51
|
Stauch KL, Villeneuve LM, Totusek S, Lamberty B, Ciborowski P, Fox HS. Quantitative Proteomics of Presynaptic Mitochondria Reveal an Overexpression and Biological Relevance of Neuronal MitoNEET in Postnatal Brain Development. Dev Neurobiol 2019; 79:370-386. [PMID: 31050203 DOI: 10.1002/dneu.22684] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 03/02/2019] [Accepted: 04/25/2019] [Indexed: 12/11/2022]
Abstract
Although it has been recognized that energy metabolism and mitochondrial structure and functional activity in the immature brain differs from that of the adult, few studies have examined mitochondria specifically at the neuronal synapse during postnatal brain development. In this study, we examined the presynaptic mitochondrial proteome in mice at postnatal day 7 and 42, a period that involves the formation and maturation of synapses. Application of two independent quantitative proteomics approaches - SWATH-MS and super-SILAC - revealed a total of 40 proteins as significantly differentially expressed in the presynaptic mitochondria. In addition to elevated levels of proteins known to be involved in ATP metabolic processes, our results identified increased levels of mitoNEET (Cisd1), an iron-sulfur containing protein that regulates mitochondrial bioenergetics. We found that mitoNEET overexpression plays a cell-type specific role in ATP synthesis and in neuronal cells promotes ATP generation. The elevated ATP levels in SH-SY5Y neuroblastoma cells were associated with increased mitochondrial membrane potential and a fragmented mitochondrial network, further supporting a role for mitoNEET as a key regulator of mitochondrial function.
Collapse
Affiliation(s)
- Kelly L Stauch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, 68198
| | - Lance M Villeneuve
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, 68198
| | - Steven Totusek
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, 68198
| | - Benjamin Lamberty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, 68198
| | - Pawel Ciborowski
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, 68198
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, 68198
| |
Collapse
|
52
|
Wood CE, Keller-Wood M. Current paradigms and new perspectives on fetal hypoxia: implications for fetal brain development in late gestation. Am J Physiol Regul Integr Comp Physiol 2019; 317:R1-R13. [PMID: 31017808 DOI: 10.1152/ajpregu.00008.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The availability of oxygen to the fetus is limited by the route taken by oxygen from the atmosphere to fetal tissues, aided or diminished by pregnancy-associated changes in maternal physiology and, ultimately, a function of atmospheric pressure and composition of the mother's inspired gas. Much of our understanding of the fetal physiological response to hypoxia comes from experiments designed to elucidate the cardiovascular and endocrine responses to transient hypoxia. Complementing this work is equally impactful research into the origins of intrauterine growth restriction in which animal models designed to restrict the transfer of oxygen from the maternal to the fetal circulation were used. A common assumption has been that outcomes measured after a period of hypoxia are related to cellular deprivation of oxygen and reoxygenation: an assumption based on a focus on what we can see "under the streetlights." Recent studies demonstrate that availability of oxygen may not tell the whole story. Transient hypoxia in the fetal sheep stimulates transcriptomics responses that mirror inflammation. This response is accompanied by the appearance of bacteria in the fetal brain and other tissues, likely resulting from a hypoxia-stimulated release of bacteria from the placenta. The appearance of bacteria in the fetus after transient hypoxia complements the recent discovery of bacterial DNA in the normal human placenta and in the tissues of fetal sheep. An understanding of the mechanism of the physiological, cellular, and molecular responses to hypoxia requires an appreciation of stimuli other than cellular oxygen deprivation: stimuli that we would have never known about without looking "between the streetlights," illuminating direct responses to the manipulated variables.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Physiology and Functional Genomics, University of Florida College of Medicine , Gainesville, Florida
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida College of Pharmacy , Gainesville, Florida
| |
Collapse
|
53
|
Qin X, Cheng J, Zhong Y, Mahgoub OK, Akter F, Fan Y, Aldughaim M, Xie Q, Qin L, Gu L, Jian Z, Xiong X, Liu R. Mechanism and Treatment Related to Oxidative Stress in Neonatal Hypoxic-Ischemic Encephalopathy. Front Mol Neurosci 2019; 12:88. [PMID: 31031592 PMCID: PMC6470360 DOI: 10.3389/fnmol.2019.00088] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/21/2019] [Indexed: 12/24/2022] Open
Abstract
Hypoxic ischemic encephalopathy (HIE) is a type of neonatal brain injury, which occurs due to lack of supply and oxygen deprivation to the brain. It is associated with a high morbidity and mortality rate. There are several therapeutic strategies that can be used to improve outcomes in patients with HIE. These include cell therapies such as marrow mesenchymal stem cells (MSCs) and umbilical cord blood stem cells (UCBCs), which are being incorporated into the new protocols for the prevention of ischemic brain damage. The focus of this review is to discuss the mechanism of oxidative stress in HIE and summarize the current available treatments for HIE. We hope that a better understanding of the relationship between oxidative stress and HIE will provide new insights on the potential therapy of this devastating condition.
Collapse
Affiliation(s)
- Xingping Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Neurosurgery, Harvard Medical School, Boston, MA, United States
| | - Jing Cheng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Omer Kamal Mahgoub
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Farhana Akter
- Department of Neurosurgery, Harvard Medical School, Boston, MA, United States.,Department of Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Yanqin Fan
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mohammed Aldughaim
- Department of Neurosurgery, Harvard Medical School, Boston, MA, United States
| | - Qiurong Xie
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lingxia Qin
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Renzhong Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
54
|
Metabolic responses in Antarctic Nototheniidae brains subjected to thermal stress. Brain Res 2019; 1708:126-137. [PMID: 30527682 DOI: 10.1016/j.brainres.2018.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/08/2018] [Accepted: 12/04/2018] [Indexed: 12/23/2022]
Abstract
Antarctic Nototheniidae is an attractive group for studying metabolic and physiological responses at high temperatures. The present work investigated the metabolic responses of the carbohydrate metabolism and antioxidant system to thermal stress at 8 °C (for 2-144 h) in the brains of Notothenia rossii and Notothenia coriiceps. In N. coriiceps, glycogenolysis was essential in the first hours of exposure (2 h) at 8 °C and, in addition to inhibiting glucose-6-phosphatase activity, was important for activating the pentose phosphate pathway. In N. rossii, anaerobic metabolism was reduced in the first hours of exposure (2 and 6 h) at 8 °C, followed by reduced hexokinase activity, suggesting energy regulation between neurons and astrocytes. The antioxidant system results indicated the importance of the actions of the glutathione-dependent antioxidant enzymes glutathione-S-transferase and glutathione peroxidase as well as those of catalase in N. coriiceps and the action of glutathione-S-transferase, glutathione peroxidase and glutathione reductase in N. rossii, especially during the first 12 h of thermal stress exposure. These results indicate tissue-specific patterns and species-specific responses to this stress.
Collapse
|
55
|
Fang J, Chavez-Valdez R, Flock DL, Avaritt O, Saraswati M, Robertson C, Martin LJ, Northington FJ. An Inhibitor of the Mitochondrial Permeability Transition Pore Lacks Therapeutic Efficacy Following Neonatal Hypoxia Ischemia in Mice. Neuroscience 2019; 406:202-211. [PMID: 30849447 DOI: 10.1016/j.neuroscience.2019.02.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 01/08/2023]
Abstract
Neonatal hypoxic ischemic (HI) brain injury causes lifelong neurologic disability. Therapeutic hypothermia (TH) is the only approved therapy that partially mitigates mortality and morbidity. Therapies specifically targeting HI-induced brain cell death are currently lacking. Intracellular calcium dysregulation, oxidative stress, and mitochondrial dysfunction through the formation of the mitochondrial permeability transition pore (mPTP) are drivers of HI cellular injury. GNX-4728, a small molecule direct inhibitor of the mPTP that increases mitochondrial calcium retention capacity, is highly effective in adult neurodegenerative disease models and could have potential as a therapy in neonatal HI. A dose of GNX-4728, equivalent to that used in animal models, 300 mg/kg, IP was highly toxic in p10 mice. We then tested the hypothesis that acute administration of 30 mg/kg, IP of GNX-4728 immediately after HI in a neonatal mouse model would provide neuroprotection. This non-lethal lower dose of GNX-4728 (30 mg/kg, IP) improved the respiratory control ratio of neonatal female HI brain tissue but not in males. Brain injury, assessed histologically with a novel metric approach at 1 and 30 days after HI, was not mitigated by GNX-4728. Our work demonstrates that a small molecule inhibitor of the mPTP has i) an age related toxicity, ii) a sex-related brain mitoprotective profile after HI but iii) this is not sufficient to attenuate forebrain HI neuropathology.
Collapse
Affiliation(s)
- Jing Fang
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Raul Chavez-Valdez
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Debbie L Flock
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Oliver Avaritt
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Manda Saraswati
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Courtney Robertson
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lee J Martin
- Department of Neuroscience and Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
56
|
Torres LH, Balestrin NT, Spelta LEW, Duro SDO, Pistis M, Marcourakis T. Exposure to tobacco smoke during the early postnatal period modifies receptors and enzymes of the endocannabinoid system in the brainstem and striatum in mice. Toxicol Lett 2019; 302:35-41. [DOI: 10.1016/j.toxlet.2018.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 11/19/2018] [Accepted: 12/12/2018] [Indexed: 01/31/2023]
|
57
|
Retinal Neuron Is More Sensitive to Blue Light-Induced Damage than Glia Cell Due to DNA Double-Strand Breaks. Cells 2019; 8:cells8010068. [PMID: 30669263 PMCID: PMC6356720 DOI: 10.3390/cells8010068] [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: 12/01/2018] [Revised: 01/06/2019] [Accepted: 01/15/2019] [Indexed: 12/12/2022] Open
Abstract
Blue light is a major component of visible light and digital displays. Over-exposure to blue light could cause retinal damage. However, the mechanism of its damage is not well defined. Here, we demonstrate that blue light (900 lux) impairs cell viability and induces cell apoptosis in retinal neurocytes in vitro. A DNA electrophoresis assay shows severe DNA damage in retinal neurocytes at 2 h after blue light treatment. γ-H2AX foci, a specific marker of DNA double-strand breaks (DSBs), is mainly located in the Map2-posotive neuron other than the glia cell. After assaying the expression level of proteins related to DNA repair, Mre11, Ligase IV and Ku80, we find that Ku80 is up-regulated in retinal neurocytes after blue light treatment. Interestingly, Ku80 is mainly expressed in glia fibrillary acidic protein (GFAP)-positive glia cells. Moreover, following blue light exposure in vivo, DNA DSBs are shown in the ganglion cell layer and only observed in Map2-positive cells. Furthermore, long-term blue light exposure significantly thinned the retina in vivo. Our findings demonstrate that blue light induces DNA DSBs in retinal neurons, and the damage is more pronounced compared to glia cells. Thus, this study provides new insights into the mechanisms of the effect of blue light on the retina.
Collapse
|
58
|
Mitra S, Bale G, Highton D, Gunny R, Uria-Avellanal C, Bainbridge A, Sokolska M, Price D, Huertas-Ceballos A, Kendall GS, Meek J, Tachtsidis I, Robertson NJ. Pressure passivity of cerebral mitochondrial metabolism is associated with poor outcome following perinatal hypoxic ischemic brain injury. J Cereb Blood Flow Metab 2019; 39:118-130. [PMID: 28949271 PMCID: PMC6311664 DOI: 10.1177/0271678x17733639] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypoxic ischemic encephalopathy (HIE) leads to significant morbidity and mortality. Impaired autoregulation after hypoxia-ischaemia has been suggested to contribute further to injury. Thalamic lactate/N-Acetylasperate (Lac/NAA) peak area ratio of > 0.3 on proton (1H) magnetic resonance spectroscopy (MRS) is associated with poor neurodevelopment outcome following HIE. Cytochrome-c-oxidase (CCO) plays a central role in mitochondrial oxidative metabolism and ATP synthesis. Using a novel broadband NIRS system, we investigated the impact of pressure passivity of cerebral metabolism (CCO), oxygenation (haemoglobin difference (HbD)) and cerebral blood volume (total haemoglobin (HbT)) in 23 term infants following HIE during therapeutic hypothermia (HT). Sixty-minute epochs of data from each infant were studied using wavelet analysis at a mean age of 48 h. Wavelet semblance (a measure of phase difference) was calculated to compare reactivity between mean arterial blood pressure (MABP) with oxCCO, HbD and HbT. OxCCO-MABP semblance correlated with thalamic Lac/NAA ( r = 0.48, p = 0.02). OxCCO-MABP semblance also differed between groups of infants with mild to moderate and severe injury measured using brain MRI score ( p = 0.04), thalamic Lac/NAA ( p = 0.04) and neurodevelopmental outcome at one year ( p = 0.04). Pressure passive changes in cerebral metabolism were associated with injury severity indicated by thalamic Lac/NAA, MRI scores and neurodevelopmental assessment at one year of age.
Collapse
Affiliation(s)
- Subhabrata Mitra
- 1 Institute for Women's Health, University College London, London, UK
| | - Gemma Bale
- 2 Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - David Highton
- 3 Neurocritical Care, National Hospital for Neurology & Neurosurgery, University College London, London, UK
| | - Roxanna Gunny
- 4 Paediatric Neuroradiology, Great Ormond Street Hospital for Children, London, UK
| | | | - Alan Bainbridge
- 5 Department of Medical Physics and Biomedical Engineering, University College London Hospital, London, UK
| | - Magdalena Sokolska
- 5 Department of Medical Physics and Biomedical Engineering, University College London Hospital, London, UK
| | - David Price
- 5 Department of Medical Physics and Biomedical Engineering, University College London Hospital, London, UK
| | | | - Giles S Kendall
- 6 Neonatal Unit, University College London Hospital, London, UK
| | - Judith Meek
- 1 Institute for Women's Health, University College London, London, UK
| | - Ilias Tachtsidis
- 2 Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | | |
Collapse
|
59
|
Luo Y, Tang H, Li H, Zhao R, Huang Q, Liu J. Recent advances in the development of neuroprotective agents and therapeutic targets in the treatment of cerebral ischemia. Eur J Med Chem 2019; 162:132-146. [DOI: 10.1016/j.ejmech.2018.11.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/30/2018] [Accepted: 11/06/2018] [Indexed: 11/25/2022]
|
60
|
Kirschen GW, Kéry R, Ge S. The Hippocampal Neuro-Glio-Vascular Network: Metabolic Vulnerability and Potential Neurogenic Regeneration in Disease. Brain Plast 2018; 3:129-144. [PMID: 30151338 PMCID: PMC6091038 DOI: 10.3233/bpl-170055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Brain metabolism is a fragile balance between nutrient/oxygen supply provided by the blood and neuronal/glial demand. Small perturbations in these parameters are necessary for proper homeostatic functioning and information processing, but can also cause significant damage and cell death if dysregulated. During embryonic and early post-natal development, massive neurogenesis occurs, a process that continues at a limited rate in adulthood in two neurogenic niches, one in the lateral ventricle and the other in the hippocampal dentate gyrus. When metabolic demand does not correspond with supply, which can occur dramatically in the case of hypoxia or ischemia, or more subtly in the case of neuropsychiatric or neurodegenerative disorders, both of these neurogenic niches can respond—either in a beneficial manner, to regenerate damaged or lost tissue, or in a detrimental fashion—creating aberrant synaptic connections. In this review, we focus on the complex relationship that exists between the cerebral vasculature and neurogenesis across development and in disease states including hypoxic-ischemic injury, hypertension, diabetes mellitus, and Alzheimer’s disease. Although there is still much to be elucidated, we are beginning to appreciate how neurogenesis may help or harm the metabolically-injured brain, in the hopes that these insights can be used to tailor novel therapeutics to regenerate damaged tissue after injury.
Collapse
Affiliation(s)
- Gregory W Kirschen
- Medical Scientist Training Program (MSTP), Stony Brook Medicine, Stony Brook, NY, USA.,Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Rachel Kéry
- Medical Scientist Training Program (MSTP), Stony Brook Medicine, Stony Brook, NY, USA.,Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Shaoyu Ge
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
61
|
Perinatal Hypoxic-Ischemic Encephalopathy and Neuroprotective Peptide Therapies: A Case for Cationic Arginine-Rich Peptides (CARPs). Brain Sci 2018; 8:brainsci8080147. [PMID: 30087289 PMCID: PMC6119922 DOI: 10.3390/brainsci8080147] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/25/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Perinatal hypoxic-ischemic encephalopathy (HIE) is the leading cause of mortality and morbidity in neonates, with survivors suffering significant neurological sequelae including cerebral palsy, epilepsy, intellectual disability and autism spectrum disorders. While hypothermia is used clinically to reduce neurological injury following HIE, it is only used for term infants (>36 weeks gestation) in tertiary hospitals and improves outcomes in only 30% of patients. For these reasons, a more effective and easily administrable pharmacological therapeutic agent, that can be used in combination with hypothermia or alone when hypothermia cannot be applied, is urgently needed to treat pre-term (≤36 weeks gestation) and term infants suffering HIE. Several recent studies have demonstrated that cationic arginine-rich peptides (CARPs), which include many cell-penetrating peptides [CPPs; e.g., transactivator of transcription (TAT) and poly-arginine-9 (R9; 9-mer of arginine)], possess intrinsic neuroprotective properties. For example, we have demonstrated that poly-arginine-18 (R18; 18-mer of arginine) and its D-enantiomer (R18D) are neuroprotective in vitro following neuronal excitotoxicity, and in vivo following perinatal hypoxia-ischemia (HI). In this paper, we review studies that have used CARPs and other peptides, including putative neuroprotective peptides fused to TAT, in animal models of perinatal HIE. We critically evaluate the evidence that supports our hypothesis that CARP neuroprotection is mediated by peptide arginine content and positive charge and that CARPs represent a novel potential therapeutic for HIE.
Collapse
|
62
|
Tucker LD, Lu Y, Dong Y, Yang L, Li Y, Zhao N, Zhang Q. Photobiomodulation Therapy Attenuates Hypoxic-Ischemic Injury in a Neonatal Rat Model. J Mol Neurosci 2018; 65:514-526. [PMID: 30032397 PMCID: PMC6109412 DOI: 10.1007/s12031-018-1121-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/11/2018] [Indexed: 12/14/2022]
Abstract
Photobiomodulation (PBM) has been demonstrated as a neuroprotective strategy, but its effect on perinatal hypoxic-ischemic encephalopathy is still unknown. The current study was designed to shed light on the potential beneficial effect of PBM on neonatal brain injury induced by hypoxia ischemia (HI) in a rat model. Postnatal rats were subjected to hypoxic-ischemic insult, followed by a 7-day PBM treatment via a continuous wave diode laser with a wavelength of 808 nm. We demonstrated that PBM treatment significantly reduced HI-induced brain lesion in both the cortex and hippocampal CA1 subregions. Molecular studies indicated that PBM treatment profoundly restored mitochondrial dynamics by suppressing HI-induced mitochondrial fragmentation. Further investigation of mitochondrial function revealed that PBM treatment remarkably attenuated mitochondrial membrane collapse, accompanied with enhanced ATP synthesis in neonatal HI rats. In addition, PBM treatment led to robust inhibition of oxidative damage, manifested by significant reduction in the productions of 4-HNE, P-H2AX (S139), malondialdehyde (MDA), as well as protein carbonyls. Finally, PBM treatment suppressed the activation of mitochondria-dependent neuronal apoptosis in HI rats, as evidenced by decreased pro-apoptotic cascade 3/9 and TUNEL-positive neurons. Taken together, our findings demonstrated that PBM treatment contributed to a robust neuroprotection via the attenuation of mitochondrial dysfunction, oxidative stress, and final neuronal apoptosis in the neonatal HI brain.
Collapse
Affiliation(s)
- Lorelei Donovan Tucker
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yujiao Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Luodan Yang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yong Li
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Ningjun Zhao
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
63
|
Lespay-Rebolledo C, Perez-Lobos R, Tapia-Bustos A, Vio V, Morales P, Herrera-Marschitz M. Regionally Impaired Redox Homeostasis in the Brain of Rats Subjected to Global Perinatal Asphyxia: Sustained Effect up to 14 Postnatal Days. Neurotox Res 2018; 34:660-676. [PMID: 29959728 DOI: 10.1007/s12640-018-9928-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/30/2022]
Abstract
The present report evaluates the effect of global perinatal asphyxia on several parameters of oxidative stress and cell viability in rat brain tissue sampled at an extended neonatal period up to 14 days, a period characterised by intensive neuritogenesis, synaptogenesis, synaptic consolidation, pruning and delayed cell death. Perinatal asphyxia was induced by immersing foetus-containing uterine horns removed by a caesarean section from on term rat dams into a water bath at 37 °C for 21 min. Asphyxia-exposed and sibling caesarean-delivered foetuses were manually resucitated and nurtured by surrogate dams for 1 to 14 postnatal (P) days. Brain samples (mesencephalon, telencephalon and hippocampus) were assayed for glutathione (reduced and oxidated levels; spectrophotometry), tissue reducing capacity (potassium ferricyanide reducing assay, FRAP), catalase (the key enzyme protecting against oxidative stress and reactive oxygen species, Western blots and ELISA) and cleaved caspase-3 (the key executioner of apoptosis, Western blots) levels. It was found that global PA produced a regionally specific and sustained increase in GSSG/GSH ratio, a regionally specific decrease in tissue reducing capacity and a regionally and time specific decrease of catalase activity and increase of cleaved caspase-3 levels. The present study provides evidence for regionally impaired redox homeostasis in the brain of rats subjected to global PA, an effect observed up to P14, mainly affecting mesencephalon and hippocampus, suggesting a sustained oxidative stress after the posthypoxia period. The oxidative stress observed postnatally can in part be associated to a respiratory apneic-like deficit, since there was a statistically significant decrease in respiration frequency in AS compared to CS neonates, also up to P14, together with the signs of a decreased peripheral blood perfusion (pink-blue skin colour in AS, compared to the pink colour observed in all CS neonates). It is proposed that PA implies a long-term metabolic insult, triggered by the length of hypoxia, the resuscitation/reoxigenation manoevres, but also by the developmental stage of the affected brain regions, and the integrity of cardiovascular and respiratory physiological functions, which are fundamental for warrantying a proper development.
Collapse
Affiliation(s)
- Carolyne Lespay-Rebolledo
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Ronald Perez-Lobos
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Andrea Tapia-Bustos
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Valentina Vio
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Paola Morales
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
- Department Neuroscience, Medical Faculty, University of Chile, Santiago, Chile
| | - Mario Herrera-Marschitz
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile.
| |
Collapse
|
64
|
Odorcyk FK, Kolling J, Sanches EF, Wyse ATS, Netto CA. Experimental neonatal hypoxia ischemia causes long lasting changes of oxidative stress parameters in the hippocampus and the spleen. J Perinat Med 2018; 46:433-439. [PMID: 28841577 DOI: 10.1515/jpm-2017-0070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/25/2017] [Indexed: 11/15/2022]
Abstract
Neonatal hypoxia ischemia (HI) is the main cause of mortality and morbidity in newborns. The mechanisms involved in its progression start immediately and persist for several days. Oxidative stress and inflammation are determinant factors of the severity of the final lesion. The spleen plays a major part in the inflammatory response to HI. This study assessed the temporal progression of HI-induced alterations in oxidative stress parameters in the hippocampus, the most affected brain structure, and in the spleen. HI was induced in Wistar rat pups in post-natal day 7. Production of reactive oxygen species (ROS), and the activity of the anti oxidant enzyme superoxide dismutase and catalase were assessed 24 h, 96 h and 38 days post-HI. Interestingly, both structures showed a similar pattern, with few alterations in the production of ROS species up to 96 h often combined with an increased activity of the anti oxidant enzymes. However, 38 days after the injury, ROS were at the highest in both structures, coupled with a decrease in the activity of the enzymes. Altogether, present results suggest that HI causes long lasting alterations in the hippocampus as well as in the spleen, suggesting a possible target for delayed treatments for HI.
Collapse
Affiliation(s)
- Felipe Kawa Odorcyk
- Post-graduation Program of Neurosciences, Departamento de Bioquímica, Instituto das Ciências da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600 anexo 90035-003, Porto Alegre, RS, Brazil, Tel./Fax: 0055-051 33085568
| | - Janaína Kolling
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Eduardo Farias Sanches
- Post-graduation Program of Neurosciences, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Angela T S Wyse
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
65
|
Amiri S, Yousefi-Ahmadipour A, Hosseini MJ, Haj-Mirzaian A, Momeny M, Hosseini-Chegeni H, Mokhtari T, Kharrazi S, Hassanzadeh G, Amini SM, Jafarinejad S, Ghazi-Khansari M. Maternal exposure to silver nanoparticles are associated with behavioral abnormalities in adulthood: Role of mitochondria and innate immunity in developmental toxicity. Neurotoxicology 2018; 66:66-77. [PMID: 29550386 DOI: 10.1016/j.neuro.2018.03.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 03/06/2018] [Accepted: 03/10/2018] [Indexed: 01/06/2023]
Abstract
Silver nanoparticles (Ag-NPs) are currently used in a wide range of consumer products. Considering the small size of Ag-NPs, they are able to pass through variety of biological barriers and exert their effects. In this regard, the unique physicochemical properties of Ag-NPs along with its high application in the industry have raised concerns about their negative effects on human health. Therefore, it investigated whether prenatal exposure to low doses of Ag-NPs is able to induce any abnormality in the cognitive and behavioral performance of adult offspring. We gavaged pregnant NMRI mice with, 1) Deionized water as vehicle, 2) Ag-NPs 10 nm (0.26 mg/kg/day), 3) Ag-NPs 30 nm (0.26 mg/kg/day), and 4) AgNO3 (0.26 mg/kg/day) from gestational day (GD) 0 until delivery day. At the postnatal day (PD) 1, our results showed that high concentration of silver is present in the brain of pups. Further, we observed mitochondrial dysfunction and upregulation of the genes relevant to innate immune system in the brain. At PD 60, results revealed that prenatal exposure to Ag-NPs provoked severe cognitive and behavioral abnormalities in male offspring. In addition, we found that prenatal exposure to Ag-NPs was associated with abnormal mitochondrial function and significant up-regulation of the genes relevant to innate immunity in the brain. Although the Ag-NPs have been considered as safe compounds at low doses, our results indicate that prenatal exposure to low doses of Ag-NPs is able to induce behavioral and cognitive abnormalities in adulthood. Also, we found that these effects are at least partly associated with hippocampal mitochondrial dysfunction and the activation of sterile inflammation during early stages of life.
Collapse
Affiliation(s)
- Shayan Amiri
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliakbar Yousefi-Ahmadipour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Tissue Engineering and Applied Cell Sciences, Department of School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir-Jamal Hosseini
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Arya Haj-Mirzaian
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Momeny
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Heshmat Hosseini-Chegeni
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Mokhtari
- Research Center of Nervous System Stem Cells, Department of Anatomy, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran; Department of Anatomy, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Sharmin Kharrazi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Amini
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jafarinejad
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Ghazi-Khansari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Iran Nanosafety Network (INSN) of Iran Nanotechnology Initiative Council (INIC), Tehran, Iran.
| |
Collapse
|
66
|
Pan S, Li S, Hu Y, Zhang H, Liu Y, Jiang H, Fang M, Li Z, Xu K, Zhang H, Lin Z, Xiao J. Resveratrol post-treatment protects against neonatal brain injury after hypoxia-ischemia. Oncotarget 2018; 7:79247-79261. [PMID: 27811363 PMCID: PMC5346711 DOI: 10.18632/oncotarget.13018] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 10/19/2016] [Indexed: 11/25/2022] Open
Abstract
Neonatal hypoxic-ischemic brain injury is a devastating disease with limited treatment options. Preventive treatment with resveratrol has indicated to be well tolerated and has lower toxicity in both experimental models and human patients. However, whether resveratrol administration post-hypoxic-ischemic protects against neonatal hypoxic-ischemic injury is not known. Here we reported that post-treatment with resveratrol significantly reduced brain damage at 7-day after the injury. We found that resveratrol reduced the expression levels of key inflammatory factors at the mRNA and protein levels, and at least partially via inhibiting microglia activation. Moreover, resveratrol exerted an anti-apoptotic effect, as assessed by TUNEL staining, and altered the expression of the apoptosis-related genes Bax, Bcl-2 and caspase3. Our data indicate that post-treatment with resveratrol protects against neonatal hypoxic-ischemic brain injury and suggest a promising therapeutic strategy to this disease.
Collapse
Affiliation(s)
- Shulin Pan
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Songlin Li
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Yingying Hu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Hao Zhang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Yanlong Liu
- The School of Pharmacy, Wenzhou Medical University, Wenzhou 325035, China
| | - Huai Jiang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Mingchu Fang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Zhengmao Li
- The School of Pharmacy, Wenzhou Medical University, Wenzhou 325035, China
| | - Kebin Xu
- The School of Pharmacy, Wenzhou Medical University, Wenzhou 325035, China
| | - Hongyu Zhang
- The School of Pharmacy, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhenlang Lin
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, China
| | - Jian Xiao
- The School of Pharmacy, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
67
|
Yang L, Xu F, Zhang M, Shang XY, Xie X, Fu T, Li JP, Li HL. Role of LncRNA MALAT-1 in hypoxia-induced PC12 cell injury via regulating p38MAPK signaling pathway. Neurosci Lett 2018; 670:41-47. [PMID: 29360503 DOI: 10.1016/j.neulet.2018.01.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To investigate the role of LncRNA MALAT-1 in hypoxia-induced cell injury. METHODS Pheochromocytoma-12 (PC12) cells were divided into seven groups: Control group, Hypoxia group (Cells treated with CoCl2), MALAT-1 group (Hypoxic cells treated with MALAT-1), NC group (Hypoxic cells treated with empty plasmid), MALAT-1 siRNA group (Hypoxic cells treated with siRNA MALAT-1), SB203580 group (Hypoxic cells treated with p38MAPK inhibitor), and MALAT-1 + SB20358 group. The content of reactive oxygen species (ROS), malondialdehyde (MDA), super oxide dismutase (SOD) and lactate dehydrogenase (LDH) was determined. Cell viability was detected by MTT assay. Apoptotic cells were observed by Hoechst 33258 and TUNEL staining assay. Mitochondrial membrane potential (MMP) was measured using JC1 vital dye. RESULTS The decreased cell viability and increased expressions of MALAT-1 and p-p38 were observed in hypoxic PC12 cells time-dependently (P < 0.05). Besides, hypoxic PC12 cells had an elevation in p-p38, ROS, MDA and LDH with the increased apoptotic cells, but a reduction in SOD and MMP, and these similar changes were more obvious in those hypoxic cells treated with MALAT-1 when compared with Controls (all P < 0.05). However, the hypoxic PC12 cells treated with SB203580 and MALAT-1 siRNA led to opposite results compared with MALAT-1 group (all P < 0.05). Importantly, SB203580 could reverse the function of MALAT-1 in aggravating the hypoxia injury of PC12 cells. CONCLUSION MALAT-1 can promote the apoptosis and oxidative stress of PC12 cells by activating p38MAPK pathway, thus aggravating the damage of PC12 cells induced by chemical hypoxia.
Collapse
Affiliation(s)
- Lin Yang
- Zunyi Medical University, Zhuhai Campus, Zhuhai, Guangdong 519041, China
| | - Fei Xu
- Department of Rehabilitation, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150036, China
| | - Miao Zhang
- The Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150001, Heilongjiang, China
| | - Xiao-Ying Shang
- Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150036, China
| | - Xin Xie
- Department of Rehabilitation, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150036, China
| | - Tao Fu
- Department of Rehabilitation, Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150036, China
| | - Jian-Ping Li
- Zunyi Medical University, Zhuhai Campus, Zhuhai, Guangdong 519041, China
| | - Hong-Lin Li
- Department of Rehabilitation, The Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, No. 411, Guogeli Street, Nangang District, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
68
|
Charriaut-Marlangue C, Besson VC, Baud O. Sexually Dimorphic Outcomes after Neonatal Stroke and Hypoxia-Ischemia. Int J Mol Sci 2017; 19:ijms19010061. [PMID: 29278365 PMCID: PMC5796011 DOI: 10.3390/ijms19010061] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 12/19/2017] [Accepted: 12/24/2017] [Indexed: 01/21/2023] Open
Abstract
Cohort studies have demonstrated a higher vulnerability in males towards ischemic and/or hypoxic-ischemic injury in infants born near- or full-term. Male sex was also associated with limited brain repair following neonatal stroke and hypoxia-ischemia, leading to increased incidence of long-term cognitive deficits compared to females with similar brain injury. As a result, the design of pre-clinical experiments considering sex as an important variable was supported and investigated because neuroprotective strategies to reduce brain injury demonstrated sexual dimorphism. While the mechanisms underlining these differences between boys and girls remain unclear, several biological processes are recognized to play a key role in long-term neurodevelopmental outcomes: gonadal hormones across developmental stages, vulnerability to oxidative stress, modulation of cell death, and regulation of microglial activation. This review summarizes the current evidence for sex differences in neonatal hypoxic-ischemic and/or ischemic brain injury, considering the major pathways known to be involved in cognitive and behavioral deficits associated with damages of the developing brain.
Collapse
Affiliation(s)
- Christiane Charriaut-Marlangue
- U1141 PROTECT, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, 48 boulevard Sérurier, 75019 Paris, France.
| | - Valérie C Besson
- U1141 PROTECT, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, 48 boulevard Sérurier, 75019 Paris, France.
- EA4475-Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Sorbonne Paris Cité, 4 Avenue de l'Observatoire, 75006 Paris, France.
| | - Olivier Baud
- U1141 PROTECT, Inserm, Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, 48 boulevard Sérurier, 75019 Paris, France.
- Division of Neonatology and Pediatric Intensive Care, Children's University Hospital of Geneva and University of Geneva, 1205 Geneva, Switzerland.
| |
Collapse
|
69
|
Thornton C, Jones A, Nair S, Aabdien A, Mallard C, Hagberg H. Mitochondrial dynamics, mitophagy and biogenesis in neonatal hypoxic-ischaemic brain injury. FEBS Lett 2017; 592:812-830. [PMID: 29265370 DOI: 10.1002/1873-3468.12943] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/22/2017] [Accepted: 12/11/2017] [Indexed: 12/13/2022]
Abstract
Hypoxic-ischaemic encephalopathy, resulting from asphyxia during birth, affects 2-3 in every 1000 term infants and depending on severity, brings about life-changing neurological consequences or death. This hypoxic-ischaemia (HI) results in a delayed neural energy failure during which the majority of brain injury occurs. Currently, there are limited treatment options and additional therapies are urgently required. Mitochondrial dysfunction acts as a focal point in injury development in the immature brain. Not only do mitochondria become permeabilised, but recent findings implicate perturbations in mitochondrial dynamics (fission, fusion), mitophagy and biogenesis. Mitoprotective therapies may therefore offer a new avenue of intervention for babies who suffer lifelong disabilities due to birth asphyxia.
Collapse
Affiliation(s)
- Claire Thornton
- Perinatal Brain Injury Group, Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Adam Jones
- Perinatal Brain Injury Group, Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Syam Nair
- Perinatal Center, Department of Physiology, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Afra Aabdien
- Perinatal Brain Injury Group, Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Carina Mallard
- Perinatal Center, Department of Physiology, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Henrik Hagberg
- Perinatal Brain Injury Group, Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK.,Perinatal Center, Department of Clinical Sciences & Physiology and Neuroscience, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
70
|
López S, Martá M, Sequeda LG, Celis C, Sutachan JJ, Albarracín SL. Cytoprotective action against oxidative stress in astrocytes and neurons by Bactris guineensis (L.) H.E. Moore (corozo) fruit extracts. Food Chem Toxicol 2017; 109:1010-1017. [DOI: 10.1016/j.fct.2017.04.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 11/28/2022]
|
71
|
Oxidative stress and endoplasmic reticulum (ER) stress in the development of neonatal hypoxic-ischaemic brain injury. Biochem Soc Trans 2017; 45:1067-1076. [PMID: 28939695 PMCID: PMC5652227 DOI: 10.1042/bst20170017] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/09/2017] [Accepted: 08/11/2017] [Indexed: 02/06/2023]
Abstract
Birth asphyxia in term neonates affects 1–2/1000 live births and results in the development of hypoxic–ischaemic encephalopathy with devastating life-long consequences. The majority of neuronal cell death occurs with a delay, providing the potential of a treatment window within which to act. Currently, treatment options are limited to therapeutic hypothermia which is not universally successful. To identify new interventions, we need to understand the molecular mechanisms underlying the injury. Here, we provide an overview of the contribution of both oxidative stress and endoplasmic reticulum stress in the development of neonatal brain injury and identify current preclinical therapeutic strategies.
Collapse
|
72
|
Sepuri NBV, Angireddy R, Srinivasan S, Guha M, Spear J, Lu B, Anandatheerthavarada HK, Suzuki CK, Avadhani NG. Mitochondrial LON protease-dependent degradation of cytochrome c oxidase subunits under hypoxia and myocardial ischemia. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2017; 1858:519-528. [PMID: 28442264 PMCID: PMC5507603 DOI: 10.1016/j.bbabio.2017.04.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 04/17/2017] [Accepted: 04/21/2017] [Indexed: 01/08/2023]
Abstract
The mitochondrial ATP dependent matrix protease, Lon, is involved in the maintenance of mitochondrial DNA nucleoids and degradation of abnormal or misfolded proteins. The Lon protease regulates mitochondrial Tfam (mitochondrial transcription factor A) level and thus modulates mitochondrial DNA (mtDNA) content. We have previously shown that hypoxic stress induces the PKA-dependent phosphorylation of cytochrome c oxidase (CcO) subunits I, IVi1, and Vb and a time-dependent reduction of these subunits in RAW 264.7 murine macrophages subjected to hypoxia and rabbit hearts subjected to ischemia/reperfusion. Here, we show that Lon is involved in the preferential turnover of phosphorylated CcO subunits under hypoxic/ischemic stress. Induction of Lon protease occurs at 6 to 12 h of hypoxia and this increase coincides with lower CcO subunit contents. Over-expression of flag-tagged wild type and phosphorylation site mutant Vb and IVi1 subunits (S40A and T52A, respectively) caused marked degradation of wild type protein under hypoxia while the mutant proteins were relatively resistant. Furthermore, the recombinant purified Lon protease degraded the phosphorylated IVi1 and Vb subunits, while the phosphorylation-site mutant proteins were resistant to degradation. 3D structural modeling shows that the phosphorylation sites are exposed to the matrix compartment, accessible to matrix PKA and Lon protease. Hypoxic stress did not alter CcO subunit levels in Lon depleted cells, confirming its role in CcO turnover. Our results therefore suggest that Lon preferentially degrades the phosphorylated subunits of CcO and plays a role in the regulation of CcO activity in hypoxia and ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Naresh B V Sepuri
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104-6009, USA
| | - Rajesh Angireddy
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104-6009, USA
| | - Satish Srinivasan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104-6009, USA
| | - Manti Guha
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104-6009, USA
| | - Joseph Spear
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104-6009, USA
| | - Bin Lu
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers The State University, New Jersey Medical School, 225 Warren Street, Newark, NJ 17103-3535, USA
| | - Hindupur K Anandatheerthavarada
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104-6009, USA
| | - Carolyn K Suzuki
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers The State University, New Jersey Medical School, 225 Warren Street, Newark, NJ 17103-3535, USA
| | - Narayan G Avadhani
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104-6009, USA.
| |
Collapse
|
73
|
Claus Henn B, Bellinger DC, Hopkins MR, Coull BA, Ettinger AS, Jim R, Hatley E, Christiani DC, Wright RO. Maternal and Cord Blood Manganese Concentrations and Early Childhood Neurodevelopment among Residents near a Mining-Impacted Superfund Site. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:067020. [PMID: 28665786 PMCID: PMC5743453 DOI: 10.1289/ehp925] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 11/20/2016] [Accepted: 11/30/2016] [Indexed: 05/21/2023]
Abstract
BACKGROUND Environmental manganese exposure has been associated with adverse neurodevelopmental outcomes among school-aged children; yet, few studies have evaluated prenatal exposure. OBJECTIVES Our study examines associations between prenatal manganese concentrations and placental transfer of manganese with neurodevelopment in 224 2-y-old children residing near the Tar Creek Superfund Site. METHODS We collected maternal and cord blood at delivery, measured manganese using inductively coupled plasma mass spectrometry, and assessed neurodevelopment using the Bayley Scales of Infant Development-II. Associations between manganese and mental (MDI) and psychomotor (PDI) development indices were estimated in multivariable models. Placental transfer, approximated by cord/maternal manganese ratio, cord/total manganese ratio (total=maternal+cord), and by joint classification according to high or low (above or below median) maternal and cord manganese, was evaluated as a predictor of neurodevelopment. RESULTS Median levels [interquartile ranges (IQR)] of manganese in maternal and cord blood, respectively, were 24.0 (19.5-29.7) and 43.1 (33.5-52.1) μg/L. Adjusting for lead, arsenic, and other potential confounders, an IQR increase in maternal manganese was associated with -3.0 (95% CI: -5.3, -0.7) points on MDI and -2.3 (95% CI: -4.1, -0.4) points on PDI. Cord manganese concentrations were not associated with neurodevelopment scores. Cord/maternal and cord/total manganese ratios were positively associated with MDI [cord/maternal: β=2.6 (95% Cl: −0.04, 5.3); cord/total: β=22.0 (95% Cl: 3.2, 40.7)] and PDI (cord/maternal: β=1.7 (95% Cl: −0.5, 3.9); cord/total: β=15.6 (95% Cl: 0.3, 20.9)). Compared to mother-child pairs with low maternal and cord manganese, associations with neurodevelopment scores were negative for pairs with either high maternal, high cord, or high maternal and cord manganese. CONCLUSIONS Maternal blood manganese concentrations were negatively associated with early childhood neurodevelopment scores in our study. Findings highlight the importance of understanding maternal exposures during pregnancy and factors influencing placental transfer. https://doi.org/10.1289/EHP925.
Collapse
Affiliation(s)
- Birgit Claus Henn
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts, USA
| | - David C Bellinger
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Neurology, Harvard Medical School and Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School and Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Marianne R Hopkins
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Brent A Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Adrienne S Ettinger
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Rebecca Jim
- Local Environmental Action Demanded (L.E.A.D.) Agency, Inc., Vinita, Oklahoma, USA
| | - Earl Hatley
- Local Environmental Action Demanded (L.E.A.D.) Agency, Inc., Vinita, Oklahoma, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Robert O Wright
- Division of Environmental Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
74
|
Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal Hypoxia Ischaemia: Mechanisms, Models, and Therapeutic Challenges. Front Cell Neurosci 2017; 11:78. [PMID: 28533743 PMCID: PMC5420571 DOI: 10.3389/fncel.2017.00078] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Neonatal hypoxia-ischaemia (HI) is the most common cause of death and disability in human neonates, and is often associated with persistent motor, sensory, and cognitive impairment. Improved intensive care technology has increased survival without preventing neurological disorder, increasing morbidity throughout the adult population. Early preventative or neuroprotective interventions have the potential to rescue brain development in neonates, yet only one therapeutic intervention is currently licensed for use in developed countries. Recent investigations of the transient cortical layer known as subplate, especially regarding subplate's secretory role, opens up a novel set of potential molecular modulators of neonatal HI injury. This review examines the biological mechanisms of human neonatal HI, discusses evidence for the relevance of subplate-secreted molecules to this condition, and evaluates available animal models. Neuroserpin, a neuronally released neuroprotective factor, is discussed as a case study for developing new potential pharmacological interventions for use post-ischaemic injury.
Collapse
Affiliation(s)
- Lancelot J. Millar
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Lei Shi
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan UniversityGuangzhou, China
| | | | - Zoltán Molnár
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| |
Collapse
|
75
|
Wang YJ, Peng QY, Deng SY, Chen CX, Wu L, Huang L, Zhang LN. Hemin protects against oxygen-glucose deprivation-induced apoptosis activation via neuroglobin in SH-SY5Y cells. Neurochem Res 2017; 42:2208-2217. [PMID: 28316021 DOI: 10.1007/s11064-017-2230-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/28/2017] [Accepted: 03/10/2017] [Indexed: 02/07/2023]
Abstract
This study aimed to investigate the mechanism underlying the neuroprotective effect of hemin in oxygen-glucose deprivation (OGD)-treated neurons. OGD-treated SH-SY5Y cells (human neuroblastoma cells) were used in the study. The cellular viability of SH-SY5Y cells was assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and the cell apoptosis rate was determined by flow cytometry analysis with Annexin V-fluorescein isothiocyanate and propidium iodide staining with or without hemin pretreatment. Cell viability and apoptotic activation were detected after hemin administration combined with neuroglobin (Nqb), thioredoxin-1, peroxiredoxin-2, or heme oxygenase-1 siRNA transient transfection. The release of cytochrome c from mitochondria and the interaction between Ngb and cytochrome c were examined with hemin pretreatment. Hemin had a neuroprotective effect in OGD-treated SH-SY5Y cells, which was mainly mediated by the upregulation of Ngb. Moreover, the release of cytochrome c from mitochondria was inhibited by hemin-induced Ngb expression through facilitating the interaction of Ngb with cytochrome c in mitochondria. The present findings provided new insights into the neuroprotective mechanisms of hemin. It was concluded that low-dose hemin pretreatment had a neuroprotective effect in OGD-treated SH-SY5Y cells, through inhibiting cell apoptosis. The neuroprotective effects of hemin following hypoxic-ischemic neuronal damage were mainly mediated by Ngb. One underlying mechanism was hemin-induced overexpression of mitochondrial Ngb, which inhibited endogenous apoptosis via the association with cytochrome c.
Collapse
Affiliation(s)
- Yun-Jia Wang
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, No 87, Xiangya Road, Changsha, 410008, China
| | - Qian-Yi Peng
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, No 87, Xiangya Road, Changsha, 410008, China
| | - Song-Yun Deng
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, No 87, Xiangya Road, Changsha, 410008, China
| | - Cai-Xia Chen
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, No 87, Xiangya Road, Changsha, 410008, China
| | - Long Wu
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, No 87, Xiangya Road, Changsha, 410008, China
| | - Li Huang
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, No 87, Xiangya Road, Changsha, 410008, China
| | - Li-Na Zhang
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, No 87, Xiangya Road, Changsha, 410008, China.
| |
Collapse
|
76
|
Sonei N, Amiri S, Jafarian I, Anoush M, Rahimi-Balaei M, Bergen H, Haj-Mirzaian A, Hosseini MJ. Mitochondrial dysfunction bridges negative affective disorders and cardiomyopathy in socially isolated rats: Pros and cons of fluoxetine. World J Biol Psychiatry 2017; 18:39-53. [PMID: 27031288 DOI: 10.3109/15622975.2016.1149218] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objectives Depression is tightly associated with cardiovascular comorbidity and accounts for high financial and social burden worldwide. Mitochondrial dysfunction contributes to the pathophysiology of depression and cardiovascular disorders; its contribution to depression-cardiovascular comorbidity has not yet been investigated. Methods Adolescent rats were subjected to 4 weeks of isolation (social isolation stress or SIS) or social conditions (control), and then they were divided into treatment (fluoxetine, 7.5 mg/kg/day for 21 days) and non-treatment groups. After different housing conditions and treatment, animals were evaluated by behavioural tests (n = 6-8) and mitochondrial assessments (n = 3) of brain and cardiac tissues. Results We found that juvenile SIS induced behavioural abnormalities and mitochondrial dysfunction in adulthood. We showed that juvenile SIS was associated with impaired respiratory chain complex, which leads to reactive oxygen species formation, oxidative damage and ATP abatement in both brain and heart. Administration of FLX (7.5 mg/kg/day) during the isolation period attenuated the effects of SIS on the brain mitochondria and behavioural abnormalities, but had little or no effect on SIS-induced mitochondrial dysfunction in cardiac tissue. Conclusions This suggests that juvenile SIS predisposes the co-occurrence of depression and cardiovascular disease through mitochondrial dysfunction and that therapeutic effect of fluoxetine is partly mediated by its effect on mitochondrial function.
Collapse
Affiliation(s)
- Nazanin Sonei
- a Zanjan Applied Pharmacology Research Center , Zanjan University of Medical Sciences , Zanjan , Iran.,b Department of Pharmacology and Toxicology, School of Pharmacy , Zanjan University of Medical Sciences , Zanjan , Iran
| | - Shayan Amiri
- c Department of Pharmacology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,d Experimental Medicine Research Center , Tehran University of Medical Sciences , Tehran , Iran
| | - Iman Jafarian
- a Zanjan Applied Pharmacology Research Center , Zanjan University of Medical Sciences , Zanjan , Iran.,b Department of Pharmacology and Toxicology, School of Pharmacy , Zanjan University of Medical Sciences , Zanjan , Iran
| | - Mahdieh Anoush
- a Zanjan Applied Pharmacology Research Center , Zanjan University of Medical Sciences , Zanjan , Iran.,b Department of Pharmacology and Toxicology, School of Pharmacy , Zanjan University of Medical Sciences , Zanjan , Iran
| | - Maryam Rahimi-Balaei
- e Department of Human Anatomy and Cell Science , College of Medicine, Faculty of Health Sciences, University of Manitoba , Winnipeg , Manitoba , Canada
| | - Hugo Bergen
- e Department of Human Anatomy and Cell Science , College of Medicine, Faculty of Health Sciences, University of Manitoba , Winnipeg , Manitoba , Canada
| | - Arya Haj-Mirzaian
- c Department of Pharmacology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,d Experimental Medicine Research Center , Tehran University of Medical Sciences , Tehran , Iran
| | - Mir-Jamal Hosseini
- a Zanjan Applied Pharmacology Research Center , Zanjan University of Medical Sciences , Zanjan , Iran.,b Department of Pharmacology and Toxicology, School of Pharmacy , Zanjan University of Medical Sciences , Zanjan , Iran
| |
Collapse
|
77
|
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a disease that occurs when the brain is subjected to hypoxia, resulting in neuronal death and neurological deficits, with a poor prognosis. The mechanisms underlying hypoxic-ischemic brain injury include excitatory amino acid release, cellular proteolysis, reactive oxygen species generation, nitric oxide synthesis, and inflammation. The molecular and cellular changes in HIE include protein misfolding, aggregation, and destruction of organelles. The apoptotic pathways activated by ischemia and hypoxia include the mitochondrial pathway, the extrinsic Fas receptor pathway, and the endoplasmic reticulum stress-induced pathway. Numerous treatments for hypoxic-ischemic brain injury caused by HIE have been developed over the last half century. Hypothermia, xenon gas treatment, the use of melatonin and erythropoietin, and hypoxic-ischemic preconditioning have proven effective in HIE patients. Molecular chaperones are proteins ubiquitously present in both prokaryotes and eukaryotes. A large number of molecular chaperones are induced after brain ischemia and hypoxia, among which the heat shock proteins are the most important. Heat shock proteins not only maintain protein homeostasis; they also exert anti-apoptotic effects. Heat shock proteins maintain protein homeostasis by helping to transport proteins to their target destinations, assisting in the proper folding of newly synthesized polypeptides, regulating the degradation of misfolded proteins, inhibiting the aggregation of proteins, and by controlling the refolding of misfolded proteins. In addition, heat shock proteins exert anti-apoptotic effects by interacting with various signaling pathways to block the activation of downstream effectors in numerous apoptotic pathways, including the intrinsic pathway, the endoplasmic reticulum-stress mediated pathway and the extrinsic Fas receptor pathway. Molecular chaperones play a key role in neuroprotection in HIE. In this review, we provide an overview of the mechanisms of HIE and discuss the various treatment strategies. Given their critical role in the disease, molecular chaperones are promising therapeutic targets for HIE.
Collapse
Affiliation(s)
- Cong Hua
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Wei-Na Ju
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Hang Jin
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xin Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Gang Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
78
|
Luo C, Jian C, Liao Y, Huang Q, Wu Y, Liu X, Zou D, Wu Y. The role of microglia in multiple sclerosis. Neuropsychiatr Dis Treat 2017; 13:1661-1667. [PMID: 28721047 PMCID: PMC5499932 DOI: 10.2147/ndt.s140634] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS). Microglia are the resident innate immune cells in the CNS; they play an important role in the processes of demyelination and remyelination in MS. Microglia can function as antigen-presenting cells and phagocytes. In the past, microglia were considered to be the same cell type as macrophages, and researchers have different opinions about the role of microglia in MS. This review focuses on the original classification of microglia and their role in the pathogenesis of MS. Moreover, we present a hypothetical model for the role of microglia in the pathogenesis of MS based on recent findings.
Collapse
Affiliation(s)
- Chun Luo
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University
| | - Chongdong Jian
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University
| | - Yuhan Liao
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University
| | - Qi Huang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University
| | - Yuejuan Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University
| | - Xixia Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University
| | - Donghua Zou
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University and The First People's Hospital of Nanning, Nanning, People's Republic of China
| | - Yuan Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University
| |
Collapse
|
79
|
Maternal creatine supplementation during pregnancy prevents acute and long-term deficits in skeletal muscle after birth asphyxia: a study of structure and function of hind limb muscle in the spiny mouse. Pediatr Res 2016; 80:852-860. [PMID: 27466898 DOI: 10.1038/pr.2016.153] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/10/2016] [Indexed: 11/08/2022]
Abstract
BACKGROUND Maternal antenatal creatine supplementation protects the brain, kidney, and diaphragm against the effects of birth asphyxia in the spiny mouse. In this study, we examined creatine's potential to prevent damage to axial skeletal muscles. METHODS Pregnant spiny mice were fed a control or creatine-supplemented diet from mid-pregnancy, and 1 d before term (39 d), fetuses were delivered by c-section with or without 7.5 min of birth asphyxia. At 24 h or 33 ± 2 d after birth, gastrocnemius muscles were obtained for ex-vivo study of twitch-tension, muscle fatigue, and structural and histochemical analysis. RESULTS Birth asphyxia significantly reduced cross-sectional area of all muscle fiber types (P < 0.05), and increased fatigue caused by repeated tetanic contractions at 24 h of age (P < 0.05). There were fewer (P < 0.05) Type I and IIa fibers and more (P < 0.05) Type IIb fibers in male gastrocnemius at 33 d of age. Muscle oxidative capacity was reduced (P < 0.05) in males at 24 h and 33 d and in females at 24 h only. Maternal creatine treatment prevented all asphyxia-induced changes in the gastrocnemius, improved motor performance. CONCLUSION This study demonstrates that creatine loading before birth protects the muscle from asphyxia-induced damage at birth.
Collapse
|
80
|
Netto CA, Sanches E, Odorcyk FK, Duran-Carabali LE, Weis SN. Sex-dependent consequences of neonatal brain hypoxia-ischemia in the rat. J Neurosci Res 2016; 95:409-421. [DOI: 10.1002/jnr.23828] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Carlos Alexandre Netto
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Eduardo Sanches
- Division of Child Development and Growth, Department of Pediatrics; University of Geneva; Geneva Switzerland
| | - Felipe Kawa Odorcyk
- Postgraduate Program of Neurosciences, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Luz Elena Duran-Carabali
- Postgraduate Program of Physiology, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Simone Nardin Weis
- Department of Cellular Biology; Universidade de Brasília; Brasilia Distrito Federal Brazil
| |
Collapse
|
81
|
Chew LJ, DeBoy CA. Pharmacological approaches to intervention in hypomyelinating and demyelinating white matter pathology. Neuropharmacology 2016; 110:605-625. [PMID: 26116759 PMCID: PMC4690794 DOI: 10.1016/j.neuropharm.2015.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 06/10/2015] [Accepted: 06/17/2015] [Indexed: 12/17/2022]
Abstract
White matter disease afflicts both developing and mature central nervous systems. Both cell intrinsic and extrinsic dysregulation result in profound changes in cell survival, axonal metabolism and functional performance. Experimental models of developmental white matter (WM) injury and demyelination have not only delineated mechanisms of signaling and inflammation, but have also paved the way for the discovery of pharmacological approaches to intervention. These reagents have been shown to enhance protection of the mature oligodendrocyte cell, accelerate progenitor cell recruitment and/or differentiation, or attenuate pathological stimuli arising from the inflammatory response to injury. Here we highlight reports of studies in the CNS in which compounds, namely peptides, hormones, and small molecule agonists/antagonists, have been used in experimental animal models of demyelination and neonatal brain injury that affect aspects of excitotoxicity, oligodendrocyte development and survival, and progenitor cell function, and which have been demonstrated to attenuate damage and improve WM protection in experimental models of injury. The molecular targets of these agents include growth factor and neurotransmitter receptors, morphogens and their signaling components, nuclear receptors, as well as the processes of iron transport and actin binding. By surveying the current evidence in non-immune targets of both the immature and mature WM, we aim to better understand pharmacological approaches modulating endogenous oligodendroglia that show potential for success in the contexts of developmental and adult WM pathology. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA.
| | - Cynthia A DeBoy
- Biology Department, Trinity Washington University, Washington, DC, USA
| |
Collapse
|
82
|
Schiavone S, Riezzo I, Turillazzi E, Trabace L. Involvement of the NADPH Oxidase NOX2-Derived Brain Oxidative Stress in an Unusual Fatal Case of Cocaine-Related Neurotoxicity Associated With Excited Delirium Syndrome. J Clin Psychopharmacol 2016; 36:513-7. [PMID: 27533346 DOI: 10.1097/jcp.0000000000000560] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Here, we investigated the possible role of the Nicotinamide Adenine Dinucleotide Phosphate oxidase NOX2-derived brain oxidative stress in a fatal case of cocaine-related neurotoxicity, associated to excited delirium syndrome. We detected a strong NOX2 immunoreactivity, mainly in cortical GABAergic neurons and astrocytes, with a minor presence in microglia, glutamatergic and dopaminergic neurons as well as a significant immunostaining for other markers of oxidative stress (8OhDG, HSP70, HSP90, and NF-κB) and apoptotic phenomena. These results support a crucial role of NOX2-derived brain oxidative stress in cocaine-induced brain dysfunctions and neurotoxicity.
Collapse
Affiliation(s)
- Stefania Schiavone
- From the Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | | | | | | |
Collapse
|
83
|
Park SM, Park CW, Lee TK, Cho JH, Park JH, Lee JC, Chen BH, Shin BN, Ahn JH, Tae HJ, Shin MC, Ohk TG, Cho JH, Won MH, Choi SY, Kim IH. Effect of ischemic preconditioning on antioxidant status in the gerbil hippocampal CA1 region after transient forebrain ischemia. Neural Regen Res 2016; 11:1081-9. [PMID: 27630689 PMCID: PMC4994448 DOI: 10.4103/1673-5374.187039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Ischemic preconditioning (IPC) is a condition of sublethal transient global ischemia and exhibits neuroprotective effects against subsequent lethal ischemic insult. We, in this study, examined the neuroprotective effects of IPC and its effects on immunoreactive changes of antioxidant enzymes including superoxide dismutase (SOD) 1 and SOD2, catalase (CAT) and glutathione peroxidase (GPX) in the gerbil hippocampal CA1 region after transient forebrain ischemia. Pyramidal neurons of the stratum pyramidale (SP) in the hippocampal CA1 region of animals died 5 days after lethal transient ischemia without IPC (8.6% (ratio of remanent neurons) of the sham-operated group); however, IPC prevented the pyramidal neurons from subsequent lethal ischemic injury (92.3% (ratio of remanent neurons) of the sham-operated group). SOD1, SOD2, CAT and GPX immunoreactivities in the sham-operated animals were easily detected in pyramidal neurons in the stratum pyramidale (SP) of the hippocampal CA1 region, while all of these immunoreactivities were rarely detected in the stratum pyramidale at 5 days after lethal transient ischemia without IPC. Meanwhile, their immunoreactivities in the sham-operated animals with IPC were similar to (SOD1, SOD2 and CAT) or higher (GPX) than those in the sham-operated animals without IPC. Furthermore, their immunoreactivities in the stratum pyramidale of the ischemia-operated animals with IPC were steadily maintained after lethal ischemia/reperfusion. Results of western blot analysis for SOD1, SOD2, CAT and GPX were similar to immunohistochemical data. In conclusion, IPC maintained or increased the expression of antioxidant enzymes in the stratum pyramidale of the hippocampal CA1 region after subsequent lethal transient forebrain ischemia and IPC exhibited neuroprotective effects in the hippocampal CA1 region against transient forebrain ischemia.
Collapse
Affiliation(s)
- Seung Min Park
- Department of Emergency Medicine, Sacred Heart Hospital, College of Medicine, Hallym University, Anyang, South Korea; Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Chan Woo Park
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Bai Hui Chen
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Bich-Na Shin
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Hyun-Jin Tae
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Soo Young Choi
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
84
|
Retta SF, Glading AJ. Oxidative stress and inflammation in cerebral cavernous malformation disease pathogenesis: Two sides of the same coin. Int J Biochem Cell Biol 2016; 81:254-270. [PMID: 27639680 PMCID: PMC5155701 DOI: 10.1016/j.biocel.2016.09.011] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022]
Abstract
CCM proteins play pleiotropic roles in various redox-sensitive signaling pathways. CCM proteins modulate the crosstalk between redox signaling and autophagy that govern cell homeostasis and stress responses. Oxidative stress and inflammation are emerging as key focal determinants of CCM lesion formation, progression and severity. The pleiotropic functions of CCM proteins may prevent vascular dysfunctions triggered by local oxidative stress and inflammatory events. The distinct therapeutic compounds proposed so far for CCM disease share the ability to modulate redox signaling and autophagy.
Cerebral Cavernous Malformation (CCM) is a vascular disease of proven genetic origin, which may arise sporadically or is inherited as an autosomal dominant condition with incomplete penetrance and highly variable expressivity. CCM lesions exhibit a range of different phenotypes, including wide inter-individual differences in lesion number, size, and susceptibility to intracerebral hemorrhage (ICH). Lesions may remain asymptomatic or result in pathological conditions of various type and severity at any age, with symptoms ranging from recurrent headaches to severe neurological deficits, seizures, and stroke. To date there are no direct therapeutic approaches for CCM disease besides the surgical removal of accessible lesions. Novel pharmacological strategies are particularly needed to limit disease progression and severity and prevent de novo formation of CCM lesions in susceptible individuals. Useful insights into innovative approaches for CCM disease prevention and treatment are emerging from a growing understanding of the biological functions of the three known CCM proteins, CCM1/KRIT1, CCM2 and CCM3/PDCD10. In particular, accumulating evidence indicates that these proteins play major roles in distinct signaling pathways, including those involved in cellular responses to oxidative stress, inflammation and angiogenesis, pointing to pathophysiological mechanisms whereby the function of CCM proteins may be relevant in preventing vascular dysfunctions triggered by these events. Indeed, emerging findings demonstrate that the pleiotropic roles of CCM proteins reflect their critical capacity to modulate the fine-tuned crosstalk between redox signaling and autophagy that govern cell homeostasis and stress responses, providing a novel mechanistic scenario that reconciles both the multiple signaling pathways linked to CCM proteins and the distinct therapeutic approaches proposed so far. In addition, recent studies in CCM patient cohorts suggest that genetic susceptibility factors related to differences in vascular sensitivity to oxidative stress and inflammation contribute to inter-individual differences in CCM disease susceptibility and severity. This review discusses recent progress into the understanding of the molecular basis and mechanisms of CCM disease pathogenesis, with specific emphasis on the potential contribution of altered cell responses to oxidative stress and inflammatory events occurring locally in the microvascular environment, and consequent implications for the development of novel, safe, and effective preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Saverio Francesco Retta
- Department of Clinical and Biological Sciences, School of Medicine and Surgery, University of Torino, Regione Gonzole 10, 10043 Orbassano, Torino, Italy; CCM Italia Research Network(1).
| | - Angela J Glading
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, 14642 Rochester, NY, USA.
| |
Collapse
|
85
|
Bakhtyukov AA, Galkina OV, Eshchenko ND. The activities of key antioxidant enzymes in the early postnatal development of rats. NEUROCHEM J+ 2016. [DOI: 10.1134/s1819712416030041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
86
|
Intratracheal transplantation of mesenchymal stem cells simultaneously attenuates both lung and brain injuries in hyperoxic newborn rats. Pediatr Res 2016; 80:415-24. [PMID: 27064241 DOI: 10.1038/pr.2016.88] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 02/20/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia is an independent risk factor for adverse neurodevelopmental outcomes in premature infants. We investigated whether attenuation of hyperoxic lung injury with intratracheal transplantation of human umbilical cord blood-derived mesenchymal stem cells (MSCs) could simultaneously mitigate brain damage in neonatal rats. METHODS Newborn Sprague-Dawley rats were exposed to hyperoxia or normoxia conditions for 14 d. MSCs (5 × 10(5) cells) were transplanted intratracheally at postnatal day (P) 5. At P14, lungs and brains were harvested for histological and biochemical analyses. RESULTS Hyperoxic lung injuries, such as impaired alveolarization evident from increased mean linear intercept (MLI) and elevated inflammatory cytokine levels were significantly alleviated with MSC transplantation. Hyperoxia decreased brain weight, increased brain cell death, and induced hypomyelination. MSC transplantation significantly ameliorated hyperoxia-induced increased terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells in the dentate gyrus and reduced myelin basic protein. In correlation analyses, brain weight and myelin basic protein (MBP) were significantly inversely correlated with lung MLI and inflammatory cytokines, while TUNEL-positive brain cell number showed a significant positive correlation with lung MLI. CONCLUSION Despite no significant improvement in short-term neurofunctional outcome, intratracheal transplantation of MSCs simultaneously attenuated hyperoxic lung and brain injuries in neonatal rats, with the extent of such attenuation being closely linked in the two tissues.
Collapse
|
87
|
Demarest TG, Waite EL, Kristian T, Puche AC, Waddell J, McKenna MC, Fiskum G. Sex-dependent mitophagy and neuronal death following rat neonatal hypoxia-ischemia. Neuroscience 2016; 335:103-13. [PMID: 27555552 DOI: 10.1016/j.neuroscience.2016.08.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/11/2016] [Accepted: 08/12/2016] [Indexed: 12/19/2022]
Abstract
Males are more susceptible than females to long-term cognitive deficits following neonatal hypoxic-ischemic encephalopathy (HIE). Mitochondrial dysfunction is implicated in the pathophysiology of cerebral hypoxia-ischemia (HI), but the influence of sex on mitochondrial quality control (MQC) after HI is unknown. Therefore, we tested the hypothesis that mitophagy is sexually dimorphic and neuroprotective 20-24h following the Rice-Vannucci model of rat neonatal HI at postnatal day 7 (PN7). Mitochondrial and lysosomal morphology and degree of co-localization were determined by immunofluorescence in the cerebral cortex. No difference in mitochondrial abundance was detected in the cortex after HI. However, net mitochondrial fission increased in both hemispheres of female brain, but was most extensive in the ipsilateral hemisphere of male brain following HI. Basal autophagy, assessed by immunoblot for the autophagosome marker LC3BI/II, was greater in males suggesting less intrinsic reserve capacity for autophagy following HI. Autophagosome formation, lysosome size, and TOM20/LAMP2 co-localization were increased in the contralateral hemisphere following HI in female, but not male brain. An accumulation of ubiquitinated mitochondrial protein was observed in male, but not female brain following HI. Moreover, neuronal cell death with NeuN/TUNEL co-staining occurred in both hemispheres of male brain, but only in the ipsilateral hemisphere of female brain after HI. In summary, mitophagy induction and neuronal cell death are sex dependent following HI. The deficit in elimination of damaged/dysfunctional mitochondria in the male brain following HI may contribute to male vulnerability to neuronal death and long-term neurobehavioral deficits following HIE.
Collapse
Affiliation(s)
- T G Demarest
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - E L Waite
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 21201, USA
| | - T Kristian
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA; Veterans Affairs Medical Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - A C Puche
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - J Waddell
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - M C McKenna
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - G Fiskum
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
88
|
Deferoxamine improves antioxidative protection in the brain of neonatal rats: The role of anoxia and body temperature. Neurosci Lett 2016; 628:116-22. [PMID: 27297770 DOI: 10.1016/j.neulet.2016.06.022] [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: 02/22/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 01/24/2023]
Abstract
After hypoxic-ischemic insult iron deposited in the brain catalyzes formation of reactive oxygen species. Newborn rats, showing reduced physiological body temperature and their hyperthermic counterparts injected with deferoxamine (DF), a chelator of iron, are protected both against iron-mediated neurotoxicity and against depletion of low-molecular antioxidants after perinatal asphyxia. Therefore, we decided to study the effects of DF on activity of antioxidant enzymes (superoxide dismutase-SOD, glutathione peroxidase-GPx and catalase-CAT) in the brain of rats exposed neonatally to a critical anoxia at body temperatures elevated to 39°C. Perinatal anoxia under hyperthermic conditions intensified oxidative stress and depleted the pool of antioxidant enzymes. Both the depletion of antioxidants and lipid peroxidation were prevented by post-anoxic DF injection. The present paper evidenced that deferoxamine may act by recovering of SOD, GPx and CAT activity to reduce anoxia-induced oxidative stress.
Collapse
|
89
|
Demarest TG, Schuh RA, Waddell J, McKenna MC, Fiskum G. Sex-dependent mitochondrial respiratory impairment and oxidative stress in a rat model of neonatal hypoxic-ischemic encephalopathy. J Neurochem 2016; 137:714-29. [PMID: 27197831 DOI: 10.1111/jnc.13590] [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: 01/08/2016] [Revised: 02/12/2016] [Accepted: 02/17/2016] [Indexed: 01/09/2023]
Abstract
Increased male susceptibility to long-term cognitive deficits is well described in clinical and experimental studies of neonatal hypoxic-ischemic encephalopathy. While cell death signaling pathways are known to be sexually dimorphic, a sex-dependent pathophysiological mechanism preceding the majority of secondary cell death has yet to be described. Mitochondrial dysfunction contributes to cell death following cerebral hypoxic-ischemia (HI). Several lines of evidence suggest that there are sex differences in the mitochondrial metabolism of adult mammals. Therefore, this study tested the hypothesis that brain mitochondrial respiratory impairment and associated oxidative stress is more severe in males than females following HI. Maximal brain mitochondrial respiration during oxidative phosphorylation was two-fold more impaired in males following HI. The endogenous antioxidant glutathione was 30% higher in the brain of sham females compared to males. Females also exhibited increased glutathione peroxidase (GPx) activity following HI injury. Conversely, males displayed a reduction in mitochondrial GPx4 protein levels and mitochondrial GPx activity. Moreover, a 3-4-fold increase in oxidative protein carbonylation was observed in the cortex, perirhinal cortex, and hippocampus of injured males, but not females. These data provide the first evidence for sex-dependent mitochondrial respiratory dysfunction and oxidative damage, which may contribute to the relative male susceptibility to adverse long-term outcomes following HI. Lower basal GSH levels, lower post-hypoxic mitochondrial glutathione peroxidase (mtGPx) activity, and mitochondrial glutathione peroxidase 4 (mtGPx4) protein levels may contribute to the susceptibility of the male brain to oxidative damage and mitochondrial dysfunction following neonatal hypoxic-ischemia (HI). Treatment of male pups with acetyl-L-carnitine (ALCAR) protects against the loss of mtGPx activity, mtGPx4 protein, and increases in protein carbonylation after HI. These findings provide novel insight into the pathophysiology of sexually dimorphic outcomes following HI.
Collapse
Affiliation(s)
- Tyler G Demarest
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, Maryland, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Rosemary A Schuh
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jaylyn Waddell
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mary C McKenna
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gary Fiskum
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, Maryland, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
90
|
Schiavone S, Neri M, Mhillaj E, Morgese MG, Cantatore S, Bove M, Riezzo I, Tucci P, Pomara C, Turillazzi E, Cuomo V, Trabace L. The NADPH oxidase NOX2 as a novel biomarker for suicidality: evidence from human post mortem brain samples. Transl Psychiatry 2016; 6:e813. [PMID: 27187235 PMCID: PMC5070044 DOI: 10.1038/tp.2016.76] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/26/2016] [Accepted: 03/17/2016] [Indexed: 12/13/2022] Open
Abstract
Recent evidence points towards a role of oxidative stress in suicidality. However, few studies were carried out on the sources of reactive oxygen species (ROS) in subjects with suicidal behaviour. We have previously demonstrated that the NADPH oxidase NOX2-derived oxidative stress has a major role in the development of neuropathological alterations observed in an animal model of psychosis. Here, we investigated the possible increase in NOX2 in post mortem brain samples of subjects who died by asphyctic suicide (AS) compared with controls (CTRL) and subjects who died by non-suicidal asphyxia (NSA). We found that NOX2 expression was significantly higher in the cortex of AS subjects than in the other two experimental groups. NOX2 immunostaining was mainly detected in GABAergic neurons, with a minor presence of NOX2-positive-stained cells in glutamatergic and dopaminergic neurons, as well as astrocytes and microglia. A sustained increase in the expression of 8-hydroxy-2'-deoxyguanosine, an indirect marker of oxidative stress, was also detected in the cortex of AS subjects, compared with CTRL and NSA subjects. A significant elevation in cortical interleukin-6 immunoreactivity in AS subjects suggested an involvement of cytokine-associated molecular pathways in NOX2 elevations. Our results suggest that the increase in NOX2-derived oxidative stress in the brain might be involved in the neuropathological pathways leading to suicidal behaviour. These results may open innovative insights in the identification of new pathogenetic and necroscopic biomarkers, predictive for suicidality and potentially useful for suicide prevention.
Collapse
Affiliation(s)
- S Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy,Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, Foggia 71122, Italy. E-mail:
| | - M Neri
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - E Mhillaj
- Department of Physiology and Pharmacology, 'Sapienza' University of Rome, Rome, Italy
| | - M G Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - S Cantatore
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - M Bove
- Department of Physiology and Pharmacology, 'Sapienza' University of Rome, Rome, Italy
| | - I Riezzo
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - P Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - C Pomara
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - E Turillazzi
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - V Cuomo
- Department of Physiology and Pharmacology, 'Sapienza' University of Rome, Rome, Italy
| | - L Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
91
|
Plasticity in the Neonatal Brain following Hypoxic-Ischaemic Injury. Neural Plast 2016; 2016:4901014. [PMID: 27047695 PMCID: PMC4800097 DOI: 10.1155/2016/4901014] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/12/2016] [Accepted: 02/07/2016] [Indexed: 12/03/2022] Open
Abstract
Hypoxic-ischaemic damage to the developing brain is a leading cause of child death, with high mortality and morbidity, including cerebral palsy, epilepsy, and cognitive disabilities. The developmental stage of the brain and the severity of the insult influence the selective regional vulnerability and the subsequent clinical manifestations. The increased susceptibility to hypoxia-ischaemia (HI) of periventricular white matter in preterm infants predisposes the immature brain to motor, cognitive, and sensory deficits, with cognitive impairment associated with earlier gestational age. In term infants HI causes selective damage to sensorimotor cortex, basal ganglia, thalamus, and brain stem. Even though the immature brain is more malleable to external stimuli compared to the adult one, a hypoxic-ischaemic event to the neonate interrupts the shaping of central motor pathways and can affect normal developmental plasticity through altering neurotransmission, changes in cellular signalling, neural connectivity and function, wrong targeted innervation, and interruption of developmental apoptosis. Models of neonatal HI demonstrate three morphologically different types of cell death, that is, apoptosis, necrosis, and autophagy, which crosstalk and can exist as a continuum in the same cell. In the present review we discuss the mechanisms of HI injury to the immature brain and the way they affect plasticity.
Collapse
|
92
|
Kletkiewicz H, Nowakowska A, Siejka A, Mila-Kierzenkowska C, Woźniak A, Caputa M, Rogalska J. Deferoxamine prevents cerebral glutathione and vitamin E depletions in asphyxiated neonatal rats: role of body temperature. Int J Hyperthermia 2016; 32:211-20. [PMID: 26794834 DOI: 10.3109/02656736.2015.1125955] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hypoxic-ischaemic brain injury involves increased oxidative stress. In asphyxiated newborns iron deposited in the brain catalyses formation of reactive oxygen species. Glutathione (GSH) and vitamin E are key factors protecting cells against such agents. Our previous investigation has demonstrated that newborn rats, showing physiological low body temperature as well as their hyperthermic counterparts injected with deferoxamine (DF) are protected against iron-mediated, delayed neurotoxicity of perinatal asphyxia. Therefore, we decided to study the effects of body temperature and DF on the antioxidant status of the brain in rats exposed neonatally to critical anoxia. Two-day-old newborn rats were exposed to anoxia in 100% nitrogen atmosphere for 10 min. Rectal temperature was kept at 33 °C (physiological to rat neonates), or elevated to the level typical of healthy adult rats (37 °C), or of febrile adult rats (39 °C). Half of the rats exposed to anoxia under extremely hyperthermic conditions (39 °C) were injected with DF. Cerebral concentrations of malondialdehyde (MDA, lipid peroxidation marker) and the levels of GSH and vitamin E were determined post-mortem, (1) immediately after anoxia, (2) 3 days, (3) 7 days, and (4) 2 weeks after anoxia. There were no post-anoxic changes in MDA, GSH and vitamin E concentrations in newborn rats kept at body temperature of 33 °C. In contrast, perinatal anoxia at elevated body temperatures intensified oxidative stress and depleted the antioxidant pool in a temperature-dependent manner. Both the depletion of antioxidants and lipid peroxidation were prevented by post-anoxic DF injection. The data support the idea that hyperthermia may extend perinatal anoxia-induced brain lesions.
Collapse
Affiliation(s)
- Hanna Kletkiewicz
- a N. Copernicus University , Department of Animal Physiology, Faculty of Biology and Environmental Protection , Toruń , Poland and
| | - Anna Nowakowska
- a N. Copernicus University , Department of Animal Physiology, Faculty of Biology and Environmental Protection , Toruń , Poland and
| | - Agnieszka Siejka
- a N. Copernicus University , Department of Animal Physiology, Faculty of Biology and Environmental Protection , Toruń , Poland and
| | | | - Alina Woźniak
- b N. Copernicus University , Department of Medical Biology, Collegium Medicum , Bydgoszcz , Poland
| | - Michał Caputa
- a N. Copernicus University , Department of Animal Physiology, Faculty of Biology and Environmental Protection , Toruń , Poland and
| | - Justyna Rogalska
- a N. Copernicus University , Department of Animal Physiology, Faculty of Biology and Environmental Protection , Toruń , Poland and
| |
Collapse
|
93
|
Rangarajan P, Karthikeyan A, Lu J, Ling EA, Dheen S. Sirtuin 3 regulates Foxo3a-mediated antioxidant pathway in microglia. Neuroscience 2015; 311:398-414. [DOI: 10.1016/j.neuroscience.2015.10.048] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 10/22/2022]
|
94
|
Berger HR, Morken TS, Vettukattil R, Brubakk AM, Sonnewald U, Widerøe M. No improvement of neuronal metabolism in the reperfusion phase with melatonin treatment after hypoxic-ischemic brain injury in the neonatal rat. J Neurochem 2015; 136:339-50. [PMID: 26526584 DOI: 10.1111/jnc.13420] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/22/2015] [Accepted: 10/23/2015] [Indexed: 01/13/2023]
Abstract
Mitochondrial impairment is a key feature underlying neonatal hypoxic-ischemic (HI) brain injury and melatonin is potentially neuroprotective through its effects on mitochondria. In this study, we have used (1) H and (13) C NMR spectroscopy after injection of [1-(13) C]glucose and [1,2-(13) C]acetate to examine neuronal and astrocytic metabolism in the early reperfusion phase after unilateral HI brain injury in 7-day-old rat pups, exploring the effects of HI on mitochondrial function and the potential protective effects of melatonin on brain metabolism. One hour after hypoxia-ischemia, astrocytic metabolism was recovered and glycolysis was normalized, whereas mitochondrial metabolism in neurons was clearly impaired. Pyruvate carboxylation was also lower in both hemispheres after HI. The transfer of glutamate from neurons to astrocytes was higher whereas the transfer of glutamine from astrocytes to neurons was lower 1 h after HI in the contralateral hemisphere. Neuronal metabolism was equally affected in pups treated with melatonin (10 mg/kg) immediately after HI as in vehicle treated pups indicating that the given dose of melatonin was not capable of protecting the neuronal mitochondria in this early phase after HI brain injury. However, any beneficial effects of melatonin might have been masked by modulatory effects of the solvent dimethyl sulfoxide on cerebral metabolism. Neuronal and astrocytic metabolism was examined by (13) C and (1) H NMR spectroscopy in the early reperfusion phase after unilateral hypoxic-ischemic brain injury and melatonin treatment in neonatal rats. One hour after hypoxia-ischemia astrocytic mitochondrial metabolism had recovered and glycolysis was normalized, whereas mitochondrial metabolism in neurons was impaired. Melatonin treatment did not show a protective effect on neuronal metabolism.
Collapse
Affiliation(s)
- Hester R Berger
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tora Sund Morken
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Ophthalmology, Trondheim University Hospital, Trondheim, Norway
| | - Riyas Vettukattil
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ann-Mari Brubakk
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ursula Sonnewald
- Department of Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marius Widerøe
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
95
|
Peng C, Rao W, Zhang L, Wang K, Hui H, Wang L, Su N, Luo P, Hao YL, Tu Y, Zhang S, Fei Z. Mitofusin 2 ameliorates hypoxia-induced apoptosis via mitochondrial function and signaling pathways. Int J Biochem Cell Biol 2015; 69:29-40. [PMID: 26434502 DOI: 10.1016/j.biocel.2015.09.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/18/2015] [Accepted: 09/30/2015] [Indexed: 02/08/2023]
Abstract
Mitochondrial dynamics play a critical role in mitochondrial function and signaling. Although mitochondria play a critical role in hypoxia/ischemia, the further mechanisms between mitochondrial dynamics and ischemia are still unclear. The current study aimed to determine the role of mitofusin 2, a key regulator of mitochondrial fusion, in a hypoxic model and to explore a novel strategy for cerebral ischemia via modulation of mitochondrial dynamics. To the best of our knowledge, this is the first study to investigate both mitochondrial function and molecular pathways to determine the role of mitofusin 2 in hypoxia-induced neuronal apoptosis. In vivo, C57BL/6 mice (male, 19-25g) underwent a permanent middle cerebral artery occlusion for 12 or 24h (n=6 per group). In vitro, cobalt chloride was used to mimic hypoxia in immortalized hippocampal neurons. Down- or up-regulation of Mfn2 was induced to investigate the role of Mfn2 in hypoxia, especially in mitochondrial function and signaling pathways. The findings demonstrated that decreased mitofusin 2 occurred both in vivo and in vitro hypoxic models; second, the anti-apoptotic effect of Mfn2 may work via restoration of mitochondrial function; third, the modulation of the B Cell Leukemia 2/Bcl-2 Associated X protein and extracellular signal-regulated kinase 1/2 signaling pathways highlight the role of Mfn2 in signaling pathways beyond fusion. In summary, depletion of mitofusin 2 would lead to apoptosis both in normal or hypoxic conditions; however, mitofusin 2 overexpression could attenuate hypoxia-induced apoptosis, which represents a potential novel strategy for neuroprotection against ischemic brain damage.
Collapse
Affiliation(s)
- Cheng Peng
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Wei Rao
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Lei Zhang
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Kai Wang
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Hao Hui
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Li Wang
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Ning Su
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Peng Luo
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Ye-lu Hao
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Yue Tu
- Department of Neurosurgery, Affiliated Hospital of Logistics, University of Chinese Armed Police Forces, Chenglin Road, Tianjin 300162, PR China
| | - Sai Zhang
- Department of Neurosurgery, Affiliated Hospital of Logistics, University of Chinese Armed Police Forces, Chenglin Road, Tianjin 300162, PR China.
| | - Zhou Fei
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| |
Collapse
|
96
|
Marcelino TB, de Lemos Rodrigues PI, Miguel PM, Netto CA, Pereira Silva LO, Matté C. Effect of maternal exercise on biochemical parameters in rats submitted to neonatal hypoxia-ischemia. Brain Res 2015; 1622:91-101. [DOI: 10.1016/j.brainres.2015.06.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 01/25/2023]
|
97
|
Maternal Oxytocin Administration Before Birth Influences the Effects of Birth Anoxia on the Neonatal Rat Brain. Neurochem Res 2015; 40:1631-43. [DOI: 10.1007/s11064-015-1645-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 05/05/2015] [Accepted: 06/19/2015] [Indexed: 01/07/2023]
|
98
|
Guan DF, Ren PY, Hu W, Zhang YL. The mGluR5 positive allosteric modulator CDPPB inhibits SO₂-induced protein radical formation and mitochondrial dysfunction through activation of Akt in mouse hippocampal HT22 cells. Cell Mol Neurobiol 2015; 35:573-83. [PMID: 25547390 PMCID: PMC11488060 DOI: 10.1007/s10571-014-0153-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/23/2014] [Indexed: 11/30/2022]
Abstract
Sulfur dioxide (SO2) is a common gas pollutant that is detrimental to many organs. Previous studies have shown that SO2 exposure is involved in neurotoxicity and increased risk of many brain disorders; however, our understanding of the mechanisms underlying SO2-induced cytotoxicity on neuronal cells remains elusive. The group I metabotropic glutamate receptor 5 (mGluR5) can modulate addiction, pain, and neuronal cell death. In the present study, we showed that SO2 derivatives exposure induced protein radical formation, mitochondrial dysfunction, and apoptotic cell death in neuronal HT22 cells. Pretreatment with 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl) (CDPPB), a positive allosteric modulator of mGluR5, significantly attenuated SO2-induced neurotoxicity, which was fully prevented by the mGluR5 antagonist MPEP. CDPPB reduced the protein radical formation and inducible nitric oxide synthase (iNOS)-derived generation of nitric oxide, and inhibited mitochondrial dysfunction in both HT22 cells and isolated mitochondria after SO2 treatment. Moreover, CDPPB increased the activation of Akt in the presence and absence of SO2 treatment. Blocking Akt activation using the selective inhibitor LY294002 partially reversed the CDPPB-induced protection against SO2-induced neurotoxicity. This study provides mechanistic experimental support for oxidative stress and mitochondrial dysfunction after SO2 exposure in neuronal cells, and also introduces a novel therapeutic approach for SO2-induced neurotoxicity.
Collapse
Affiliation(s)
- Dong-Feng Guan
- Xi’an Jiaotong University Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061 China
- Linfen People’s Hospital, Linfen, Shanxi 041000 China
| | - Peng-Yu Ren
- Xi’an Jiaotong University Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi 710061 China
| | - Wei Hu
- Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068 China
| | - Yue-Lin Zhang
- Department of Neurosurgery, Shaanxi Provincial People’s Hospital, The Third Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710068 China
| |
Collapse
|
99
|
Hamel MS, Hughes BL, Rouse DJ. Whither oxygen for intrauterine resuscitation? Am J Obstet Gynecol 2015; 212:461-2. 461.e1. [PMID: 25659471 DOI: 10.1016/j.ajog.2015.01.057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 01/30/2015] [Indexed: 10/24/2022]
Abstract
Oxygen is frequently administered to women in labor in the hope of improving fetal status. However, there is a paucity of outcome data to support this practice. Although maternal oxygen administration may make physiological sense, unwarranted faith in maternal oxygen therapy may delay the indicated intervention or result in continued labor stimulation when neither is in the best interests of the fetus. A properly designed clinical trial would help answer whether maternal oxygen supplementation in labor should be considered an indicated intervention for nonreassuring fetal status.
Collapse
|
100
|
Baburamani AA, Miyakuni Y, Vontell R, Supramaniam VG, Svedin P, Rutherford M, Gressens P, Mallard C, Takeda S, Thornton C, Hagberg H. Does Caspase-6 Have a Role in Perinatal Brain Injury? Dev Neurosci 2015; 37:321-37. [PMID: 25823427 PMCID: PMC4876595 DOI: 10.1159/000375368] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 01/19/2015] [Indexed: 12/31/2022] Open
Abstract
Apoptotic mechanisms are centre stage for the development of injury in the immature brain, and caspases have been shown to play a pivotal role during brain development and in response to injury. The inhibition of caspases using broad-spectrum agents such as Q-VD-OPh is neuroprotective in the immature brain. Caspase-6, an effector caspase, has been widely researched in neurodevelopmental disorders and found to be important following adult stroke, but its function in the neonatal brain has yet to be detailed. Furthermore, caspases may be important in microglial activation; microglia are required for optimal brain development and following injury, and their close involvement during neuronal cell death suggests that apoptotic cues such as caspase activation may be important in microglial activation. Therefore, in this study we aimed to investigate the possible apoptotic and non-apoptotic functions caspase-6 may have in the immature brain in response to hypoxia-ischaemia. We examined whether caspases are involved in microglial activation. We assessed cleaved caspase-6 expression following hypoxia-ischaemia and conducted primary microglial cultures to assess whether the broad-spectrum inhibitor Q-VD-OPh or caspase-6 gene deletion affected lipopolysaccharide (LPS)-mediated microglial activation and phenotype. We observed cleaved caspase-6 expression to be low but present in the cell body and cell processes in both a human case of white matter injury and 72 h following hypoxia-ischaemia in the rat. Gene deletion of caspase-6 did not affect the outcome of brain injury following mild (50 min) or severe (60 min) hypoxia-ischaemia. Interestingly, we did note that cleaved caspase-6 was co-localised with microglia that were not of apoptotic morphology. We observed that mRNA of a number of caspases was modulated by low-dose LPS stimulation of primary microglia. Q-VD-OPh treatment and caspase-6 gene deletion did not affect microglial activation but modified slightly the M2b phenotype response by changing the time course of SOCS3 expression after LPS administration. Our results suggest that the impact of active caspase-6 in the developing brain is subtle, and we believe there are predominantly other caspases (caspase-2, −3, −8, −9) that are essential for the cell death processes in the immature brain.
Collapse
Affiliation(s)
- Ana A Baburamani
- Perinatal Center, Institute of Neuroscience and Physiology, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|