se proteins involved in the response to Se compounds as sensors and transducers of anticancer signals, i.e., the pharmacoproteome of Se compounds, are described and include critical elements in the different phases of cancer onset and progression from initiation and escape of immune surveillance to tumor growth, angiogenesis, and metastasis. The efficacy and mode of action on these compounds vary depending on the inorganic and organic form of Se used as either supplement or pharmacological agent. In this regard, differences in experimental/clinical protocols provide options for either chemoprevention or therapy in different human cancers.
Collapse
Affiliation(s)
| | | | - Andreza Fabro de Bem
- Center of Biological Sciences (CCB), Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil; Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Kenneth D Tew
- Medical University of South Carolina, Charleston, SC, United States
| | | | - Rafael Radi
- Center for Free Radical and Biomedical Research (CEINBIO), Universidad de la República, Montevideo, Uruguay
| | | | | |
Collapse
|
43
|
Subramanyam M, Goyal J. Translational biomarkers: from discovery and development to clinical practice. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 21-22:3-10. [PMID: 27978985 DOI: 10.1016/j.ddtec.2016.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/12/2016] [Accepted: 10/31/2016] [Indexed: 06/06/2023]
Abstract
The refinement of disease taxonomy utilizing molecular phenotypes has led to significant improvements in the precision of disease diagnosis and customization of treatment options. This has also spurred efforts to identify novel biomarkers to understand the impact of therapeutically altering the underlying molecular network on disease course, and to support decision-making in drug discovery and development. However, gaps in knowledge regarding disease heterogeneity, combined with the inadequacies of surrogate disease model systems, make it challenging to demonstrate the unequivocal association of molecular and physiological biomarkers to disease pathology. This article will discuss the current landscape in biomarker research and highlight strategies being adopted to increase the likelihood of transitioning biomarkers from discovery to medical practice to enable more objective decision making, and to improve health outcome.
Collapse
Affiliation(s)
- Meena Subramanyam
- Global Biomarker Discovery and Development, Biogen, 300 Binney St., Cambridge, MA 02142, United States.
| | - Jaya Goyal
- Value Based Medicine, Biogen, 300 Binney St., Cambridge, MA 02142, United States
| |
Collapse
|