151
|
Shamoon L, Romero A, De la Cuesta F, Sánchez-Ferrer CF, Peiró C. Angiotensin-(1-7), a protective peptide against vascular aging. Peptides 2022; 152:170775. [PMID: 35231551 DOI: 10.1016/j.peptides.2022.170775] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/09/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022]
Abstract
Vascular aging is a complex and multifaceted process that provokes profound molecular, structural, and functional changes in the vasculature. Eventually, these profound aging alterations make arteries more prone to vascular disease, including hypertension, atherosclerosis and other arterial complications that impact the organism beyond the cardiovascular system and accelerate frailty. For these reasons, preventing or delaying the hallmarks of vascular aging is nowadays a major health goal, especially in our aged societies. In this context, angiotensin(Ang)-(1-7), a major player of the protective branch of the renin-angiotensin system, has gained relevance over recent years as growing knowledge on its anti-aging properties is being unveiled. Here, we briefly review the main actions of Ang-(1-7) against vascular aging. These include protection against vascular cell senescence, anti-inflammatory and antioxidant effects together with the induction of cytoprotective systems. Ang-(1-7) further ameliorates endothelial dysfunction, a hallmark of vascular aging and disease, attenuates fibrosis and calcification and promotes protective angiogenesis and repair. Although further research is needed to better understand the anti-aging properties of Ang-(1-7) on the vasculature, this heptapeptide arises as a promising pharmacological tool for preventing vascular aging and frailty.
Collapse
Affiliation(s)
- L Shamoon
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Paz, IdIPAZ, Madrid, Spain
| | - A Romero
- German Center for the Study of Diabetes, Düsseldorf, Germany
| | - F De la Cuesta
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Spain.
| | - C F Sánchez-Ferrer
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Paz, IdIPAZ, Madrid, Spain.
| | - C Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Paz, IdIPAZ, Madrid, Spain.
| |
Collapse
|
152
|
Tian JJ, Levy M, Zhang X, Sinnott R, Maddela R. Counteracting Health Risks by Modulating Homeostatic Signaling. Pharmacol Res 2022; 182:106281. [PMID: 35661711 DOI: 10.1016/j.phrs.2022.106281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/14/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
Abstract
Homeostasis was initially conceptualized by Bernard and Cannon around a century ago as a steady state of physiological parameters that vary within a certain range, such as blood pH, body temperature, and heart rate1,2. The underlying mechanisms that maintain homeostasis are explained by negative feedbacks that are executed by the neuronal, endocrine, and immune systems. At the cellular level, homeostasis, such as that of redox and energy steady state, also exists and is regulated by various cell signaling pathways. The induction of homeostatic mechanism is critical for human to adapt to various disruptive insults (stressors); while on the other hand, adaptation occurs at the expense of other physiological processes and thus runs the risk of collateral damages, particularly under conditions of chronic stress. Conceivably, anti-stress protection can be achieved by stressor-mimicking medicinals that elicit adaptive responses prior to an insult and thereby serve as health risk countermeasures; and in situations where maladaptation may occur, downregulating medicinals could be used to suppress the responses and prevent subsequent pathogenesis. Both strategies are preemptive interventions particularly suited for individuals who carry certain lifestyle, environmental, or genetic risk factors. In this article, we will define and characterize a new modality of prophylactic intervention that forestalls diseases via modulating homeostatic signaling. Moreover, we will provide evidence from the literature that support this concept and distinguish it from other homeostasis-related interventions such as adaptogen, hormesis, and xenohormesis.
Collapse
Affiliation(s)
- Junqiang J Tian
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA.
| | - Mark Levy
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA
| | - Xuekai Zhang
- Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing100029, China; US Center for Chinese Medicine, 14801 Physicians lane, 171 A 2nd Floor, #281, Rockville MD 20850, USA
| | - Robert Sinnott
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA
| | - Rolando Maddela
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA
| |
Collapse
|
153
|
5-Methyltetrahydrofolate Attenuates Oxidative Stress and Improves Kidney Function in Acute Kidney Injury through Activation of Nrf2 and Antioxidant Defense. Antioxidants (Basel) 2022; 11:antiox11061046. [PMID: 35739943 PMCID: PMC9219715 DOI: 10.3390/antiox11061046] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/10/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023] Open
Abstract
Oxidative stress is a major mediator of adverse outcomes in acute kidney injury (AKI). Deficiency of micronutrients, such as folate, is common in AKI. Our previous study reported that AKI impaired kidney reabsorption of folate and decreased plasma folate level in rats. The present study investigated the effect of 5-methyltetrahydrofolate (5-MTHF), a biologically active form of folate/folic acid, on AKI-impaired kidney function and oxidative stress. Sprague-Dawley rats developed AKI after kidney ischemia (45 min) and reperfusion (24 h). Injection of 5-MTHF (3 µg/kg body weight) improved kidney function and attenuated oxidative stress with a restoration of glutathione and a reduction of lipid peroxidation in the kidney. Injection of 5-MTHF activated transcription factor Nrf2 and increased the expression of glutathione synthesizing enzymes, superoxide dismutase-1 and heme oxygenase-1 in the kidney. Simulated ischemia-reperfusion through hypoxia-reoxygenation increased oxidative stress in proximal tubular cells. Incubation of cells with 5-MTHF alleviated cell injury and increased antioxidant enzyme expression and intracellular glutathione levels. Inhibition of Nrf2 expression through siRNA transfection abolished the effect of 5-MTHF against oxidative stress. These results suggest that low-dose folic acid can improve kidney function through activation of Nrf2 and restoration of antioxidant defence. Micronutrient supplements may improve clinical outcomes in AKI.
Collapse
|
154
|
Zhou Q, Zhang N, Hu T, Xu H, Duan X, Liu B, Chen F, Wang M. Dietary phenolic-type Nrf2-activators: implications in the control of toxin-induced hepatic disorders. Food Funct 2022; 13:5480-5497. [PMID: 35411358 DOI: 10.1039/d1fo04237h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Numerous studies have exemplified the importance of nuclear factor erythroid 2-related factor 2 (Nrf2) activation in the alleviation of toxin-induced hepatic disorders primarily through eliminating oxidative stress. Whereafter, increasingly more efforts have been contributed to finding Nrf2-activators, especially from dietary polyphenols. The present review summarized the phenolic-type Nrf2-activators published in the past few decades, analyzed their effectiveness based on their structural characteristics and outlined their related mechanisms. It turns out that flavonoids are the largest group of phenolic-type Nrf2-activators, followed by nonflavonoids and phenolic acids. When counting on subgroups, the top three types are flavonols, flavones, and hydroxycinnamic acids, with curcuminoids having the highest effective doses. Moreover, most polyphenols work through the phosphorylation of Nrf2. Besides, mitogen-activated protein kinases (MAPKs) and protein kinase B (Akt) are the frequent targets of these Nrf2-activators, which indirectly mediate the behavior of Nrf2. However, current data are not sufficient to conclude any structure-activity relationship.
Collapse
Affiliation(s)
- Qian Zhou
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| | - Nana Zhang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Tingyan Hu
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| | - Hui Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| | - Xinxing Duan
- Schlegel Research Institute for Aging & Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada
| | - Bin Liu
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| | - Feng Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| | - Mingfu Wang
- Institute for Advanced Study, Shenzhen University, Shenzhen, China. .,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China.
| |
Collapse
|
155
|
Liu X, Dilxat T, Shi Q, Qiu T, Lin J. The combination of nicotinamide mononucleotide and lycopene prevents cognitive impairment and attenuates oxidative damage in D-galactose induced aging models via Keap1-Nrf2 signaling. Gene X 2022; 822:146348. [PMID: 35183682 DOI: 10.1016/j.gene.2022.146348] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/03/2022] [Accepted: 02/15/2022] [Indexed: 12/30/2022] Open
Abstract
Aging is referred to progressive dysfunction of body organs, including the brain. This study aims to explore the anti-aging effect of combing nicotinamide mononucleotide (NMN) and lycopene (Lyco) (NMN + Lyco) on aging rats and senescent PC12 cells. Both in vivo and in vitro aging models were established using D-galactose (D-gal). The combination showed a trend to superiority over monotherapy in preventing aging in vivo and in vitro. Morris water maze test showed that NMN + Lyco effectively improved the ability of spatial location learning and memory of aging model rats. NMN + Lyco mitigated the oxidative stress of rat brains, livers, and PC12 cells by elevating the levels of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), GSH, as well as total antioxidant capacity (T-AOC), and reducing malondialdehyde (MDA) content. CCK-8 assay, senescence-associated β-galactosidase staining, and flow cytometer confirmed the cellular senescence of PC12 cells after exposing D-gal, and indicated the anti-senescence effect of NMN + Lyco in vitro. Moreover, NMN + Lyco effectively down-regulated the expressions of p53, p21, and p16 (senescence-related genes), and activated Keap1-Nrf2 signaling in both in vivo and in vitro aging models. In total, NMN + Lyco protected rats and PC12 cells from cognitive impairment and cellular senescence induced by D-gal, of which effects might be linked to the reduction of oxidative stress and the activation of Keap1-Nrf2 signaling.
Collapse
Affiliation(s)
- Xuxin Liu
- Xinjiang Agricultural Vocational Technical College, Changji, Xinjiang, China.
| | - Tursunay Dilxat
- Xinjiang Agricultural Vocational Technical College, Changji, Xinjiang, China.
| | - Qiang Shi
- Xinjiang Agricultural Vocational Technical College, Changji, Xinjiang, China.
| | - Taoyu Qiu
- Xinjiang Agricultural Vocational Technical College, Changji, Xinjiang, China.
| | - Junping Lin
- Xinjiang Changji National High-Tech Industrial Development Zone, Changji, Xinjiang, China.
| |
Collapse
|
156
|
Protective Effects against the Development of Alzheimer’s Disease in an Animal Model through Active Immunization with Methionine-Sulfoxide Rich Protein Antigen. Antioxidants (Basel) 2022; 11:antiox11040775. [PMID: 35453459 PMCID: PMC9029927 DOI: 10.3390/antiox11040775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/09/2022] [Accepted: 04/10/2022] [Indexed: 02/04/2023] Open
Abstract
The brain during Alzheimer’s disease (AD) is under severe oxidative attack by reactive oxygen species that may lead to methionine oxidation. Oxidation of the sole methionine (Met35) of beta-amyloid (Aβ), and possibly methionine residues of other extracellular proteins, may be one of the earliest events contributing to the toxicity of Aβ and other proteins in vivo. In the current study, we immunized transgenic AD (APP/PS1) mice at 4 months of age with a recombinant methionine sulfoxide (MetO)-rich protein from Zea mays (antigen). This treatment induced the production of anti-MetO antibody in blood-plasma that exhibits a significant titer up to at least 10 months of age. Compared to the control mice, the antigen-injected mice exhibited the following significant phenotypes at 10 months of age: better short and long memory capabilities; reduced Aβ levels in both blood-plasma and brain; reduced Aβ burden and MetO accumulations in astrocytes in hippocampal and cortical regions; reduced levels of activated microglia; and elevated antioxidant capabilities (through enhanced nuclear localization of the transcription factor Nrf2) in the same brain regions. These data collected in a preclinical AD model are likely translational, showing that active immunization could give a possibility of delaying or preventing AD onset. This study represents a first step toward the complex way of starting clinical trials in humans and conducting the further confirmations that are needed to go in this direction.
Collapse
|
157
|
Chen J, Chen Y, Zheng Y, Zhao J, Yu H, Zhu J. Relationship between Neuroprotective Effects and Structure of Procyanidins. Molecules 2022; 27:molecules27072308. [PMID: 35408708 PMCID: PMC9000754 DOI: 10.3390/molecules27072308] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 02/04/2023] Open
Abstract
This study evaluated the relationship between the neuroprotective effects of procyanidins and their structural characteristics. In vitro, a rat pheochromocytoma cell line (PC12) was exposed to the grape seed-derived procyanidin monomers: catechin (C), epicatechin (EC), and epicatechin gallate (ECG); the procyanidin dimers: procyanidin B1 (B1), procyanidin B2 (B2), procyanidin B3 (B3), procyanidin B4 (B4), procyanidin B1-3-O-gallate (B1-G), and procyanidin B2-3-O-gallate (B2-G); and the procyanidin trimers: procyanidin C1 (C1) and N-acetyl-l-cysteine (NAC) for 24 h. Cells were then incubated with 200 μM H2O2 for 24 h. In vivo, zebrafish larvae (AB strain) 3 days post-fertilization were incubated with NAC or procyanidins (C, EC, ECG, B1, B2, B3, B4, B1-G, B2-G, C1) in 300 µM H2O2 for 4 days. Different grape seed procyanidins increased the survival of PC12 cells challenged with H2O2, improved the movement behavior disorder of zebrafish caused by H2O2, inhibited the increase of ROS and MDA and the decrease of GSH-Px, CAT, and SOD activities, and up-regulated the Nrf2/ARE pathway. The neuroprotective effects of the procyanidin trimer C1 treatment group were greater than the other treatment groups. These results suggest that the neuroprotective effect of procyanidins is positively correlated with their degree of polymerization.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiajin Zhu
- Correspondence: ; Tel./Fax: +86-571-8898-2191
| |
Collapse
|
158
|
Ordonez AA, Bullen CK, Villabona-Rueda AF, Thompson EA, Turner ML, Merino VF, Yan Y, Kim J, Davis SL, Komm O, Powell JD, D'Alessio FR, Yolken RH, Jain SK, Jones-Brando L. Sulforaphane exhibits antiviral activity against pandemic SARS-CoV-2 and seasonal HCoV-OC43 coronaviruses in vitro and in mice. Commun Biol 2022; 5:242. [PMID: 35304580 PMCID: PMC8933402 DOI: 10.1038/s42003-022-03189-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 02/24/2022] [Indexed: 12/31/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), has incited a global health crisis. Currently, there are limited therapeutic options for the prevention and treatment of SARS-CoV-2 infections. We evaluated the antiviral activity of sulforaphane (SFN), the principal biologically active phytochemical derived from glucoraphanin, the naturally occurring precursor present in high concentrations in cruciferous vegetables. SFN inhibited in vitro replication of six strains of SARS-CoV-2, including Delta and Omicron, as well as that of the seasonal coronavirus HCoV-OC43. Further, SFN and remdesivir interacted synergistically to inhibit coronavirus infection in vitro. Prophylactic administration of SFN to K18-hACE2 mice prior to intranasal SARS-CoV-2 infection significantly decreased the viral load in the lungs and upper respiratory tract and reduced lung injury and pulmonary pathology compared to untreated infected mice. SFN treatment diminished immune cell activation in the lungs, including significantly lower recruitment of myeloid cells and a reduction in T cell activation and cytokine production. Our results suggest that SFN should be explored as a potential agent for the prevention or treatment of coronavirus infections.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - C Korin Bullen
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andres F Villabona-Rueda
- Division of Pulmonology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth A Thompson
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mitchell L Turner
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vanessa F Merino
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Yan
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Kim
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephanie L Davis
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Oliver Komm
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan D Powell
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Franco R D'Alessio
- Division of Pulmonology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanjay K Jain
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lorraine Jones-Brando
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
159
|
Chondronasiou D, Gill D, Mosteiro L, Urdinguio RG, Berenguer‐Llergo A, Aguilera M, Durand S, Aprahamian F, Nirmalathasan N, Abad M, Martin‐Herranz DE, Stephan‐Otto Attolini C, Prats N, Kroemer G, Fraga MF, Reik W, Serrano M. Multi-omic rejuvenation of naturally aged tissues by a single cycle of transient reprogramming. Aging Cell 2022; 21:e13578. [PMID: 35235716 PMCID: PMC8920440 DOI: 10.1111/acel.13578] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 12/01/2022] Open
Abstract
The expression of the pluripotency factors OCT4, SOX2, KLF4, and MYC (OSKM) can convert somatic differentiated cells into pluripotent stem cells in a process known as reprogramming. Notably, partial and reversible reprogramming does not change cell identity but can reverse markers of aging in cells, improve the capacity of aged mice to repair tissue injuries, and extend longevity in progeroid mice. However, little is known about the mechanisms involved. Here, we have studied changes in the DNA methylome, transcriptome, and metabolome in naturally aged mice subject to a single period of transient OSKM expression. We found that this is sufficient to reverse DNA methylation changes that occur upon aging in the pancreas, liver, spleen, and blood. Similarly, we observed reversion of transcriptional changes, especially regarding biological processes known to change during aging. Finally, some serum metabolites and biomarkers altered with aging were also restored to young levels upon transient reprogramming. These observations indicate that a single period of OSKM expression can drive epigenetic, transcriptomic, and metabolomic changes toward a younger configuration in multiple tissues and in the serum.
Collapse
Affiliation(s)
- Dafni Chondronasiou
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Diljeet Gill
- Epigenetics ProgrammeBabraham InstituteCambridgeUK
| | | | - Rocio G. Urdinguio
- Cancer Epigenetics and Nanomedicine LaboratoryNanomaterials and Nanotechnology Research Center (CINN CSIC)OviedoSpain
- Health Research Institute of Asturias (ISPA)OviedoSpain
- Institute of Oncology of Asturias (IUOPA)University of OviedoOviedoSpain
- Department of Organisms and Systems Biology (BOS)University of OviedoOviedoSpain
- CIBER of Rare Diseases (CIBERER)OviedoSpain
| | - Antonio Berenguer‐Llergo
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Mònica Aguilera
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Sylvere Durand
- Metabolomics and Cell Biology PlatformsInstitut Gustave RoussyVillejuifFrance
- Centre de Recherche des CordeliersEquipe Labellisée par la Ligue Contre le CancerUniversité de ParisSorbonne UniversitéParisFrance
- Inserm U1138Institut Universitaire de FranceParisFrance
| | - Fanny Aprahamian
- Metabolomics and Cell Biology PlatformsInstitut Gustave RoussyVillejuifFrance
- Centre de Recherche des CordeliersEquipe Labellisée par la Ligue Contre le CancerUniversité de ParisSorbonne UniversitéParisFrance
- Inserm U1138Institut Universitaire de FranceParisFrance
| | - Nitharsshini Nirmalathasan
- Metabolomics and Cell Biology PlatformsInstitut Gustave RoussyVillejuifFrance
- Centre de Recherche des CordeliersEquipe Labellisée par la Ligue Contre le CancerUniversité de ParisSorbonne UniversitéParisFrance
- Inserm U1138Institut Universitaire de FranceParisFrance
| | - Maria Abad
- Vall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
| | | | - Camille Stephan‐Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Neus Prats
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Guido Kroemer
- Metabolomics and Cell Biology PlatformsInstitut Gustave RoussyVillejuifFrance
- Centre de Recherche des CordeliersEquipe Labellisée par la Ligue Contre le CancerUniversité de ParisSorbonne UniversitéParisFrance
- Inserm U1138Institut Universitaire de FranceParisFrance
- Pôle de BiologieHôpital Européen Georges PompidouAP‐HPParisFrance
- Suzhou Institute for Systems MedicineChinese Academy of Medical SciencesSuzhouChina
- Department of Women's and Children's HealthKarolinska InstituteKarolinska University HospitalStockholmSweden
| | - Mario F. Fraga
- Cancer Epigenetics and Nanomedicine LaboratoryNanomaterials and Nanotechnology Research Center (CINN CSIC)OviedoSpain
- Health Research Institute of Asturias (ISPA)OviedoSpain
- Institute of Oncology of Asturias (IUOPA)University of OviedoOviedoSpain
- Department of Organisms and Systems Biology (BOS)University of OviedoOviedoSpain
- CIBER of Rare Diseases (CIBERER)OviedoSpain
| | - Wolf Reik
- Epigenetics ProgrammeBabraham InstituteCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
- Wellcome Trust Sanger InstituteCambridgeUK
- Altos Labs Cambridge InstituteCambridgeUK
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
| |
Collapse
|
160
|
Arslanbaeva L, Bisaglia M. Activation of the Nrf2 Pathway as a Therapeutic Strategy for ALS Treatment. Molecules 2022; 27:1471. [PMID: 35268572 PMCID: PMC8911691 DOI: 10.3390/molecules27051471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 12/19/2022] Open
Abstract
Amyotrophic lateral sclerosis is a progressive and fatal disease that causes motoneurons degeneration and functional impairment of voluntary muscles, with limited and poorly efficient therapies. Alterations in the Nrf2-ARE pathway are associated with ALS pathology and result in aberrant oxidative stress, making the stimulation of the Nrf2-mediated antioxidant response a promising therapeutic strategy in ALS to reduce oxidative stress. In this review, we first introduce the involvement of the Nrf2 pathway in the pathogenesis of ALS and the role played by astrocytes in modulating such a protective pathway. We then describe the currently developed activators of Nrf2, used in both preclinical animal models and clinical studies, taking into consideration their potentialities as well as the possible limitations associated with their use.
Collapse
Affiliation(s)
| | - Marco Bisaglia
- Department of Biology, University of Padua, 35131 Padua, Italy
- Center Study for Neurodegeneration (CESNE), University of Padua, 35131 Padua, Italy
| |
Collapse
|
161
|
Christopher E, Loan JJM, Samarasekera N, McDade K, Rose J, Barrington J, Hughes J, Smith C, Al-Shahi Salman R. Nrf2 activation in the human brain after stroke due to supratentorial intracerebral haemorrhage: a case–control study. BMJ Neurol Open 2022; 4:e000238. [PMID: 35265844 PMCID: PMC8860052 DOI: 10.1136/bmjno-2021-000238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/17/2022] [Indexed: 01/05/2023] Open
Abstract
Aims Pharmacological activation of the antioxidative transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) improves outcomes in experimental models of intracerebral haemorrhage (ICH). However, the Nrf2 pathway has not been previously studied in humans after ICH. Our study aims to address this gap. Methods We selected cases with fatal ICH from a prospective community-based inception cohort study and age-matched and sex-matched controls who died suddenly of non-neurological disease. We used immunohistochemistry to quantify Nrf2 (% total area stained overall and % of nuclei stained) and CD68 expression in controls and perihaematomal, ipsilateral and contralateral brain tissue from cases. We measured downstream haem oxygenase-1 (HMOX1) and NAD(P)H dehydrogenase quinone 1 [NQO1] expression using RNA in situ hybridisation. Results 26 ICH cases (median age: 82 (IQR 76–86); 13 (50%) male) and eight controls (median age: 79 (IQR 77–80); 3 (37.5%) male) were included. We found no significant differences in overall % of Nrf2 staining between ICH cases and controls. However, the mean % of nuclei staining for Nrf2 seemed higher in perihaematomal compared with contralateral regions, although this was only statistically significant >60 days after ICH (25% (95% CI 17% to 33%) vs 14% (95% CI 11% to 17%), p=0.029). The percentage of perihaematomal tissue staining for CD68 was higher >60 days after ICH (6.75%, 95% CI 2.78% to 10.73%) compared with contralateral tissue (1.45%, 95% CI 0.93% to 1.96%, p=0.027) and controls (1.08%, 95% CI 0.20% to 1.97%, p=0.0008). RNA in situ hybridisation suggested increased abundance of HMOX1 and NQO1 transcripts in perihaematomal versus distant ipsilateral brain tissue obtained <7 days from onset of ICH. Conclusions We found evidence of Nrf2 activation in human brain tissue after ICH. Pharmacological augmentation of Nrf2 activation after ICH might be a promising therapeutic approach.
Collapse
Affiliation(s)
- Edward Christopher
- The University of Edinburgh College of Medicine and Veterinary Medicine, Edinburgh, UK
| | - James J M Loan
- Division of Clinical Neurosciences, NHS Lothian, Edinburgh, UK
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Neshika Samarasekera
- Division of Clinical Neurosciences, NHS Lothian, Edinburgh, UK
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Karina McDade
- Academic Neuropathology, The University of Edinburgh, Edinburgh, UK
| | - Jamie Rose
- Academic Neuropathology, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Jack Barrington
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Jeremy Hughes
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Colin Smith
- Academic Neuropathology, The University of Edinburgh, Edinburgh, UK
| | - Rustam Al-Shahi Salman
- Division of Clinical Neurosciences, NHS Lothian, Edinburgh, UK
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
162
|
Liu C, Li Y, Wang X. TDAG51-Deficiency Podocytes are Protected from High-Glucose-Induced Damage Through Nrf2 Activation via the AKT-GSK-3β Pathway. Inflammation 2022; 45:1520-1533. [PMID: 35175494 DOI: 10.1007/s10753-022-01638-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 11/25/2022]
Abstract
T cell death-associated gene 51 (TDAG51) has been implicated in the development of various pathological conditions. However, whether TDAG51 plays a role in diabetic renal disease remains unknown. The current work investigated the possible function of TDAG51 in diabetic renal disease using high-glucose (HG)-stimulated podocytes in vitro. The elevation of TDAG51 was observed in podocytes in response to HG exposure and the glomeruli of diabetic mice. The siRNAs targeting TDAG51 were applied to deplete TDAG51 in HG-stimulated podocytes. Crucially, TDAG51 deficiency was sufficient to decrease the apoptosis, oxidative stress, and inflammation caused by HG. Mechanically, the inhibition of TDAG51 was capable of enhancing the activation of nuclear factor E2-related factor 2 (Nrf2) associated with the upregulation of AKT-glycogen synthase kinase-3β (GSK-3β) pathway. The reduction of AKT abolished the activation of Nrf2 elicited by TDAG51 deficiency. Additionally, the reduction of Nrf2 diminished the anti-HG injury effect elicited by TDAG51 deficiency. Overall, these data demonstrate that TDAG51 deficiency defends against HG-induced podocyte damage through Nrf2 activation by regulating AKT-GSK-3β pathway. This study suggests that TDAG1 may have a potential role in diabetic renal disease by affecting HG-induced podocyte damage.
Collapse
Affiliation(s)
- Chuntian Liu
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, 710004, Shaanxi, China.
| | - Yanling Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Xiaojuan Wang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an, 710004, Shaanxi, China
| |
Collapse
|
163
|
Zhang M, Yu X, Li D, Ma N, Wei Z, Ci X, Zhang S. Nrf2 Signaling Pathway Mediates the Protective Effects of Daphnetin Against D-Galactose Induced-Premature Ovarian Failure. Front Pharmacol 2022; 13:810524. [PMID: 35153783 PMCID: PMC8832979 DOI: 10.3389/fphar.2022.810524] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/05/2022] [Indexed: 12/20/2022] Open
Abstract
Oxidative damage can lead to severe ovarian dysfunctions and even premature ovarian failure. Nrf2, a significant transcription factor that regulates the oxidative stress response of cells, declines with age. Daphnetin, as a kind of natural Chinese herbal medicine, can activate Nrf2 and further promote the antioxidant defense of cells. However, whether Daphnetin treatment can protect ovary from premature ovarian failure and the specific mechanism involved are not understood. This study aimed to investigate the protective function of Daphnetin against the ovarian aging induced by D-galactose in wild-type and Nrf2−/− mice. Female C57BL/6 mice with Wild-type and Nrf2−/− were divided into five groups separately and the premature ovarian failure model were established by D-galactose and then Daphnetin and VE were given for treatment. After 42 days, ovaries tissue and serum were collected for biochemical determination, H&E staining, Immunohistochemical staining and western blot analysis. In the WT-POF group, ovarian function was broke, and the expression of the ovarian senescence-associated protein P16 and the level of oxidative stress were significantly increased, while the expression of the anti-senescence protein klotho was significantly decreased. In addition, the expression of Nrf2 and the antioxidases GCLC, HO-1 and NQO1 were decreased, but TXNIP and NLRP3 were significantly increased. Furthermore, the characteristics of premature ovarian failure were more significant in Nrf2 knockout mice than in wild-type mice, especially the expression of NLRP3 and TXNIP. Moreover, daphnetin, an Nrf2 activator, rescued d-gal-induced POF in a dose-dependent manner, while the protective effect was weakened or even lost in Nrf2 knockout mice. Our results suggested that daphnetin is likely to be a candidate drug for premature ovarian failure treatment and it is mostly possible referred to the molecular mechanism of increasing Nrf2 expression and inhibiting NLRP3 activation in the ovarian aging process.
Collapse
Affiliation(s)
- Mengwen Zhang
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, China
| | - Xiaowei Yu
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, China
| | - Danjie Li
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, China
| | - Ning Ma
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, China
| | - Zhentong Wei
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Xinxin Ci, ; Songling Zhang,
| | - Songling Zhang
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Xinxin Ci, ; Songling Zhang,
| |
Collapse
|
164
|
Atyah M, Zhou C, Zhou Q, Chen W, Weng J, Wang P, Shi Y, Dong Q, Ren N. The Age-Specific Features and Clinical Significance of NRF2 and MAPK10 Expression in HCC Patients. Int J Gen Med 2022; 15:737-748. [PMID: 35082522 PMCID: PMC8786352 DOI: 10.2147/ijgm.s351263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/12/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Nuclear factor (erythroid-derived 2)-like 2 (NRF2) functions decline with age; however, cancer cells can hijack its pathways to ensure survival and aggressiveness. Yet, the role of NRF2 in hepatocellular carcinoma (HCC) is rarely investigated in an age-specific manner. This study investigates the expression of NRF2 and its activator (MAPK10) in different age groups of HCC patients, in addition to the age-specific features of NRF2 and MAPK10 interaction and their clinical significance. METHODS Tumor and near-tumor tissue samples of 181 HCC patients were used to complete a protein expression analysis of NRF2 and MAPK10. Patients' survival and clinical data were collected for clinical analysis. Global databases (TCGA, ICGC) were used to collect MAPK10 genetic mutation and mRNA expression data in patients with HCC, colorectal, stomach, and pancreatic cancers. RESULTS Our findings revealed an increase in NRF2 protein expression but only in younger HCC patients, along with a decline in MAPK10 ability to activate NRF2 in older patients. We also found an increased MAPK10 genetic mutation rate and decreased mRNA expression in older patients. Low MAPK10 and NRF2 expression levels were associated with shorter survival and poorer prognosis due to positive correlation with microvascular invasion, tumor thrombus, elevated AFP levels, and larger tumor size. CONCLUSION NRF2 expression and oxidative stress mechanism in HCC patients are influenced by age. This magnifies the need to consider patients' age in treatment strategies and guidelines and re-evaluates the application of studies' age-standardized findings in older patients who are usually excluded from relevant research.
Collapse
Affiliation(s)
- Manar Atyah
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, People’s Republic of China
| | - Chenhao Zhou
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, People’s Republic of China
| | - Qiang Zhou
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, People’s Republic of China
| | - Wanyong Chen
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, People’s Republic of China
| | - Jialei Weng
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, People’s Republic of China
| | - Pengcheng Wang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, People’s Republic of China
| | - Yi Shi
- Biomedical Research Centre, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Qiongzhu Dong
- Institute of Fudan Minhang Academic Health System, and Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, 201199, People’s Republic of China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Ning Ren
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Fudan Minhang Academic Health System, and Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, 201199, People’s Republic of China
| |
Collapse
|
165
|
Lee JM, Lee JH, Song MK, Kim YJ. NXP032 Ameliorates Aging-Induced Oxidative Stress and Cognitive Impairment in Mice through Activation of Nrf2 Signaling. Antioxidants (Basel) 2022; 11:antiox11010130. [PMID: 35052634 PMCID: PMC8772799 DOI: 10.3390/antiox11010130] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/31/2021] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
Aging is a neurodegenerative disease that leads to cognitive impairment, and an increase in oxidative stress as a major cause is an important factor. It has been reported that aging-related cognitive impairment is associated with increased oxidative damage in several brain regions during aging. As a powerful antioxidant, vitamin C plays an important role in preventing oxidative stress, but due to its unstable chemical properties, it is easily oxidized and thus the activity of antioxidants is reduced. In order to overcome this easily oxidized vulnerability, we developed NXP032 (vitamin C/DNA aptamer complex) that can enhance the antioxidant efficacy of vitamin C using an aptamer. We developed NXP032 (vitamin C/DNA Aptamin C320 complex) that can enhance the antioxidant efficacy of vitamin C using an aptamer. In the present study, we evaluated the neuroprotective effects of NXP032 on aging-induced cognitive decline, oxidative stress, and neuronal damage in 17-month-old female mice. NXP032 was orally administered at 200 mg/kg of ascorbic acid and 4 mg/kg of DNA aptamer daily for eight weeks. Before the sacrifice, a cognitive behavioral test was performed. Administration of NXP032 alleviated cognitive impairment, neuronal damage, microglia activity, and oxidative stress due to aging. We found that although aging decreases the Nrf2-ARE pathway, NXP032 administration activates the Nrf2-ARE pathway to increase the expression of SOD-1 and GSTO1/2. The results suggest that the new aptamer complex NXP032 may be a therapeutic intervention to alleviate aging-induced cognitive impairment and oxidative stress.
Collapse
Affiliation(s)
- Jae-Min Lee
- College of Nursing Science, Kyung Hee University, Seoul 02447, Korea;
| | - Joo Hee Lee
- Department of Nursing, Graduate School, Kyung Hee University, Seoul 02447, Korea;
| | - Min Kyung Song
- Robert Wood Johnson Medical School, Institute for Neurological Therapeutics, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA;
| | - Youn-Jung Kim
- College of Nursing Science, Kyung Hee University, Seoul 02447, Korea;
- Correspondence: ; Tel.: +82-2-961-0311
| |
Collapse
|
166
|
Calpain Inhibitors as Potential Therapeutic Modulators in Neurodegenerative Diseases. Neurochem Res 2022; 47:1125-1149. [PMID: 34982393 DOI: 10.1007/s11064-021-03521-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023]
Abstract
It is considered a significant challenge to understand the neuronal cell death mechanisms with a suitable cure for neurodegenerative disorders in the coming years. Calpains are one of the best-considered "cysteine proteases activated" in brain disorders. Calpain is an important marker and mediator in the pathophysiology of neurodegeneration. Calpain activation being the essential neurodegenerative factor causing apoptotic machinery activation, it is crucial to develop reliable and effective approaches to prevent calpain-mediated apoptosis in degenerating neurons. It has been recently seen that the "inhibition of calpain activation" has appeared as a possible therapeutic target for managing neurodegenerative diseases. A systematic literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was conducted. The present article reviews the basic pathobiology and role of selective calpain inhibitors used in various neurodegenerative diseases as a therapeutic target.
Collapse
|
167
|
Mokrane N, Snabi Y, Cens T, Guiramand J, Charnet P, Bertaud A, Menard C, Rousset M, de Jesus Ferreira MC, Thibaud JB, Cohen-Solal C, Vignes M, Roussel J. Manipulations of Glutathione Metabolism Modulate IP 3-Mediated Store-Operated Ca 2+ Entry on Astroglioma Cell Line. Front Aging Neurosci 2022; 13:785727. [PMID: 34975458 PMCID: PMC8719003 DOI: 10.3389/fnagi.2021.785727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/01/2021] [Indexed: 02/03/2023] Open
Abstract
The regulation of the redox status involves the activation of intracellular pathways as Nrf2 which provides hormetic adaptations against oxidative stress in response to environmental stimuli. In the brain, Nrf2 activation upregulates the formation of glutathione (GSH) which is the primary antioxidant system mainly produced by astrocytes. Astrocytes have also been shown to be themselves the target of oxidative stress. However, how changes in the redox status itself could impact the intracellular Ca2+ homeostasis in astrocytes is not known, although this could be of great help to understand the neuronal damage caused by oxidative stress. Indeed, intracellular Ca2+ changes in astrocytes are crucial for their regulatory actions on neuronal networks. We have manipulated GSH concentration in astroglioma cells with selective inhibitors and activators of the enzymes involved in the GSH cycle and analyzed how this could modify Ca2+ homeostasis. IP3-mediated store-operated calcium entry (SOCE), obtained after store depletion elicited by Gq-linked purinergic P2Y receptors activation, are either sensitized or desensitized, following GSH depletion or increase, respectively. The desensitization may involve decreased expression of the proteins STIM2, Orai1, and Orai3 which support SOCE mechanism. The sensitization process revealed by exposing cells to oxidative stress likely involves the increase in the activity of Calcium Release-Activated Channels (CRAC) and/or in their membrane expression. In addition, we observe that GSH depletion drastically impacts P2Y receptor-mediated changes in membrane currents, as evidenced by large increases in Ca2+-dependent K+ currents. We conclude that changes in the redox status of astrocytes could dramatically modify Ca2+ responses to Gq-linked GPCR activation in both directions, by impacting store-dependent Ca2+-channels, and thus modify cellular excitability under purinergic stimulation.
Collapse
Affiliation(s)
- Nawfel Mokrane
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Yassin Snabi
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Thierry Cens
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Janique Guiramand
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Pierre Charnet
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Anaïs Bertaud
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Claudine Menard
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Matthieu Rousset
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France
| | - Marie-Céleste de Jesus Ferreira
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | | | - Catherine Cohen-Solal
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Michel Vignes
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| | - Julien Roussel
- UMR 5247 Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,Department of Biological Sciences, Université de Montpellier, Montpellier, France
| |
Collapse
|
168
|
Seminotti B, Grings M, Tucci P, Leipnitz G, Saso L. Nuclear Factor Erythroid-2-Related Factor 2 Signaling in the Neuropathophysiology of Inherited Metabolic Disorders. Front Cell Neurosci 2021; 15:785057. [PMID: 34955754 PMCID: PMC8693715 DOI: 10.3389/fncel.2021.785057] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/05/2021] [Indexed: 01/14/2023] Open
Abstract
Inherited metabolic disorders (IMDs) are rare genetic conditions that affect multiple organs, predominantly the central nervous system. Since treatment for a large number of IMDs is limited, there is an urgent need to find novel therapeutical targets. Nuclear factor erythroid-2-related factor 2 (Nrf2) is a transcription factor that has a key role in controlling the intracellular redox environment by regulating the expression of antioxidant enzymes and several important genes related to redox homeostasis. Considering that oxidative stress along with antioxidant system alterations is a mechanism involved in the neuropathophysiology of many IMDs, this review focuses on the current knowledge about Nrf2 signaling dysregulation observed in this group of disorders characterized by neurological dysfunction. We review here Nrf2 signaling alterations observed in X-linked adrenoleukodystrophy, glutaric acidemia type I, hyperhomocysteinemia, and Friedreich’s ataxia. Additionally, beneficial effects of different Nrf2 activators are shown, identifying a promising target for treatment of patients with these disorders. We expect that this article stimulates research into the investigation of Nrf2 pathway involvement in IMDs and the use of potential pharmacological modulators of this transcription factor to counteract oxidative stress and exert neuroprotection.
Collapse
Affiliation(s)
- Bianca Seminotti
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mateus Grings
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Guilhian Leipnitz
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Postgraduate Program in Biological Sciences: Physiology, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
169
|
Xiong Y, Xiong Y, Zhang H, Zhao Y, Han K, Zhang J, Zhao D, Yu Z, Geng Z, Wang L, Wang Y, Luan X. hPMSCs-Derived Exosomal miRNA-21 Protects Against Aging-Related Oxidative Damage of CD4 + T Cells by Targeting the PTEN/PI3K-Nrf2 Axis. Front Immunol 2021; 12:780897. [PMID: 34887868 PMCID: PMC8649962 DOI: 10.3389/fimmu.2021.780897] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs)-derived exosomes were considered a novel therapeutic approach in many aging-related diseases. This study aimed to clarify the protective effects of human placenta MSCs-derived exosomes (hPMSC-Exo) in aging-related CD4+ T cell senescence and identified the underlying mechanisms using a D-gal induced mouse aging model. Senescent T cells were detected SA-β-gal stain. The degree of DNA damage was evaluated by detecting the level of 8-OH-dG. The superoxide dismutase (SOD) and total antioxidant capacity (T-AOC) activities were measured. The expression of aging-related proteins and senescence-associated secretory phenotype (SASP) were detected by Western blot and RT-PCR. We found that hPMSC-Exo treatment markedly decreased oxidative stress damage (ROS and 8-OH-dG), SA-β-gal positive cell number, aging-related protein expression (p53 and γ-H2AX), and SASP expression (IL-6 and OPN) in senescent CD4+ T cells. Additionally, hPMSC-Exo containing miR-21 effectively downregulated the expression of PTEN, increased p-PI3K and p-AKT expression, and Nrf2 nuclear translocation and the expression of downstream target genes (NQO1 and HO-1) in senescent CD4+ T cells. Furthermore, in vitro studies uncovered that hPMSC-Exo attenuated CD4+ T cell senescence by improving the PTEN/PI3K-Nrf2 axis by using the PTEN inhibitor bpV (HOpic). We also validated that PTEN was a target of miR-21 by using a luciferase reporter assay. Collectively, the obtained results suggested that hPMSC-Exo attenuates CD4+ T cells senescence via carrying miRNA-21 and activating PTEN/PI3K-Nrf2 axis mediated exogenous antioxidant defenses.
Collapse
Affiliation(s)
- Yanlian Xiong
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Yanlei Xiong
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hengchao Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Yaxuan Zhao
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Kaiyue Han
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Jiashen Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Dongmei Zhao
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Zhenhai Yu
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Ziran Geng
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Longfei Wang
- Department of Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Yueming Wang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Xiying Luan
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| |
Collapse
|
170
|
Huang Y, Shen Z, Huang C, Lin C, Tsai T. Cisd2 slows down liver aging and attenuates age-related metabolic dysfunction in male mice. Aging Cell 2021; 20:e13523. [PMID: 34811857 PMCID: PMC8672792 DOI: 10.1111/acel.13523] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/18/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023] Open
Abstract
The liver plays a pivotal role in mammalian aging. However, the mechanisms underlying liver aging remain unclear. Cisd2 is a pro‐longevity gene in mice. Cisd2 mediates lifespan and healthspan via regulation of calcium homeostasis and mitochondrial functioning. Intriguingly, the protein level of Cisd2 is significantly decreased by about 50% in the livers of old male mice. This down‐regulation of Cisd2 may result in the aging liver exhibiting non‐alcoholic fatty liver disease (NAFLD) phenotype. Here, we use Cisd2 transgenic mice to investigate whether maintaining Cisd2 protein at a persistently high level is able to slow down liver aging. Our study identifies four major discoveries. Firstly, that Cisd2 expression attenuates age‐related dysregulation of lipid metabolism and other pathological abnormalities. Secondly, revealed by RNA sequencing analysis, the livers of old male mice undergo extensive transcriptomic alterations, and these are associated with steatosis, hepatitis, fibrosis, and xenobiotic detoxification. Intriguingly, a youthful transcriptomic profile, like that of young 3‐month‐old mice, was found in old Cisd2 transgenic male mice at 26 months old. Thirdly, Cisd2 suppresses the age‐associated dysregulation of various transcription regulators (Nrf2, IL‐6, and Hnf4a), which keeps the transcriptional network in a normal pattern. Finally, a high level of Cisd2 protein protects the liver from oxidative stress, and this is associated with a reduction in mitochondrial DNA deletions. These findings demonstrate that Cisd2 is a promising target for the development of therapeutic agents that, by bringing about an effective enhancement of Cisd2 expression, will slow down liver aging.
Collapse
Affiliation(s)
- Yi‐Long Huang
- Department of Life Sciences and Institute of Genome Sciences National Yang Ming Chiao Tung University Taipei Taiwan
- Aging and Health Research Center National Yang Ming Chiao Tung University Taipei Taiwan
| | - Zhao‐Qing Shen
- Department of Life Sciences and Institute of Genome Sciences National Yang Ming Chiao Tung University Taipei Taiwan
| | - Chen‐Hua Huang
- Department of Life Sciences and Institute of Genome Sciences National Yang Ming Chiao Tung University Taipei Taiwan
| | - Chao‐Hsiung Lin
- Department of Life Sciences and Institute of Genome Sciences National Yang Ming Chiao Tung University Taipei Taiwan
- Aging and Health Research Center National Yang Ming Chiao Tung University Taipei Taiwan
| | - Ting‐Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences National Yang Ming Chiao Tung University Taipei Taiwan
- Aging and Health Research Center National Yang Ming Chiao Tung University Taipei Taiwan
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Taiwan
| |
Collapse
|
171
|
Zhang H, Xu J, Peng C, Qiu J, Wang Z, Regenstein JM, Yang X, Zheng Z. Anti-fatigue liquid formulations made from fruits. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
172
|
Li Y, Qiu X, Lu Z, Zhan F, Yang M, Sarath Babu V, Li J, Qin Z, Lin L. Molecular and functional characterization of MST2 in grass carp during bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2021; 119:19-30. [PMID: 34560286 DOI: 10.1016/j.fsi.2021.09.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 06/13/2023]
Abstract
The regulation of host redox homeostasis is critically important in the immune response to pathogens. The "mammalian sterile 20-like" kinase 2 (MST2) has been shown to play a role in apoptosis, cell proliferation, and cancer; however, few studies have examined its ability to modulate redox homeostasis during innate immunity, especially in teleost fish. In this study, we cloned the MST2 gene of Ctenopharyngodon idella (CiMST2) and analyzed its tissue distribution. CiMST2 was present in most of the studied tissues, and it was most highly expressed in brain tissue. Expression patterns analysis revealed that MST2 mRNA and protein were significantly up-regulated under bacterial infection, suggesting that it is involved in the immune response. Bacterial stimulation significantly increased the level of antioxidases. To explore the interplay between CiMST2 and antioxidant regulation, we examined the effects of CiMST2 overexpression and conducted RNA interference assays in vitro. CiMST2 overexpression significantly increased the expression levels of nuclear factor E2-related factor 2 (Nrf2) and other antioxidases and vice versa, revealing that CiMST2 regulated host redox homeostasis via Nrf2-antioxidant responsive element (ARE) signaling. Overall, our findings provide a new perspective on the role of MST2 in innate immunity in teleosts as well as insights that will aid the prevention and control of disease in the grass carp farming industry.
Collapse
Affiliation(s)
- Yanan Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, USA
| | - Xiaolong Qiu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, USA
| | - Zhijie Lu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, USA
| | - Fanbin Zhan
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, USA
| | - Minxuan Yang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, USA
| | - V Sarath Babu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, USA
| | - Jun Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, USA; School of Biological Sciences, Lake Superior State University, Sault Ste. Marie, MI, 49783, USA
| | - Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, USA.
| | - Li Lin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, USA.
| |
Collapse
|
173
|
Kim MN, Moon JH, Cho YM. Sodium-glucose cotransporter-2 inhibition reduces cellular senescence in the diabetic kidney by promoting ketone body-induced NRF2 activation. Diabetes Obes Metab 2021; 23:2561-2571. [PMID: 34318973 DOI: 10.1111/dom.14503] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/07/2021] [Accepted: 07/25/2021] [Indexed: 01/10/2023]
Abstract
AIMS To evaluate whether sodium-glucose cotransporter-2 (SGLT2) inhibition reduces cellular senescence in the kidney and to investigate the molecular pathways involved in the renoprotective effect. MATERIALS AND METHODS Dapagliflozin (1 mg/kg), glimepiride (2.5 mg/kg) or vehicle was administered daily via oral gavage for 8 weeks in db/db mice. Expression levels of ageing marker genes (p21, p16, and p53) and oxidative stress were measured in the kidney using real-time RT-PCR, immunohistochemistry, and Western blot analysis. For in vitro analysis, HK-2 cells, a human renal tubular epithelial cell line, were pretreated with H2 O2 to induce cellular senescence, and the levels of ageing markers were measured after treatment with β-hydroxybutyrate (β-HB) or NRF2-specific siRNA. RESULTS Expression levels of ageing marker genes (p21, p16 and p53) and senescence-associated secretory phenotypes of the kidney were increased in the vehicle-treated db/db (db/db + vehicle) group compared with the db/+ group, and this increase was markedly reversed in the dapagliflozin-treated db/db (db/db + SGLT2 inhibitor) group, but not in the glimepiride-treated db/db (db/db + sulphonylurea [SU]) group. In the kidneys of mice in the db/db + SGLT2 inhibitor group, oxidative stress and DNA damage were also reduced compared with those of mice in the db/db + vehicle and db/db + SU groups. Dapagliflozin increased plasma β-HB, which reduced H2 O2 -induced DNA damage and senescence in HK-2 cells. β-HB-induced NRF2 nuclear translocation mediated anti-senescent effects by inducing antioxidant pathways. CONCLUSIONS Dapagliflozin prevented the progression of diabetic kidney disease by inhibiting cellular senescence and oxidative stress via ketone-induced NRF2 activation.
Collapse
Affiliation(s)
- Mi Na Kim
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Joon Ho Moon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Young Min Cho
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Institute on Aging, Seoul National University, Seoul, South Korea
| |
Collapse
|
174
|
Alì S, Davinelli S, Accardi G, Aiello A, Caruso C, Duro G, Ligotti ME, Pojero F, Scapagnini G, Candore G. Healthy ageing and Mediterranean diet: A focus on hormetic phytochemicals. Mech Ageing Dev 2021; 200:111592. [PMID: 34710375 DOI: 10.1016/j.mad.2021.111592] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/06/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022]
Abstract
Mediterranean diet (MedDiet) is rich in fruits and vegetables associated with longevity and a reduced risk of several age-related diseases. It is demonstrated that phytochemicals in these plant products enhance the positive effects of MedDiet by acting on the inflammatory state and reducing oxidative stress. Evidence support that these natural compounds act as hormetins, triggering one or more adaptive stress-response pathways at low doses. Activated stress-response pathways increase the expression of cytoprotective proteins and multiple genes that act as lifespan regulators, essential for the ageing process. In these ways, the hormetic response by phytochemicals such as resveratrol, ferulic acid, and several others in MedDiet might enhance cells' ability to cope with more severe challenges, resist diseases, and promote longevity. This review discusses the role of MedDiet phytochemicals in healthy ageing and the prevention of age-related diseases.
Collapse
Affiliation(s)
- Sawan Alì
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Sergio Davinelli
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Italy.
| | - Giovanni Duro
- Institute for Research and Biomedical Innovation, National Research Council, Palermo, Italy
| | - Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Italy; Institute for Research and Biomedical Innovation, National Research Council, Palermo, Italy
| | - Fanny Pojero
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Italy
| |
Collapse
|
175
|
Chen J, Liang JQ, Zhen YF, Chang L, Zhou ZT, Shen XJ. DCAF1-targeting microRNA-3175 activates Nrf2 signaling and inhibits dexamethasone-induced oxidative injury in human osteoblasts. Cell Death Dis 2021; 12:1024. [PMID: 34716304 PMCID: PMC8556244 DOI: 10.1038/s41419-021-04300-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/29/2022]
Abstract
Activation of nuclear-factor-E2-related factor 2 (Nrf2) signaling can protect human osteoblasts from dexamethasone-induced oxidative injury. DDB1 and CUL4 associated factor 1 (DCAF1) is a novel ubiquitin E3 ligase for Nrf2 protein degradation. We identified a novel DCAF1-targeting miRNA, miR-3175. RNA pull-down, Argonaute 2 RNA-immunoprecipitation, and RNA fluorescent in situ hybridization results confirmed a direct binding between miR-3175 and DCAF1 mRNA in primary human osteoblasts. DCAF1 3'-untranslated region luciferase activity and its expression were significantly decreased after miR-3175 overexpression but were augmented with miR-3175 inhibition in human osteoblasts and hFOB1.19 osteoblastic cells. miR-3175 overexpression activated Nrf2 signaling, causing Nrf2 protein stabilization, antioxidant response (ARE) activity increase, and transcription activation of Nrf2-dependent genes in human osteoblasts and hFOB1.19 cells. Furthermore, dexamethasone-induced oxidative injury and apoptosis were largely attenuated by miR-3175 overexpression in human osteoblasts and hFOB1.19 cells. Importantly, shRNA-induced silencing or CRISPR/Cas9-mediated Nrf2 knockout abolished miR-3175 overexpression-induced osteoblast cytoprotection against dexamethasone. Conversely, DFAC1 knockout, by the CRISPR/Cas9 method, activated the Nrf2 cascade and inhibited dexamethasone-induced cytotoxicity in hFOB1.19 cells. Importantly, miR-3175 expression was decreased in necrotic femoral head tissues of dexamethasone-taking patients, where DCAF1 mRNA was upregulated. Together, silencing DCAF1 by miR-3175 activated Nrf2 signaling to inhibit dexamethasone-induced oxidative injury and apoptosis in human osteoblasts.
Collapse
Affiliation(s)
- Jing Chen
- Department of Endocrinology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Jin-Qian Liang
- Department of Orthopaedics, Peking Union Medical College Hospital, Beijing, China
| | - Yun-Fang Zhen
- The Center of Diagnosis and Treatment for Children's Bone Diseases, The Children's Hospital of Soochow University, Suzhou, China
| | - Lei Chang
- Department of Spine Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Zhen-Tao Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Xiong-Jie Shen
- Department of Spine Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China.
| |
Collapse
|
176
|
Liang JQ, Zhou ZT, Bo L, Tan HN, Hu JH, Tan MS. Phosphoglycerate kinase 1 silencing by a novel microRNA microRNA-4523 protects human osteoblasts from dexamethasone through activation of Nrf2 signaling cascade. Cell Death Dis 2021; 12:964. [PMID: 34667156 PMCID: PMC8526604 DOI: 10.1038/s41419-021-04250-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/16/2021] [Accepted: 09/29/2021] [Indexed: 12/26/2022]
Abstract
Nuclear-factor-E2-related factor 2 (Nrf2) cascade activation can ameliorate dexamethasone (DEX)-induced oxidative injury and death in human osteoblasts. Phosphoglycerate kinase 1 (PGK1) depletion is shown to efficiently activate Nrf2 signaling by inducing methylglyoxal modification of Kelch-like ECH-associated protein 1 (Keap1). We here identified a novel PGK1-targeting microRNA: microRNA-4523 (miR-4523). RNA fluorescent in situ hybridization, RNA pull-down, and Argonaute-2 RNA immunoprecipitation results confirmed a direct binding between miR-4523 and PGK1 mRNA in primary human osteoblasts and hFOB1.19 osteoblastic cells. Forced overexpression of miR-4523, using a lentiviral construct, robustly decreased PGK1 3'-UTR (untranslated region) luciferase activity and downregulated its expression in human osteoblasts and hFOB1.19 cells. Furthermore, miR-4523 overexpression activated the Nrf2 signaling cascade, causing Keap1-Nrf2 disassociation, Nrf2 protein stabilization, and its nuclear translocation as well as transcription activation of Nrf2-dependent genes (NQO1, GCLC, and HO1) in human osteoblasts. By expressing a UTR-null PGK1 construct, miR-4523 overexpression-induced Nrf2 cascade activation was however largely inhibited. Importantly, DEX-induced reactive oxygen species production, oxidative injury, and cell apoptosis were significantly attenuated by miR-4523 overexpression in human osteoblasts and hFOB1.19 cells. Such actions by miR-4523 were abolished by Nrf2 shRNA or knockout, but mimicked by PGK1 knockout (using CRISPR/Cas9 method). In PGK1 knockout human osteoblasts, miR-4523 overexpression failed to further increase Nrf2 cascade activation and offer osteoblast cytoprotection against DEX. Significantly, miR-4523 is downregulated in human necrotic femoral head tissues of DEX-taking patients. Together, PGK1 silencing by miR-4523 protected human osteoblasts from DEX through activation of the Nrf2 signaling cascade.
Collapse
Affiliation(s)
- Jin-Qian Liang
- Department of Orthopaedics, Peking Union Medical College Hospital, Beijing, China
| | - Zhen-Tao Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Lin Bo
- Department of Rheumatology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hai-Ning Tan
- Department of Orthopaedics, Peking Union Medical College Hospital, Beijing, China
| | - Jian-Hua Hu
- Department of Orthopaedics, Peking Union Medical College Hospital, Beijing, China.
| | - Ming-Sheng Tan
- Spinal Surgery, Sino-Japanese Friendship Hospital, Beijing, China.
| |
Collapse
|
177
|
Jiang Y, Gu C, Xu H, Shi F, Zhang X, Wang F. iKeap1 activates Nrf2 signaling to protect myocardial cells from oxygen glucose deprivation/re-oxygenation-induced oxidative injury. Biochem Biophys Res Commun 2021; 574:110-117. [PMID: 34461498 DOI: 10.1016/j.bbrc.2021.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 11/24/2022]
Abstract
Nuclear factor E2-related factor 2 (Nrf2) activation could efficiently protect myocardial cells from oxygen glucose deprivation/re-oxygenation (OGDR). An ultra-large structure-based virtual screening has discovered iKeap1 as a novel, direct and potent Keap1 inhibitor. Here we found that iKeap1 efficiently activated Nrf2 signaling in H9c2 myocardial cells and primary murine myocardiocytes. iKeap1 induced Keap1-Nrf2 disassociation, cytosol Nrf2 protein stabilization and nuclear translocation. The antioxidant response element (ARE) activity and expression of Nrf2 cascade genes (HO1, NQO1 and GCLC) were increased in iKeap1-treated myocardial cells. In H9c2 cells and murine myocardiocytes, iKeap1 potently inhibited OGDR-induced oxidative injury by inhibiting reactive oxygen species (ROS) production, mitochondrial depolarization, lipid peroxidation and DNA damage. In addition, OGDR-induced myocardial cell death and apoptosis were largely ameliorated after pretreatment with the novel Keap1 inhibitor. Significantly, in H9c2 cells iKeap1-induced myocardial cytoprotection against OGDR was abolished with Nrf2 silencing or knockout (using CRISPR/Cas9 method). Moreover, CRISPR/Cas9-induced Keap1 knockout led to constitutive activation of Nrf2 cascade and inhibited OGDR-induced oxidative injury. Importantly, iKeap1 was unable to further protect Keap1-knockout H9c2 cells from OGDR. Together, iKeap1 activated Nrf2 signaling to protect myocardial cells from OGDR-induced oxidative injury and cell death.
Collapse
Affiliation(s)
- Yicheng Jiang
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, China
| | - Cheng Gu
- Department of Critical Care Medicine, Changshu No.2 People's Hospital, Changshu, China
| | - Hai Xu
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, China
| | - Feiya Shi
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, China
| | - Xiwen Zhang
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, China.
| | - Fang Wang
- Department of Cardiology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China.
| |
Collapse
|
178
|
Zhu F, Shao J, Tian Y, Xu Z. Sulfiredoxin-1 protects retinal ganglion cells from high glucose-induced oxidative stress and inflammatory injury by potentiating Nrf2 signaling via the Akt/GSK-3β pathway. Int Immunopharmacol 2021; 101:108221. [PMID: 34653733 DOI: 10.1016/j.intimp.2021.108221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/17/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
Sulfiredoxin-1 (Srxn1) has been acknowledged as a remarkable pro-survival factor in the protection of cells against stress-induced damage. The persistent exposure of retinal ganglion cells (RGCs) to high glucose (HG) in diabetes induces cellular damage, which contributes to the onset of diabetic retinopathy, a severe complication of diabetes. So far, little is known about the role of Srxn1 in regulating HG-induced injury of RGCs. The goals of this work were to evaluate the possible relevance of Srxn1 in the modulation of HG-induced apoptosis, oxidative stress and inflammation of RGCs in vitro. Our data showed that HG exposure caused a marked decrease in Srxn1 expression in RGCs. The up-regulation of Srxn1 markedly decreased HG-evoked apoptosis, reactive oxygen species (ROS) generation and pro-inflammatory cytokine release in RGCs. On the contrary, the depletion of Srxn1 rendered RGCs more susceptible to HG-induced injury. Further data demonstrated that Srnx1 enhanced the activation of nuclear factor erythroid-2 (E2)-related factor 2 (Nrf2) signaling in HG-exposed RGCs associated with up-regulating the phosphorylation of Akt and glucogen synthase kinase-3β (GSK-3β). Notably, the inhibition of Akt abolished Srnx1-overexpression-mediated Nrf2 activation, while GSK-3β inhibition reversed Srnx1-depletion-mediated inactivation of Nrf2. In addition, Nrf2 inhibition partially abrogated Srnx1-mediated protective effects against HG-induced injury of RGCs. In summary, these data demonstrate that the overexpression of Srxn1 protects RGCs from the HG-induced injury of RGCs by enhancing Nrf2 signaling via modulation of Akt/GSK-3β axis. Our work highlights that the Srxn1-mediated Akt/GSK-3β/Nrf2 axis may exert a possible role in regulating RGC injury of diabetic retinopathy.
Collapse
Affiliation(s)
- Fei Zhu
- Ophthalmology, Yulin Hospital of Traditional Chinese Medicine, Yulin 719000, China
| | - Juan Shao
- Ophthalmology, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, China.
| | - Yunlin Tian
- Ophthalmology, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, China
| | - Zhiguo Xu
- Ophthalmology, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
179
|
Cuadrado A. Brain-Protective Mechanisms of Transcription Factor NRF2: Toward a Common Strategy for Neurodegenerative Diseases. Annu Rev Pharmacol Toxicol 2021; 62:255-277. [PMID: 34637322 DOI: 10.1146/annurev-pharmtox-052220-103416] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neurodegenerative diseases are characterized by the loss of homeostatic functions that control redox and energy metabolism, neuroinflammation, and proteostasis. The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is a master controller of these functions, and its overall activity is compromised during aging and in these diseases. However, NRF2 can be activated pharmacologically and is now being considered a common therapeutic target. Many gaps still exist in our knowledge of the specific role that NRF2 plays in specialized brain cell functions or how these cells respond to the hallmarks of these diseases. This review discusses the relevance of NRF2 to several hallmark features of neurodegenerative diseases and the current status of pharmacological activators that might pass through the blood-brain barrier and provide a disease-modifying effect. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid, Madrid 28049, Spain.,Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid 28029, Spain.,Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid 28046, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid 28031, Spain;
| |
Collapse
|
180
|
Zhao G, Qi L, Wang Y, Li X, Li Q, Tang X, Wang X, Wu C. Antagonizing effects of curcumin against mercury-induced autophagic death and trace elements disorder by regulating PI3K/AKT and Nrf2 pathway in the spleen. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 222:112529. [PMID: 34293585 DOI: 10.1016/j.ecoenv.2021.112529] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/27/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
Mercury is a naturally occurring element and highly toxic to humans even at a low dosage. Curcumin is a polyphenol found in turmeric (Curcuma longa), widely used as a treatment strategy to improve antioxidant and anti-inflammatory properties. The purpose of this study was to investigate the potential protective mechanisms of curcumin in spleen damage induced by HgCl2. The mice were given curcumin by intragastric administration 2 h before HgCl2 injection for 24 h. At first, splenic transcriptome analysis showed that 3334 genes (2134 up and 1200 down) were differently expressed in HgCl2-induced spleen damage model. Notably, KEGG enrichment showed phosphatidylinositol 3-kinase (PI3K)-AKT might be a key signaling pathways in HgCl2-induced spleen damage. Furthermore, our data demonstrated that HgCl2 could induce autophagic cell death, evidenced by increases the protein expression of PI3K, AKT, LC3-II and p62 and the number of apoptotic cells. Furthermore, we found that curcumin significantly combated autophagic cell death, sodium overload and calcium leak induced by HgCl2. Simultaneously, further studies demonstrated that curcumin significantly activated nuclear factor (erythroid-derived-2)-like 2 (Nrf2) signaling pathway, and subsequent enhancing antioxidant defenses. Taken together, our data indicated that inorganic mercury could result in autophagic cell death, which may be related to the regulation of PI3K-AKT signaling cascades. Furthermore, Nrf2-mediated antioxidant defenses may be the target of curcumin to confers an adaptive survival response to resist spleen damage induced by HgCl2. The present study perfects the mechanism theory of HgCl2-induced spleen damage and provides a way for pharmacological intervention to prevent spleen injury.
Collapse
Affiliation(s)
- Guifang Zhao
- Department of Core Medical Laboratory, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, PR China
| | - Ling Qi
- Department of Core Medical Laboratory, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, PR China
| | - Yanling Wang
- Department of Pathophysiology, College of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan Province, PR China
| | - Xinlian Li
- Department of Pathophysiology, College of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan Province, PR China
| | - Qiuyue Li
- Department of Pathophysiology, College of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan Province, PR China
| | - Xiaoqing Tang
- Department of Pathophysiology, College of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan Province, PR China
| | - Xiali Wang
- Department of Pathophysiology, College of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan Province, PR China
| | - Chunling Wu
- Department of Pathophysiology, College of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan Province, PR China.
| |
Collapse
|
181
|
Sumida K, Pierre JF, Han Z, Mims TS, Potukuchi PK, Yuzefpolskaya M, Colombo PC, Demmer RT, Datta S, Kovesdy CP. Circulating Microbial Signatures and Cardiovascular Death in Patients With ESRD. Kidney Int Rep 2021; 6:2617-2628. [PMID: 34622101 PMCID: PMC8484116 DOI: 10.1016/j.ekir.2021.07.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Patients with end-stage renal disease (ESRD) experience disproportionately high cardiovascular morbidity and mortality. Accumulating evidence suggests a role for the circulating microbiome in the pathogenesis of cardiovascular disease; however, little is known about its association with premature cardiovascular mortality in ESRD. METHODS In a pilot case-control study of 17 hemodialysis patients who died of a cardiovascular event and 17 matched hemodialysis controls who remained alive during a median follow-up of 2.0 years, we compared the levels and composition of circulating microbiome, including Bacteria, Archaea, and Fungi, in serum samples by quantitative polymerase chain reaction and 16S or Internal Transcribed Spacer (ITS) ribosomal RNA (rRNA) sequencing, respectively. Associations of the circulating cell-free microbial signatures with clinical parameters and cardiovascular death were examined using the Spearman rank correlation and multivariable conditional logistic regression, respectively. RESULTS Both 16S and ITS rRNA were detectable in all (except 3 for ITS) examined patients' serum samples. Despite no significant difference in 16S rRNA levels and α diversity between cases and controls, taxonomic analysis demonstrated differential community membership between groups, with significantly greater Actinobacteria and less Proteobacteria observed in cases than in controls at the phylum level. Proportions of Actinobacteria and Proteobacteria phyla were significantly correlated with plasma nuclear factor erythroid 2-related factor 2 (Nrf2) levels (rho = -0.41 and 0.42, P = 0.015 and 0.013, respectively) and marginally associated with risk of cardiovascular death (adjusted odds ratios [95% confidence intervals] = 1.12 [0.98-1.29] and 0.88 [0.76-1.02] for 1% increase, respectively). CONCLUSION Alterations of the circulating cell-free microbial signatures may be associated with higher premature cardiovascular mortality in ESRD.
Collapse
Affiliation(s)
- Keiichi Sumida
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Joseph F. Pierre
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Zhongji Han
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Tahliyah S. Mims
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Praveen Kumar Potukuchi
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Melana Yuzefpolskaya
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Columbia University, New York, New York, USA
| | - Paolo C. Colombo
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Columbia University, New York, New York, USA
| | - Ryan T. Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Susmita Datta
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Csaba P. Kovesdy
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Nephrology Section, Memphis VA Medical Center, Memphis, Tennessee, USA
| |
Collapse
|
182
|
Chen J, Chen Y, Zheng Y, Zhao J, Yu H, Zhu J, Li D. Protective Effects and Mechanisms of Procyanidins on Parkinson's Disease In Vivo and In Vitro. Molecules 2021; 26:5558. [PMID: 34577027 PMCID: PMC8464719 DOI: 10.3390/molecules26185558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022] Open
Abstract
This research assessed the molecular mechanism of procyanidins (PCs) against neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and its metabolite 1-methyl-4-phenylpyridinium (MPP+) induced Parkinson's disease (PD) models. In vitro, PC12 cells were incubated with PCs or deprenyl for 24 h, and then exposed to 1.5 mM MPP+ for 24 h. In vivo, zebrafish larvae (AB strain) 3 days post-fertilization (dpf) were incubated with deprenyl or PCs in 400 μM MPTP for 4 days. Compared with MPP+/MPTP alone, PCs significantly improved antioxidant activities (e.g., glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), catalase (CAT)), and decreased levels of reactive oxygen species (ROS) and malondialdehyde (MDA). Furthermore, PCs significantly increased nuclear Nrf2 accumulation in PC12 cells and raised the expression of NQO1, HO-1, GCLM, and GCLC in both PC12 cells and zebrafish compared to MPP+/MPTP alone. The current study shows that PCs have neuroprotective effects, activate the nuclear factor-erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway and alleviate oxidative damage in MPP+/MPTP-induced PD models.
Collapse
Affiliation(s)
- Juan Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Yixuan Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Yangfan Zheng
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Jiawen Zhao
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Huilin Yu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Jiajin Zhu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
| | - Duo Li
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310000, China; (J.C.); (Y.C.); (Y.Z.); (J.Z.); (H.Y.)
- Institute of Nutrition & Health, Qingdao University, Qingdao 266000, China;
| |
Collapse
|
183
|
Fu J, Ni C, Ni H, Xu L, He Q, Pan H, Huang D, Sun Y, Luo G, Liu M, Yao M. Spinal Nrf2 translocation may inhibit neuronal NF-κB activation and alleviate allodynia in a rat model of bone cancer pain. J Neurochem 2021; 158:1110-1130. [PMID: 34254317 PMCID: PMC9292887 DOI: 10.1111/jnc.15468] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/23/2021] [Accepted: 07/02/2021] [Indexed: 01/11/2023]
Abstract
Bone cancer pain (BCP) is a clinical pathology that urgently needs to be solved, but research on the mechanism of BCP has so far achieved limited success. Nuclear factor erythroid 2 (NFE2)-related factor 2 (Nrf2) has been shown to be involved in pain, but its involvement in BCP and the specific mechanism have yet to be examined. This study aimed to test the hypothesis that BCP induces the transfer of Nrf2 from the cytoplasm to the nucleus and further promotes nuclear transcription to activate heme oxygenase-1 (HO-1) and inhibit the activation of nuclear factor-kappa B (NF-κB) signalling, ultimately regulating the neuroinflammatory response. Von-Frey was used for behavioural analysis in rats with BCP, whereas western blotting, real-time quantitative PCR (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) were used to detect molecular expression changes, and immunofluorescence was used to detect cellular localization. We demonstrated that BCP induced increased Nrf2 nuclear protein expression with decreased cytoplasmic protein expression in the spinal cord. Further increases in Nrf2 nuclear protein expression can alleviate hyperalgesia and activate HO-1 to inhibit the expression of NF-κB nuclear protein and inflammatory factors. Strikingly, intrathecal administration of the corresponding siRNA reversed the above effects. In addition, the results of double immune labelling revealed that Nrf2 and NF-κB were coexpressed in spinal cord neurons of rats with BCP. In summary, these findings suggest that the entry of Nrf2 into the nucleus promotes the expression of HO-1, inhibiting activation of the NF-κB signalling pathway, reducing neuroinflammation and ultimately exerting an anti-nociceptive effect.
Collapse
Affiliation(s)
- Jie Fu
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Chaobo Ni
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Hua‐Dong Ni
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Long‐Sheng Xu
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Qiu‐Li He
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Huan Pan
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Dong‐Dong Huang
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Yan‐Bao Sun
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Ge Luo
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Ming‐Juan Liu
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Ming Yao
- Department of Anesthesiology and Pain Research CenterThe First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing UniversityJiaxingChina
| |
Collapse
|
184
|
Emoto MC, Sato-Akaba H, Hamaue N, Kawanishi K, Koshino H, Shimohama S, Fujii HG. Early detection of redox imbalance in the APPswe/PS1dE9 mouse model of Alzheimer's disease by in vivo electron paramagnetic resonance imaging. Free Radic Biol Med 2021; 172:9-18. [PMID: 34058322 DOI: 10.1016/j.freeradbiomed.2021.05.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/14/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that causes progressive cognitive decline. Deposition of amyloid-β (Aβ) peptides is the most important pathophysiological hallmark of AD. Oxidative stress induced by the generation of reactive oxygen species (ROS) is a prominent phenomenon in AD and is known to occur early in its course. Several reports have suggested a relationship between changes in redox status and AD pathology, including progressive Aβ deposition, glial cell activation, and inflammation. In the present study, we employed a newly designed three-dimensional continuous-wave digital electron paramagnetic resonance (EPR) imager with a blood-brain barrier (BBB)-permeable redox-sensitive piperidine nitroxide probe, 4-oxo-2,2,6,6-tetramethyl-piperidine-d16-1-oxyl, for early detection of changed brain redox status. Using this system, we noninvasively compared age-matched 7-month-old AD model mice with normal littermates (WT mice). The obtained brain redox images of AD and WT mice clearly showed impaired brain redox status of AD mice compared to WT, suggesting that oxidative damage had already increased in 7-month-old AD mice compared with age-matched WT mice. The pathological changes in 7-month-old mice in this study were detected earlier than in previous studies in which only AD mice older than 9 months of age could be imaged. Since EPR images suggested that oxidative damage was already increased in 7-month-old AD mice compared to age-matched WT mice, we also evaluated antioxidant levels and the activity of superoxide dismutase (SOD) in brain tissue homogenates of 7-month-old AD and WT mice. Compared to WT mice, decreased levels of glutathione and mitochondrial SOD activity were found in AD mice, which supports the EPR imaging results indicating impaired brain redox status. These results indicate that the EPR imaging method developed in this study is useful for early noninvasive detection of altered brain redox status due to oxidative disease.
Collapse
Affiliation(s)
- Miho C Emoto
- Department of Clinical Laboratory Science, School of Medical Technology, Health Sciences University of Hokkaido, Sapporo, Hokkaido, 002-8072, Japan
| | - Hideo Sato-Akaba
- Department of Systems Innovation, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka, 560-8531, Japan
| | - Naoya Hamaue
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari, Hokkaido, 061-0293, Japan
| | - Katsuya Kawanishi
- Department of Removable Prosthodontics, School of Dentistry, Health Sciences University of Hokkaido, Ishikari, Hokkaido, 061-0293, Japan
| | - Hisashi Koshino
- Department of Removable Prosthodontics, School of Dentistry, Health Sciences University of Hokkaido, Ishikari, Hokkaido, 061-0293, Japan
| | - Shun Shimohama
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, 060-8556, Japan
| | - Hirotada G Fujii
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari, Hokkaido, 061-0293, Japan.
| |
Collapse
|
185
|
Yu H, Jiang X, Dong F, Zhang F, Ji X, Xue M, Yang F, Chen J, Hu X, Bao Z. Lipid accumulation-induced hepatocyte senescence regulates the activation of hepatic stellate cells through the Nrf2-antioxidant response element pathway. Exp Cell Res 2021; 405:112689. [PMID: 34107274 DOI: 10.1016/j.yexcr.2021.112689] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/12/2021] [Accepted: 06/03/2021] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most prevalent chronic liver disease globally. Elderly individuals are at a higher risk of developing NAFLD with severe clinical outcomes. Although NAFLD is closely related to liver aging, the role of hepatocyte senescence in the progression of NAFLD, especially in the development of fibrosis, is still unclear. The early stage of NAFLD is mainly characterized by lipid accumulation in hepatocytes, which could lead to severe oxidative stress, causing cellular senescence. In the present study, hepatocytes cultured in the presence of free fatty acids to induce lipid deposition were used as a hepatocyte senescence model in vitro. Senescent hepatocytes significantly increased the activation of co-cultured primary hepatic stellate cells (HSCs) and the expression of pro-fibrosis molecules. Moreover, the antioxidant regulator nuclear factor erythroid 2-related factor 2 (Nrf2) that was upregulated in senescent hepatocytes was found to be related to the activation of co-cultured HSCs. The Nrf2 agonist sulforaphane, which upregulated the transcriptional activity of the Nrf2-antioxidant response element (ARE) pathway, remarkably inhibited hepatocyte senescence and its activation effect on HSCs. However, the liver tissue obtained from non-alcoholic steatohepatitis (NASH) mice with Nrf2 knockdown showed decreased antioxidation and significant liver senescence and fibrosis. In conclusion, this study confirmed that lipid accumulation induces hepatocyte senescence, which leads to HSC activation and development of hepatic fibrosis. Increasing the activity of the Nrf2-ARE antioxidant pathway in senescent hepatocytes elicited the opposite effect, suggesting that targeting Nrf2 may prevent or delay the progression of aging-related liver fibrosis in NASH.
Collapse
Affiliation(s)
- Huiyuan Yu
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Xin Jiang
- Department of Gerontology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Fangyuan Dong
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Fan Zhang
- Department of Gerontology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Xueying Ji
- Department of Gerontology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Mengjuan Xue
- Department of Gerontology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Fan Yang
- Department of Gerontology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Jie Chen
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China.
| | - Xiaona Hu
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Department of Gerontology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China.
| | - Zhijun Bao
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Department of Gerontology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
186
|
The Impact of Melatonin Supplementation and NLRP3 Inflammasome Deletion on Age-Accompanied Cardiac Damage. Antioxidants (Basel) 2021; 10:antiox10081269. [PMID: 34439517 PMCID: PMC8389221 DOI: 10.3390/antiox10081269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 08/06/2021] [Indexed: 12/16/2022] Open
Abstract
To investigate the role of NLRP3 inflammasome in cardiac aging, we evaluate here morphological and ultrastructural age-related changes of cardiac muscles fibers in wild-type and NLRP3-knockout mice, as well as studying the beneficial effect of melatonin therapy. The results clarified the beginning of the cardiac sarcopenia at the age of 12 months, with hypertrophy of cardiac myocytes, increased expression of β-MHC, appearance of small necrotic fibers, decline of cadiomyocyte number, destruction of mitochondrial cristae, appearance of small-sized residual bodies, and increased apoptotic nuclei ratio. These changes were progressed in the cardiac myocytes of 24 old mice, accompanied by excessive collagen deposition, higher expressions of IL-1α, IL-6, and TNFα, complete mitochondrial vacuolation and damage, myofibrils disorganization, multivesicular bodies formation, and nuclear fragmentation. Interestingly, cardiac myocytes of NLRP3-/- mice showed less detectable age-related changes compared with WT mice. Oral melatonin therapy preserved the normal cardiomyocytes structure, restored cardiomyocytes number, and reduced β-MHC expression of cardiac hypertrophy. In addition, melatonin recovered mitochondrial architecture, reduced apoptosis and multivesicular bodies' formation, and decreased expressions of β-MHC, IL-1α, and IL-6. Fewer cardiac sarcopenic changes and highly remarkable protective effects of melatonin treatment detected in aged cardiomyocytes of NLRP3-/- mice compared with aged WT animals, confirming implication of the NLRP3 inflammasome in cardiac aging. Thus, NLRP3 suppression and melatonin therapy may be therapeutic approaches for age-related cardiac sarcopenia.
Collapse
|
187
|
Hou Y, Li J, Wu JC, Wu QX, Fang J. Activation of Cellular Antioxidant Defense System by Naturally Occurring Dibenzopyrone Derivatives Confers Neuroprotection against Oxidative Insults. ACS Chem Neurosci 2021; 12:2798-2809. [PMID: 34297534 DOI: 10.1021/acschemneuro.1c00023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Seven dibenzopyrone phenolic derivatives, i.e., alternariol (1), alternariol 5-O-methyl ether (2), altenusin B (3), dehydroaltenusin (4), altenuene (5), altenusin (6), and alterlactone (7), were isolated from endophytic fungi Alternaria alternata extract, and these compounds' structures were elucidated based on various spectroscopic data. Compound 3, a diphenic acid derivative, was determined as a new compound. In this study, compounds 3, 4, 6, and 7 displayed remarkable neuroprotective effects against oxidative injuries by acting as potent activators of nuclear factor-erythroid derived 2-like 2 (Nrf2) in PC12 cells. A mechanistic study indicated that these compounds induced the nuclear accumulation of Nrf2, promoted the expression of Nrf2-governed cytoprotective genes, and increased the cellular antioxidant capacity. More importantly, genetic silence of Nrf2 expression deprived the observed cytoprotection, highlighting the important role of Nrf2 in the protection of these compounds.
Collapse
Affiliation(s)
- Yanan Hou
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jie Li
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jun-Chen Wu
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Quan-Xiang Wu
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
188
|
Hou Y, Peng S, Song Z, Bai F, Li X, Fang J. Oat polyphenol avenanthramide-2c confers protection from oxidative stress by regulating the Nrf2-ARE signaling pathway in PC12 cells. Arch Biochem Biophys 2021; 706:108857. [PMID: 33781769 DOI: 10.1016/j.abb.2021.108857] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/08/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023]
Abstract
Accumulating evidence has demonstrated that cellular antioxidant systems play essential roles in retarding oxidative stress-related diseases, such as Parkinson's disease. Because nuclear factor erythroid 2-related factor 2 (Nrf2) is a chief regulator of cellular antioxidant systems, small molecules with Nrf2-activating ability may be promising neuroprotective agents. Avenanthramide-2c (Aven-2c), avenanthramide-2f (Aven-2f) and avenanthramide-2p (Aven-2p) are the most abundant avenanthramides in oats, and they have been documented to possess multiple pharmacological benefits. In this work, we synthesized these three compounds and evaluated their cytoprotective effect against oxidative stress-induced PC12 cell injuries. Aven-2c displayed the best protective potency among them. Aven-2c conferred protection on PC12 cells by scavenging free radicals and activating the Nrf2-ARE signaling pathway. Pretreatment of PC12 cells with Aven-2c efficiently enhanced Nrf2 nuclear accumulation and evoked the expression of a set of cytoprotective molecules. The mechanistic study also supports that Nrf2 activation is the molecular basis for the cellular action of Aven-2c. Collectively, this study demonstrates that Aven-2c is a potent Nrf2 agonist, shedding light on the potential usage of Aven-2c in the treatment of neuroprotective diseases.
Collapse
Affiliation(s)
- Yanan Hou
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Shoujiao Peng
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Zilong Song
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Feifei Bai
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
189
|
Wandt VK, Winkelbeiner N, Lossow K, Kopp JF, Schwarz M, Alker W, Nicolai MM, Simon L, Dietzel C, Hertel B, Pohl G, Ebert F, Schomburg L, Bornhorst J, Haase H, Kipp AP, Schwerdtle T. Ageing-associated effects of a long-term dietary modulation of four trace elements in mice. Redox Biol 2021; 46:102083. [PMID: 34371368 PMCID: PMC8358688 DOI: 10.1016/j.redox.2021.102083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/24/2021] [Accepted: 07/25/2021] [Indexed: 01/11/2023] Open
Abstract
Trace elements (TEs) are essential for diverse processes maintaining body function and health status. The complex regulation of the TE homeostasis depends among others on age, sex, and nutritional status. If the TE homeostasis is disturbed, negative health consequences can result, e.g., caused by impaired redox homeostasis and genome stability maintenance. Based on age-related shifts in TEs which have been described in mice well-supplied with TEs, we aimed to understand effects of a long-term feeding with adequate or suboptimal amounts of four TEs in parallel. As an additional intervention, we studied mice which received an age-adapted diet with higher concentrations of selenium and zinc to counteract the age-related decline of both TEs. We conducted comprehensive analysis of diverse endpoints indicative for the TE and redox status, complemented by analysis of DNA (hydroxy)methylation and markers denoting genomic stability maintenance. TE concentrations showed age-specific alterations which were relatively stable and independent of their nutritional supply. In addition, hepatic DNA hydroxymethylation was significantly increased in the elderly mice and markers indicative for the redox status were modulated. The reduced nutritional supply with TEs inconsistently affected their status, with most severe effects regarding Fe deficiency. This may have contributed to the sex-specific differences observed in the alterations related to the redox status and DNA repair activity. Overall, our results highlight the complexity of factors impacting on the TE status and its physiological consequences. Alterations in TE supply, age, and sex proved to be important determinants that need to be taken into account when considering TE interventions for improving general health and supporting convalescence in the clinics. Trace element profiles differ by age and sex under moderately modulated TE supply. Maintenance of age-related trace element shifts through all feeding groups. Cu/Zn ratio and DNA hydroxymethylation emerge as appropriate murine ageing markers.
Collapse
Affiliation(s)
- Viktoria K Wandt
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany.
| | - Nicola Winkelbeiner
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany.
| | - Kristina Lossow
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 24, 07743, Jena, Germany; German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
| | - Johannes F Kopp
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany.
| | - Maria Schwarz
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 24, 07743, Jena, Germany.
| | - Wiebke Alker
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; Chair of Food Chemistry and Toxicology, Technische Universität Berlin, Straße des 17. Juni 135, 10623, Berlin, Germany.
| | - Merle M Nicolai
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstr. 20, 42119, Wuppertal, Germany.
| | - Luise Simon
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany.
| | - Caroline Dietzel
- Chair of Food Chemistry and Toxicology, Technische Universität Berlin, Straße des 17. Juni 135, 10623, Berlin, Germany.
| | - Barbara Hertel
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
| | - Gabriele Pohl
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
| | - Franziska Ebert
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany.
| | - Lutz Schomburg
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Julia Bornhorst
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstr. 20, 42119, Wuppertal, Germany.
| | - Hajo Haase
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; Chair of Food Chemistry and Toxicology, Technische Universität Berlin, Straße des 17. Juni 135, 10623, Berlin, Germany.
| | - Anna P Kipp
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 24, 07743, Jena, Germany.
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| |
Collapse
|
190
|
Role of Nrf2 in Synaptic Plasticity and Memory in Alzheimer's Disease. Cells 2021; 10:cells10081884. [PMID: 34440653 PMCID: PMC8391447 DOI: 10.3390/cells10081884] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important transcription factor that reduces oxidative stress. When reactive oxygen species (ROS) or reactive nitrogen species (RNS) are detected, Nrf2 translocates from the cytoplasm into the nucleus and binds to the antioxidant response element (ARE), which regulates the expression of antioxidant and anti-inflammatory genes. Nrf2 impairments are observed in the majority of neurodegenerative disorders, including Alzheimer’s disease (AD). The classic hallmarks of AD include β-amyloid (Aβ) plaques, and neurofibrillary tangles (NFTs). Oxidative stress is observed early in AD and is a novel therapeutic target for the treatment of AD. The nuclear translocation of Nrf2 is impaired in AD compared to controls. Increased oxidative stress is associated with impaired memory and synaptic plasticity. The administration of Nrf2 activators reverses memory and synaptic plasticity impairments in rodent models of AD. Therefore, Nrf2 activators are a potential novel therapeutic for neurodegenerative disorders including AD.
Collapse
|
191
|
A Review on Recent Advancement on Age-Related Hearing Loss: The Applications of Nanotechnology, Drug Pharmacology, and Biotechnology. Pharmaceutics 2021; 13:pharmaceutics13071041. [PMID: 34371732 PMCID: PMC8309044 DOI: 10.3390/pharmaceutics13071041] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 11/30/2022] Open
Abstract
Aging is considered a contributing factor to many diseases such as cardiovascular disease, Alzheimer’s disease, and hearing loss. Age-related hearing loss, also termed presbycusis, is one of the most common sensory impairments worldwide, affecting one in five people over 50 years of age, and this prevalence is growing annually. Associations have emerged between presbycusis and detrimental health outcomes, including social isolation and mental health. It remains largely untreatable apart from hearing aids, and with no globally established prevention strategies in the clinical setting. Hence, this review aims to explore the pathophysiology of presbycusis and potential therapies, based on a recent advancement in bile acid-based bio-nanotechnologies. A comprehensive online search was carried out using the following keywords: presbycusis, drugs, hearing loss, bile acids, nanotechnology, and more than 150 publications were considered directly relevant. Evidence of the multifaceted oxidative stress and chronic inflammation involvement in cellular damage and apoptosis that is associated with a loss of hair cells, damaged and inflamed stria vascularis, and neuronal signalling loss and apoptosis continues to emerge. New robust and effective therapies require drug delivery deeper into the various layers of the cochlea. Bile acid-based nanotechnology has gained wide interest in its permeation-enhancing ability and potential for numerous applications in treating presbycusis.
Collapse
|
192
|
Zheng YH, Yang JJ, Tang PJ, Zhu Y, Chen Z, She C, Chen G, Cao P, Xu XY. A novel Keap1 inhibitor iKeap1 activates Nrf2 signaling and ameliorates hydrogen peroxide-induced oxidative injury and apoptosis in osteoblasts. Cell Death Dis 2021; 12:679. [PMID: 34226516 PMCID: PMC8257690 DOI: 10.1038/s41419-021-03962-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 02/08/2023]
Abstract
An ultra-large structure-based virtual screening has discovered iKeap1 as a direct Keap1 inhibitor that can efficiently activate Nrf2 signaling. We here tested its potential effect against hydrogen peroxide (H2O2)-induced oxidative injury in osteoblasts. In primary murine and human osteoblasts, iKeap1 robustly activated Nrf2 signaling at micromole concentrations. iKeap1 disrupted Keap1-Nrf2 association, causing Nrf2 protein stabilization, cytosol accumulation and nuclear translocation in murine and human osteoblasts. The anti-oxidant response elements (ARE) activity and transcription of Nrf2-ARE-dependent genes (including HO1, NQO1 and GCLC) were increased as well. Significantly, iKeap1 pretreatment largely ameliorated H2O2-induced reactive oxygen species production, lipid peroxidation and DNA damage as well as cell apoptosis and programmed necrosis in osteoblasts. Moreover, dexamethasone- and nicotine-induced oxidative injury and apoptosis were alleviated by iKeap1. Importantly, Nrf2 shRNA or CRISPR/Cas9-induced Nrf2 knockout completely abolished iKeap1-induced osteoblast cytoprotection against H2O2. Conversely, CRISPR/Cas9-induced Keap1 knockout induced Nrf2 cascade activation and mimicked iKeap1-induced cytoprotective actions in murine osteoblasts. iKeap1 was ineffective against H2O2 in the Keap1-knockout murine osteoblasts. Collectively, iKeap1 activated Nrf2 signaling cascade to inhibit H2O2-induced oxidative injury and death of osteoblasts.
Collapse
Affiliation(s)
- Yue-huan Zheng
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-jun Yang
- grid.412538.90000 0004 0527 0050Department of Orthopedics, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Pei-jun Tang
- grid.490559.4Department of Pulmonary, The Affiliated Infectious Diseases Hospital of Soochow University, The Fifth People’s Hospital of Suzhou, Suzhou, China
| | - Yuan Zhu
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhe Chen
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chang She
- grid.452666.50000 0004 1762 8363Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Chen
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Cao
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang-yang Xu
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
193
|
Anandhan A, Nguyen N, Syal A, Dreher LA, Dodson M, Zhang DD, Madhavan L. NRF2 Loss Accentuates Parkinsonian Pathology and Behavioral Dysfunction in Human α-Synuclein Overexpressing Mice. Aging Dis 2021; 12:964-982. [PMID: 34221542 PMCID: PMC8219498 DOI: 10.14336/ad.2021.0511] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (NRF2) is a central regulator of cellular stress responses and its transcriptional activation promotes multiple cellular defense and survival mechanisms. The loss of NRF2 has been shown to increase oxidative and proteotoxic stress, two key pathological features of neurodegenerative disorders such as Parkinson's disease (PD). Moreover, compromised redox homeostasis and protein quality control can cause the accumulation of pathogenic proteins, including alpha-synuclein (α-Syn) which plays a key role in PD. However, despite this link, the precise mechanisms by which NRF2 may regulate PD pathology is not clear. In this study, we generated a humanized mouse model to study the importance of NRF2 in the context of α-Syn-driven neuropathology in PD. Specifically, we developed NRF2 knockout and wild-type mice that overexpress human α-Syn (hα-Syn+/Nrf2-/- and hα-Syn+/Nrf2+/+ respectively) and tested changes in their behavior through nest building, challenging beam, and open field tests at three months of age. Cellular and molecular alterations in α-Syn, including phosphorylation and subsequent oligomerization, as well as changes in oxidative stress, inflammation, and autophagy were also assessed across multiple brain regions. It was observed that although monomeric α-Syn levels did not change, compared to their wild-type counterparts, hα-Syn+/Nrf2-/- mice exhibited increased phosphorylation and oligomerization of α-Syn. This was associated with a loss of tyrosine hydroxylase expressing dopaminergic neurons in the substantia nigra, and more pronounced behavioral deficits reminiscent of early-stage PD, in the hα-Syn+/Nrf2-/- mice. Furthermore, hα-Syn+/Nrf2-/- mice showed significantly amplified oxidative stress, greater expression of inflammatory markers, and signs of increased autophagic burden, especially in the midbrain, striatum and cortical brain regions. These results support an important role for NRF2, early in PD progression. More broadly, it indicates NRF2 biology as fundamental to PD pathogenesis and suggests that targeting NRF2 activation may delay the onset and progression of PD.
Collapse
Affiliation(s)
- Annadurai Anandhan
- Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
| | - Nhat Nguyen
- Physiology Undergraduate Program, Tucson, AZ, USA.
| | - Arjun Syal
- Neuroscience and Cognitive Science Undergraduate Program, Tucson, AZ, USA.
| | - Luke A Dreher
- Ecology and Evolutionary Biology Undergraduate Program, Tucson, AZ, USA.
| | - Matthew Dodson
- Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
| | - Donna D Zhang
- Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
| | - Lalitha Madhavan
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- Evelyn F McKnight Brain Institute and Bio5 Institute, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
194
|
Weinhouse C. The roles of inducible chromatin and transcriptional memory in cellular defense system responses to redox-active pollutants. Free Radic Biol Med 2021; 170:85-108. [PMID: 33789123 PMCID: PMC8382302 DOI: 10.1016/j.freeradbiomed.2021.03.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/17/2022]
Abstract
People are exposed to wide range of redox-active environmental pollutants. Air pollution, heavy metals, pesticides, and endocrine disrupting chemicals can disrupt cellular redox status. Redox-active pollutants in our environment all trigger their own sets of specific cellular responses, but they also activate a common set of general stress responses that buffer the cell against homeostatic insults. These cellular defense system (CDS) pathways include the heat shock response, the oxidative stress response, the hypoxia response, the unfolded protein response, the DNA damage response, and the general stress response mediated by the stress-activated p38 mitogen-activated protein kinase. Over the past two decades, the field of environmental epigenetics has investigated epigenetic responses to environmental pollutants, including redox-active pollutants. Studies of these responses highlight the role of chromatin modifications in controlling the transcriptional response to pollutants and the role of transcriptional memory, often referred to as "epigenetic reprogramming", in predisposing previously exposed individuals to more potent transcriptional responses on secondary challenge. My central thesis in this review is that high dose or chronic exposure to redox-active pollutants leads to transcriptional memories at CDS target genes that influence the cell's ability to mount protective responses. To support this thesis, I will: (1) summarize the known chromatin features required for inducible gene activation; (2) review the known forms of transcriptional memory; (3) discuss the roles of inducible chromatin and transcriptional memory in CDS responses that are activated by redox-active environmental pollutants; and (4) propose a conceptual framework for CDS pathway responsiveness as a readout of total cellular exposure to redox-active pollutants.
Collapse
Affiliation(s)
- Caren Weinhouse
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, 97214, USA.
| |
Collapse
|
195
|
Calabrese EJ, Kozumbo WJ, Kapoor R, Dhawan G, Lara PC, Giordano J. Nrf2 activation putatively mediates clinical benefits of low-dose radiotherapy in COVID-19 pneumonia and acute respiratory distress syndrome (ARDS): Novel mechanistic considerations. Radiother Oncol 2021; 160:125-131. [PMID: 33932453 PMCID: PMC8080499 DOI: 10.1016/j.radonc.2021.04.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022]
Abstract
Novel mechanistic insights are discussed herein that link a single, nontoxic, low-dose radiotherapy (LDRT) treatment (0.5-1.0 Gy) to (1) beneficial subcellular effects mediated by the activation of nuclear factor erythroid 2-related transcription factor (Nrf2) and to (2) favorable clinical outcomes for COVID-19 pneumonia patients displaying symptoms of acute respiratory distress syndrome (ARDS). We posit that the favorable clinical outcomes following LDRT result from potent Nrf2-mediated antioxidant responses that rebalance the oxidatively skewed redox states of immunological cells, driving them toward anti-inflammatory phenotypes. Activation of Nrf2 by ionizing radiation is highly dose dependent and conforms to the features of a biphasic (hormetic) dose-response. At the cellular and subcellular levels, hormetic doses of <1.0 Gy induce polarization shifts in the predominant population of lung macrophages, from an M1 pro-inflammatory to an M2 anti-inflammatory phenotype. Together, the Nrf2-mediated antioxidant responses and the subsequent shifts to anti-inflammatory phenotypes have the capacity to suppress cytokine storms, resolve inflammation, promote tissue repair, and prevent COVID-19-related mortality. Given these mechanistic considerations-and the historical clinical success of LDRT early in the 20th century-we opine that LDRT should be regarded as safe and effective for use at almost any stage of COVID-19 infection. In theory, however, optimal life-saving potential is thought to occur when LDRT is applied prior to the cytokine storms and before the patients are placed on mechanical oxygen ventilators. The administration of LDRT either as an intervention of last resort or too early in the disease progression may be far less effective in saving the lives of ARDS patients.
Collapse
Affiliation(s)
| | | | - Rachna Kapoor
- Saint Francis Hospital and Medical Center, Hartford, USA
| | - Gaurav Dhawan
- Sri Guru Ram Das University of Health Sciences, Amritsar, India.
| | - Pedro C Lara
- Department of Radiation Oncology, Hospital Universitario San Roque, Universidad Fernando Pessoa Canarias, Las Palmas Gran Canaria, Spain.
| | - James Giordano
- Departments of Neurology and Biochemistry, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
196
|
Nrf2 as a potential target for Parkinson's disease therapy. J Mol Med (Berl) 2021; 99:917-931. [PMID: 33844027 DOI: 10.1007/s00109-021-02071-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/28/2021] [Accepted: 03/29/2021] [Indexed: 02/08/2023]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder featuring both motor and nonmotor symptoms associated with a progressive loss of dopaminergic neurons in the substantia nigra pars compacta. Conventionally, PD treatment options have focused on dopamine replacement and provide only symptomatic relief. However, disease-modifying therapies are still unavailable. Mechanistically, genetic and environmental factors can produce oxidative stress which has been implicated as a core contributor to the initiation and progression of PD through the degeneration of dopaminergic neurons. Importantly, nuclear factor erythroid 2-related factor 2 (Nrf2) is essential for maintaining redox homeostasis by binding to the antioxidant response element which exists in the promoter regions of most genes coding for antioxidant enzymes. Furthermore, protein kinase C, mitogen-activated protein kinases, and phosphotidylinositol 3-kinase have been implicated in the regulation of Nrf2 activity during PD. Here, we review the evidence supporting the regulation of Nrf2 through Keap1-dependent and Keap1-independent mechanisms. We also address that targeting Nrf2 may provide a therapeutic option to mitigate oxidative stress-associated PD. Finally, we discuss currently known classes of small molecule activators of Nrf2, including Nrf2-activating compounds in PD.
Collapse
|
197
|
Piloni NE, Vargas R, Fernández V, Videla LA, Puntarulo S. Effects of acute iron overload on Nrf2-related glutathione metabolism in rat brain. Biometals 2021; 34:1017-1027. [PMID: 34136984 DOI: 10.1007/s10534-021-00324-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/08/2021] [Indexed: 02/08/2023]
Abstract
Iron (Fe) overload triggers free radical production and lipid peroxidation processes that may lead to cell death (ferroptosis). The hypothesis of this work was that acute Fe-dextran treatment triggers Nrf2-mediated antioxidant regulation in rat brain involving glutathione (GSH) metabolism. Over the initial 8 h after Fe-dextran administration (single dose of 500 mg Fe-dextran/kg), total Fe, malondialdehyde (MDA) content, glutathione peroxidase (GPx), GPx-Se dependent (GPx-Se) and glutathione S-transferases (GST) activities were increased in rat whole brain. The content of GSH and the activity of glutathione reductase (GR) showed decreases (p < 0.05) after 6 and 8 h post injection in cortex. A significant increase in nuclear Nrf2 protein levels over control values was achieved after 6 h of Fe-dextran administration, while no significant differences were observed in the cytosolic fraction. Nuclear Nrf2/cytosolic Nrf2 ratios showed enhancement (p < 0.05) after 6 h of Fe overload, suggesting a greater translocation of the factor to the nucleus. No significant differences were observed in the expression of Keap1 in nuclear or cytosolic extracts. It is concluded that acute Fe overload induces oxidative stress in rat brain with the concomitant lipid peroxidation increase and GSH depletion, leading to the elevation of Nrf2-controlled GPx, GPx-Se and GST protein expression as a protective adaptive response. Further studies are required to fully comprehend the complex network of interrelated processes keeping the balance of GSH functions as chelator, antioxidant and redox buffer in the understanding of the ferroptotic and hormetic mechanisms triggered by Fe overload in brain.
Collapse
Affiliation(s)
- Natacha E Piloni
- Fisicoquímica-IBIMOL, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, CAAD1113, Buenos Aires, Argentina.,Instituto de Bioquímica y Medicina Molecular (IBIMOL), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Romina Vargas
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Virginia Fernández
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Susana Puntarulo
- Fisicoquímica-IBIMOL, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, CAAD1113, Buenos Aires, Argentina. .,Instituto de Bioquímica y Medicina Molecular (IBIMOL), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
198
|
Dodson M, Anandhan A, Zhang DD, Madhavan L. An NRF2 Perspective on Stem Cells and Ageing. FRONTIERS IN AGING 2021; 2:690686. [PMID: 36213179 PMCID: PMC9536878 DOI: 10.3389/fragi.2021.690686] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/03/2021] [Indexed: 04/24/2023]
Abstract
Redox and metabolic mechanisms lie at the heart of stem cell survival and regenerative activity. NRF2 is a major transcriptional controller of cellular redox and metabolic homeostasis, which has also been implicated in ageing and lifespan regulation. However, NRF2's role in stem cells and their functioning with age is only just emerging. Here, focusing mainly on neural stem cells, which are core to adult brain plasticity and function, we review recent findings that identify NRF2 as a fundamental player in stem cell biology and ageing. We also discuss NRF2-based molecular programs that may govern stem cell state and function with age, and implications of this for age-related pathologies.
Collapse
Affiliation(s)
- Matthew Dodson
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
| | - Annadurai Anandhan
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
- Department of Neurology, University of Arizona, Tucson, AZ, United States
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
| | - Lalitha Madhavan
- Department of Neurology, University of Arizona, Tucson, AZ, United States
- Evelyn F. McKnight Brain Institute and Bio5 Institute, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
199
|
Luu Quoc Q, Cao Thi Bich T, Kim SH, Park HS, Shin YS. Administration of vitamin E attenuates airway inflammation through restoration of Nrf2 in a mouse model of asthma. J Cell Mol Med 2021; 25:6721-6732. [PMID: 34089243 PMCID: PMC8278095 DOI: 10.1111/jcmm.16675] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 04/16/2021] [Accepted: 05/10/2021] [Indexed: 12/21/2022] Open
Abstract
Accumulating evidence reveals that ROS is one of the key mediators that contribute to the development of asthma. Studies on antioxidants have shown to have beneficial effects on asthma management. However, we still do not know the precise mechanism, and the effects depend on age. This study was conducted to assess the levels of ROS and the effect of antioxidants in younger and older mice using an eosinophilic asthma model. We analyzed airway hyperresponsiveness (AHR), cytokines in bronchoalveolar lavage fluid (BALF), inflammatory cell counts, and the expression levels of NFκB, Nrf2, EPx, and EDN in the lung tissue, as well as the level of ROS in the lung tissue and BALF. The degree of eosinophilia and the levels of IL-5, ROS, and NFκB were significantly increased, whereas the endogenous levels of vitamin E and Nrf2 were decreased in the lung and BALF in the older mice compared to younger mice. The administration of vitamin E attenuated AHR, airway inflammation, and the level of IL-13 and ROS and enhanced the Nrf2 level in the older mice compared to the younger mice. Taken together, vitamin E treatment may have the therapeutic potential through restoration of the Nrf2 level, especially in elderly asthma.
Collapse
Affiliation(s)
- Quang Luu Quoc
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | - Tra Cao Thi Bich
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | - Seo-Hee Kim
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | - Yoo Seob Shin
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
200
|
Anandhan A, Kirwan KR, Corenblum MJ, Madhavan L. Enhanced NRF2 expression mitigates the decline in neural stem cell function during aging. Aging Cell 2021; 20:e13385. [PMID: 34128307 PMCID: PMC8208782 DOI: 10.1111/acel.13385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/10/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
Although it is known that aging affects neural stem progenitor cell (NSPC) biology in fundamental ways, the underlying dynamics of this process are not fully understood. Our previous work identified a specific critical period (CP) of decline in NSPC activity and function during middle age (13–15 months), and revealed the reduced expression of the redox‐sensitive transcription factor, NRF2, as a key mediator of this process. Here, we investigated whether augmenting NRF2 expression could potentially mitigate the NSPC decline across the identified CP. NRF2 expression in subventricular zone (SVZ) NSPCs was upregulated via GFP tagged recombinant adeno‐associated viral vectors (AAV‐NRF2‐eGFP), and its cellular and behavioral effects compared to animals that received control vectors (AAV‐eGFP). The vectors were administered into the SVZs of aging rats, at time points either before or after the CP. Results indicate that animals that had received AAV‐NRF2‐eGFP, prior to the CP (11 months of age), exhibited substantially improved behavioral function (fine olfactory discrimination and motor tasks) in comparison to those receiving control viruses. Further analysis revealed that NSPC proliferation, self‐renewal, neurogenesis, and migration to the olfactory bulb had significantly increased upon NRF2 upregulation. On the other hand, increasing NRF2 after the CP (at 20 months of age) produced no notable changes in NSPC activity at either cellular or behavioral levels. These results, for the first time, indicate NRF2 pathway modulation as a means to support NSPC function with age and highlight a critical time‐dependency for activating NRF2 to enhance NSPC function.
Collapse
Affiliation(s)
- Annadurai Anandhan
- Department of Neurology University of Arizona Tucson AZ USA
- Pharmacology and Toxicology University of Arizona Tucson AZ USA
| | - Konner R. Kirwan
- Neuroscience and Cognitive Science Undergraduate Program Tucson AZ USA
| | | | - Lalitha Madhavan
- Department of Neurology University of Arizona Tucson AZ USA
- Bio5 Institute University of ArizonaTucsonAZUSA
- Evelyn F McKnight Brain Institute University of Arizona Tucson AZ USA
| |
Collapse
|