1
|
Zelelew D, Endale M, Melaku Y, Geremew T, Eswaramoorthy R, Tufa LT, Choi Y, Lee J. Ultrasonic-Assisted Synthesis of Heterocyclic Curcumin Analogs as Antidiabetic, Antibacterial, and Antioxidant Agents Combined with in vitro and in silico Studies. Adv Appl Bioinform Chem 2023; 16:61-91. [PMID: 37533689 PMCID: PMC10392906 DOI: 10.2147/aabc.s403413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/11/2023] [Indexed: 08/04/2023] Open
Abstract
Background Heterocyclic analogs of curcumin have a wide range of therapeutic potential and the ability to control the activity of a variety of metabolic enzymes. Methods 1H-NMR and 13C-NMR spectroscopic techniques were used to determine the structures of synthesized compounds. The agar disc diffusion method and α-amylase inhibition assay were used to examine the antibacterial and anti-diabetic potential of the compounds against α-amylase enzyme inhibitory activity, respectively. DPPH-free radical scavenging and lipid peroxidation inhibition assays were used to assess the in vitro antioxidant potential. Results and Discussion In this work, nine heterocyclic analogs derived from curcumin precursors under ultrasonic irradiation were synthesized in excellent yields (81.4-93.7%) with improved reaction time. Results of antibacterial activities revealed that compounds 8, and 11 displayed mean inhibition zone of 13.00±0.57, and 19.66±00 mm, respectively, compared to amoxicillin (12.87±1.41 mm) at 500 μg/mL against E. coli, while compounds 8, 11 and 16 displayed mean inhibition zone of 17.67±0.57, 14.33±0.57 and 23.33±00 mm, respectively, compared to amoxicillin (13.75±1.83 mm) at 500 μg/mL against P. aeruginosa. Compound 11 displayed a mean inhibition zone of 11.33±0.57 mm compared to amoxicillin (10.75±1.83 mm) at 500 μg/mL against S. aureus. Compound 11 displayed higher binding affinities of -7.5 and -8.3 Kcal/mol with penicillin-binding proteins (PBPs) and β-lactamases producing bacterial strains, compared to amoxicillin (-7.2 and -7.9 Kcal/mol, respectively), these results are in good agreement with the in vitro antibacterial activities. In vitro antidiabetic potential on α-amylase enzyme revealed that compounds 11 (IC50=7.59 µg/mL) and 16 (IC50=4.08 µg/mL) have higher inhibitory activities than acarbose (IC50=8.0 µg/mL). Compound 8 showed promising antioxidant inhibition efficacy of DPPH (IC50 = 2.44 g/mL) compared to ascorbic acid (IC50=1.24 g/mL), while compound 16 revealed 89.9±20.42% inhibition of peroxide generation showing its potential in reducing the development of lipid peroxides. In silico molecular docking analysis, results are in good agreement with in vitro biological activity. In silico ADMET profiles suggested the adequate oral drug-likeness potential of the compounds without adverse effects. Conclusion According to our findings, both biological activities and in silico computational studies results demonstrated that compounds 8, 11, and 16 are promising α-amylase inhibitors and antibacterial agents against E. coli, P. aeruginosa, and S. aureus, whereas compound 8 was found to be a promising antioxidant agent.
Collapse
Affiliation(s)
- Demis Zelelew
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
| | - Milkyas Endale
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
| | - Yadessa Melaku
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
| | - Teshome Geremew
- Department of Applied Biology, School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
| | | | - Lemma Teshome Tufa
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
- Research Institute of Materials Chemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Youngeun Choi
- Department of Chemistry, Department of Chemistry Engineering and Applied Chemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Jaebeom Lee
- Department of Chemistry, Department of Chemistry Engineering and Applied Chemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| |
Collapse
|
2
|
Peroxiredoxin-5 Knockdown Accelerates Pressure Overload-Induced Cardiac Hypertrophy in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5067544. [PMID: 35132351 PMCID: PMC8817848 DOI: 10.1155/2022/5067544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/21/2021] [Indexed: 11/29/2022]
Abstract
A recent study showed that peroxiredoxins (Prxs) play an important role in the development of pathological cardiac hypertrophy. However, the involvement of Prx5 in cardiac hypertrophy remains unclear. Therefore, this study is aimed at investigating the role and mechanisms of Prx5 in pathological cardiac hypertrophy and dysfunction. Transverse aortic constriction (TAC) surgery was performed to establish a pressure overload-induced cardiac hypertrophy model. In this study, we found that Prx5 expression was upregulated in hypertrophic hearts and cardiomyocytes. In addition, Prx5 knockdown accelerated pressure overload-induced cardiac hypertrophy and dysfunction in mice by activating oxidative stress and cardiomyocyte apoptosis. Importantly, heart deterioration caused by Prx5 knockdown was related to mitogen-activated protein kinase (MAPK) pathway activation. These findings suggest that Prx5 could be a novel target for treating cardiac hypertrophy and heart failure.
Collapse
|
3
|
Agborbesong E, Zhou JX, Li LX, Calvet JP, Li X. Antioxidant enzyme peroxiredoxin 5 regulates cyst growth and ciliogenesis via modulating Plk1 stability. FASEB J 2022; 36:e22089. [PMID: 34888938 PMCID: PMC9060392 DOI: 10.1096/fj.202101270rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023]
Abstract
Oxidative stress is emerging as a contributing factor to the homeostasis in cystic diseases. However, the role antioxidant enzymes play in the pathogenesis of autosomal dominant polycystic kidney disease (ADPKD) remains elusive. Peroxiredoxin 5 (Prdx5) is an antioxidant enzyme that catalyzes the reduction of H2 O2 and alkyl hydroperoxide and plays an important role in different biological processes. In this study, we show that Prdx5 is downregulated in a PKD mutant mouse model and ADPKD patient kidneys. Knockdown of Prdx5 resulted in the formation of cysts in a three-dimensional mouse inner medullar collecting duct (IMCD) cell Matrigel culture system. The mechanisms of Prdx5 deficiency mediated cyst growth include: (1) induction of oxidative stress as indicated by increased mRNA expression of heme oxygenase-1, an oxidant stress marker; (2) activation of Erk, S6 and mTORC1, which contribute to cystic renal epithelial cell proliferation and cyst growth; (3) abnormal centrosome amplification and multipolar spindle formation which result in genome instability; (4) upregulation of Polo-like kinase 1 (Plk1) and Aurora kinase A, important mitotic kinases involved in cell proliferation and ciliogenesis; (5) impaired formation of primary cilia in mouse IMCD3 and retinal pigment epithelial cells, which could be rescued by inhibiting Plk1 activity; and (6) restraining the effect of Wnt3a and Wnt5a ligands on primary cilia in mouse IMCD3 cells, while regulating the activity of the canonical and non-canonical Wnt signaling in a separate cilia independent mechanism, respectively. Importantly, we found that targeting Plk1 with its inhibitor, volasertib, delayed cyst growth in Pkd1 conditional knockout mouse kidneys. Together, these findings indicate that Prdx5 is an important antioxidant that regulates cyst growth via diverse mechanisms, in particular, the Prdx5-Plk1 axis, and that induction and activation of Prdx5, alone or together with inhibition of Plk1, represent a promising strategy for combatting ADPKD.
Collapse
Affiliation(s)
- Ewud Agborbesong
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Julie Xia Zhou
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Linda Xiaoyan Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - James P. Calvet
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
4
|
Tiklová K, Gillberg L, Volakakis N, Lundén-Miguel H, Dahl L, Serrano GE, Adler CH, Beach TG, Perlmann T. Disease Duration Influences Gene Expression in Neuromelanin-Positive Cells From Parkinson's Disease Patients. Front Mol Neurosci 2021; 14:763777. [PMID: 34867188 PMCID: PMC8632647 DOI: 10.3389/fnmol.2021.763777] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
Analyses of gene expression in cells affected by neurodegenerative disease can provide important insights into disease mechanisms and relevant stress response pathways. Major symptoms in Parkinson’s disease (PD) are caused by the degeneration of midbrain dopamine (mDA) neurons within the substantia nigra. Here we isolated neuromelanin-positive dopamine neurons by laser capture microdissection from post-mortem human substantia nigra samples recovered at both early and advanced stages of PD. Neuromelanin-positive cells were also isolated from individuals with incidental Lewy body disease (ILBD) and from aged-matched controls. Isolated mDA neurons were subjected to genome-wide gene expression analysis by mRNA sequencing. The analysis identified hundreds of dysregulated genes in PD. Results showed that mostly non-overlapping genes were differentially expressed in ILBD, subjects who were early after diagnosis (less than five years) and those autopsied at more advanced stages of disease (over five years since diagnosis). The identity of differentially expressed genes suggested that more resilient, stably surviving DA neurons were enriched in samples from advanced stages of disease, either as a consequence of positive selection of a less vulnerable long-term surviving mDA neuron subtype or due to up-regulation of neuroprotective gene products.
Collapse
Affiliation(s)
- Katarína Tiklová
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.,Ludwig Institute for Cancer Research, Stockholm, Sweden
| | - Linda Gillberg
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.,Ludwig Institute for Cancer Research, Stockholm, Sweden
| | | | | | - Lina Dahl
- Ludwig Institute for Cancer Research, Stockholm, Sweden
| | - Geidy E Serrano
- Banner Sun Health Research Institute, Sun City, AZ, United States
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, United States
| | - Thomas G Beach
- Banner Sun Health Research Institute, Sun City, AZ, United States
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Role of the Redox State of Human Peroxiredoxin-5 on Its TLR4-Activating DAMP Function. Antioxidants (Basel) 2021; 10:antiox10121902. [PMID: 34943005 PMCID: PMC8750366 DOI: 10.3390/antiox10121902] [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: 11/12/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Human peroxiredoxin-5 (PRDX5) is a unique redox-sensitive protein that plays a dual role in brain ischemia-reperfusion injury. While intracellular PRDX5 has been reported to act as a neuroprotective antioxidative enzyme by scavenging peroxides, once released extracellularly from necrotic brain cells, the protein aggravates neural cell death by inducing expression of proinflammatory cytokines in macrophages through activation of Toll-like receptor (TLR) 2 (TLR2) and 4 (TLR4). Although recent evidence showed that PRDX5 was able to interact directly with TLR4, little is known regarding the role of the cysteine redox state of PRDX5 on its DAMP function. To gain insights into the role of PRDX5 redox-active cysteine residues in the TLR4-dependent proinflammatory activity of the protein, we used a recombinant human PRDX5 in the disulfide (oxidized) form and a mutant version lacking the peroxidatic cysteine, as well as chemically reduced and hyperoxidized PRDX5 proteins. We first analyzed the oxidation state and oligomerization profile by Western blot, mass spectrometry, and SEC-MALS. Using ELISA, we demonstrate that the disulfide bridge between the enzymatic cysteines is required to allow improved TLR4-dependent IL-8 secretion. Moreover, single-molecule force spectroscopy experiments revealed that TLR4 alone is not sufficient to discriminate the different PRDX5 redox forms. Finally, flow cytometry binding assays show that disulfide PRDX5 has a higher propensity to bind to the surface of living TLR4-expressing cells than the mutant protein. Taken together, these results demonstrate the importance of the redox state of PRDX5 cysteine residues on TLR4-induced inflammation.
Collapse
|
6
|
Chhunchha B, Kubo E, Kompella UB, Singh DP. Engineered Sumoylation-Deficient Prdx6 Mutant Protein-Loaded Nanoparticles Provide Increased Cellular Defense and Prevent Lens Opacity. Antioxidants (Basel) 2021; 10:antiox10081245. [PMID: 34439493 PMCID: PMC8389307 DOI: 10.3390/antiox10081245] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Aberrant Sumoylation-mediated protein dysfunction is involved in a variety of oxidative and aging pathologies. We previously reported that Sumoylation-deficient Prdx6K(lysine)122/142R(Arginine) linked to the TAT-transduction domain gained stability and protective efficacy. In the present study, we formulated wild-type TAT-HA-Prdx6WT and Sumoylation-deficient Prdx6-loaded poly-lactic-co-glycolic acid (PLGA) nanoparticles (NPs) to further enhance stability, protective activities, and sustained delivery. We found that in vitro and subconjuctival delivery of Sumoylation-deficient Prdx6-NPs provided a greater protection of lens epithelial cells (LECs) derived from human and Prdx6-/--deficient mouse lenses against oxidative stress, and it also delayed the lens opacity in Shumiya cataract rats (SCRs) than TAT-HA-Prdx6WT-NPs. The encapsulation efficiencies of TAT-HA-Prdx6-NPs were ≈56%-62%. Dynamic light scattering (DLS) and atomic force microscopy (AFM) analyses showed that the NPs were spherical, with a size of 50-250 nm and a negative zeta potential (≈23 mV). TAT-HA-Prdx6 analog-NPs released bioactive TAT-HA-Prdx6 (6%-7%) within 24 h. Sumoylation-deficient TAT-HA-Prdx6-NPs provided 35% more protection by reducing the oxidative load of LECs exposed to H2O2 compared to TAT-HA-Prdx6WT-NPs. A subconjuctival delivery of TAT-HA-Prdx6 analog-NPs demonstrated that released TAT-HA-Prdx6K122/142R could reduce lens opacity by ≈60% in SCRs. Collectively, our results demonstrate for the first time that the subconjuctival delivery of Sumoylation-deficient Prdx6-NPs is efficiently cytoprotective and provide a proof of concept for potential use to delay cataract and oxidative-related pathobiology in general.
Collapse
Affiliation(s)
- Bhavana Chhunchha
- Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence: (B.C.); (D.P.S.)
| | - Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 9200265, Ishikawa, Japan;
| | - Uday B. Kompella
- Departments of Pharmaceutical Sciences, Ophthalmology, and Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Dhirendra P. Singh
- Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence: (B.C.); (D.P.S.)
| |
Collapse
|
7
|
Dincel ED, Gürsoy E, Yilmaz-Ozden T, Ulusoy-Güzeldemirci N. Antioxidant activity of novel imidazo[2,1-b]thiazole derivatives: Design, synthesis, biological evaluation, molecular docking study and in silico ADME prediction. Bioorg Chem 2020; 103:104220. [DOI: 10.1016/j.bioorg.2020.104220] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
|
8
|
Peroxiredoxin 5 Silencing Sensitizes Dopaminergic Neuronal Cells to Rotenone via DNA Damage-Triggered ATM/p53/PUMA Signaling-Mediated Apoptosis. Cells 2019; 9:cells9010022. [PMID: 31861721 PMCID: PMC7016837 DOI: 10.3390/cells9010022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
Peroxiredoxins (Prxs) are a family of thioredoxin peroxidases. Accumulating evidence suggests that changes in the expression of Prxs may be involved in neurodegenerative diseases pathology. However, the expression and function of Prxs in Parkinson’s disease (PD) remains unclear. Here, we showed that Prx5 was the most downregulated of the six Prx subtypes in dopaminergic (DA) neurons in rotenone-induced cellular and rat models of PD, suggesting possible roles in regulating their survival. Depletion of Prx5 sensitized SH-SY5Y DA neuronal cells to rotenone-induced apoptosis. The extent of mitochondrial membrane potential collapse, cytochrome c release, and caspase activation was increased by Prx5 loss. Furthermore, Prx5 knockdown enhanced the induction of PUMA by rotenone through a p53-dependent mechanism. Using RNA interference approaches, we demonstrated that the p53/PUMA signaling was essential for Prx5 silencing-exacerbated mitochondria-driven apoptosis. Additionally, downregulation of Prx5 augmented rotenone-induced DNA damage manifested as induction of phosphorylated histone H2AX (γ-H2AX) and activation of ataxia telangiectasia mutated (ATM) kinase. The pharmacological inactivation of ATM revealed that ATM was integral to p53 activation by DNA damage. These findings provided a novel link between Prx5 and DNA damage-triggered ATM/p53/PUMA signaling in a rotenone-induced PD model. Thus, Prx5 might play an important role in protection against rotenone-induced DA neurodegeneration.
Collapse
|
9
|
Kang JB, Park DJ, Koh PO. Identification of proteins differentially expressed by glutamate treatment in cerebral cortex of neonatal rats. Lab Anim Res 2019; 35:24. [PMID: 32257912 PMCID: PMC7081608 DOI: 10.1186/s42826-019-0026-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 11/12/2019] [Indexed: 11/22/2022] Open
Abstract
Glutamate leads to neuronal cell damage by generating neurotoxicity during brain development. The objective of this study is to identify proteins that differently expressed by glutamate treatment in neonatal cerebral cortex. Sprague-Dawley rat pups (post-natal day 7) were intraperitoneally injected with vehicle or glutamate (10 mg/kg). Brain tissues were isolated 4 h after drug treatment and fixed for morphological study. Moreover, cerebral cortices were collected for protein study. Two-dimensional gel electrophoresis and mass spectrometry were carried out to identify specific proteins. We observed severe histopathological changes in glutamate-exposed cerebral cortex. We identified various proteins that differentially expressed by glutamate exposure. Identified proteins were thioredoxin, peroxiredoxin 5, ubiquitin carboxy-terminal hydrolase L1, proteasome subunit alpha proteins, isocitrate dehydrogenase, and heat shock protein 60. Heat shock protein 60 was increased in glutamate exposed condition. However, other proteins were decreased in glutamate-treated animals. These proteins are related to anti-oxidant, protein degradation, metabolism, signal transduction, and anti-apoptotic function. Thus, our findings can suggest that glutamate leads to neonatal cerebral cortex damage by regulation of specific proteins that mediated with various functions.
Collapse
Affiliation(s)
- Ju-Bin Kang
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| | - Dong-Ju Park
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| | - Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju, 52828 South Korea
| |
Collapse
|
10
|
Abruzzo PM, Matté A, Bolotta A, Federti E, Ghezzo A, Guarnieri T, Marini M, Posar A, Siciliano A, De Franceschi L, Visconti P. Plasma peroxiredoxin changes and inflammatory cytokines support the involvement of neuro-inflammation and oxidative stress in Autism Spectrum Disorder. J Transl Med 2019; 17:332. [PMID: 31578139 PMCID: PMC6775664 DOI: 10.1186/s12967-019-2076-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/21/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND It has been established that children with Autism Spectrum Disorders (ASD) are affected by oxidative stress, the origin of which is still under investigation. In the present work, we evaluated inflammatory and pro-oxidant soluble signature in non-syndromic ASD and age-matched typically developing (TD) control children. METHODS We analyzed leukocyte gene expression of inflammatory cytokines and inflammation/oxidative-stress related molecules in 21 ASD and 20 TD children. Moreover, in another-comparable-group of non-syndromic ASD (N = 22) and TD (N = 21) children, we analyzed for the first time the protein expression of the four members of the antioxidant enzyme family of peroxiredoxins (Prx) in both erythrocyte membranes and in plasma. RESULTS The gene expression of IL6 and of HSP70i, a stress protein, was increased in ASD children. Moreover, gene expression of many inflammatory cytokines and inflammation/oxidative stress-related proteins correlated with clinical features, and appeared to be linked by a complex network of inter-correlations involving the Aryl Hydrocarbon Receptor signaling pathway. In addition, when the study of inter-correlations within the expression pattern of these molecules was extended to include the healthy subjects, the intrinsic physiological relationships of the inflammatory/oxidative stress network emerged. Plasma levels of Prx2 and Prx5 were remarkably increased in ASD compared to healthy controls, while no significant differences were found in red cell Prx levels. CONCLUSIONS Previous findings reported elevated inflammatory cytokines in the plasma of ASD children, without clearly pointing to the presence of neuro-inflammation. On the other hand, the finding of microglia activation in autoptic specimens was clearly suggesting the presence of neuro-inflammation in ASD. Given the role of peroxiredoxins in the protection of brain cells against oxidative stress, the whole of our results, using peripheral data collected in living patients, support the involvement of neuro-inflammation in ASD, and generate a rational for neuro-inflammation as a possible therapeutic target and for plasma Prx5 as a novel indicator of ASD severity.
Collapse
Affiliation(s)
- P M Abruzzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, Bologna, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Via A. Capecelatro, 66, 20148, Milan, Italy
| | - A Matté
- Department of Medicine, University of Verona Medical School, Verona, Italy
| | - A Bolotta
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, Bologna, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Via A. Capecelatro, 66, 20148, Milan, Italy
| | - E Federti
- Department of Medicine, University of Verona Medical School, Verona, Italy
| | - A Ghezzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, Bologna, Italy
| | - T Guarnieri
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - M Marini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, Bologna, Italy. .,IRCCS Fondazione Don Carlo Gnocchi, Via A. Capecelatro, 66, 20148, Milan, Italy.
| | - A Posar
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Ugo Foscolo 7, 40123, Bologna, Italy.,Child Neurology and Psychiatry Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura, 3, 40139, Bologna, Italy
| | - A Siciliano
- Department of Medicine, University of Verona Medical School, Verona, Italy
| | - L De Franceschi
- Department of Medicine, University of Verona Medical School, Verona, Italy
| | - P Visconti
- Child Neurology and Psychiatry Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura, 3, 40139, Bologna, Italy
| |
Collapse
|
11
|
Wagner JT, Knapp MJ, Podrabsky JE. Antioxidant capacity and anoxia-tolerance in Austrofundulus limnaeus embryos. J Exp Biol 2019; 222:jeb.204347. [DOI: 10.1242/jeb.204347] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/28/2019] [Indexed: 12/19/2022]
Abstract
Embryos of Austrofundulus limnaeus can tolerate extreme environmental stresses by entering into a state of metabolic and developmental arrest known as diapause. Oxidative stress is ubiquitous in aerobic organisms and the unique biology and ecology of A. limnaeus likely results in frequent and repeated exposures to oxidative stress during development. Antioxidant capacity of A. limnaeus was explored during development by measuring antioxidant capacity due to small molecules and several enzymatic antioxidant systems. Diapause II embryos can survive for several days in 1% hydrogen peroxide without indications of negative effects. Surprisingly, both small and large molecule antioxidant systems are highest during early development and may be due to maternal provisioning. Antioxidant capacity is largely invested in small molecules during early development and in enzymatic systems during late development. The switch in antioxidant mechanisms and decline in small molecule antioxidants during development correlates with the loss of extreme anoxia tolerance.
Collapse
Affiliation(s)
- Josiah T. Wagner
- Department of Biology, Portland State University, P.O. Box 751, Portland, OR 97207, USA
- Knight Cancer Institute Cancer Early Detection Advanced Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mailcode: KR-CEDR, Portland, OR 97239, USA
| | - Michael J. Knapp
- Department of Biology, Portland State University, P.O. Box 751, Portland, OR 97207, USA
| | - Jason E. Podrabsky
- Department of Biology, Portland State University, P.O. Box 751, Portland, OR 97207, USA
| |
Collapse
|
12
|
Detienne G, De Haes W, Mergan L, Edwards SL, Temmerman L, Van Bael S. Beyond ROS clearance: Peroxiredoxins in stress signaling and aging. Ageing Res Rev 2018; 44:33-48. [PMID: 29580920 DOI: 10.1016/j.arr.2018.03.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 03/21/2018] [Indexed: 12/12/2022]
Abstract
Antioxidants were long predicted to have lifespan-promoting effects, but in general this prediction has not been well supported. While some antioxidants do seem to have a clear effect on longevity, this may not be primarily as a result of their role in the removal of reactive oxygen species, but rather mediated by other mechanisms such as the modulation of intracellular signaling. In this review we discuss peroxiredoxins, a class of proteinaceous antioxidants with redox signaling and chaperone functions, and their involvement in regulating longevity and stress resistance. Peroxiredoxins have a clear role in the regulation of lifespan and survival of many model organisms, including the mouse, Caenorhabditis elegans and Drosophila melanogaster. Recent research on peroxiredoxins - in these models and beyond - has revealed surprising new insights regarding the interplay between peroxiredoxins and longevity signaling, which will be discussed here in detail. As redox signaling is emerging as a potentially important player in the regulation of longevity and aging, increased knowledge of these fascinating antioxidants and their mode(s) of action is paramount.
Collapse
Affiliation(s)
- Giel Detienne
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| | - Wouter De Haes
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| | - Lucas Mergan
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| | - Samantha L Edwards
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| | - Liesbet Temmerman
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| | - Sven Van Bael
- Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| |
Collapse
|
13
|
Shen GN, Liu L, Feng L, Jin Y, Jin MH, Han YH, Jin CH, Jin YZ, Lee DS, Kwon TH, Cui YD, Sun HN. Knockdown of peroxiredoxin V increases glutamate‑induced apoptosis in HT22 hippocampal neuron cells. Mol Med Rep 2018; 17:7827-7834. [PMID: 29620243 DOI: 10.3892/mmr.2018.8826] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/15/2018] [Indexed: 11/06/2022] Open
Abstract
High concentrations of glutamate may mediate neuronal cell apoptosis by increasing intracellular reactive oxygen species (ROS) levels. Peroxiredoxin V (Prx V), a member of the Prx family, serves crucial roles in protecting cells from oxidative stress. The present study investigated the regulatory effect of Prx V on glutamate‑induced effects on viability and apoptosis in HT22 cells. Western blotting was used for protein expression analysis and Annexin V/PI staining and flow cytometry for determination of apoptosis. The results demonstrated that glutamate may ROS‑dependently increase HT22 cell apoptosis and upregulate Prx V protein levels. Furthermore, knockdown of Prx V protein expression with a lentivirus significantly enhanced HT22 cell apoptosis mediated by glutamate, which was reversed by inhibition of ROS with N‑acetyl‑L‑cysteine. Inhibiting the extracellular signal‑regulated kinase (ERK) signaling pathway with PD98059, a specific inhibitor for ERK phosphorylation, markedly decreased glutamate‑induced HT22 cell apoptosis in Prx V knockdown cells, indicating the potential involvement of ERK signaling in glutamate‑induced HT22 cell apoptosis. In addition, an increase in nuclear apoptosis‑inducing factor was observed in Prx V knockdown HT22 cells following glutamate treatment, compared with mock cells, whereas no differences in B‑cell lymphoma‑2 and cleaved‑caspase‑3 protein expression levels were observed between mock and Prx V knockdown cells. The results of the present study indicated that Prx V may have potential as a therapeutic molecular target for glutamate‑induced neuronal cell death and provide novel insight into the role of Prx V in oxidative‑stress induced neuronal cell death.
Collapse
Affiliation(s)
- Gui-Nan Shen
- Department of Disease Model Animal Research Center, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Lei Liu
- Department of Disease Model Animal Research Center, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Li Feng
- Pharmaron Beijing Co., Ltd., Beijing 100176, P.R. China
| | - Yu Jin
- Laboratory of Anatomy and Histology, Yanbian University Health Science Center, Yanji, Jilin 133000, P.R. China
| | - Mei-Hua Jin
- Department of Disease Model Animal Research Center, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hao Han
- Department of Disease Model Animal Research Center, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Cheng-Hao Jin
- Department of Disease Model Animal Research Center, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yong-Zhe Jin
- Laboratory of Anatomy and Histology, Yanbian University Health Science Center, Yanji, Jilin 133000, P.R. China
| | - Dong-Soek Lee
- Laboratory of Molecular Neurobiology, School of Life Sciences, KNU Creative Bio Research Group (BK21 Plus Project), Kyungpook National University, Daegu 41566, Republic of Korea
| | - Tae Ho Kwon
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju, Jeju 63243, Republic of Korea
| | - Yu-Dong Cui
- Department of Disease Model Animal Research Center, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hu-Nan Sun
- Department of Disease Model Animal Research Center, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| |
Collapse
|
14
|
Kubo E, Chhunchha B, Singh P, Sasaki H, Singh DP. Sulforaphane reactivates cellular antioxidant defense by inducing Nrf2/ARE/Prdx6 activity during aging and oxidative stress. Sci Rep 2017; 7:14130. [PMID: 29074861 PMCID: PMC5658327 DOI: 10.1038/s41598-017-14520-8] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/11/2017] [Indexed: 12/21/2022] Open
Abstract
Upon oxidative stress and aging, Nrf2 (NFE2-related factor2) triggers antioxidant defense genes to defends against homeostatic failure. Using human(h) or rat(r) lens epithelial cells (LECs) and aging human lenses, we showed that a progressive increase in oxidative load during aging was linked to a decline in Prdx6 expression. DNA binding experiments using gel-shift and ChIP assays demonstrated a progressive reduction in Nrf2/ARE binding (-357/-349) of Prdx6 promoter. The promoter (-918) with ARE showed a marked reduction in young vs aged hLECs, which was directly correlated to decreased Nrf2/ARE binding. A Nrf2 activator, Sulforaphane (SFN), augmented Prdx6, catalase and GSTπ expression in dose-dependent fashion, and halted Nrf2 dysregulation of these antioxidants. SFN reinforced Nrf2/DNA binding and increased promoter activities by enhancing expression and facilitating Nrf2 translocalization in nucleus. Conversely, promoter mutated at ARE site did not respond to SFN, validating the SFN-mediated restoration of Nrf2/ARE signaling. Furthermore, SFN rescued cells from UVB-induced toxicity in dose-dependent fashion, which was consistent with SFN's dose-dependent activation of Nrf2/ARE interaction. Importantly, knockdown of Prdx6 revealed that Prdx6 expression was prerequisite for SFN-mediated cytoprotection. Collectively, our results suggest that loss of Prdx6 caused by dysregulation of ARE/Nrf2 can be attenuated through a SFN, to combat diseases associated with aging.
Collapse
Affiliation(s)
- Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa, Japan.
| | - Bhavana Chhunchha
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, NE, Omaha, USA
| | - Prerna Singh
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, NE, Omaha, USA
| | - Hiroshi Sasaki
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa, Japan
| | - Dhirendra P Singh
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, NE, Omaha, USA.
| |
Collapse
|
15
|
Park J, Kim B, Chae U, Lee DG, Kam MK, Lee SR, Lee S, Lee HS, Park JW, Lee DS. Peroxiredoxin 5 Decreases Beta-Amyloid-Mediated Cyclin-Dependent Kinase 5 Activation Through Regulation of Ca 2+-Mediated Calpain Activation. Antioxid Redox Signal 2017; 27:715-726. [PMID: 28358580 DOI: 10.1089/ars.2016.6810] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
AIMS Aberrant Cdk5 (cyclin-dependent kinase 5) and oxidative stress are crucial components of diverse neurodegenerative disorders, including Alzheimer's disease (AD). We previously reported that a change in peroxiredoxin (Prx) expression is associated with protection from neuronal death. The aim of the current study was to analyze the role of Prx in regulating Cdk5 activation in AD. RESULTS We found that of the six Prx subtypes, Prx5 was increased the most in cellular (N2a-APPswe cells) model of AD. Prx5 in the brain of APP (amyloid precursor protein) transgenic mouse (Tg2576) was more increased than a nontransgenic mouse. We evaluated Prx5 function by using overexpression (Prx5-WT), a mutation in the catalytic residue (Prx5-C48S), and knockdown. Increased neuronal death and Cdk5 activation by amyloid beta oligomer (AβO) were rescued by Prx5-WT expression, but not by Prx5-C48S or Prx5 knockdown. Prx5 plays a role in Cdk5 regulation by inhibiting the conversion of p35 to p25, which is increased by AβO accumulation. Prx5 is also upregulated in both the cytosol and mitochondria and it protects cells from AβO-mediated oxidative stress by eliminating intracellular and mitochondrial reactive oxygen species. Moreover, Prx5 regulates Ca2+ and Ca2+-mediated calpain activation, which are key regulators of p35 cleavage to p25. Innovation and Conclusion: Our study represents the first demonstration that Prx5 induction is a key factor in the suppression of Cdk5-related neuronal death in AD and we show that it functions via regulation of Ca2+-mediated calpain activation. Antioxid. Redox Signal. 27, 715-726.
Collapse
Affiliation(s)
- Junghyung Park
- 1 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University , Daegu, Republic of Korea.,2 College of Natural Sciences, Kyungpook National University , Daegu, Republic of Korea
| | - Bokyung Kim
- 1 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University , Daegu, Republic of Korea.,2 College of Natural Sciences, Kyungpook National University , Daegu, Republic of Korea
| | - Unbin Chae
- 1 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University , Daegu, Republic of Korea.,2 College of Natural Sciences, Kyungpook National University , Daegu, Republic of Korea
| | - Dong Gil Lee
- 1 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University , Daegu, Republic of Korea.,2 College of Natural Sciences, Kyungpook National University , Daegu, Republic of Korea
| | - Min Kyoung Kam
- 1 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University , Daegu, Republic of Korea.,2 College of Natural Sciences, Kyungpook National University , Daegu, Republic of Korea
| | - Sang-Rae Lee
- 3 National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB) , Cheongju, Republic of Korea
| | - Seunghoon Lee
- 4 Animal Biotechnology Division, National Institute of Animal Science , Jeonju, Republic of Korea
| | - Hyun-Shik Lee
- 1 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University , Daegu, Republic of Korea.,2 College of Natural Sciences, Kyungpook National University , Daegu, Republic of Korea
| | - Jeen-Woo Park
- 1 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University , Daegu, Republic of Korea.,2 College of Natural Sciences, Kyungpook National University , Daegu, Republic of Korea
| | - Dong-Seok Lee
- 1 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University , Daegu, Republic of Korea.,2 College of Natural Sciences, Kyungpook National University , Daegu, Republic of Korea
| |
Collapse
|
16
|
Daher I, Le Dieu-Lugon B, Dourmap N, Lecuyer M, Ramet L, Gomila C, Ausseil J, Marret S, Leroux P, Roy V, El Mestikawy S, Daumas S, Gonzalez B, Leroux-Nicollet I, Cleren C. Magnesium Sulfate Prevents Neurochemical and Long-Term Behavioral Consequences of Neonatal Excitotoxic Lesions: Comparison Between Male and Female Mice. J Neuropathol Exp Neurol 2017; 76:883-897. [PMID: 28922852 DOI: 10.1093/jnen/nlx073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Magnesium sulfate (MgSO4) administration to mothers at risk of preterm delivery is proposed as a neuroprotective strategy against neurological alterations such as cerebral palsy in newborns. However, long-term beneficial or adverse effects of MgSO4 and sex-specific sensitivity remain to be investigated. We conducted behavioral and neurochemical studies of MgSO4 effects in males and females, from the perinatal period to adolescence in a mouse model of cerebral neonatal lesion. The lesion was produced in 5-day-old (P5) pups by ibotenate intracortical injection. MgSO4 (600 mg/kg, i.p.) prior to ibotenate prevented lesion-induced sensorimotor alterations in both sexes at P6 and P7. The lesion increased glutamate level at P10 in the prefrontal cortex, which was prevented by MgSO4 in males. In neonatally lesioned adolescent mice, males exhibited more sequelae than females in motor and cognitive functions. In the perirhinal cortex of adolescent mice, the neonatal lesion induced an increase in vesicular glutamate transporter 1 density in males only, which was negatively correlated with cognitive scores. Long-term sequelae were prevented by neonatal MgSO4 administration. MgSO4 never induced short- or long-term deleterious effect on its own. These results also strongly suggest that sex-specific neuroprotection should be foreseen in preterm infants.
Collapse
Affiliation(s)
- Ismaël Daher
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Bérénice Le Dieu-Lugon
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Nathalie Dourmap
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Matthieu Lecuyer
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Lauriane Ramet
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Cathy Gomila
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Jérôme Ausseil
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Stéphane Marret
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Philippe Leroux
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Vincent Roy
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Salah El Mestikawy
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Stéphanie Daumas
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Bruno Gonzalez
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Isabelle Leroux-Nicollet
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| | - Carine Cleren
- Department of Neonatal Pediatrics and Intensive Care - Neuropediatrics, Normandie Univ, UNIROUEN, INSERM U1245, and Rouen University Hospital, Rouen, France; Normal and Pathological Glutamatergic Systems, Neuroscience Paris Seine, IBPS, INSERM U1130, CNRS UMR 8246 Université Pierre et Marie Curie, Paris, France; INSERM U1088, Laboratoire de Biochimie, Centre de Biologie Humaine, Amiens-Picardie University Hospital, Amiens, France; Normandie Univ, UNIROUEN, PSY-NCA, Rouen, France
| |
Collapse
|
17
|
Chhunchha B, Kubo E, Fatma N, Singh DP. Sumoylation-deficient Prdx6 gains protective function by amplifying enzymatic activity and stability and escapes oxidative stress-induced aberrant Sumoylation. Cell Death Dis 2017; 8:e2525. [PMID: 28055018 PMCID: PMC5386354 DOI: 10.1038/cddis.2016.424] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/17/2016] [Accepted: 11/14/2016] [Indexed: 01/08/2023]
Abstract
Aberrant Sumoylation of protein(s) in response to oxidative stress or during aging is known to be involved in etiopathogenesis of many diseases. Upon oxidative stress, Peroxiredoxin (Prdx) 6 is aberrantly Sumoylated by Sumo1, resulting in loss of functions and cell death. We identified lysines (K) 122 and 142 as the major Sumo1 conjugation sites in Prdx6. Intriguingly, the mutant Prdx6 K122/142 R (arginine) gained protective efficacy, increasing in abundance and promoting glutathione (GSH) peroxidase and acidic calcium-independent phospholipase A2 (aiPLA2) activities. Using lens epithelial cells derived from targeted inactivation of Prdx6−/− gene and relative enzymatic and stability assays, we discovered dramatic increases in GSH-peroxidase (30%) and aiPLA2 (37%) activities and stability in the K122/142 R mutant, suggesting Sumo1 destabilized Prdx6 integrity. Prdx6−/−LECs with EGFP-Sumo1 transduced or co-expressed with mutant TAT-HA-Prdx6K122/142 R or pGFP-Prdx6K122/142 R were highly resistant to oxidative stress, demonstrating mutant protein escaped and interrupted the Prdx6 aberrant Sumoylation-mediated cell death pathway. Mutational analysis of functional sites showed that both peroxidase and PLA2 active sites were necessary for mutant Prdx6 function, and that Prdx6 phosphorylation (at T177 residue) was essential for optimum PLA2 activity. Our work reveals the involvement of oxidative stress-induced aberrant Sumoylation in dysregulation of Prdx6 function. Mutant Prdx6 at its Sumo1 sites escapes and abates this adverse process by maintaining its integrity and gaining function. We propose that the K122/142R mutant of Prdx6 in the form of a TAT-fusion protein may be an easily applicable intervention for pathobiology of cells related to aberrant Sumoylation signaling in aging or oxidative stress.
Collapse
Affiliation(s)
- Bhavana Chhunchha
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa, Ishikawa, Japan
| | - Nigar Fatma
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dhirendra P Singh
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
18
|
Liu DL, Zhao LX, Zhang S, Du JR. Peroxiredoxin 1-mediated activation of TLR4/NF-κB pathway contributes to neuroinflammatory injury in intracerebral hemorrhage. Int Immunopharmacol 2016; 41:82-89. [PMID: 27821296 DOI: 10.1016/j.intimp.2016.10.025] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/09/2016] [Accepted: 10/29/2016] [Indexed: 10/20/2022]
Abstract
The proinflammatory properties of extracellular peroxiredoxins (Prxs) via induction of Toll-like receptor 4 (TLR4) activation have been gradually revealed under diverse stress conditions, including cerebral ischemia but not hemorrhage. Prx1 is proposed to be a major hemorrhagic stress-inducible isoform of Prxs during acute and subacute phases of intracerebral hemorrhage (ICH). However, the potential of Prx1 in the neuroinflammatory injury after ICH remains unclear. This study investigated the proinflammatory effect and underlying mechanism of extracellular Prx1 in cultured murine macrophages and a collagenase-induced mouse ICH model. The current results show that incubation of exogenous Prx1 (0-50nM) with murine RAW264.7 macrophages resulted in increased expression of TLR4, nuclear translocation of nuclear factor κB (NF-κB) p65 and production of proinflammatory mediators (NO, TNF-a and IL-6) in a concentration-dependent manner. In addition, ICH induced murine neurological deficits, cerebral edema and neuropathological alterations, such as neuron injury, astrocyte and microglia/macrophage activation, and neutrophil and T lymphocyte invasion up to 72h after ICH. Moreover, ICH stimulated Prx1 expression and extracellular release, TLR4/NF-κB signaling activation, reflected by increases in TLR4 expression, extracellular signal-regulated kinase (ERK) 1/2 and NF-κB activation, and production of cytokines (TNF-α, IL-6 and IL-17). Taken together, these findings suggest that extracellular Prx1-mediated TLR4/NF-κB pathway activation probably contributes to neuroinflammatory injury after ICH, and thus blocking Prx1-TLR4 signaling might provide a novel anti-neuroinflammatory strategy with extended therapeutic window for hemorrhagic stroke.
Collapse
Affiliation(s)
- Dong-Ling Liu
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Li-Xue Zhao
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Shuang Zhang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
19
|
Zhao LX, Du JR, Zhou HJ, Liu DL, Gu MX, Long FY. Differences in Proinflammatory Property of Six Subtypes of Peroxiredoxins and Anti-Inflammatory Effect of Ligustilide in Macrophages. PLoS One 2016; 11:e0164586. [PMID: 27716839 PMCID: PMC5055302 DOI: 10.1371/journal.pone.0164586] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 09/27/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Peroxiredoxins (Prxs) are proposed to function as damage-associated molecular patterns (DAMPs) and contribute to post-ischemic neuroinflammation and brain injury by activating Toll-like receptor (TLR) 4 at the acute and subacute phases after ischemic stroke. However, there are few studies concerning the inflammatory profiles of six distinct subtypes of Prxs (Prx1-Prx6). Our previous study demonstrated that the protective effect of ligustilide (LIG) against cerebral ischemia was associated with inhibition of neuroinflammatory response and Prx/TLR4 signaling in rats. Herein, the present study explored the inflammatory members of Prxs and the effect of LIG on their inflammatory responses in macrophages. METHODOLOGY/PRINCIPAL FINDINGS The murine RAW264.7 macrophages were treated with each of exogenous recombinant Prxs at a range of 1 to 50 nM for 24 h. The WST-1 test showed that Prx3 exhibited a significant cytotoxicity, whereas the rest five Prxs did not affect cellular viability. The quantitative measurements with spectrometry or ELISA indicated that three subtypes, Prx1, Prx2 and Prx4, increased production of proinflammatory mediators, including nitric oxide (NO) metabolites, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in a concentration-dependent manner. Immunostaining demonstrated that 20 nM Prx1, Prx2 or Prx4 significantly increased expression of TLR4 and iNOS and nuclear translocation of NF-κB p65. However, Prx5 and Prx6 showed no poinflammatory effect in macrophages. Remarkably, LIG treatment effectively inhibited the inflammatory response induced by Prx1, Prx2 and Prx4. CONCLUSION Three members of Prxs, Prx1, Prx2 and Prx4, are inflammatory DAMPs that induce TLR4 activation and inflammatory response in macrophages, which is effectively inhibited by LIG. These results suggest that inflammatory Prxs-activated macrophages may provide a novel cellular model for screening the potential inhibitors of DAMPs-associated inflammatory diseases such as stroke. Moreover, selective blocking strategies targeting the inflammatory subtypes of Prxs probably provide promising therapeutic approaches with a prolonged time window for stroke.
Collapse
Affiliation(s)
- Li-Xue Zhao
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
- * E-mail:
| | - Hong-Jing Zhou
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Dong-Ling Liu
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Man-Xia Gu
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Fang-Yi Long
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Patergnani S, Fossati V, Bonora M, Giorgi C, Marchi S, Missiroli S, Rusielewicz T, Wieckowski MR, Pinton P. Mitochondria in Multiple Sclerosis: Molecular Mechanisms of Pathogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 328:49-103. [PMID: 28069137 DOI: 10.1016/bs.ircmb.2016.08.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mitochondria, the organelles that function as the powerhouse of the cell, have been increasingly linked to the pathogenesis of many neurological disorders, including multiple sclerosis (MS). MS is a chronic inflammatory demyelinating disease of the central nervous system (CNS) and a leading cause of neurological disability in young adults in the western world. Its etiology remains unknown, and while the inflammatory component of MS has been heavily investigated and targeted for therapeutic intervention, the failure of remyelination and the process of axonal degeneration are still poorly understood. Recent studies suggest a role of mitochondrial dysfunction in the neurodegenerative aspects of MS. This review is focused on mitochondrial functions under physiological conditions and the consequences of mitochondrial alterations in various CNS disorders. Moreover, we summarize recent findings linking mitochondrial dysfunction to MS and discuss novel therapeutic strategies targeting mitochondria-related pathways as well as emerging experimental approaches for modeling mitochondrial disease.
Collapse
Affiliation(s)
- S Patergnani
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - V Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - M Bonora
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - C Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - S Marchi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - S Missiroli
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - T Rusielewicz
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - M R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - P Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
21
|
Abstract
An adverse outcome is still encountered in 45% of full-term neonates with perinatal asphyxia who are treated with moderate hypothermia. At present pharmacologic therapies are developed to be added to hypothermia. In the present article, these potential neuroprotective interventions are described based on the molecular pathways set in motion during fetal hypoxia and following reoxygenation and reperfusion after birth. These pathways include excessive production of excitotoxins with subsequent over-stimulation of NMDA receptors and calcium influx in neuronal cells, excessive production of reactive oxygen and nitrogen species, activation of inflammation leading to inappropriate apoptosis, and loss of neurotrophic factors. Possibilities for pharmacologic combination therapy, where each drug will be administered based on the optimal point of time in the cascade of destructive molecular reactions, may further reduce brain damage due to perinatal asphyxia.
Collapse
Affiliation(s)
- Frank van Bel
- Department of Neonatology, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands.
| | - Floris Groenendaal
- Department of Neonatology, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| |
Collapse
|
22
|
Kim B, Park J, Chang KT, Lee DS. Peroxiredoxin 5 prevents amyloid-beta oligomer-induced neuronal cell death by inhibiting ERK-Drp1-mediated mitochondrial fragmentation. Free Radic Biol Med 2016; 90:184-94. [PMID: 26582373 DOI: 10.1016/j.freeradbiomed.2015.11.015] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/19/2015] [Accepted: 11/10/2015] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder, is caused by amyloid-beta oligomers (AβOs). AβOs induce cell death by triggering oxidative stress and mitochondrial dysfunction. A recent study showed that AβO-induced oxidative stress is associated with extracellular signal-regulated kinase (ERK)-dynamin related protein 1 (Drp1)-mediated mitochondrial fission. Reactive oxygen species (ROS) are regulated by antioxidant enzymes, especially peroxiredoxins (Prxs) that scavenge H2O2. These enzymes inhibit neuronal cell death induced by various neurotoxic reagents. However, it is unclear whether Prx5, which is specifically expressed in neuronal cells, protects these cells from AβO-induced damage. In this study, we found that Prx5 expression was upregulated by AβO-induced oxidative stress and that Prx5 decreased ERK-Drp1-mediated mitochondrial fragmentation and apoptosis of HT-22 neuronal cells. Prx5 expression was affected by AβO, and amelioration of oxidative stress by N-acetyl-L-cysteine decreased AβO-induced Prx5 expression. Prx5 overexpression reduced ROS as well as RNS and apoptotic cell death but Prx5 knockdown did not. In addition, Prx5 overexpression ameliorated ERK-Drp1-mediated mitochondrial fragmentation but Prx5 knockdown did not. These results indicated that inducible Prx5 expression by AβO plays a key role in inhibiting both ERK-Drp1-induced mitochondrial fragmentation and neuronal cell death by regulating oxidative stress. Thus, Prx5 may be a new therapeutic agent for treating AD.
Collapse
Affiliation(s)
- Bokyung Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Junghyung Park
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Kyu-Tae Chang
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungcheongbuk-do, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701, Republic of Korea.
| |
Collapse
|
23
|
Valek L, Kanngießer M, Häussler A, Agarwal N, Lillig CH, Tegeder I. Redoxins in peripheral neurons after sciatic nerve injury. Free Radic Biol Med 2015; 89:581-92. [PMID: 26456799 DOI: 10.1016/j.freeradbiomed.2015.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 01/24/2023]
Abstract
Peripheral nerve injury causes redox stress in injured neurons by upregulations of pro-oxidative enzymes, but most neurons survive suggesting an activation of endogenous defense against the imbalance. As potential candidates we assessed thioredoxin-fold proteins, called redoxins, which maintain redox homeostasis by reduction of hydrogen peroxide or protein dithiol-disulfide exchange. Using a histologic approach, we show that the peroxiredoxins (Prdx1-6), the glutaredoxins (Glrx1, 2, 3 and 5), thioredoxin (Txn1 and 2) and their reductases (Txnrd1 and 2) are expressed in neurons, glial and/or vascular cells of the dorsal root ganglia (DRGs) and in the spinal cord. They show distinct cellular and subcellular locations in agreement with the GO terms for "cellular component". The expression and localization of Glrx, Txn and Txnrd proteins was not affected by sciatic nerve injury but peroxiredoxins were upregulated in the DRGs, Prdx1 and Prdx6 mainly in non-neuronal cells and Prdx4 and Prdx5 in DRG neurons, the latter associated with an increase of respective mRNAs and protein accumulation in peripheral and/or central fibers. The upregulation of Prdx4 and Prdx5 in DRG neurons was reduced in mice with a cre-loxP mediated deficiency of hypoxia inducible factor 1 alpha (HIF1α) in these neurons. The results identify Prdx4 and Prdx5 as endogenous HIF1α-dependent, transcriptionally regulated defenders of nerve injury evoked redox stress that may be important for neuronal survival and regeneration.
Collapse
Affiliation(s)
- Lucie Valek
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Maike Kanngießer
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Annett Häussler
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Nitin Agarwal
- Institute of Pharmacology, Medical Faculty, University of Heidelberg, Germany
| | - Christopher Horst Lillig
- Institute for Medical Biochemistry and Molecular Biology, Medical Faculty of the Ernst-Moritz Arndt-University, Greifswald, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany.
| |
Collapse
|
24
|
Shahaduzzaman MD, Mehta V, Golden JE, Rowe DD, Green S, Tadinada R, Foran EA, Sanberg PR, Pennypacker KR, Willing AE. Human umbilical cord blood cells induce neuroprotective change in gene expression profile in neurons after ischemia through activation of Akt pathway. Cell Transplant 2015; 24:721-35. [PMID: 25413246 DOI: 10.3727/096368914x685311] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human umbilical cord blood (HUCB) cell therapies have shown promising results in reducing brain infarct volume and most importantly in improving neurobehavioral function in rat permanent middle cerebral artery occlusion, a model of stroke. In this study, we examined the gene expression profile in neurons subjected to oxygen-glucose deprivation (OGD) with or without HUCB treatment and identified signaling pathways (Akt/MAPK) important in eliciting HUCB-mediated neuroprotective responses. Gene chip microarray analysis was performed using RNA samples extracted from the neuronal cell cultures from four experimental groups: normoxia, normoxia+HUCB, OGD, and OGD+HUCB. Both quantitative RT-PCR and immunohistochemistry were carried out to verify the microarray results. Using the Genomatix software program, promoter regions of selected genes were compared to reveal common transcription factor-binding sites and, subsequently, signal transduction pathways. Under OGD condition, HUCB cells significantly reduced neuronal loss from 68% to 44% [one-way ANOVA, F(3, 16)=11, p=0.0003]. Microarray analysis identified mRNA expression of Prdx5, Vcam1, CCL20, Alcam, and Pax6 as being significantly altered by HUCB cell treatment. Inhibition of the Akt pathway significantly abolished the neuroprotective effect of HUCB cells [one-way ANOVA, F(3, 11)=8.663, p=0.0031]. Our observations show that HUCB neuroprotection is dependent on the activation of the Akt signaling pathway that increases transcription of the Prdx5 gene. We concluded that HUCB cell therapy would be a promising treatment for stroke and other forms of brain injury by modifying acute gene expression to promote neural cell protection.
Collapse
Affiliation(s)
- M D Shahaduzzaman
- Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Shah FA, Gim SA, Kim MO, Koh PO. Proteomic identification of proteins differentially expressed in response to resveratrol treatment in middle cerebral artery occlusion stroke model. J Vet Med Sci 2014; 76:1367-74. [PMID: 24998396 PMCID: PMC4221170 DOI: 10.1292/jvms.14-0169] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Resveratrol has a
neuroprotective effect against cerebral ischemia. The objective of this study was to
identify proteins that are differentially expressed in the cerebral cortex of vehicle- and
resveratrol-treated animals during ischemic injury. Focal cerebral ischemia was induced as
middle cerebral artery occlusion (MCAO) in male rats. Rats were treated with vehicle or
resveratrol before MCAO, and cerebral cortex was collected 24 hr after MCAO. Cerebral
cortex proteins were identified by two-dimensional gel electrophoresis and mass
spectrometry. Several proteins were identified as differentially expressed between
vehicle- and resveratrol-treated animals. Among these proteins, expression of
peroxiredoxin-5, isocitrate dehydrogenase [NAD+], apolipoprotein A-I and
ubiquitin carboxy terminal hydrolase L1 was decreased in the vehicle-treated group,
whereas resveratrol attenuated the injury-induced decrease in expression of these
proteins. However, expression of collapsing response mediator protein 2 was increased in
the vehicle-treated group, whereas resveratrol prevented the injury-induced increase in
the expression of this protein. These findings suggest that resveratrol modulates the
expression of various proteins that associated with oxidative stress and energy metabolism
in focal cerebral ischemia.
Collapse
Affiliation(s)
- Fawad-Ali Shah
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | | | | | | |
Collapse
|
26
|
Kuang X, Wang LF, Yu L, Li YJ, Wang YN, He Q, Chen C, Du JR. Ligustilide ameliorates neuroinflammation and brain injury in focal cerebral ischemia/reperfusion rats: involvement of inhibition of TLR4/peroxiredoxin 6 signaling. Free Radic Biol Med 2014; 71:165-175. [PMID: 24681253 DOI: 10.1016/j.freeradbiomed.2014.03.028] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 03/18/2014] [Accepted: 03/18/2014] [Indexed: 12/21/2022]
Abstract
Blocking TLR4/peroxiredoxin (Prx6) signaling is proposed to be a novel therapeutic strategy for ischemic stroke because extracellular Prx6 released from ischemic cells may act as an endogenous ligand for TLR4 and initiate destructive immune responses in ischemic brain. Our previous studies showed that ligustilide (LIG) exerted antineuroinflammatory and neuroprotective effects against ischemic insult, but the underlying mechanisms remain unclear. This study investigated whether the TLR4/Prx6 pathway is involved in the protective effect of LIG against postischemic neuroinflammation and brain injury induced by transient middle cerebral artery occlusion (MCAO) in rats. Intraperitoneal LIG administration (20 and 40 mg/kg/day) at reperfusion onset after MCAO resulted in a reduction of brain infarct size and improved neurological outcome over 72 h. LIG-induced neuroprotection was accompanied by improvement of neuropathological alterations, including neuron loss, astrocyte and microglia/macrophage activation, neutrophil and T-lymphocyte invasion, and regulation of inflammatory mediators expression. Moreover, LIG significantly inhibited the expression and extracellular release of Prx6 and activation of TLR4 signaling, reflected by decreased TLR4 expression, extracellular signal-regulated kinase 1/2 phosphorylation, and transcriptional activity of NF-κB and signal transducer and activator of transcription 3 in the ischemic brain. Our results demonstrate that LIG may provide an early and direct neuroprotection by inhibiting TLR4/Prx6 signaling and subsequent immunity and neuroinflammation after cerebral ischemia. These findings support the translational potential of blocking TLR4/Prx6 signaling for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Xi Kuang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Liang-Fen Wang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lu Yu
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yong-Jie Li
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yan-Nan Wang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Qian He
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chu Chen
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
27
|
Markham A, Bains R, Franklin P, Spedding M. Changes in mitochondrial function are pivotal in neurodegenerative and psychiatric disorders: how important is BDNF? Br J Pharmacol 2014; 171:2206-29. [PMID: 24720259 PMCID: PMC3976631 DOI: 10.1111/bph.12531] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 11/08/2013] [Accepted: 11/11/2013] [Indexed: 12/13/2022] Open
Abstract
The brain is at the very limit of its energy supply and has evolved specific means of adapting function to energy supply, of which mitochondria form a crucial link. Neurotrophic and inflammatory processes may not only have opposite effects on neuroplasticity, but also involve opposite effects on mitochondrial oxidative phosphorylation and glycolytic processes, respectively, modulated by stress and glucocorticoids, which also have marked effects on mood. Neurodegenerative processes show marked disorders in oxidative metabolism in key brain areas, sometimes decades before symptoms appear (Parkinson's and Alzheimer's diseases). We argue that brain-derived neurotrophic factor couples activity to changes in respiratory efficiency and these effects may be opposed by inflammatory cytokines, a key factor in neurodegenerative processes.
Collapse
Affiliation(s)
- A Markham
- Department of Pharmacy, Health & Well Being, Faculty of Applied Sciences, University of SunderlandSunderland, UK
| | - R Bains
- University of PortsmouthPortsmouth, UK
| | - P Franklin
- Department of Pharmacy, Health & Well Being, Faculty of Applied Sciences, University of SunderlandSunderland, UK
| | - M Spedding
- Spedding Research Solutions SARLLe Vesinet, France
| |
Collapse
|
28
|
Jeong HJ, Yoo DY, Kim DW, Yeo HJ, Cho SB, Hyeon J, Park JH, Park J, Eum WS, Hwang HS, Won MH, Hwang IK, Choi SY. Neuroprotective effect of PEP-1-peroxiredoxin2 on CA1 regions in the hippocampus against ischemic insult. Biochim Biophys Acta Gen Subj 2014; 1840:2321-30. [PMID: 24631653 DOI: 10.1016/j.bbagen.2014.03.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 03/01/2014] [Accepted: 03/04/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND Oxidative stress is a leading cause of various diseases, including ischemia and inflammation. Peroxiredoxin2 (PRX2) is one of six mammalian isoenzymes (PRX1-6) that can reduce hydrogen peroxide (H2O2) and organic hydroperoxides to water and alcohols. METHODS We produced PEP-1-PRX2 transduction domain (PTD)-fused protein and investigated the effect of PEP-1-PRX2 on oxidative stress-induced neuronal cell death by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, Western blot, immunofluorescence microscopy, and immunohistochemical analysis. RESULTS Our data showed that PEP-1-PRX2, which can effectively transduce into various types of cells and brain tissues, could be implicated in suppressing generation of reactive oxygen species, preventing depolarization of the mitochondrial membrane, and inhibiting the apoptosis pathway in H2O2-stimulated HT22, murine hippocampal neuronal cells, likely resulting in protection of HT22 cells against H2O2-induced toxicity. In addition, we found that in a transient forebrain ischemia model, PEP-1-PRX2 inhibited the activation of astrocytes and microglia in the CA1 region of the hippocampus and lipid peroxidation and also prevented neuronal cell death against ischemic damage. CONCLUSIONS These findings suggest that the transduced PEP-1-PRX2 has neuroprotective functions against oxidative stress-induced cell death in vitro and in vivo. GENERAL SIGNIFICANCE PEP-1-PRX2 could be a potential therapeutic agent for oxidative stress-induced brain diseases such as ischemia.
Collapse
Affiliation(s)
- Hoon Jae Jeong
- Department of Biomedical Sciences, Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Republic of Korea
| | - Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 151-742, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Kangnung-Wonju National University, Gangneung 210-702, Republic of Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Sciences, Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Republic of Korea
| | - Su Bin Cho
- Department of Biomedical Sciences, Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Republic of Korea
| | - Jiye Hyeon
- Department of Biomedical Sciences, Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Republic of Korea
| | - Jung Hwan Park
- Department of Biomedical Sciences, Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Republic of Korea
| | - Jinseu Park
- Department of Biomedical Sciences, Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Republic of Korea
| | - Won Sik Eum
- Department of Biomedical Sciences, Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Republic of Korea
| | - Hyun Sook Hwang
- Department of Biomedical Sciences, Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul 151-742, Republic of Korea.
| | - Soo Young Choi
- Department of Biomedical Sciences, Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Republic of Korea.
| |
Collapse
|
29
|
Van der Eecken V, Clippe A, Dekoninck S, Goemaere J, Walbrecq G, Van Veldhoven PP, Knoops B. Abolition of peroxiredoxin-5 mitochondrial targeting during canid evolution. PLoS One 2013; 8:e72844. [PMID: 24023783 PMCID: PMC3759418 DOI: 10.1371/journal.pone.0072844] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 07/16/2013] [Indexed: 11/19/2022] Open
Abstract
In human, the subcellular targeting of peroxiredoxin-5 (PRDX5), a thioredoxin peroxidase, is dependent on the use of multiple alternative transcription start sites and two alternative in-frame translation initiation sites, which determine whether or not the region encoding a mitochondrial targeting sequence (MTS) is translated. In the present study, the abolition of PRDX5 mitochondrial targeting in dog is highlighted and the molecular mechanism underlying the loss of mitochondrial PRDX5 during evolution is examined. Here, we show that the absence of mitochondrial PRDX5 is generalized among the extant canids and that the first events leading to PRDX5 MTS abolition in canids involve a mutation in the more 5′ translation initiation codon as well as the appearance of a STOP codon. Furthermore, we found that PRDX5 MTS functionality is maintained in giant panda and northern elephant seal, which are phylogenetically closely related to canids. Also, the functional consequences of the restoration of mitochondrial PRDX5 in dog Madin-Darby canine kidney (MDCK) cells were investigated. The restoration of PRDX5 mitochondrial targeting in MDCK cells, instead of protecting, provokes deleterious effects following peroxide exposure independently of its peroxidase activity, indicating that mitochondrial PRDX5 gains cytotoxic properties under acute oxidative stress in MDCK cells. Altogether our results show that, although mitochondrial PRDX5 cytoprotective function against oxidative stress has been clearly demonstrated in human and rodents, PRDX5 targeting to mitochondria has been evolutionary lost in canids. Moreover, restoration of mitochondrial PRDX5 in dog MDCK cells, instead of conferring protection against peroxide exposure, makes them more vulnerable.
Collapse
Affiliation(s)
- Valérie Van der Eecken
- Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - André Clippe
- Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Sophie Dekoninck
- Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Julie Goemaere
- Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Geoffroy Walbrecq
- Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Paul P. Van Veldhoven
- Department of Cellular and Molecular Medicine, LIPIT, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Bernard Knoops
- Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve, Belgium
- * E-mail:
| |
Collapse
|
30
|
De Simoni S, Linard D, Hermans E, Knoops B, Goemaere J. Mitochondrial peroxiredoxin-5 as potential modulator of mitochondria-ER crosstalk in MPP+-induced cell death. J Neurochem 2013; 125:473-85. [PMID: 23216451 DOI: 10.1111/jnc.12117] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 11/30/2012] [Accepted: 12/04/2012] [Indexed: 12/11/2022]
Abstract
Peroxiredoxin-5 (PRDX5) is an antioxidant enzyme which differs from the other peroxiredoxins with regards to its enzymatic mechanism, its high affinity for organic peroxides and peroxynitrite and its wide subcellular distribution. In particular, the mitochondrial isoform of PRDX5 confers a remarkable cytoprotection toward oxidative stress to mammalian cells. Mitochondrial dysfunction and disruption of Ca²⁺ homeostasis are implicated in neurodegeneration. Growing evidence supports that endoplasmic reticulum (ER) could operate in tandem with mitochondria to regulate intracellular Ca²⁺ fluxes in neurodegenerative processes. Here, we overexpressed mitochondrial PRDX5 in SH-SY5Y cells to dissect the role of this enzyme in 1-methyl-4-phenylpyridinium (MPP)⁺-induced cell death. Our data show that mitochondria-dependent apoptosis triggered by MPP⁺, assessed by the measurement of caspase-9 activation and mitochondrial DNA damage, is prevented by mitochondrial PRDX5 overexpression. Moreover, PRDX5 overexpression blocks the increase in intracellular Ca²⁺, Ca²⁺-dependent activation of calpains and Bax cleavage. Finally, using Ca²⁺ channel inhibitors (Nimodipine, Dantrolene and 2-APB), we show that Ca²⁺ release arises essentially from ER stores through 1,4,5-inositol-trisphosphate receptors (IP3 R). Altogether, our results suggest that the MPP⁺ mitochondrial pathway of apoptosis is regulated by mitochondrial PRDX5 in a process that could involve redox modulation of Ca²⁺ transporters via a crosstalk between mitochondria and ER.
Collapse
Affiliation(s)
- Stéphanie De Simoni
- Group of Cell Biology, Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | | | | | | | | |
Collapse
|
31
|
Haapasalo T, Nordfors K, Järvelä S, Kok E, Sallinen P, Kinnula VL, Haapasalo HK, Soini Y. Peroxiredoxins and their expression in ependymomas. J Clin Pathol 2012; 66:12-7. [PMID: 23076395 DOI: 10.1136/jclinpath-2012-201048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
AIMS Peroxiredoxins I-VI (Prxs) have recently been shown to have a role in the tumorigenesis of astrocytic brain tumours. In some tumour types they are associated with Nrf2 (transcription factor NF-E2-related factor), a sensor of oxidative stress, and DJ-1 (also known as PARK7), a protein known to stabilise Nrf2. METHODS We investigated the immunohistochemical expression of Prxs I-VI, Nrf2 and DJ-1 in a total of 76 ependymomas and their relationship with clinicopathological features of these tumours. RESULTS There was a significant expression of all Prxs except Prx IV in the ependymomas. Strong nuclear and cytoplasmic expression of Nrf2 could be detected in these tumours. Prx I expression was significantly associated with cytoplasmic and nuclear Nrf2 expression. Prx I expression was also associated with tumour site, with cerebellar ependymomas having a lower expression of Prx I than other tumours. DJ-1 did not associate with Prxs but nuclear DJ-1 had an inverse association with nuclear Nrf2. Cytoplasmic DJ-1 associated with worse survival in ependymoma patients. CONCLUSIONS This study indicates that oxidative mechanisms as reflected by Nrf2 expression are highly activated in ependymomas. Prxs, especially Prx I, were associated with Nrf2 expression, suggesting a role for Nrf2 in Prx I synthesis in ependymomas. While DJ-1 did not associate with any of the Prxs, its expression was associated with worsened patient survival and could have a role as a prognostic marker in ependymomas.
Collapse
Affiliation(s)
- Toomas Haapasalo
- Department of Pathology, Fimlab Laboratories, Tampere University Hospital, and University of Tampere, Tampere, Finland
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Xi L, Zhu SG, Hobbs DC, Kukreja RC. Identification of protein targets underlying dietary nitrate-induced protection against doxorubicin cardiotoxicity. J Cell Mol Med 2012; 15:2512-24. [PMID: 21251210 PMCID: PMC3110615 DOI: 10.1111/j.1582-4934.2011.01257.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
We recently demonstrated protective effect of chronic oral nitrate supplementation against cardiomyopathy caused by doxorubicin (DOX), a highly effective anticancer drug. The present study was designed to identify novel protein targets related to nitrate-induced cardioprotection. Adult male CF-1 mice received cardioprotective regimen of nitrate (1 g NaNO3 per litre of drinking water) for 7 days before DOX injection (15 mg/kg, i.p.) and continued for 5 days after DOX treatment. Subsequently the heart samples were collected for proteomic analysis with two-dimensional differential in-gel electrophoresis with 3 CyDye labelling. Using 1.5 cut-off ratio, we identified 36 proteins that were up-regulated by DOX in which 32 were completely reversed by nitrate supplementation (89%). Among 19 proteins down-regulated by DOX, 9 were fully normalized by nitrate (47%). The protein spots were further identified with Matrix Assisted Laser Desorption/Ionization-Time-of-Flight (MALDI-TOF)/TOF tandem mass spectrometry. Three mitochondrial antioxidant enzymes were altered by DOX, i.e. up-regulation of manganese superoxide dismutase and peroxiredoxin 3 (Prx3), and down-regulation of Prx5, which were reversed by nitrate. These results were further confirmed by Western blots. Nitrate supplementation also significantly improved animal survival rate from 80% in DOX alone group to 93% in Nitrate + DOX group 5 days after the DOX treatment. In conclusion, the proteomic analysis has identified novel protein targets underlying nitrate-induced cardioprotection. Up-regulation of Prx5 by nitrate may explain the observed enhancement of cardiac antioxidant defence by nitrate supplementation.
Collapse
Affiliation(s)
- Lei Xi
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298–0204, USA.
| | | | | | | |
Collapse
|
33
|
Dietary inorganic nitrate alleviates doxorubicin cardiotoxicity: mechanisms and implications. Nitric Oxide 2012; 26:274-84. [PMID: 22484629 DOI: 10.1016/j.niox.2012.03.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 02/11/2012] [Accepted: 03/22/2012] [Indexed: 11/21/2022]
Abstract
Doxorubicin (DOX) is one of the most powerful and widely prescribed chemotherapeutic agents to treat divergent human cancers. However, the clinical use of DOX is restricted due to its severe cardiotoxic side-effects. There has been ongoing search for cardioprotectants against DOX toxicity. Inorganic nitrate has emerged as a bioactive compound that can be reduced into nitrite and nitric oxide in vivo and in turn plays a therapeutic role in diseases associated with nitric oxide insufficiency or dysregulation. In this review, we describe a novel concept of using dietary supplementation of inorganic nitrate to reduce DOX-induced cardiac cellular damage and dysfunction, based on our recent promising studies in a mouse model of DOX cardiotoxicity. Our data show that chronic oral ingestion of sodium nitrate, at a dose equivalent to ~400% of the Acceptable Daily Intake of the World Health Organization, alleviated DOX-induced left ventricular dysfunction and mitochondrial respiratory chain damage. Such cardioprotective effects were associated with reduction of cardiomyocyte necrosis/apoptosis, tissue lipid peroxidation, and mitochondrial H(2)O(2) generation following DOX treatment. Furthermore, proteomic studies revealed enhanced cardiac expression of mitochondrial antioxidant enzyme - peroxiredoxin 5 in the nitrate-treated animals. These studies suggest that inorganic nitrate could be an inexpensive therapeutic agent for long-term oral administration in preventing DOX-induced cardiac toxicity and myopathy during the prolonged pathological process. Future clinical trials in the cancer patients undergoing DOX chemotherapy are warranted to translate these experimental findings into an effective new therapy in preventing the DOX-induced cardiomyopathy.
Collapse
|
34
|
Robertson NJ, Tan S, Groenendaal F, van Bel F, Juul SE, Bennet L, Derrick M, Back SA, Valdez RC, Northington F, Gunn AJ, Mallard C. Which neuroprotective agents are ready for bench to bedside translation in the newborn infant? J Pediatr 2012; 160:544-552.e4. [PMID: 22325255 PMCID: PMC4048707 DOI: 10.1016/j.jpeds.2011.12.052] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 12/02/2011] [Accepted: 12/30/2011] [Indexed: 02/07/2023]
|
35
|
Zhu H, Santo A, Li Y. The antioxidant enzyme peroxiredoxin and its protective role in neurological disorders. Exp Biol Med (Maywood) 2012; 237:143-9. [PMID: 22302711 DOI: 10.1258/ebm.2011.011152] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Peroxiredoxin (Prx) represents a family of sulfhydryl-dependent peroxidases that reduce hydrogen peroxide and organic hydroperoxides to water and alcohols, respectively. There are six known mammalian isozymes (Prx1-6), classified as typical 2-Cys, atypical 2-Cys, or 1-Cys Prxs. In addition to their well-established peroxide-scavenging activity, Prxs also participate in the regulation of various cell signaling pathways. Experimental studies provide substantial evidence for a protective role of Prxs in various neurological disorders involving oxidative and inflammatory stress. There is also evidence suggesting a potential benefit of Prxs in certain neurological diseases in human subjects. This review first describes the biochemical properties and molecular regulation of Prxs, then summarizes the major findings on the neuroprotective functions of Prxs and finally discusses the feasibility of using natural compounds, including those from herbal remedies to augment Prx expression to counteract oxidative neurological disorders.
Collapse
Affiliation(s)
- Hong Zhu
- Laboratory of Molecular Pharmacology and Toxicology, Department of Pharmacology, Edward Via College of Osteopathic Medicine, Virginia Tech Corporate Research Center, Blacksburg, VA 24060, USA.
| | | | | |
Collapse
|
36
|
Markham A, Cameron I, Bains R, Franklin P, Kiss JP, Schwendimann L, Gressens P, Spedding M. Brain-derived neurotrophic factor-mediated effects on mitochondrial respiratory coupling and neuroprotection share the same molecular signalling pathways. Eur J Neurosci 2012; 35:366-74. [DOI: 10.1111/j.1460-9568.2011.07965.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
37
|
Rowe DD, Leonardo CC, Recio JA, Collier LA, Willing AE, Pennypacker KR. Human umbilical cord blood cells protect oligodendrocytes from brain ischemia through Akt signal transduction. J Biol Chem 2011; 287:4177-87. [PMID: 22158864 DOI: 10.1074/jbc.m111.296434] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human umbilical cord blood (HUCB) cells protect the brain against ischemic injury, yet the mechanism of protection remains unclear. Using both in vitro and in vivo paradigms, this study examined the role of Akt signaling and peroxiredoxin 4 expression in human umbilical cord blood cell-mediated protection of oligodendrocytes from ischemic conditions. As previously reported, the addition of HUCB cells to oligodendrocyte cultures prior to oxygen glucose deprivation significantly enhanced oligodendrocyte survival. The presence of human umbilical cord blood cells also increased Akt phosphorylation and elevated peroxiredoxin 4 expression in oligodendrocytes. Blocking either Akt or peroxiredoxin 4 activity with Akt Inhibitor IV or a peroxiredoxin 4-neutralizing antibody, respectively, negated the protective effects of human umbilical cord blood cells. In vivo, systemic administration of human umbilical cord blood cells 48 h after middle cerebral artery occlusion increased Akt phosphorylation and peroxiredoxin 4 protein expression while reducing proteolytic cleavage of caspase 3 in oligodendrocytes residing in the ipsilateral external capsule. Moreover, human umbilical cord blood cells protected striatal white matter bundles from degeneration following middle cerebral artery occlusion. These results suggest that the soluble factors released from human umbilical cord blood cells converge on Akt to elevate peroxiredoxin 4 levels, and these effects contribute to oligodendrocyte survival.
Collapse
Affiliation(s)
- Derrick D Rowe
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, College of Medicine, University of South Florida, Tampa, Florida 33612, USA
| | | | | | | | | | | |
Collapse
|
38
|
Mishra M, Manavalan A, Sze SK, Heese K. Neuronal p60TRP expression modulates cardiac capacity. J Proteomics 2011; 75:1600-17. [PMID: 22172954 DOI: 10.1016/j.jprot.2011.11.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 11/20/2011] [Accepted: 11/28/2011] [Indexed: 01/08/2023]
Abstract
Heart failure, including myocardial infarction, is the leading cause for death and the incidence of cardiovascular diseases is predicted to continue to rise worldwide. In the present study we investigated the whole heart proteome profile of transgenic p60-Transcription Regulator Protein (p60TRP) mice to gain an insight into the molecular events caused by the long-term effect of neural p60TRP over-expression on cardiac proteome changes and its potential implication for cardiovascular functions. Using an iTRAQ (isobaric tags for relative and absolute quantitation)-based proteomics research approach, we identified 1148 proteins, out of which 116 were found to be significantly altered in the heart of neural transgenic p60TRP mice. Based on the observed data, we conclude that in vivo neural over-expression of transgenic p60TRP with its neuroprotective therapeutic potential significantly affects cardiovascular capacities.
Collapse
Affiliation(s)
- Manisha Mishra
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | | | | | | |
Collapse
|
39
|
Song IS, Kim HK, Jeong SH, Lee SR, Kim N, Rhee BD, Ko KS, Han J. Mitochondrial peroxiredoxin III is a potential target for cancer therapy. Int J Mol Sci 2011; 12:7163-85. [PMID: 22072940 PMCID: PMC3211031 DOI: 10.3390/ijms12107163] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 09/30/2011] [Accepted: 10/20/2011] [Indexed: 01/01/2023] Open
Abstract
Mitochondria are involved either directly or indirectly in oncogenesis and the alteration of metabolism in cancer cells. Cancer cells contain large numbers of abnormal mitochondria and produce large amounts of reactive oxygen species (ROS). Oxidative stress is caused by an imbalance between the production of ROS and the antioxidant capacity of the cell. Several cancer therapies, such as chemotherapeutic drugs and radiation, disrupt mitochondrial homeostasis and release cytochrome c, leading to apoptosome formation, which activates the intrinsic pathway. This is modulated by the extent of mitochondrial oxidative stress. The peroxiredoxin (Prx) system is a cellular defense system against oxidative stress, and mitochondria in cancer cells are known to contain high levels of Prx III. Here, we review accumulating evidence suggesting that mitochondrial oxidative stress is involved in cancer, and discuss the role of the mitochondrial Prx III antioxidant system as a potential target for cancer therapy. We hope that this review will provide the basis for new strategic approaches in the development of effective cancer treatments.
Collapse
Affiliation(s)
- In-Sung Song
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 614-7-5, Korea; E-Mails: (I.-S.S.); (H.-K.K.); (S.-H.J.); (S.-R.L.); (N.K.); (B.D.R.); (K.S.K.)
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Gressens P, Le Verche V, Fraser M, Rousset CI, Schwendimann L, Bennet L, George SA, Wang X, Mallard C, Tilley BC, Dournaud P, Gunn AJ, Hagberg H, Levison SW. Pitfalls in the quest of neuroprotectants for the perinatal brain. Dev Neurosci 2011; 33:189-98. [PMID: 21968048 DOI: 10.1159/000333109] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 09/14/2011] [Indexed: 11/19/2022] Open
Abstract
Sick preterm and term newborns are highly vulnerable to neural injury, and thus there has been a major search for new, safe and efficacious neuroprotective interventions in recent decades. Preclinical studies are essential to select candidate drugs for clinical trials in humans. This article focuses on 'negative' preclinical studies, i.e. studies where significant differences cannot be detected. Such findings are critical to inform both clinical and preclinical investigators, but historically they have been difficult to publish. A significant amount of time and resources is lost when negative results or nonpromising therapeutics are replicated in separate laboratories because these negative results were not shared with the research community in an open and accessible format. In this article, we discuss approaches to strengthen conclusions from negative preclinical studies and, conversely, to reduce false-negative preclinical evaluations of potential therapeutic compounds. Without being exhaustive, we address three major issues in conducting and interpreting preclinical experiments, including: (a) the choice of animal models, (b) the experimental design, and (c) issues concerning statistical analyses of the experiments. This general introduction is followed by synopses of negative data obtained from studies of three potential therapeutics for perinatal brain injury: (1) the somatostatin analog octreotide, (2) an AMPA/kainate receptor antagonist, topiramate, and (3) a pyruvate derivative, ethyl pyruvate.
Collapse
|
41
|
Knoops B, Goemaere J, Van der Eecken V, Declercq JP. Peroxiredoxin 5: structure, mechanism, and function of the mammalian atypical 2-Cys peroxiredoxin. Antioxid Redox Signal 2011; 15:817-29. [PMID: 20977338 DOI: 10.1089/ars.2010.3584] [Citation(s) in RCA: 171] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Peroxiredoxin 5 (PRDX5) was the last member to be identified among the six mammalian peroxiredoxins. It is also the unique atypical 2-Cys peroxiredoxin in mammals. Like the other five members, PRDX5 is widely expressed in tissues but differs by its surprisingly large subcellular distribution. In human cells, it has been shown that PRDX5 can be addressed to mitochondria, peroxisomes, the cytosol, and the nucleus. PRDX5 is a peroxidase that can use cytosolic or mitochondrial thioredoxins to reduce alkyl hydroperoxides or peroxynitrite with high rate constants in the 10(6) to 10(7) M(-1)s(-1) range, whereas its reaction with hydrogen peroxide is more modest, in the 10(5) M(-1)s(-1) range. PRDX5 crystal structures confirmed the proposed enzymatic mechanisms based on biochemical data but revealed also some specific unexpected structural features. So far, PRDX5 has been viewed mainly as a cytoprotective antioxidant enzyme acting against endogenous or exogenous peroxide attacks rather than as a redox sensor. Accordingly, overexpression of the enzyme in different subcellular compartments protects cells against death caused by nitro-oxidative stresses, whereas gene silencing makes them more vulnerable. Thus, more than 10 years after its molecular cloning, mammalian PRDX5 appears to be a unique peroxiredoxin exhibiting specific functional and structural features.
Collapse
Affiliation(s)
- Bernard Knoops
- Institut des Sciences de Vie, Université catholique de Louvain, Louvain-la-Neuve, Belgium.
| | | | | | | |
Collapse
|
42
|
van Horssen J, Witte ME, Schreibelt G, de Vries HE. Radical changes in multiple sclerosis pathogenesis. Biochim Biophys Acta Mol Basis Dis 2011; 1812:141-50. [DOI: 10.1016/j.bbadis.2010.06.011] [Citation(s) in RCA: 225] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 06/08/2010] [Accepted: 06/16/2010] [Indexed: 12/20/2022]
|
43
|
Chou JLJ, Wu CHY, Tsai CY, Chang AYW, Chan SHH. Proteomic investigation of a neural substrate intimately related to brain death. Proteomics 2010; 11:239-48. [PMID: 21204251 DOI: 10.1002/pmic.201000438] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 10/15/2010] [Accepted: 10/18/2010] [Indexed: 01/18/2023]
Abstract
Our current understanding on brain death remains limited despite its clinical importance. This study evaluated whether the proteome expressed in the rat rostral ventrolateral medulla (RVLM), a neural substrate that our laboratory identified previously to be intimately related to brain death, is uniquely different from other brain areas, using the cerebral cortex, which is defunct under persistent vegetative state for comparison. We found that a group of antioxidant proteins, including members of the peroxiredoxin (Prx) family (Prx-1, Prx-2, Prx-5, Prx-6), thioredoxin and mitochondrial manganese superoxide dismutase, exhibited significantly higher protein and mRNA expression levels in RVLM when compared to cerebral cortex. Tissue oxygen, ATP contents and ATP synthase subunits α and β in RVLM were also significantly elevated. On the other hand, protein and mRNA levels of members of the ubiquitin-proteasome system, including proteasome subunit α type-1, ubiquitin, uniquitin-conjugating enzyme E2 N, ubiquitin carboxyl-terminal hydrolase isozyme L1 and L3, were comparable in both brain regions. We conclude that a significantly elevated level of antioxidant proteins and mRNA in RVLM is consistent with the exhibition of higher tissue oxygen tension and metabolic energy production in this neural substrate, which together constitute a safeguard mechanism against brain death.
Collapse
Affiliation(s)
- Jimmy Li-Jer Chou
- Center for Translational Research in Biomedical Sciences, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung County, Taiwan, Republic of China
| | | | | | | | | |
Collapse
|
44
|
Yang HY, Kwon J, Cho EJ, Choi HI, Park C, Park HR, Park SH, Chung KJ, Ryoo ZY, Cho KO, Lee TH. Proteomic analysis of protein expression affected by peroxiredoxin V knock-down in hypoxic kidney. J Proteome Res 2010; 9:4003-15. [PMID: 20553050 DOI: 10.1021/pr100190b] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Peroxiredoxin V, an atypical thioredoxin peroxidase, is widely expressed in mammalian tissues. In addition, Prdx V is localized in mitochondria, peroxisome, cytosol, and the nucleus. Prdx V has been reported to protect a wide range of cellular environments as an antioxidant enzyme, and its dysfunctions may be implicated in several diseases, such as cancer, inflammation, and neurodegenerative disease. Identification and relative quantification of proteins affected by Prdx V may help identify novel signaling mechanisms that are important for oxidative stress response. However, the role of Prdx V in the modulation of hypoxia-related cellular response is not studied yet. To examine the function of endogenous Prdx V in hypoxic condition in vivo, we generated a transgenic mouse model with Prdx V siRNA expression controlled by U6 promoter. Of many tissues, the knockdown of Prdx V expression was displayed in the kidney, lung, and liver but not the spleen and skin. We conducted on the basis of nano-UPLC-MS(E) proteomic study to identify the Prdx V-affected protein networks in hypoxic kidneys. In this study, we identified protein networks associated with oxidative stress, fatty acid metabolism, and mitochondrial dysfunction. Our results indicated that Prdx V affected to regulation of kidney homeostasis under hypoxia stress.
Collapse
Affiliation(s)
- Hee-Young Yang
- Department of Oral Biochemistry, Dental Science Research Institute, The second Stage of Brain Korea 21 for Dental School, Chonnam National University, Gwangju, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wu J, Tang Q, Shen J, Yao A, Wang F, Pu L, Yu Y, Li X, Li G, Zhang F, Sun B, Kong L, Li D, Zhang Y, Guo X, Wang X. Comparative proteome profile during the early period of small-for-size liver transplantation in rats revealed the protective role of Prdx5. J Hepatol 2010; 53:73-83. [PMID: 20451279 DOI: 10.1016/j.jhep.2010.01.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Revised: 01/12/2010] [Accepted: 01/12/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS In living-donor liver transplantation (LDLT), "small-for-size graft (SFSG) syndrome" is a complex process resulting primarily from ischemia-reperfusion injury (IRI) and portal hypertension associated with size mismatch between graft and recipient. In the early period of LDLT, molecular events related to subsequent apoptosis, necrosis, proliferation and regeneration appeared in specific protein expression patterns. METHODS We used 2D-PAGE and MALDI-TOF/TOF technology to construct a comparative proteome profile for small-for-size liver grafts (SFSGs) during the early period of LDLT in rats (ischemia 1h, and 2, 6, 24, 48 h post-reperfusion); sham-operated liver was the control. Western blotting was used to confirm the proteomics results and immunohistochemistry was carried out to explore the cellular localization of selected proteins. We further performed cluster and bioinformatics analyses of differential proteins. Lastly, we overexpressed Prdx5 in liver grafts using an adenoviral vector to evaluate its protective role. RESULTS We identified 314 differential protein spots corresponding to 259 different proteins. Cluster analyses revealed six expression patterns, and bioinformatics analyses revealed that each pattern was related to many specific cell processes. We also showed that Prdx5 overexpression could attenuate injury to SFSGs and increase survival in recipients. CONCLUSIONS Taken together, these results reveal an important proteome profile that is functional in SFSGs during early period of LDLT, and provide a strong basis for further research.
Collapse
Affiliation(s)
- Jindao Wu
- Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, Department of Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Järvelä S, Rantala I, Rodriguez A, Kallio H, Parkkila S, Kinnula VL, Soini Y, Haapasalo H. Specific expression profile and prognostic significance of peroxiredoxins in grade II-IV astrocytic brain tumors. BMC Cancer 2010; 10:104. [PMID: 20307276 PMCID: PMC2858108 DOI: 10.1186/1471-2407-10-104] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Accepted: 03/22/2010] [Indexed: 11/29/2022] Open
Abstract
Background Peroxiredoxins (Prxs) have recently been suggested to have a role in tumorigenesis. Methods We studied the expression of Prx I-VI and their relationship to patient survival in 383 grade II-IV diffuse astrocytic brain tumors. Results Prx I positivity was found in 68%, Prx II in 84%, Prx III in 90%, Prx IV in 5%, Prx V in 4% and Prx VI in 47% of the tumors. Prx I and Prx II expression decreased significantly with increasing malignancy grade (p < 0.001 and p < 0.001). Patients with Prx I or Prx II positive tumors were significantly younger than the average age of all the patients (p = 0.014 and p = 0.005). A lower proliferation rate was associated with Prx I and Prx VI positive tumors (p = 0.019 and p = 0.033), and a lower apoptotic rate was found within Prx I and Prx II positive tumors (p < 0.001 and p = 0.007). Patients with Prx I and Prx II positive tumors had a significantly better survival rate than their Prx-negative counterparts (p = 0.0052 and p = 0.0002). Conclusion The expression of Prx I and Prx II correlates with astrocytic tumor features, such as grade and patient age and proliferation activity (Prx I), and accordingly with patient survival.
Collapse
Affiliation(s)
- Sally Järvelä
- Department of Pathology, Centre for Laboratory Medicine, Tampere University Hospital, Tampere, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Sun HN, Kim SU, Huang SM, Kim JM, Park YH, Kim SH, Yang HY, Chung KJ, Lee TH, Choi HS, Min JS, Park MK, Kim SK, Lee SR, Chang KT, Lee SH, Yu DY, Lee DS. Microglial peroxiredoxin V acts as an inducible anti-inflammatory antioxidant through cooperation with redox signaling cascades. J Neurochem 2010; 114:39-50. [PMID: 20345759 DOI: 10.1111/j.1471-4159.2010.06691.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Reactive oxygen species (ROS) actively participate in microglia-mediated pathogenesis as pro-inflammatory molecules. However, little is known about the involvement of specific antioxidants in maintaining the microglial oxidative balance. We demonstrate that microglial peroxiredoxin (Prx) 5 expression is up-regulated by lipopolysaccharide (LPS) through activation of the ROS-sensitive signaling pathway and is involved in attenuation of both microglial activation and nitric oxide (NO) generation. Unlike in stimulation of oxidative insults with paraquat and hydrogen peroxide, Prx V expression is highly sensitive to LPS-stimulation in microglia. Reduction of ROS level by treatment with either NADPH oxidase inhibitor or antioxidant ablates LPS-mediated Prx V up-regulation in BV-2 microglial cells and is closely associated with the activation of the c-jun N-terminal kinase (JNK) signaling pathway. This suggests the involvement of ROS/JNK signaling in LPS-mediated Prx V induction. Furthermore, NO induces Prx V up-regulation that is ablated by the addition of inducible nitric oxide synthase inhibitor or deleted mutation of inducible nitric oxide synthase in LPS-stimulated microglia. Therefore, these results suggest that Prx V is induced by cooperative action among the ROS, RNS, and JNK signaling cascades. Interestingly, knockdown of Prx V expression causes the acceleration of microglia activation, including augmented ROS generation and JNK-dependent NO production. In summary, we demonstrate that Prx V plays a key role in the microglial activation process through modulation of the balance between ROS/NO generation and the corresponding JNK cascade activation.
Collapse
Affiliation(s)
- Hu-Nan Sun
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-806, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Injury to the premature brain is a major contributor to infant mortality and morbidity, often leading to mental retardation and sensory-motor impairment. The disease process is believed to be caused, sustained, and aggravated by multiple perinatal factors that team up in a multi-hit fashion. Clinical, epidemiological, and experimental studies have revealed that key factors such as inflammation, excitotoxicity, and oxidative stress contribute considerably to white- and gray-matter injury in premature infants, whose brains are particularly susceptible to damage. Depending on the timing, lesions of the immature brain may influence developmental events in their natural sequence and redirect subsequent development. We review current concepts on molecular mechanisms underlying injury to the premature brain.
Collapse
Affiliation(s)
- Angela M. Kaindl
- Université Paris 7, Faculté de Medecine Denis Diderot, Paris, France, PremUP, Paris, France, Inserm, U676, Paris, France
| | - Geraldine Favrais
- Université Paris 7, Faculté de Medecine Denis Diderot, Paris, France, PremUP, Paris, France, Inserm, U676, Paris, France
| | - Pierre Gressens
- Université Paris 7, Faculté de Medecine Denis Diderot, Paris, France, , PremUP, Paris, France, AP HP, Hôpital Robert Debré, Service de Neurologie Pédiatrique, Paris, France, Inserm, U676, Paris, France
| |
Collapse
|
49
|
Kubo E, Singh DP, Fatma N, Akagi Y. TAT-mediated peroxiredoxin 5 and 6 protein transduction protects against high-glucose-induced cytotoxicity in retinal pericytes. Life Sci 2009; 84:857-64. [PMID: 19351539 DOI: 10.1016/j.lfs.2009.03.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 03/19/2009] [Accepted: 03/28/2009] [Indexed: 01/13/2023]
Abstract
AIMS Hyperglycemia-induced oxidative stress is implicated in pericyte apoptosis seen in diabetic retinopathy. The six mammalian Peroxiredoxins (PRDXs) comprise a novel family of antioxidative proteins that negatively regulate oxidative stress-induced apoptosis by controlling reactive oxygen species (ROS) levels. MAIN METHODS Sprague-Dawley rats were used to detect the retinal expressions of PRDXs1-6. Pig pericytes cultured in high-glucose medium were used to monitor the protective effect of PRDX5 and 6 against high-glucose-associated change. Recombinant PRDX5 and 6 proteins were linked to the Trans-Activating Transduction (TAT) domain from HIV-1 TAT protein for their efficient delivery into cells/tissues. KEY FINDINGS We found higher expression of PRDX5 and 6 mRNAs and PRDX5 and 6 proteins in retina than the other Prdxs (Prdx1-4). Western blotting affirmed the intracellular presence of TAT-linked proteins and revealed the efficient transduction of TAT-HA-PRDX5 and 6 in these cells. Extrinsic supply of TAT-HA-PRDX5 and 6 proteins inhibited the oxidative stress-induced DNA damage after high-glucose exposure in pig pericytes. The cell survival and apoptosis assay revealed that extrinsic supply of TAT-HA-PRDX5 and 6 proteins was responsible for inhibiting hyperglycemia-induced pericyte apoptosis. SIGNIFICANCE Results suggest that delivery of PRDX5 and 6 might protect hyperglycemia-induced pericyte loss to inhibit oxidative stress.
Collapse
Affiliation(s)
- Eri Kubo
- Department of Ophthalmology, University of Fukui, Fukui, Japan.
| | | | | | | |
Collapse
|
50
|
Schubert D, Herrera F, Cumming R, Read J, Low W, Maher P, Fischer WH. Neural cells secrete a unique repertoire of proteins. J Neurochem 2009; 109:427-35. [PMID: 19200335 DOI: 10.1111/j.1471-4159.2009.05968.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Proteins that are released from cells consist of those in the extracellular matrix, as well as extracellular signaling and adhesion molecules. The majority of these extracellular proteins are, however, unknown. To determine their identity, we have used a proteomics approach to define proteins released from neurons, astrocytes and neural precursor cells. Using two-dimensional gels and liquid chromatography/mass spectrometry technology, it is shown that while astrocytes release a relatively small number of proteins, neurons and neuronal precursor cells release a larger number of proteins with more functional diversity. Although there is overlap between the different cell types, the exact composition of the extracellular protein pool is unique for each cell population. The various subsets of extracellular neural proteins include those involved in cellular Redox regulation and chaperones. In addition, many proteolytic enzymes are found outside of the cell. These data show that the extracellular space within the nervous system has a more diverse protein composition than previously thought.
Collapse
Affiliation(s)
- David Schubert
- The Salk Institute for Biological Studies, La Jolla, California 92037-1099, USA
| | | | | | | | | | | | | |
Collapse
|