1
|
Eaton L, Wang T, Roy M, Pamenter ME. Naked Mole-Rat Cortex Maintains Reactive Oxygen Species Homeostasis During In Vitro Hypoxia or Ischemia and Reperfusion. Curr Neuropharmacol 2023; 21:1450-1461. [PMID: 35339183 PMCID: PMC10324332 DOI: 10.2174/1570159x20666220327220929] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/07/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022] Open
Abstract
Neuronal injury during acute hypoxia, ischemia, and following reperfusion are partially attributable to oxidative damage caused by deleterious fluctuations of reactive oxygen species (ROS). In particular, mitochondrial superoxide (O2•-) production is believed to upsurge during lowoxygen conditions and also following reperfusion, before being dismutated to H2O2 and released into the cell. However, disruptions of redox homeostasis may be beneficially attenuated in the brain of hypoxia-tolerant species, such as the naked mole-rat (NMR, Heterocephalus glaber). As such, we hypothesized that ROS homeostasis is better maintained in the brain of NMRs during severe hypoxic/ ischemic insults and following reperfusion. We predicted that NMR brain would not exhibit substantial fluctuations in ROS during hypoxia or reoxygenation, unlike previous reports from hypoxiaintolerant mouse brain. To test this hypothesis, we measured cortical ROS flux using corrected total cell fluorescence measurements from live brain slices loaded with the MitoSOX red superoxide (O2•-) indicator or chloromethyl 2',7'-dichlorodihydrofluorescein diacetate (CM-H2-DCFDA; which fluoresces with whole-cell hydrogen peroxide (H2O2) production) during various low-oxygen treatments, exogenous oxidative stress, and reperfusion. We found that NMR cortex maintained ROS homeostasis during low-oxygen conditions, while mouse cortex exhibited a ~40% increase and a ~30% decrease in mitochondrial O2•- and cellular H2O2 production, respectively. Mitochondrial ROS homeostasis in NMRs was only disrupted following sodium cyanide application, which was similarly observed in mice. Our results suggest that NMRs have evolved strategies to maintain ROS homeostasis during acute bouts of hypoxia and reoxygenation, potentially as an adaptation to life in an intermittently hypoxic environment.
Collapse
Affiliation(s)
- Liam Eaton
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Tina Wang
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Maria Roy
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Matthew E. Pamenter
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
2
|
Berkowitz BA, Podolsky RH, Childers KL, Gow A, Schneider BL, Lloyd SC, Bosse KE, Conti AC, Roberts R, Berri AM, Graffice E, Sinan K, Eliwat W, Shen Y. Age-related murine hippocampal CA1 laminae oxidative stress measured in vivo by QUEnch-assiSTed (QUEST) MRI: impact of isoflurane anesthesia. GeroScience 2020; 42:563-574. [PMID: 31981008 PMCID: PMC7205849 DOI: 10.1007/s11357-020-00162-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Age-related impairments in spatial learning and memory often precede non-familial neurodegenerative disease. Ex vivo studies suggest that physiologic age-related oxidative stress in hippocampus area CA1 may contribute to prodromal spatial disorientation and to morbidity. Yet, conventional blood or cerebrospinal fluid assays appear insufficient for early detection or management of oxidative stress within CA1 sub-regions in vivo. Here, we address this biomarker problem using a non-invasive MRI index of CA1 laminae oxidative stress based on reduction in R1 (= 1/T1) after anti-oxidant administration. An R1 reduction reflects quenching of continuous and excessive production of endogenous paramagnetic free radicals. Careful motion-correction image acquisition, and avoiding repeated exposure to isoflurane, facilitates detection of hippocampus CA1 laminae oxidative stress with QUEnch-assiSTed (QUEST) MRI. Intriguingly, age- and isoflurane-related oxidative stress is localized to the stratum lacunosum of the CA1 region. Our data raise the possibility of using QUEST MRI and FDA-approved anti-oxidants to remediate spatial disorientation and later neurodegeneration with age in animals and humans.
Collapse
Affiliation(s)
- Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA.
| | - Robert H Podolsky
- Beaumont Research Institute, Beaumont Health, Royal Oak, MI, 48073, USA
| | | | - Alexander Gow
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Brandy L Schneider
- John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Deptarment of Neurosurgery, School of Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Scott C Lloyd
- John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Deptarment of Neurosurgery, School of Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Kelly E Bosse
- John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Deptarment of Neurosurgery, School of Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Alana C Conti
- John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Deptarment of Neurosurgery, School of Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Robin Roberts
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Ali M Berri
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Emma Graffice
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Kenan Sinan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Waleed Eliwat
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Yimin Shen
- Department of Radiology, School of Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
3
|
Köles L, Szepesy J, Berekméri E, Zelles T. Purinergic Signaling and Cochlear Injury-Targeting the Immune System? Int J Mol Sci 2019; 20:ijms20122979. [PMID: 31216722 PMCID: PMC6627352 DOI: 10.3390/ijms20122979] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 02/06/2023] Open
Abstract
Hearing impairment is the most common sensory deficit, affecting more than 400 million people worldwide. Sensorineural hearing losses currently lack any specific or efficient pharmacotherapy largely due to the insufficient knowledge of the pathomechanism. Purinergic signaling plays a substantial role in cochlear (patho)physiology. P2 (ionotropic P2X and the metabotropic P2Y) as well as adenosine receptors expressed on cochlear sensory and non-sensory cells are involved mostly in protective mechanisms of the cochlea. They are implicated in the sensitivity adjustment of the receptor cells by a K+ shunt and can attenuate the cochlear amplification by modifying cochlear micromechanics. Cochlear blood flow is also regulated by purines. Here, we propose to comprehend this field with the purine-immune interactions in the cochlea. The role of harmful immune mechanisms in sensorineural hearing losses has been emerging in the horizon of cochlear pathologies. In addition to decreasing hearing sensitivity and increasing cochlear blood supply, influencing the immune system can be the additional avenue for pharmacological targeting of purinergic signaling in the cochlea. Elucidating this complexity of purinergic effects on cochlear functions is necessary and it can result in development of new therapeutic approaches in hearing disabilities, especially in the noise-induced ones.
Collapse
Affiliation(s)
- László Köles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
| | - Judit Szepesy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
| | - Eszter Berekméri
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
- Department of Ecology, University of Veterinary Medicine, H-1078 Budapest, Hungary.
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, H-1083 Budapest, Hungary.
| |
Collapse
|
4
|
Yuan Y, Zheng Z. Geniposide protects PC-12 cells against oxygen and glucose deprivation-induced injury by up-regulation of long-noncoding RNA H19. Life Sci 2018; 216:176-182. [PMID: 30472296 DOI: 10.1016/j.lfs.2018.11.047] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/16/2018] [Accepted: 11/21/2018] [Indexed: 01/09/2023]
Abstract
AIMS Hypoxic-ischemic encephalopathy (HIE) is a common brain injury disease in neonates, which can lead to neonatal disability and death. Geniposide (GEN) is a main ingredient of Gardenia jasminoides, whose anti-tumor, anti-inflammatory and anti-apoptotic effects have been reported in various diseases. However, the effect of GEN on HIE remains uninvestigated. This study aimed to clarify the protective effect of GEN on PC-12 cells against oxygen and glucose deprivation (OGD)-induced injury. MAIN METHODS PC-12 cells were subjected to OGD treatment, cell viability, cell cycle-associated factors, apoptosis and apoptosis-associated factors were then determined. The different concentrations of GEN were used to stimulate PC-12 cells, and the effects of GEN on cell proliferation and apoptosis in OGD-treatment cells were assessed. Subsequently, relative expression level of H19 was analyzed in PC-12 cells after treatment with GEN. After this, si-H19 was transfected into PC-12 cells to explore the regulatory effect of H19 on PC-12 cells after treatment with GEN and OGD. Besides, PI3K/AKT and Wnt/β-catenin pathways were examined by western blot assay. KEY FINDINGS OGD significantly inhibited cell viability, decreased CyclinD1, CDK4 and CDK6 expression, induced apoptosis and up-regulated Cleaved-Caspase-9/-7/-3 expression in PC-12 cells. GEN treatment obviously alleviated OGD-induced cell injury. Additionally, H19 expression was up-regulated by GEN, and H19 knockdown reversed the protective effect of GEN on PC-12 cells against OGD-induced injury. Finally, GEN activated PI3K/AKT and Wnt/β-catenin pathways by regulating H19 in OGD-insulted PC-12 cells. SIGNIFICANCE The findings suggested that GEN protected PC-12 cells against OGD-induced injury by up-regulation of H19.
Collapse
Affiliation(s)
- Yanran Yuan
- Department of Children Rehabilitation, Jining No.1 People's Hospital, Jining 272011, China; Jining No.1 People's Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Zebao Zheng
- Department of Children Rehabilitation, Jining No.1 People's Hospital, Jining 272011, China; Jining No.1 People's Hospital of Jining Medical University, Jining Medical University, Jining 272000, China.
| |
Collapse
|
5
|
Yin B, Barrionuevo G, Weber SG. Mitochondrial GSH Systems in CA1 Pyramidal Cells and Astrocytes React Differently during Oxygen-Glucose Deprivation and Reperfusion. ACS Chem Neurosci 2018; 9:738-748. [PMID: 29172440 DOI: 10.1021/acschemneuro.7b00369] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Pyramidal cells and astrocytes have differential susceptibility to oxygen-glucose deprivation and reperfusion (OGD-RP). It is known that excessive reactive oxygen species (ROS) in mitochondria initiates cell death, while glutathione (GSH) is one of the major defenses against ROS. Although it is known that astrocytes contain a higher concentration of GSH than neurons, and that astrocytes can provide neurons with GSH, we are unaware of a detailed and quantitative examination of the dynamic changes in the mitochondrial GSH system in the two cell types during OGD-RP. Here, we determined mitochondrial membrane potential and the degrees of oxidation of the mitochondrially targeted roGFP-based sensors for hydrogen peroxide (OxDP) and GSH (OxDG). We also developed a method to estimate the mitochondrial GSH (mGSH) concentration in single cells in the CA1 region of organotypic hippocampal slice cultures at several time-points during OGD-RP. We find that mitochondrial membrane potential drops in pyramidal cells during OGD while it is relatively stable in astrocytes. In both types of cell, the mitochondrial membrane potential decreases during RP. During OGD-RP, mitochondrial peroxide levels are the same. Astrocytic mGSH is more than four times higher than pyramidal cell mGSH (3.2 vs 0.7 mM). Astrocytic mGSH is drained from mitochondria during OGD, whereas in pyramidal cells it remains fairly constant. OxDGSH prior to and during OGD is lower (less oxidized) in pyramidal cells than in astrocytes, but the two nearly converge during RP. The larger changes of redox status in the GSH system in pyramidal cells than astrocytes is an upstream sign of the higher mortality of the pyramidal cells after facing an insult. The pattern of [mGSH] changes in the two cell types could be recognized as another mechanism by which astrocytes protect neurons from transient, extreme conditions.
Collapse
Affiliation(s)
- Bocheng Yin
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Germán Barrionuevo
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Stephen G. Weber
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
6
|
Abstract
Baicalin, a plant-derived flavonoid, has been reported to exert neuroprotective effects on ischemia-like or excitotoxic injury. To confirm this function and explore the possible mechanism, we investigated the protective effect of baicalin on an in-vitro model of ischemia (oxygen-glucose deprivation-treated endothelial cell). In the present study, we found that baicalin (100 μM) inhibited cell death, reduced cell membrane damage, and maintained the integrity of the nucleus. Flow cytometric analysis and Hoechst 33258/propidium iodide double staining results showed that the necroptosis ratio decreased with baicalin treatment. Western blot analysis showed that baicalin regulated the expression of RIP-1 and RIP-3 in bEnd.3 cells and the use of detection kits showed that baicalin inhibited the production of reactive oxygen species and malondialdehyde, and increased the activity of superoxide dismutase in oxygen-glucose deprivation-treated bEnd.3 cells. These results indicated that baicalin effectively alleviated the oxidative stress, decreased the proportion of cells undergoing necrosis, and reduced cell damage.
Collapse
|
7
|
Ma J, Yang Q, Wei Y, Yang Y, Ji C, Hu X, Mai S, Kuang S, Tian X, Luo Y, Liang G, Yang J. Effect of the PGD2-DP signaling pathway on primary cultured rat hippocampal neuron injury caused by aluminum overload. Sci Rep 2016; 6:24646. [PMID: 27089935 PMCID: PMC4835855 DOI: 10.1038/srep24646] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/01/2016] [Indexed: 12/16/2022] Open
Abstract
In the present study, the agonists and antagonists of DP receptor were used to examine whether the PGD2-DP signaling pathway affects neuronal function. Primary cultured hippocampal neuron was prepared and treated with aluminum maltolate (100 μM) to establish the neuronal damage model. PGD2 and cAMP content was detected by ELISA. L-PGDS and DPs mRNA and protein expression were measured by RT-PCR and Western blotting, respectively. The aluminium-load neuron was treated with the DP1 agonist BW245C, the DP1 antagonist BWA868C, the DP2 agonist DK-PGD2, and the DP2 antagonist CAY10471, respectively. Neuronal pathomorphology was observed using H-E staining. The cell viability and the lactate dehydrogenase leakage rates of neurons were measured with MTT and LDH kit, respectively. Ca2+ level was detected by Fluo-3/AM. In the model group, the MTT values obviously decreased; LDH leakage rates and PGD2 content increased significantly; L-PGDS, DP1 mRNA and protein expressions increased, and DP2 level decreased. BW245C reduced the Ca2+ fluorescence intensity and protected the neurons. DK-PGD2 increased the intensity of Ca2+ fluorescence, while CAY10471 had the opposite effect. In conclusion, contrary to the effect of DP2, the PGD2-DP1 signaling pathway protects against the primary cultured rat hippocampal neuronal injury caused by aluminum overload.
Collapse
Affiliation(s)
- Jie Ma
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Qunfang Yang
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Yuling Wei
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Yang Yang
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Chaonan Ji
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Xinyue Hu
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Shaoshan Mai
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Shengnan Kuang
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Xiaoyan Tian
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Ying Luo
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Guojuan Liang
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Junqing Yang
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| |
Collapse
|
8
|
Maixner DW, Yan X, Hooks SB, Weng HR. AMPKα1 knockout enhances nociceptive behaviors and spinal glutamatergic synaptic activities via production of reactive oxygen species in the spinal dorsal horn. Neuroscience 2016; 326:158-169. [PMID: 27058143 DOI: 10.1016/j.neuroscience.2016.03.061] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/25/2016] [Accepted: 03/28/2016] [Indexed: 12/15/2022]
Abstract
Emerging studies have shown that pharmacological activation of adenosine monophosphate-activated protein kinase (AMPK) produces potent analgesic effects in different animal pain models. Currently, the spinal molecular and synaptic mechanism by which AMPK regulates the pain signaling system remains unclear. To address this issue, we utilized the Cre-LoxP system to conditionally knockout the AMPKα1 gene in the nervous system of mice. We demonstrated that AMPKα1 is imperative for maintaining normal nociception, and mice deficient for AMPKα1 exhibit mechanical allodynia. This is concomitantly associated with increased glutamatergic synaptic activities in neurons located in the superficial spinal dorsal horn, which results from the increased glutamate release from presynaptic terminals and function of ligand-gated glutamate receptors at the postsynaptic neurons. Additionally, AMPKα1 knockout mice have increased activities of extracellular signal-regulated kinases (ERK) and p38 mitogen-activated protein kinases (p38), as well as elevated levels of interleukin-1β (IL-1β), reactive oxygen species (ROS), and heme oxygenase 1 (HO-1) in the spinal dorsal horn. Systemic administration of a non-specific ROS scavenger (phenyl-N-tert-butylnitrone, PBN) or a HO-1 activator (Cobalt protoporphyrin IX, CoPP) attenuated allodynia in AMPKα1 knockout mice. Bath-perfusion of the ROS scavenger or HO-1 activator effectively attenuated the increased ROS levels and glutamatergic synaptic activities in the spinal dorsal horn. Our findings suggest that ROS are the key down-stream signaling molecules mediating the behavioral hypersensitivity in AMPKα1 knockout mice. Thus, targeting AMPKα1 may represent an effective approach for the treatment of pathological pain conditions associated with neuroinflammation at the spinal dorsal horn.
Collapse
Affiliation(s)
- Dylan W Maixner
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia 30602, USA
| | - Xisheng Yan
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia 30602, USA.,Department of Cardiovascular Medicine, The Third Hospital of Wuhan, Wuhan 430074, Hubei Province, China.,Department of Endocrinology and Metabolism, Shanghai Tenth People's hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Shelley B Hooks
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia 30602, USA
| | - Han-Rong Weng
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia 30602, USA
| |
Collapse
|
9
|
Yin B, Barrionuevo G, Weber SG. Optimized real-time monitoring of glutathione redox status in single pyramidal neurons in organotypic hippocampal slices during oxygen-glucose deprivation and reperfusion. ACS Chem Neurosci 2015; 6:1838-48. [PMID: 26291433 DOI: 10.1021/acschemneuro.5b00186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A redox-sensitive Grx1-roGFP2 fusion protein was introduced by transfection into single pyramidal neurons in the CA1 subfield of organotypic hippocampal slice cultures (OHSCs). We assessed changes in the GSH system in neuronal cytoplasm and mitochondria during oxygen-glucose deprivation and reperfusion (OGD/RP), an in vitro model of stroke. Pyramidal cells in a narrow range of depths below the surface of the OHSC were transfected by gene gun or single-cell electroporation with cyto- or mito-Grx1-roGFP2. To mimic the conditions of acute stroke, we developed an optimized superfusion system with the capability of rapid and reproducible exchange of the solution bathing the OHSCs. Measurements of pO2 as a function of tissue depth show that in the region containing the transfected cells, the pO2 is well-controlled. We also found that the pO2 changes on the same time scale as changes in intracranial pressure, cerebral blood flow, and pO2 during acute stroke. Determining the reduction potential, EGSH, from the ratiometric fluorescence signal requires an absolute intensity measurement during calibration of the Grx1-roGFP2. Using the signal from cotransfected tdTomato as an internal standard during calibration improves quantitative measurements of Grx1-roGFP2 redox status and allows EGSH to be determined. EGSH becomes more reducing during OGD and more oxidizing during RP in mitochondria while changes in cytoplasm are not significant compared with controls.
Collapse
Affiliation(s)
- Bocheng Yin
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Germán Barrionuevo
- Department
of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Stephen G. Weber
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
10
|
Brassai A, Suvanjeiev RG, Bán EG, Lakatos M. Role of synaptic and nonsynaptic glutamate receptors in ischaemia induced neurotoxicity. Brain Res Bull 2015; 112:1-6. [DOI: 10.1016/j.brainresbull.2014.12.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/10/2014] [Accepted: 12/12/2014] [Indexed: 11/17/2022]
|
11
|
Lim DC, Brady DC, Po P, Chuang LP, Marcondes L, Kim EY, Keenan BT, Guo X, Maislin G, Galante RJ, Pack AI. Simulating obstructive sleep apnea patients' oxygenation characteristics into a mouse model of cyclical intermittent hypoxia. J Appl Physiol (1985) 2014; 118:544-57. [PMID: 25429097 DOI: 10.1152/japplphysiol.00629.2014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mouse models of cyclical intermittent hypoxia (CIH) are used to study the consequences of both hypoxia and oxidative stress in obstructive sleep apnea (OSA). Whether or not a mouse model of CIH that simulates OSA patients' oxygenation characteristics would translate into improved patient care remains unanswered. First we identified oxygenation characteristics using the desaturation and resaturation time in 47 OSA subjects from the Molecular Signatures of Obstructive Sleep Apnea Cohort (MSOSA). We observe that a cycle of intermittent hypoxia is not sinusoidal; specifically, desaturation time increases in an almost linear relationship to the degree of hypoxia (nadir), whereas resaturation time is somewhat constant (∼15 s), irrespective of the nadir. Second, we modified the Hycon mouse model of CIH to accommodate a 15-s resaturation time. Using this modified CIH model, we explored whether a short resaturation schedule (15 s), which includes the characteristics of OSA patients, had a different effect on levels of oxidative stress (i.e., urinary 8,12-iso-iPF2α-VI levels) compared with sham and a long resaturation schedule (90 s), a schedule that is not uncommon in rodent models of CIH. Results suggest that shorter resaturation time may result in a higher level of 8,12-iso-iPF2α-VI compared with long resaturation or sham conditions. Therefore, simulating the rodent model of CIH to reflect this and other OSA patients' oxygenation characteristics may be worthy of consideration to better understand the effects of hypoxia, oxidative stress, and their interactions.
Collapse
Affiliation(s)
- Diane C Lim
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania;
| | - Daniel C Brady
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Pengse Po
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Li Pang Chuang
- Department of Thoracic Medicine and Department of Sleep Center, Chang Gung Memorial Hospital, Taipei, Taiwan and Graduate Institute of Clinical Medical Sciences, Chang Gung University, Tauyan, Taiwan; and
| | | | - Emily Y Kim
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brendan T Keenan
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaofeng Guo
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Greg Maislin
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Raymond J Galante
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Allan I Pack
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
12
|
Huang T, Gao D, Jiang X, Hu S, Zhang L, Fei Z. Resveratrol inhibits oxygen-glucose deprivation-induced MMP-3 expression and cell apoptosis in primary cortical cells via the NF-κB pathway. Mol Med Rep 2014; 10:1065-71. [PMID: 24840287 DOI: 10.3892/mmr.2014.2239] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 03/18/2014] [Indexed: 11/05/2022] Open
Abstract
Resveratrol (Res) or trans-3,4',5-trihydroxystilbene, has been proven to exert neuroprotective effects in cerebral ischemia. The aim of the present study was to investigate whether Res has neuroprotective effects in primary cortical neurons subjected to transient oxygen-glucose deprivation (OGD) via inhibiting the expression of the gene encoding stromelysin-1, also known as matrix metalloproteinase-3 (MMP-3), and via inhibiting cell apoptosis. Primary cortical cells were exposed to OGD, followed by reoxygenation to induce transient ischemia. Res (50 µM) was added into the culture medium during transient ischemia in the presence or absence of the nuclear factor (NF)-κB inhibitor pyrrolidine dithiocarbamate (PDTC; 10 µM) or 500 µM of the nitric oxide (NO) donor NOC-18. Cell viability was assessed using the tetrazolium reduction (MTT) assay. Cell apoptosis was evaluated by flow cytometry. MMP-3 expression was analyzed by western blot and reverse transcription-polymerase chain reaction (RT-PCR), while the levels of inducible NO synthase (iNOS), NF-κB, caspase-3, cleaved caspase-3, B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (Bax) were assayed by western blot. NO was detected using a spectrophotometric method. We found that the cellular viability was significantly reduced by transient OGD and that this effect was reversed by Res treatment. In addition, OGD was shown to induce cell apoptosis, the expression of Bax and the activation of caspase-3, and inhibit the expression of Bcl-2, and these effects were also reversed by Res treatment. Res treatment significantly reduced the level of MMP-3 that was induced by transient OGD, via inhibition of NF-κB expression. In addition, Res inhibited iNOS expression and NO synthesis that were induced by OGD. MMP-3 expression induced by NO was attenuated by Res treatment and was partially restored by exogenous NO using NOC-18. Taken together, these findings indicate that OGD induces apoptosis through canonical apoptosis signaling and by modulating the expression of MMP-3; Res can reverse the OGD-induced MMP-3 expression and cell apoptosis via the NF-κB-iNOS/NO pathway. Therefore, Res may be a promising agent for the treatment of neuronal injury associated with stroke.
Collapse
Affiliation(s)
- Tao Huang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Dakuan Gao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Shijie Hu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
13
|
Navarro-Yepes J, Zavala-Flores L, Anandhan A, Wang F, Skotak M, Chandra N, Li M, Pappa A, Martinez-Fong D, Del Razo LM, Quintanilla-Vega B, Franco R. Antioxidant gene therapy against neuronal cell death. Pharmacol Ther 2014; 142:206-30. [PMID: 24333264 PMCID: PMC3959583 DOI: 10.1016/j.pharmthera.2013.12.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022]
Abstract
Oxidative stress is a common hallmark of neuronal cell death associated with neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, as well as brain stroke/ischemia and traumatic brain injury. Increased accumulation of reactive species of both oxygen (ROS) and nitrogen (RNS) has been implicated in mitochondrial dysfunction, energy impairment, alterations in metal homeostasis and accumulation of aggregated proteins observed in neurodegenerative disorders, which lead to the activation/modulation of cell death mechanisms that include apoptotic, necrotic and autophagic pathways. Thus, the design of novel antioxidant strategies to selectively target oxidative stress and redox imbalance might represent important therapeutic approaches against neurological disorders. This work reviews the evidence demonstrating the ability of genetically encoded antioxidant systems to selectively counteract neuronal cell loss in neurodegenerative diseases and ischemic brain damage. Because gene therapy approaches to treat inherited and acquired disorders offer many unique advantages over conventional therapeutic approaches, we discussed basic research/clinical evidence and the potential of virus-mediated gene delivery techniques for antioxidant gene therapy.
Collapse
Affiliation(s)
- Juliana Navarro-Yepes
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; Department of Toxicology, CINVESTAV-IPN, Mexico City, Mexico
| | - Laura Zavala-Flores
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Annadurai Anandhan
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Fang Wang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Maciej Skotak
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Namas Chandra
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Ming Li
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, Alexandroupolis, Greece
| | - Daniel Martinez-Fong
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City, Mexico
| | | | | | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| |
Collapse
|
14
|
Koncz I, Szász BK, Szabó SI, Kiss JP, Mike A, Lendvai B, Sylvester Vizi E, Zelles T. The tricyclic antidepressant desipramine inhibited the neurotoxic, kainate-induced [Ca(2+)]i increases in CA1 pyramidal cells in acute hippocampal slices. Brain Res Bull 2014; 104:42-51. [PMID: 24742525 DOI: 10.1016/j.brainresbull.2014.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/24/2014] [Accepted: 04/01/2014] [Indexed: 12/18/2022]
Abstract
Kainate (KA), used for modelling neurodegenerative diseases, evokes excitotoxicity. However, the precise mechanism of KA-evoked [Ca(2+)]i increase is unexplored, especially in acute brain slice preparations. We used [Ca(2+)]i imaging and patch clamp electrophysiology to decipher the mechanism of KA-evoked [Ca(2+)]i rise and its inhibition by the tricyclic antidepressant desipramine (DMI) in CA1 pyramidal cells in rat hippocampal slices and in cultured hippocampal cells. The effect of KA was dose-dependent and relied totally on extracellular Ca(2+). The lack of effect of dl-2-amino-5-phosphonopentanoic acid (AP-5) and abolishment of the response by 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) suggested the involvement of non-N-methyl-d-aspartate receptors (non-NMDARs). The predominant role of the Ca(2+)-impermeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptors (AMPARs) in the initiation of the Ca(2+) response was supported by the inhibitory effect of the selective AMPAR antagonist GYKI 53655 and the ineffectiveness of 1-naphthyl acetylspermine (NASPM), an inhibitor of the Ca(2+)-permeable AMPARs. The voltage-gated Ca(2+) channels (VGCC), blocked by ω-Conotoxin MVIIC+nifedipine+NiCl2, contributed to the [Ca(2+)]i rise. VGCCs were also involved, similarly to AMPAR current, in the KA-evoked depolarisation. Inhibition of voltage-gated Na(+) channels (VGSCs; tetrodotoxin, TTX) did not affect the depolarisation of pyramidal cells but blocked the depolarisation-evoked action potential bursts and reduced the Ca(2+) response. The tricyclic antidepressant DMI inhibited the KA-evoked [Ca(2+)]i rise in a dose-dependent manner. It directly attenuated the AMPA-/KAR current, but its more potent inhibition on the Ca(2+) response supports additional effect on VGCCs, VGSCs and Na(+)/Ca(2+) exchangers. The multitarget action on decisive players of excitotoxicity holds out more promise in clinical therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- István Koncz
- Department of Pharmacology & Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Bernadett K Szász
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Szilárd I Szabó
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | | | - Arpád Mike
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Balázs Lendvai
- Gedeon Richter Plc., Pharmacology and Drug Safety Department, Budapest, Hungary
| | - E Sylvester Vizi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
15
|
Zhang J, Malik A, Choi H, Ko R, Dissing-Olesen L, MacVicar B. Microglial CR3 Activation Triggers Long-Term Synaptic Depression in the Hippocampus via NADPH Oxidase. Neuron 2014; 82:195-207. [DOI: 10.1016/j.neuron.2014.01.043] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2014] [Indexed: 12/28/2022]
|
16
|
Maraula G, Traini C, Mello T, Coppi E, Galli A, Pedata F, Pugliese AM. Effects of oxygen and glucose deprivation on synaptic transmission in rat dentate gyrus: role of A2A adenosine receptors. Neuropharmacology 2012; 67:511-20. [PMID: 23261865 DOI: 10.1016/j.neuropharm.2012.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 11/29/2012] [Accepted: 12/03/2012] [Indexed: 01/01/2023]
Abstract
The hippocampus is comprised of two distinct subfields that show different responses to hypoxic-ischemic brain injury: the CA1 region is particularly susceptible whereas the dentate gyrus (DG) is quite resistant. Our aim was to determine the synaptic and proliferative response of the DG to severe oxygen and glucose deprivation (OGD) in acute rat hippocampal slices and to investigate the contribution of A(2A) adenosine receptor antagonism to recovery of synaptic activity after OGD. Extracellular recordings of field excitatory post-synaptic potentials (fEPSPs) in granule cells of the DG in brain slices prepared from male Wistar rats were used. A 9-min OGD is needed in the DG to always induce the appearance of anoxic depolarization (AD) and the irreversible block of synaptic activity, as recorded up to 24 h from the end of the insult, whereas only 7-min OGD is required in the CA1 region. Selective antagonism of A(2A) adenosine receptors by ZM241385 significantly prevents or delays the appearance of AD and protects from the irreversible block of neurotransmission induced by 9-min OGD in the DG. The effects of 9-min OGD on proliferation and maturation of cells localized in the subgranular zone of DG in slices prepared from 5-bromo-2'-deoxyuridine (BrdU) treated rats was investigated. Slices were further incubated with an immature neuronal marker, doublecortin (DCX). The number of BrdU(+) cells was significantly decreased 6 h after 9-min OGD and this effect was antagonized by ZM241385. After 24 h from the end of 9-min OGD, the number of BrdU(+) cells returned to that found before OGD and increased arborization of tertiary dendrites of DCX(+) cells was observed. The adenosine A(2A) antagonist ZM241385 protects from synaptic failure and from decreased proliferation of immature neuronal cells at a precocious time after OGD.
Collapse
Affiliation(s)
- Giovanna Maraula
- Department of Preclinical and Clinical Pharmacology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | | | | | | | | | | | | |
Collapse
|
17
|
Lin YP, Chen TY, Tseng HW, Lee MH, Chen ST. Chemical and biological evaluation of nephrocizin in protecting nerve growth factor-differentiated PC12 cells by 6-hydroxydopamine-induced neurotoxicity. PHYTOCHEMISTRY 2012; 84:102-115. [PMID: 22954731 DOI: 10.1016/j.phytochem.2012.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 07/12/2012] [Accepted: 07/25/2012] [Indexed: 06/01/2023]
Abstract
The neurotoxin 6-hydroxydopamine (6-OHDA) has been widely used to generate an experimental model of Parkinson's disease. This model is crucial in the search for compounds that diminish 6-OHDA-induced nerve growth factor (NGF)-differentiated PC12 cell death. Nephrocizin (luteolin-7-O-β-D-glucopyranoside), a flavone glycoside, was isolated from widely distributed plants. The protective effects of pre-treatment with nephrocizin on the induced neurotoxicity in PC12 cells by 6-OHDA and its oxidative products, H₂O₂-, and p-quinone, were evaluated herein. Nephrocizin promoted cell viability, scavenged ROS-related products, increased cellular glutathione (GSH) levels, and reduced caspase-3 and -8 activities in 6-OHDA-, H₂O₂-, or p-quinone-treated PC12 cells. Furthermore, nephrocizin-conjugated metabolites in PC12 cells were identified with the boronate-affinity method and LC-MS technology, and preferential regioselectivity at the C2' and C5' positions by the nephrocizin-GSH (or NAC) adduct method was observed. These lines of evidence established that nephrocizin could form a dimer to diminish the intracellular ROS. These results demonstrate the first neuroprotective mechanism of nephrocizin against 6-OHDA-, H₂O₂- or p-quinone-induced cytotoxicity in PC12 cells via chemical and biological studies. These dietary antioxidants are potential candidates for use in intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Pei Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan, ROC
| | | | | | | | | |
Collapse
|
18
|
Laranjinha J, Santos RM, Lourenço CF, Ledo A, Barbosa RM. Nitric oxide signaling in the brain: translation of dynamics into respiration control and neurovascular coupling. Ann N Y Acad Sci 2012; 1259:10-8. [DOI: 10.1111/j.1749-6632.2012.06582.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
19
|
Yang EJ, Kim SI, Park SY, Bang HY, Jeong JH, So JH, Rhee IK, Song KS. Fermentation enhances the in vitro antioxidative effect of onion (Allium cepa) via an increase in quercetin content. Food Chem Toxicol 2012; 50:2042-8. [DOI: 10.1016/j.fct.2012.03.065] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 03/21/2012] [Accepted: 03/22/2012] [Indexed: 11/16/2022]
|
20
|
Martínez-Romero R, Cañuelo A, Siles E, Oliver FJ, Martínez-Lara E. Nitric oxide modulates hypoxia-inducible factor-1 and poly(ADP-ribose) polymerase-1 cross talk in response to hypobaric hypoxia. J Appl Physiol (1985) 2012; 112:816-23. [DOI: 10.1152/japplphysiol.00898.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The physiological response to hypobaric hypoxia represents a complex network of biochemical pathways in which the nitrergic system plays an important role. Previous studies have provided evidence for an interplay between the hypoxia-inducible factor-1 (HIF-1) and poly(ADP-ribose) polymerase-1 (PARP-1) under hypoxia. Here, we evaluate the potential involvement of nitric oxide (NO) in the cross talk between these two proteins. With this aim, we studied comparatively the effect of pharmacological inhibitors of NO production or PARP activity in the response of the mouse cerebral cortex to 4 h of exposure to a simulated altitude of 31,000 ft. Particularly, we analyzed the NO and reactive oxygen species production, the expression of NO synthase (NOS) isoforms, PARP-1 activity, HIF-1α expression and HIF-1 transcriptional activity, the protein level of the factor inhibiting HIF, and, finally, beclin-1 and fractin expression, as markers of cellular damage. Our results demonstrate that the reduction of NO level did not affect reactive oxygen species production but significantly 1) dampened the posthypoxic increase in neuronal NOS and inducible NOS expression without altering endothelial NOS protein level; 2) prevented PARP activation; 3) decreased HIF-1α response to hypoxia; 4) achieved a higher long-term HIF-1 transcriptional activity by reducing factor inhibiting HIF expression; and 5) reduced hypoxic damage. The pharmacological inhibition of PARP reproduced the NOS expression pattern and the HIF-1α response observed in NOS-inhibited mice, supporting its involvement in the NO-dependent regulation of hypoxia. As a whole, these results provide new data about the molecular mechanism underlying the beneficial effects of controlling NO production under hypobaric hypoxic conditions.
Collapse
Affiliation(s)
| | - Ana Cañuelo
- Department of Experimental Biology, University of Jaén, Jaén; and
| | - Eva Siles
- Department of Experimental Biology, University of Jaén, Jaén; and
| | - F. Javier Oliver
- Institute of Parasitology and Biomedicine, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | | |
Collapse
|
21
|
Lendvai B, Halmos GB, Polony G, Kapocsi J, Horváth T, Aller M, Sylvester Vizi E, Zelles T. Chemical neuroprotection in the cochlea: The modulation of dopamine release from lateral olivocochlear efferents. Neurochem Int 2011; 59:150-8. [DOI: 10.1016/j.neuint.2011.05.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 05/12/2011] [Accepted: 05/17/2011] [Indexed: 01/16/2023]
|
22
|
Cordycepin protects against cerebral ischemia/reperfusion injury in vivo and in vitro. Eur J Pharmacol 2011; 664:20-8. [PMID: 21554870 DOI: 10.1016/j.ejphar.2011.04.052] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2010] [Revised: 04/12/2011] [Accepted: 04/18/2011] [Indexed: 12/23/2022]
Abstract
Cordycepin, (3'-deoxyadenosine), a bioactive compound of Cordyceps militaris, has been shown to exhibit many pharmacological actions, such as anti-inflammatory, antioxidative and anticancer activities. Little is known about the neuroprotective action of cordycepin as well as its molecular mechanisms. In this study, cordycepin was investigated for its neuroprotective potential in mice with ischemia following 15 min of the bilateral common carotid artery occlusion and 4h of reperfusion. The effect of cordycepin was also studied in mice brain slices treated with oxygen-glucose deprivation (OGD) injury. Our results showed that cordycepin was able to prevent postischemic neuronal degeneration and brain slice injury. Excitatory amino acids such as glutamate and aspartate in brain homogenized supernatant, which were increased in ischemia/reperfusion group, were detected by high performance liquid chromatography (HPLC). The results showed that cordycepin was able to decrease the extracellular level of glutamate and aspartate significantly. Moreover, cordycepin was able to increase the activity of superoxide dismutase (SOD) and decrease the level of malondialdehyde (MDA), ameliorating the extent of oxidation. Furthermore, matrix metalloproteinase-3(MMP-3), a key enzyme involved in inflammatory reactions, was markedly increased after ischemia reperfusion, whereas cordycepin was able to inhibit its expression obviously. In conclusion, our in vivo and in vitro study showed that cordycepin was able to exert a potent neuroprotective function after cerebral ischemia/reperfusion.
Collapse
|
23
|
Sandhir R, Mehrotra A, Kamboj SS. Lycopene prevents 3-nitropropionic acid-induced mitochondrial oxidative stress and dysfunctions in nervous system. Neurochem Int 2010; 57:579-87. [DOI: 10.1016/j.neuint.2010.07.005] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 07/11/2010] [Accepted: 07/12/2010] [Indexed: 01/13/2023]
|
24
|
Hwang IK, Yoo KY, Kim DW, Lee CH, Choi JH, Kwon YG, Kim YM, Choi SY, Won MH. Changes in the expression of mitochondrial peroxiredoxin and thioredoxin in neurons and glia and their protective effects in experimental cerebral ischemic damage. Free Radic Biol Med 2010; 48:1242-51. [PMID: 20156553 DOI: 10.1016/j.freeradbiomed.2010.02.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Revised: 01/25/2010] [Accepted: 02/07/2010] [Indexed: 11/19/2022]
Abstract
We observed chronological changes in the mitochondrial-specific antioxidant enzymes peroxiredoxin 3 (Prx3) and thioredoxin 2 (Trx2) and their neuroprotective effects in the hippocampal CA1 region after 5 min of transient cerebral ischemia in gerbils. In the sham-operated group, weak Prx3 and Trx2 immunoreactivity was detected in the stratum pyramidale. Prx3 immunoreactivity was increased in pyramidal neurons and expressed in microglia 1 and 3 days, respectively, after ischemia/reperfusion (I/R). Trx2 immunoreactivity in pyramidal neurons increased 30 min and 1 day after I/R and decreased 6 h after I/R. Trx2 immunoreaction was expressed in astrocytes at 3 days postischemia. The intraventricular administration of Prx3 or Prx3/Trx2 (16 microg/20 microl, icv) using an osmotic pump significantly reduced ischemia-induced hyperactivity in a spontaneous motor test and protected CA1 pyramidal neurons from the ischemic damage. In addition, the activation of astrocytes and microglia was decreased in the ischemic CA1 region after Prx3/Trx2 treatment. In addition, treatment with Prx3 or Prx3/Trx2 significantly reduced lipid peroxidation and the release of cytochrome c from mitochondria and cytoplasm in the ischemic CA1 region. These results suggest that changes in the expression of Prx3 and Trx2 in the hippocampal CA1 region after I/R may be associated with the delayed neuronal death of CA1 pyramidal cells induced by transient cerebral ischemia, and that treatment with Prx3 or Prx3/Trx2 in ischemic brains shows a potent neuroprotective effect against ischemic damage by reducing lipid peroxidation and mitochondrial-mediated apoptosis by I/R.
Collapse
Affiliation(s)
- In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University, Seoul 151-742, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Li Y, Hawkins BE, DeWitt DS, Prough DS, Maret W. The relationship between transient zinc ion fluctuations and redox signaling in the pathways of secondary cellular injury: relevance to traumatic brain injury. Brain Res 2010; 1330:131-41. [PMID: 20303343 DOI: 10.1016/j.brainres.2010.03.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 03/05/2010] [Accepted: 03/11/2010] [Indexed: 10/19/2022]
Abstract
A major obstacle that hampers the design of drug therapy for traumatic brain injury is the incomplete understanding of the biochemical pathways that lead to secondary cellular injury and contribute to cell death. One such pathway involves reactive species that generate potentially cytotoxic zinc ion fluctuations as a major executor of neuronal, and possibly glial, cell death. Whether zinc ions released during traumatic brain injury are toxic or protective is controversial but can be approached by investigating the exact concentrations of free zinc ions, the thresholds of compromised zinc buffering capacity, and the mechanism of cellular homeostatic control of zinc. Rapidly stretch-injured rat pheochromocytoma (PC12) cells express cellular zinc ion fluctuations that depend on the production of nitric oxide. Chelation of cellular zinc ions after rapid stretch injury, however, increases cellular reactive oxygen species. In a rat model of traumatic brain injury, parasagittal fluid percussion, analysis of the metal load of metallothionein was used as an indicator of changes in cellular zinc ion concentrations. The combined results from the cellular and in vivo investigations caution against interpreting zinc ion fluctuations in the early phase (24h) after injury as a primarily cytotoxic event.
Collapse
Affiliation(s)
- Yuan Li
- Division of Human Nutrition, Department of Preventive Medicine and Community Health, The University of Texas Medical Branch, Galveston, TX 77555, USA.
| | | | | | | | | |
Collapse
|
26
|
Lourenço CF, Santos R, Barbosa RM, Gerhardt G, Cadenas E, Laranjinha J. In vivo modulation of nitric oxide concentration dynamics upon glutamatergic neuronal activation in the hippocampus. Hippocampus 2010; 21:622-30. [DOI: 10.1002/hipo.20774] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2009] [Indexed: 11/07/2022]
|
27
|
Vizi ES, Fekete A, Karoly R, Mike A. Non-synaptic receptors and transporters involved in brain functions and targets of drug treatment. Br J Pharmacol 2010; 160:785-809. [PMID: 20136842 DOI: 10.1111/j.1476-5381.2009.00624.x] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Beyond direct synaptic communication, neurons are able to talk to each other without making synapses. They are able to send chemical messages by means of diffusion to target cells via the extracellular space, provided that the target neurons are equipped with high-affinity receptors. While synaptic transmission is responsible for the 'what' of brain function, the 'how' of brain function (mood, attention, level of arousal, general excitability, etc.) is mainly controlled non-synaptically using the extracellular space as communication channel. It is principally the 'how' that can be modulated by medicine. In this paper, we discuss different forms of non-synaptic transmission, localized spillover of synaptic transmitters, local presynaptic modulation and tonic influence of ambient transmitter levels on the activity of vast neuronal populations. We consider different aspects of non-synaptic transmission, such as synaptic-extrasynaptic receptor trafficking, neuron-glia communication and retrograde signalling. We review structural and functional aspects of non-synaptic transmission, including (i) anatomical arrangement of non-synaptic release sites, receptors and transporters, (ii) intravesicular, intra- and extracellular concentrations of neurotransmitters, as well as the spatiotemporal pattern of transmitter diffusion. We propose that an effective general strategy for efficient pharmacological intervention could include the identification of specific non-synaptic targets and the subsequent development of selective pharmacological tools to influence them.
Collapse
Affiliation(s)
- E S Vizi
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | |
Collapse
|
28
|
Fekete A, Franklin L, Ikemoto T, Rózsa B, Lendvai B, Sylvester Vizi E, Zelles T. Mechanism of the persistent sodium current activator veratridine-evoked Ca elevation: implication for epilepsy. J Neurochem 2009; 111:745-56. [PMID: 19719824 DOI: 10.1111/j.1471-4159.2009.06368.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although the role of Na(+) in several aspects of Ca(2+) regulation has already been shown, the exact mechanism of intracellular Ca(2+) concentration ([Ca(2+)](i)) increase resulting from an enhancement in the persistent, non-inactivating Na(+) current (I(Na,P)), a decisive factor in certain forms of epilepsy, has yet to be resolved. Persistent Na(+) current, evoked by veratridine, induced bursts of action potentials and sustained membrane depolarization with monophasic intracellular Na(+) concentration ([Na(+)](i)) and biphasic [Ca(2+)](i) increase in CA1 pyramidal cells in acute hippocampal slices. The Ca(2+) response was tetrodotoxin- and extracellular Ca(2+)-dependent and ionotropic glutamate receptor-independent. The first phase of [Ca(2+)](i) rise was the net result of Ca(2+) influx through voltage-gated Ca(2+) channels and mitochondrial Ca(2+) sequestration. The robust second phase in addition involved reverse operation of the Na(+)-Ca(2+) exchanger and mitochondrial Ca(2+) release. We excluded contribution of the endoplasmic reticulum. These results demonstrate a complex interaction between persistent, non-inactivating Na(+) current and [Ca(2+)](i) regulation in CA1 pyramidal cells. The described cellular mechanisms are most likely part of the pathomechanism of certain forms of epilepsy that are associated with I(Na,P). Describing the magnitude, temporal pattern and sources of Ca(2+) increase induced by I(Na,P) may provide novel targets for antiepileptic drug therapy.
Collapse
Affiliation(s)
- Adám Fekete
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Szigony, Hungary
| | | | | | | | | | | | | |
Collapse
|
29
|
Li CT, Zhang WP, Lu YB, Fang SH, Yuan YM, Qi LL, Zhang LH, Huang XJ, Zhang L, Chen Z, Wei EQ. Oxygen-glucose deprivation activates 5-lipoxygenase mediated by oxidative stress through the p38 mitogen-activated protein kinase pathway in PC12 cells. J Neurosci Res 2009; 87:991-1001. [DOI: 10.1002/jnr.21913] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
30
|
Rolseth V, Rundén-Pran E, Neurauter CG, Yndestad A, Luna L, Aukrust P, Ottersen OP, Bjørås M. Base excision repair activities in organotypic hippocampal slice cultures exposed to oxygen and glucose deprivation. DNA Repair (Amst) 2008; 7:869-78. [PMID: 18406215 DOI: 10.1016/j.dnarep.2008.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Revised: 02/07/2008] [Accepted: 02/12/2008] [Indexed: 01/22/2023]
Abstract
The capacity for DNA repair is likely to be one of the factors that determine the vulnerability of neurons to ischemic stress and may influence the pathological outcome of stroke. In this report, initiation of base excision repair (BER) was assessed by analysis of enzyme activity and gene expression level of DNA glycosylases and AP-endonucleases in rat organotypic hippocampal slice cultures exposed to oxygen and glucose deprivation (OGD) - an in vitro model of stroke. Under basal conditions, AP-endonuclease activity and base removal of ethenoadenine and 8-oxoguanine (8-oxoG) were higher (by approximately 20-35 %) in CA3/fascia dentata (FD) than in CA1. Base removal of uracil did not differ between the two hippocampal regions, while removal of 5-hydroxyuracil (5-OHU) was slightly less efficient in CA3/FD than in CA1. Analyses performed immediately after 30 min of OGD revealed a decreased AP-endonuclease activity (by approximately 20%) in CA1 as well as CA3/FD, and an increased ethenoadenine activity (by approximately 25%) in CA1. Activities for 8-oxoG, 5-OHU and uracil showed no significant changes at this time point. At 8h after OGD, none of the enzyme activities differed from control values. Real-time RT-PCR showed that transcription of DNA glycosylases, including Ogg1, Nth1, Ung, Aag, Neil1 and Neil2 were not changed in response to OGD treatment (t=0 h). The hippocampal expression of Neil2 was low compared with the other DNA glycosylases. These data indicate that CA1 has a lower capacity than CA3/FD for removal of base lesions under basal conditions. The relatively low capacity for BER in basal conditions and the apparent failure to upregulate repair of oxidative damage after OGD might contribute to the high vulnerability of CA1 to ischemic injury.
Collapse
Affiliation(s)
- Veslemøy Rolseth
- Centre for Molecular Biology and Neuroscience, Institute of Medical Microbiology, University of Oslo, Rikshospitalet, N-0027 Oslo, Norway
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Milusheva E, Baranyi M, Kittel A, Fekete A, Zelles T, Vizi ES, Sperlágh B. Modulation of dopaminergic neurotransmission in rat striatum upon in vitro and in vivo diclofenac treatment. J Neurochem 2007; 105:360-8. [PMID: 18036194 PMCID: PMC2324205 DOI: 10.1111/j.1471-4159.2007.05141.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Diclofenac (DCF) is a widely used non-steroidal anti-inflammatory drug, which also act as a mitochondrial toxin. As it is known that selective mitochondrial complex I inhibition combined with mild oxidative stress causes striatal dopaminergic dysfunction, we tested whether DCF also compromise dopaminergic function in the striatum. [3H]Dopamine ([3H]DA) release was measured from rat striatal slices after in vitro (2 h, 10–25 μmol/L) or in vivo (3 mg/kg i.v. for 28 days) DCF treatment. In vitro treatment significantly decreased [3H]DA uptake and dopamine (DA) content of the slices. H2O2 (0.1 mmol/L)-evoked DA release was enhanced. Intracellular reactive oxygen species production was not significantly changed in the presence of DCF. After in vivo DCF treatment no apparent decrease in striatal DA content was observed and the uptake of [3H]DA into slices was increased. The intensity of tyrosine hydroxylase immunoreactivity in the striatum was highly variable, and both decrease and increase were observed in individual rats. The H2O2-evoked [3H]DA release was significantly decreased and the effluent contained a significant amount of [3H]octopamine, [3H]tyramine, and [3H]β-phenylethylamine. The ATP content and adenylate energy charge were decreased. In conclusion, whereas in vitro DCF pre-treatment resembles the effect of the mitochondrial toxin rotenone, in vivo it rather counteracts than aggravates dopaminergic dysfunction. J. Neurochem. (2008) 105, 360–368.
Collapse
Affiliation(s)
- Elisaveta Milusheva
- Institute of Neurobiology (former Institute of Physiology), Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | | | | | | | | | | |
Collapse
|