1
|
Tavassoli A, McDermott A. Hypoxia-inducing transcription factors: architects of tumorigenesis and targets for anticancer drug discovery. Transcription 2024:1-32. [PMID: 39470609 DOI: 10.1080/21541264.2024.2417475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024] Open
Abstract
Hypoxia-inducible factors (HIFs) play a pivotal role as master regulators of tumor survival and growth, controlling a wide array of cellular processes in response to hypoxic stress. Clinical data correlates upregulated HIF-1 and HIF-2 levels with an aggressive tumor phenotype and poor patient outcome. Despite extensive validation as a target in cancer, pharmaceutical targeting of HIFs, particularly the interaction between α and βsubunits that forms the active transcription factor, has proved challenging. Nonetheless, many indirect inhibitors of HIFs have been identified, targeting diverse parts of this pathway. Significant strides have also been made in the development of direct inhibitors of HIF-2, exemplified by the FDA approval of Belzutifan for the treatment of metastatic clear cell renal carcinoma. While efforts to target HIF-1 using various therapeutic modalities have shown promise, no clinical candidates have yet emerged. This review aims to provide insights into the intricate and extensive role played by HIFs in cancer, and the ongoing efforts to develop therapeutic agents against this target.
Collapse
Affiliation(s)
- Ali Tavassoli
- School of Chemistry, University of Southampton, Southampton, UK
| | | |
Collapse
|
2
|
Duck SA, Nazareth M, Fassinger A, Pinto C, Elmore G, Nugent M, St Pierre M, Vannucci SJ, Chavez-Valdez R. Blood glucose and β-hydroxybutyrate predict significant brain injury after hypoxia-ischemia in neonatal mice. Pediatr Res 2024:10.1038/s41390-024-03461-4. [PMID: 39181984 DOI: 10.1038/s41390-024-03461-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/13/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The Vannucci procedure is widely used to model cerebral hypoxic-ischemic (HI) injury in neonatal rodents. Identifying minimally invasive biomarkers linked to brain injury would improve stratification of pups to experimental treatments. We hypothesized that extreme blood glucose (BG) and β-hydroxybutyrate (bHB) levels immediately after HI will correlate with severity of brain injury in this model. METHODS C57BL6 mice of both sexes underwent the Vannucci procedure with BG and bHB measured immediately after hypoxia. GFAP and α-fodrin were measured to assess injury severity at 4h, P11, P18 and P40. Open field (OF), Y-maze (YM), and Object-location task (OLT) were tested at P40. RESULTS Clinical seizures-like stereotypies during hypoxia were associated with lower post-hypoxia BG in HI-injured mice. Low BG after HI was related to higher GFAP expression, higher α-fodrin breakdown, lower residual regional volume, and worse working memory. BG was superior to bHB in ROC analysis with BG threshold of <111 mg/dL providing 100% specificity with 72% sensitivity for hippocampal HI-injury. CONCLUSIONS Post-hypoxic BG is a minimally invasive screening tool to identify pups with significant HI brain injury in the Vannucci model modified for mice improving our ability to stratify pups to experimental treatments to assess effectiveness. IMPACT End hypoxic-ischemic blood glucose levels are a reliable and inexpensive biomarker to detect hypoxic-ischemic brain injury in mice. Screening with blood glucose levels post-hypoxia allows appropriate stratification of those mouse pups most likely to be injured to experimental treatments improving validity and translatability of the results. These findings provide biological plausibility to the clinical observation that extreme blood glucose levels relate to worse outcomes after hypoxia-ischemia.
Collapse
Affiliation(s)
- Sarah Ann Duck
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University - School of Medicine, Baltimore, MD, USA
| | - Michelle Nazareth
- Department of Neuroscience, Johns Hopkins University Krieger School of Arts and Sciences, Baltimore, MD, USA
| | - Abigail Fassinger
- Department of Neuroscience, Johns Hopkins University Krieger School of Arts and Sciences, Baltimore, MD, USA
| | - Charles Pinto
- Department of Human Biology, University of Toronto, Toronto, ON, Canada
| | - Genesis Elmore
- Department of Neuroscience, Johns Hopkins University Krieger School of Arts and Sciences, Baltimore, MD, USA
| | - Michael Nugent
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University - School of Medicine, Baltimore, MD, USA
| | - Mark St Pierre
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University - School of Medicine, Baltimore, MD, USA
| | - Susan J Vannucci
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Raul Chavez-Valdez
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University - School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Talati CP, Lee JW, Lu S, Ojeda NB, Prakash V, Dankhara N, Nielson TC, Sandifer SP, Bidwell GL, Pang Y, Fan LW, Bhatt AJ. Intranasal insulin attenuates hypoxia-ischemia-induced short-term sensorimotor behavioral disturbances, neuronal apoptosis, and brain damage in neonatal rats. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 6:100123. [PMID: 38235171 PMCID: PMC10793091 DOI: 10.1016/j.crneur.2023.100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/19/2024] Open
Abstract
There is a significant need for additional therapy to improve outcomes for newborns with acute Hypoxic-ischemic (HI) encephalopathy (HIE). New evidence suggests that insulin could be neuroprotective. This study aimed to investigate whether intranasal insulin attenuates HI-induced brain damage and neurobehavioral dysfunction in neonatal rats. Postnatal day 10 (P10), Sprague-Dawley rat pups were randomly divided into Sham + Vehicle, Sham + Insulin, HI + Vehicle, and HI + Insulin groups with equal male-to-female ratios. Pups either had HI by permanent ligation of the right common carotid artery followed by 90 min of hypoxia (8% O2) or sham surgery followed by room air exposure. Immediately after HI or Sham, pups were given fluorescence-tagged insulin (Alex-546-insulin)/vehicle, human insulin (25 μg), or vehicle in each nare under anesthesia. Shortly after administration, widespread Alex-546-insulin-binding cells were detected in the brain, primarily co-localized with neuronal nuclei-positive neurons on double-immunostaining. In the hippocampus, phospho-Akt was activated in a subset of Alex-546-insulin double-labeled cells, suggesting activation of the Akt/PI3K pathway in these neurons. Intranasal insulin (InInsulin) reduced HI-induced sensorimotor behavioral disturbances at P11. InInsulin prevented HI-induced increased Fluoro-Jade C+ degenerated neurons, cleaved caspase 3+ neurons, and volume loss in the ipsilateral brain at P11. There was no sex-specific response to HI or insulin. The findings confirm that intranasal insulin provides neuroprotection against HI brain injury in P10 rats associated with activation of intracellular cell survival signaling. If further pre-clinical research shows long-term benefits, intranasal insulin has the potential to be a promising non-invasive therapy to improve outcomes for newborns with HIE.
Collapse
Affiliation(s)
- Chirag P. Talati
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Jonathan W. Lee
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Silu Lu
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Norma B. Ojeda
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Varsha Prakash
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Nilesh Dankhara
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Tanner C. Nielson
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Sara P. Sandifer
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Gene L. Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Yi Pang
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Abhay J. Bhatt
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| |
Collapse
|
4
|
Yang F, Xie T, Hu Z, Chu Z, Lu H, Wu Q, Qin D, Sun S, Luo Z, Luo F. Exploration on anti-hypoxia properties of peptides: a review. Crit Rev Food Sci Nutr 2023:1-16. [PMID: 38116946 DOI: 10.1080/10408398.2023.2291824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Peptides are important components of human nutrition and health, and considered as safe, nontoxic, and easily absorbed potential drugs. Anti-hypoxia peptides are a kind of peptides that can prevent hypoxia or hypoxia damage. In this paper, the sources, preparations, and molecular mechanisms of anti-hypoxia peptides were systemically reviewed. The combination of bioinformatics, chemical synthesis, enzymatic hydrolysis, and microbial fermentation are recommended for efficient productions of anti-hypoxic peptides. The mechanisms of anti-hypoxic peptides include interference with glycolytic process and HIF-1α pathway, mitochondrial apoptosis, and inflammatory response. In addition, bioinformatics analysis, including virtual screening and molecular docking, provides an alternative or auxiliary method for exploring the potential anti-hypoxic activities and mechanisms of peptides. The potential challenges and prospects of anti-hypoxic peptides are also discussed. This paper can provide references for researchers in this field and promote further research and clinical applications of anti-hypoxic peptides in the future.
Collapse
Affiliation(s)
- Feiyan Yang
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Tiantian Xie
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Zuomin Hu
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Zhongxing Chu
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Han Lu
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Qi Wu
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Dandan Qin
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Shuguo Sun
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Zhang Luo
- College of Food Science, Tibet Agriculture & Animal Husbandry University, Nyingchi, Tibet, China
| | - Feijun Luo
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| |
Collapse
|
5
|
Lin W, Zhao XY, Cheng JW, Li LT, Jiang Q, Zhang YX, Han F. Signaling pathways in brain ischemia: Mechanisms and therapeutic implications. Pharmacol Ther 2023; 251:108541. [PMID: 37783348 DOI: 10.1016/j.pharmthera.2023.108541] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Ischemic stroke occurs when the arteries supplying blood to the brain are narrowed or blocked, inducing damage to brain tissue due to a lack of blood supply. One effective way to reduce brain damage and alleviate symptoms is to reopen blocked blood vessels in a timely manner and reduce neuronal damage. To achieve this, researchers have focused on identifying key cellular signaling pathways that can be targeted with drugs. These pathways include oxidative/nitrosative stress, excitatory amino acids and their receptors, inflammatory signaling molecules, metabolic pathways, ion channels, and other molecular events involved in stroke pathology. However, evidence suggests that solely focusing on protecting neurons may not yield satisfactory clinical results. Instead, researchers should consider the multifactorial and complex mechanisms underlying stroke pathology, including the interactions between different components of the neurovascular unit. Such an approach is more representative of the actual pathological process observed in clinical settings. This review summarizes recent research on the multiple molecular mechanisms and drug targets in ischemic stroke, as well as recent advances in novel therapeutic strategies. Finally, we discuss the challenges and future prospects of new strategies based on the biological characteristics of stroke.
Collapse
Affiliation(s)
- Wen Lin
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiang-Yu Zhao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jia-Wen Cheng
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Li-Tao Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Quan Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yi-Xuan Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China.
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China; Institute of Brain Science, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
6
|
Jung HY, Kwon HJ, Kim W, Hahn KR, Moon SM, Yoon YS, Kim DW, Hwang IK. The neuroprotective effects of phosphoglycerate mutase 5 are mediated by decreasing oxidative stress in HT22 hippocampal cells and gerbil hippocampus. Neurochem Int 2022; 157:105346. [PMID: 35513204 DOI: 10.1016/j.neuint.2022.105346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/14/2022] [Accepted: 04/25/2022] [Indexed: 10/18/2022]
Abstract
Phosphoglycerate mutase 5 (PGAM5), a glycolytic enzyme, plays an important role in cell death and regulation of mitochondrial dynamics. In this study, we investigated the effects of PGAM5 on oxidative stress in HT22 hippocampal cells and ischemic damage in the gerbil hippocampus to elucidate the role of PGAM5 in oxidative and ischemic stress. Constructs were designed with a PEP-1 expression vector to facilitate the intracellular delivery of PGAM5 proteins. We observed time- and concentration-dependent increases in the intracellular delivery of the PEP-1-PGAM5 protein, but not its control protein (PGAM5), in HT22 cells, and morphologically demonstrated the localization of the transduced protein, which was stably expressed in the cytoplasm after 12 h of PEP-1-PGAM5 treatment. PEP-1-PGAM5 treatment significantly ameliorated cell death, reactive oxygen species formation, DNA fragmentation, and the reduction of cell proliferation induced by H2O2 treatment in HT22 cells. In addition, PEP-1-PGAM5 was effectively delivered to the gerbil hippocampus 8 h after treatment, and ischemia-induced hyperlocomotion and neuronal death in the hippocampal CA1 region were significantly alleviated 1 and 4 days after ischemia, respectively. Ischemia-induced microglial activation was also mitigated by treatment with 1.0 mg/kg PEP-1-PGAM5. At 3 h after ischemia, PEP-1-PGAM5 treatment significantly ameliorated the increase in lipid peroxidation, as assessed by malondialdehyde and hydroperoxide levels, and decreased glutathione levels (increases in glutathione disulfide, the oxidized form of glutathione) in the hippocampus. Two days after ischemia, treatment with PEP-1-PGAM5 significantly alleviated the ischemia-induced reduction in glutathione peroxidase activity and further increased superoxide dismutase activity in the hippocampus. The neuroprotective effects of PEP-1-PGAM5 are partially mediated by a reduction in oxidative stress, such as the formation of reactive oxygen species, and increases in the activity of antioxidants such as glutathione peroxidase and superoxide dismutase.
Collapse
Affiliation(s)
- Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea; Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, South Korea
| | - Hyun Jung Kwon
- Department of Biomedical Sciences, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea; Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - Woosuk Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea; Department of Anatomy, College of Veterinary Medicine, Veterinary Science Research Institute, Konkuk University, Seoul, 05030, South Korea
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Seung Myung Moon
- Department of Neurosurgery, Dongtan Sacred Heart Hospital, College of Medicine, Hallym University, Hwaseong, 18450, South Korea; Research Institute for Complementary & Alternative Medicine, Hallym University, Chuncheon, 24253, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea.
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
7
|
Lorain P, Bower A, Gottardi E, Dommergues M, Foix L'Helias L, Guellec I, Kayem G. Risk factors for hypoxic-ischemic encephalopathy in cases of severe acidosis: A case-control study. Acta Obstet Gynecol Scand 2022; 101:471-478. [PMID: 35338480 DOI: 10.1111/aogs.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The aim of the study was to identify the obstetric risk factors for hypoxic-ischemic encephalopathy (HIE) in infants with asphyxia at birth. MATERIAL AND METHODS This multicenter case-control study covered the 5-year period from 2014 through 2018 and included newborns ≥36 weeks of gestation with an umbilical pH at birth ≤7.0. Cases were newborns who developed moderate or severe HIE; they were matched with controls with pH ≤7.0 at birth over the same period without moderate or severe HIE. The factors studied were maternal, gestational, intrapartum, delivery-related, and neonatal characteristics. A multivariable analysis was performed to study the maternal, obstetric, and neonatal factors independently associated with moderate or severe HIE. RESULTS Our review of the records identified 41 cases and 98 controls. Compared with controls, children with moderate or severe HIE had a lower 5-min Apgar score, lower umbilical artery pH, and higher cord lactate levels at birth and at 1 h of life. Obstetric factors associated with moderate or severe HIE were the occurrence of an acute event (adjusted odds ratio [aOR] 6.4; 95% confidence interval [CI] 1.8-22.5), maternal fever (aOR 3.5; 95% CI 1.0-11.9), and thick meconium during labor (aOR 2.9; 95% CI 1.0-8.6). CONCLUSIONS HIE is associated with a lower 5-min Apgar score and with the severity of acidosis at birth and at 1 h of life. In newborns with a pH <7.0 at birth, the occurrence of an acute obstetric event, maternal fever, and thick meconium are independent factors associated with moderate or severe HIE.
Collapse
Affiliation(s)
- Perrine Lorain
- Department of Gynecology and Obstetrics, Trousseau Hospital - Assistance Publique-Hopitaux de Paris, Sorbonne University, Paris, France
| | - Alexandra Bower
- Department of Neonatology, Trousseau Hospital - Assistance Publique-Hopitaux de Paris, Sorbonne University, Paris, France
| | - Elsa Gottardi
- Department of Gynecology and Obstetrics, Trousseau Hospital - Assistance Publique-Hopitaux de Paris, Sorbonne University, Paris, France
| | - Marc Dommergues
- Department of Gynecology and Obstetrics, Pitié Salpétrière Hospital - Assistance Publique-Hopitaux de Paris, Sorbonne University, Paris, France
| | - Laurence Foix L'Helias
- Department of Neonatology, Trousseau Hospital - Assistance Publique-Hopitaux de Paris, Sorbonne University, Paris, France.,Inserm UMR 1153, Obstetrical, Perinatal and Pediatric Epidemiology Research Team (EPOPé), Research Center for Epidemiology and BioStatistics Sorbonne Paris Cité (CRESS), DHU Risks in pregnancy, Paris Descartes University, Paris, France
| | - Isabelle Guellec
- Inserm UMR 1153, Obstetrical, Perinatal and Pediatric Epidemiology Research Team (EPOPé), Research Center for Epidemiology and BioStatistics Sorbonne Paris Cité (CRESS), DHU Risks in pregnancy, Paris Descartes University, Paris, France.,Neonatal Intensive Care Unit, Trousseau Hospital - Assistance Publique-Hopitaux de Paris, Sorbonne University, Paris, France
| | - Gilles Kayem
- Department of Gynecology and Obstetrics, Trousseau Hospital - Assistance Publique-Hopitaux de Paris, Sorbonne University, Paris, France.,Inserm UMR 1153, Obstetrical, Perinatal and Pediatric Epidemiology Research Team (EPOPé), Research Center for Epidemiology and BioStatistics Sorbonne Paris Cité (CRESS), DHU Risks in pregnancy, Paris Descartes University, Paris, France
| |
Collapse
|
8
|
Guo S, Wehbe A, Syed S, Wills M, Guan L, Lv S, Li F, Geng X, Ding Y. Cerebral Glucose Metabolism and Potential Effects on Endoplasmic Reticulum Stress in Stroke. Aging Dis 2022; 14:450-467. [PMID: 37008060 PMCID: PMC10017147 DOI: 10.14336/ad.2022.0905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
Ischemic stroke is an extremely common pathology with strikingly high morbidity and mortality rates. The endoplasmic reticulum (ER) is the primary organelle responsible for conducting protein synthesis and trafficking as well as preserving intracellular Ca2+ homeostasis. Mounting evidence shows that ER stress contributes to stroke pathophysiology. Moreover, insufficient circulation to the brain after stroke causes suppression of ATP production. Glucose metabolism disorder is an important pathological process after stroke. Here, we discuss the relationship between ER stress and stroke and treatment and intervention of ER stress after stroke. We also discuss the role of glucose metabolism, particularly glycolysis and gluconeogenesis, post-stroke. Based on recent studies, we speculate about the potential relationship and crosstalk between glucose metabolism and ER stress. In conclusion, we describe ER stress, glycolysis, and gluconeogenesis in the context of stroke and explore how the interplay between ER stress and glucose metabolism contributes to the pathophysiology of stroke.
Collapse
Affiliation(s)
- Sichao Guo
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Alexandra Wehbe
- Department of Neurosurgery, Wayne State University School of Medicine, USA
- Harvard T.H. Chan School of Public Health, USA
| | - Shabber Syed
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Melissa Wills
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Longfei Guan
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
- Department of Neurosurgery, Wayne State University School of Medicine, USA
| | - Shuyu Lv
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China
- Department of Neurosurgery, Wayne State University School of Medicine, USA
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China
- Correspondence should be addressed to: Dr. Xiaokun Geng, Beijing Luhe Hospital, Capital Medical University, Beijing, China. E-mail: ; Dr. Yuchuan Ding, Wayne State University School of Medicine, Detroit, MI 48201, USA. E-mail:
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, USA
- Correspondence should be addressed to: Dr. Xiaokun Geng, Beijing Luhe Hospital, Capital Medical University, Beijing, China. E-mail: ; Dr. Yuchuan Ding, Wayne State University School of Medicine, Detroit, MI 48201, USA. E-mail:
| |
Collapse
|
9
|
Guo S, Cosky E, Li F, Guan L, Ji Y, Wei W, Peng C, Geng X, Ding Y. An inhibitory and beneficial effect of chlorpromazine and promethazine (C + P) on hyperglycolysis through HIF-1α regulation in ischemic stroke. Brain Res 2021; 1763:147463. [PMID: 33811844 DOI: 10.1016/j.brainres.2021.147463] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND After ischemic stroke, the increased catabolism of glucose (hyperglycolysis) results in the production of reactive oxygen species (ROS) via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX). A depressive or hibernation-like effect of C + P on brain activity was reported to induce neuroprotection. The current study assesses the effect of C + P on hyperglycolysis and NOX activation. METHODS Adult male Sprague-Dawley rats were subjected to 2 h of middle cerebral artery occlusion (MCAO) followed by 6 or 24 h of reperfusion. At the onset of reperfusion, rats received C + P with or without temperature control, or phloretin [glucose transporter (GLUT)-1 inhibitor], or cytochalasin B (GLUT-3 inhibitor). We detected brain ROS, apoptotic cell death, and ATP levels along with HIF-1α expression. Cerebral hyperglycolysis was measured by glucose, protein expression of GLUT-1/3, and phosphofructokinase-1 (PFK-1), as well as lactate and lactate dehydrogenase (LDH) at 6 and 24 h of reperfusion. The enzymatic activity of NOX and protein expression of its subunits (gp91phox) were detected. Neural SHSY5Y cells were placed under 2 h of oxygen-glucose deprivation (OGD) followed by reoxygenation for 6 and 24 h with C + P treatment. Cell viability and protein levels of HIF-1α, GLUT-1/3, PFK-1, LDH, and gp91phox were measured. A HIF-1α overexpression vector was transfected into the cells, and then protein levels of HIF-1α, GLUT-1/3, PFK-1, and LDH were quantitated. In sham-operated rats and control cells, the protein levels of HIF-1α, GLUT-1/3, PFK-1, LDH, and gp91phox were measured at 6 and 24 h after C + P administration. RESULTS C + P reduced the protein elevations after stroke in HIF-1α, glycolytic enzymes, as well as in ROS, cell death, glucose and lactate, but raised ATP levels in the brain. In ischemic rats exposed to GLUT-1/3 inhibitors, ROS, cell death, glucose, and lactate were all decreased, as well as GLUT-1, GLUT-3, LDH, and PFK-1 protein levels. C + P decreased ischemia-induced NOX activation by reducing the enzymatic activity and protein expression of the NOX subunit gp91phox, as was observed in the presence of GLUT-1/3 inhibitors. These markers were significantly decreased following C + P administration with the induced hypothermia, while C + P administration with temperature control at 37 °C induced lesser protection after ischemia stroke. In the OGD/reoxygenation model, C + P treatment increased cell viability and diminished protein levels of HIF-1α, GLUT-1, GLUT-3, PFK-1, LDH, and gp91phox. However, in OGD with HIF-1α overexpression, C + P was unable to effectively reduce the upregulated GLUT-1, GLUT-3, and LDH. In normal conditions, C + P reduced HIF-1α and the levels of key glycolytic enzymes depending on its pharmacological effect. CONCLUSION C + P, partially depending on hypothermia, attenuates hyperglycolysis and NOX activation through HIF-1α regulation.
Collapse
Affiliation(s)
- Sichao Guo
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Eric Cosky
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Fengwu Li
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China
| | - Longfei Guan
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Yu Ji
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA; Department of General Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China
| | - Wenjing Wei
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA; China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Changya Peng
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Xiaokun Geng
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 48201, USA.
| |
Collapse
|
10
|
Mitochondrial Dysfunction and Permeability Transition in Neonatal Brain and Lung Injuries. Cells 2021; 10:cells10030569. [PMID: 33807810 PMCID: PMC7999701 DOI: 10.3390/cells10030569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 01/16/2023] Open
Abstract
This review discusses the potential mechanistic role of abnormally elevated mitochondrial proton leak and mitochondrial bioenergetic dysfunction in the pathogenesis of neonatal brain and lung injuries associated with premature birth. Providing supporting evidence, we hypothesized that mitochondrial dysfunction contributes to postnatal alveolar developmental arrest in bronchopulmonary dysplasia (BPD) and cerebral myelination failure in diffuse white matter injury (WMI). This review also analyzes data on mitochondrial dysfunction triggered by activation of mitochondrial permeability transition pore(s) (mPTP) during the evolution of perinatal hypoxic-ischemic encephalopathy. While the still cryptic molecular identity of mPTP continues to be a subject for extensive basic science research efforts, the translational significance of mitochondrial proton leak received less scientific attention, especially in diseases of the developing organs. This review is focused on the potential mechanistic relevance of mPTP and mitochondrial dysfunction to neonatal diseases driven by developmental failure of organ maturation or by acute ischemia-reperfusion insult during development.
Collapse
|
11
|
Dong X, Zhuang S, Huang Y, Yang X, Fu Y, Yu L, Zhao Y. Expression profile of circular RNAs in the peripheral blood of neonates with hypoxic‑ischemic encephalopathy. Mol Med Rep 2020; 22:87-96. [PMID: 32468058 PMCID: PMC7248490 DOI: 10.3892/mmr.2020.11091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 02/24/2020] [Indexed: 12/17/2022] Open
Abstract
Circular RNAs (circRNAs) are a class of non-coding RNAs that participate in various biological processes. However, the function of circRNAs in neonatal hypoxic‑ischemic encephalopathy (HIE) is not fully understood. In the present study, the differentially expressed circRNAs in the peripheral blood of neonates with HIE and control samples were characterized by a microarray assay. A total of 456 circRNAs were significantly differentially expressed in the peripheral blood of neonates with HIE, with 250 upregulated and 206 downregulated circRNAs in HIE compared with the control samples. Reverse transcription‑quantitative PCR was used to investigate specific circRNAs. Gene Ontology, and Kyoto Encyclopedia of Genes and Genomes pathway analyses were used to determine the function of the parent genes of the dysregulated circRNAs. In addition, microRNAs that may be associated with specific circRNAs were predicted using miRanda. Collectively, the present results indicated the potential importance of circRNAs in the peripheral blood of neonates with HIE.
Collapse
Affiliation(s)
- Xiaohua Dong
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang, Jiangsu 214500, P.R. China
| | - Sisi Zhuang
- Department of Pediatrics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210036, P.R. China
| | - Yun Huang
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang, Jiangsu 214500, P.R. China
| | - Xiaojing Yang
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang, Jiangsu 214500, P.R. China
| | - Yanrong Fu
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Lingling Yu
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang, Jiangsu 214500, P.R. China
| | - Yingmin Zhao
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang, Jiangsu 214500, P.R. China
| |
Collapse
|
12
|
Metabolic Phenotypes of Hypoxic-Ischemic Encephalopathy with Normal vs. Pathologic Magnetic Resonance Imaging Outcomes. Metabolites 2020; 10:metabo10030109. [PMID: 32183365 PMCID: PMC7143850 DOI: 10.3390/metabo10030109] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/12/2020] [Accepted: 03/12/2020] [Indexed: 01/09/2023] Open
Abstract
Hypoxic-Ischemic Encephalopathy (HIE) is one of the most relevant contributors to neurological disability in term infants. We hypothesized that clinical outcomes of newborns with (HIE) can be associated with changes at plasma metabolic level enabling the detection of brain injury. Plasma samples of a cohort of 55 asphyxiated infants who evolved to moderate/severe HIE were collected between birth and completion of therapeutic hypothermia (TH). Samples were analyzed employing a quantitative gas chromatography–mass spectrometry method for the determination of lactate and pyruvate and an untargeted liquid chromatography–time-of-flight mass spectrometry method for metabolic fingerprinting. Brain injury was assessed employing magnetic resonance imaging (MRI). A critical assessment of the usefulness of lactate, pyruvate, and pyruvate/lactate for outcome prediction was carried out. Besides, metabolic fingerprinting identified a dynamic perturbation of eleven metabolic pathways, including amino acid and purine metabolism, and the steroid hormone biosynthesis, in newborns with pathologic MRI outcomes. Although data suggest the usefulness of lactate and pyruvate monitoring during 72 h for discerning outcomes, only the steroid hormone biosynthesis pathway was significantly altered in early plasma samples (i.e., before the initiation of TH). This study highlights pathways that might potentially be targeted for biomarker discovery or adjuvant therapies to be combined with TH.
Collapse
|
13
|
Koyama M, Furukawa F, Koga Y, Funayama S, Furukawa S, Baba O, Lin CC, Hwang PP, Moriyama S, Okumura SI. Gluconeogenesis and glycogen metabolism during development of Pacific abalone, Haliotis discus hannai. Am J Physiol Regul Integr Comp Physiol 2020; 318:R619-R633. [PMID: 31994899 DOI: 10.1152/ajpregu.00211.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In lecithotrophic larvae, egg yolk nutrients are essential for development. Although yolk proteins and lipids are the major nutrient sources for most animal embryos and larvae, the contribution of carbohydrates to development has been less understood. In this study, we assessed glucose and glycogen metabolism in developing Pacific abalone, a marine gastropod mollusc caught and cultured in east Asia. We found that glucose and glycogen content gradually elevated in developing abalone larvae, and coincident expression increases of gluconeogenic genes and glycogen synthase suggested abalone larvae had activated gluconeogenesis and glycogenesis during this stage. At settling, however, glycogen sharply decreased, with concomitant increases in glucose content and expression of Pyg and G6pc, suggesting the settling larvae had enhanced glycogen conversion to glucose. A liquid chromatography-mass spectrometry (LC/MS)-based metabolomic approach that detected intermediates of these pathways further supported active metabolism of glycogen. Immunofluorescence staining and in situ hybridization suggested the digestive gland has an important role as glycogen storage tissue during settlement, while many other tissues also showed a capacity to metabolize glycogen. Finally, inhibition of glycolysis affected survival of the settling veliger larvae, revealing that glucose is, indeed, an important nutrient source in settling larvae. Our results suggest glucose and glycogen are required for proper energy balance in developing abalone and especially impact survival during settling.
Collapse
Affiliation(s)
- Mugen Koyama
- School of Marine Biosciences, Kitasato University, Kanagawa, Japan
| | - Fumiya Furukawa
- School of Marine Biosciences, Kitasato University, Kanagawa, Japan
| | - Yuka Koga
- School of Marine Biosciences, Kitasato University, Kanagawa, Japan
| | - Shohei Funayama
- School of Marine Biosciences, Kitasato University, Kanagawa, Japan
| | | | - Otto Baba
- Oral and Maxillofacial Anatomy, Tokushima University Graduate School, Tokushima, Japan
| | - Ching-Chun Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | | | - Sei-Ichi Okumura
- School of Marine Biosciences, Kitasato University, Kanagawa, Japan
| |
Collapse
|
14
|
Mikrogeorgiou A, Xu D, Ferriero DM, Vannucci SJ. Assessing Cerebral Metabolism in the Immature Rodent: From Extracts to Real-Time Assessments. Dev Neurosci 2019; 40:463-474. [PMID: 30991389 DOI: 10.1159/000496921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/09/2019] [Indexed: 12/27/2022] Open
Abstract
Brain development is an energy-expensive process. Although glucose is irreplaceable, the developing brain utilizes a variety of substrates such as lactate and the ketone bodies, β-hydroxybutyrate and acetoacetate, to produce energy and synthesize the structural components necessary for cerebral maturation. When oxygen and nutrient supplies to the brain are restricted, as in neonatal hypoxia-ischemia (HI), cerebral energy metabolism undergoes alterations in substrate use to preserve the production of adenosine triphosphate. These changes have been studied by in situ biochemical methods that yielded valuable quantitative information about high-energy and glycolytic metabolites and established a temporal profile of the cerebral metabolic response to hypoxia and HI. However, these analyses relied on terminal experiments and averaging values from several animals at each time point as well as challenging requirements for accurate tissue processing.More recent methodologies have focused on in vivo longitudinal analyses in individual animals. The emerging field of metabolomics provides a new investigative tool for studying cerebral metabolism. Magnetic resonance spectroscopy (MRS) has enabled the acquisition of a snapshot of the metabolic status of the brain as quantifiable spectra of various intracellular metabolites. Proton (1H) MRS has been used extensively as an experimental and diagnostic tool of HI in the pursuit of markers of long-term neurodevelopmental outcomes. Still, the interpretation of the metabolite spectra acquired with 1H MRS has proven challenging, due to discrepancies among studies, regarding calculations and timing of measurements. As a result, the predictive utility of such studies is not clear. 13C MRS is methodologically more challenging, but it provides a unique window on living tissue metabolism via measurements of the incorporation of 13C label from substrates into brain metabolites and the localized determination of various metabolic fluxes. The newly developed hyperpolarized 13C MRS is an exciting method for assessing cerebral metabolism in vivo, that bears the advantages of conventional 13C MRS but with a huge gain in signal intensity and much shorter acquisition times. The first part of this review article provides a brief description of the findings of biochemical and imaging methods over the years as well as a discussion of their associated strengths and pitfalls. The second part summarizes the current knowledge on cerebral metabolism during development and HI brain injury.
Collapse
Affiliation(s)
- Alkisti Mikrogeorgiou
- Department of Pediatrics, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Duan Xu
- Department of Radiology and Biomedical Imaging, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Donna M Ferriero
- Department of Pediatrics, University of California San Francisco School of Medicine, San Francisco, California, USA.,Department of Neurology, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Susan J Vannucci
- Department of Pediatrics and Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA,
| |
Collapse
|
15
|
Narang R, Carter K, Muncie C, Pang Y, Fan LWW, Feng Y, Ojeda NB, Bhatt AJ. Intrauterine growth restriction and neonatal hypoxic ischemic brain injury causes sex-specific long-term neurobehavioral abnormalities in rats. J Neurosci Res 2019; 97:661-672. [PMID: 30843634 DOI: 10.1002/jnr.24389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 12/28/2018] [Accepted: 01/02/2019] [Indexed: 12/22/2022]
Abstract
There is a lack of knowledge of factors preventing an adequate response to moderate hypothermia after hypoxic ischemic (HI) brain injury. We hypothesized that growth restriction from reduced intrauterine perfusion would predispose neonatal rats to have a worse outcome with HI brain injury. IUGR was induced by placental insufficiency in dams at 14 days of gestation. HI was induced at postnatal day (P) 10 by permanent right carotid artery ligation followed by 90 min of hypoxia (8% oxygen). Tests for early brain injury and neurobehavioral outcomes were subsequently done. All statistical analysis was done using Two-way ANOVA; post hoc Holm-Sidak test. HI in control and IUGR groups decreased the success rate of the contralateral vibrissa-elicited forelimb test, increased response latency in movement initiation test and increased the time to finish elevated beam walk test at P40 and P60. IUGR augmented HI-induced abnormality in vibrissa-elicited forelimb test at P40 but showed higher success rate when compared to HI only group at P60. IUGR's negative effect on HI-induced changes on the elevated beam walk test was sex-specific and exaggerated in P60 males. Increased TUNEL positive cells in the cortex were noted at 72 h after in HI in control but not in IUGR groups. In conclusion, the consequences of IUGR on subsequent neonatal HI varied based on age, sex and outcomes examined, and overall, male sex and IUGR had worse effects on the long-term neurobehavioral outcomes following HI.
Collapse
Affiliation(s)
- Radhika Narang
- Division of Newborn Medicine, Valley Children's Healthcare, Madera, California
| | - Kathleen Carter
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Colin Muncie
- Department of Surgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yi Pang
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lir-Wan W Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yangzheng Feng
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Norma B Ojeda
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Abhay J Bhatt
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
16
|
Herrera-Marschitz M, Perez-Lobos R, Lespay-Rebolledo C, Tapia-Bustos A, Casanova-Ortiz E, Morales P, Valdes JL, Bustamante D, Cassels BK. Targeting Sentinel Proteins and Extrasynaptic Glutamate Receptors: a Therapeutic Strategy for Preventing the Effects Elicited by Perinatal Asphyxia? Neurotox Res 2018; 33:461-473. [PMID: 28844085 PMCID: PMC5766721 DOI: 10.1007/s12640-017-9795-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/29/2022]
Abstract
Perinatal asphyxia (PA) is a relevant cause of death at the time of labour, and when survival is stabilised, associated with short- and long-term developmental disabilities, requiring inordinate care by health systems and families. Its prevalence is high (1 to 10/1000 live births) worldwide. At present, there are few therapeutic options, apart from hypothermia, that regrettably provides only limited protection if applied shortly after the insult.PA implies a primary and a secondary insult. The primary insult relates to the lack of oxygen, and the secondary one to the oxidative stress triggered by re-oxygenation, formation of reactive oxygen (ROS) and reactive nitrogen (RNS) species, and overactivation of glutamate receptors and mitochondrial deficiencies. PA induces overactivation of a number of sentinel proteins, including hypoxia-induced factor-1α (HIF-1α) and the genome-protecting poly(ADP-ribose) polymerase-1 (PARP-1). Upon activation, PARP-1 consumes high amounts of ATP at a time when this metabolite is scarce, worsening in turn the energy crisis elicited by asphyxia. The energy crisis also impairs ATP-dependent transport, including glutamate re-uptake by astroglia. Nicotinamide, a PARP-1 inhibitor, protects against the metabolic cascade elicited by the primary stage, avoiding NAD+ exhaustion and the energetic crisis. Upon re-oxygenation, however, oxidative stress leads to nuclear translocation of the NF-κB subunit p65, overexpression of the pro-inflammatory cytokines IL-1β and TNF-α, and glutamate-excitotoxicity, due to impairment of glial-glutamate transport, extracellular glutamate overflow, and overactivation of NMDA receptors, mainly of the extrasynaptic type. This leads to calcium influx, mitochondrial impairment, and inactivation of antioxidant enzymes, increasing further the activity of pro-oxidant enzymes, thereby making the surviving neonate vulnerable to recurrent metabolic insults whenever oxidative stress is involved. Here, we discuss evidence showing that (i) inhibition of PARP-1 overactivation by nicotinamide and (ii) inhibition of extrasynaptic NMDA receptor overactivation by memantine can prevent the short- and long-term consequences of PA. These hypotheses have been evaluated in a rat preclinical model of PA, aiming to identify the metabolic cascades responsible for the long-term consequences induced by the insult, also assessing postnatal vulnerability to recurrent oxidative insults. Thus, we present and discuss evidence demonstrating that PA induces long-term changes in metabolic pathways related to energy and oxidative stress, priming vulnerability of cells with both the neuronal and the glial phenotype. The effects induced by PA are region dependent, the substantia nigra being particularly prone to cell death. The issue of short- and long-term consequences of PA provides a framework for addressing a fundamental issue referred to plasticity of the CNS, since the perinatal insult triggers a domino-like sequence of events making the developing individual vulnerable to recurrent adverse conditions, decreasing his/her coping repertoire because of a relevant insult occurring at birth.
Collapse
Affiliation(s)
- Mario Herrera-Marschitz
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Ronald Perez-Lobos
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
- Escuela de Tecnologia Medica, Facultad de Medicina, Universidad Andres Bello, PO Box 8370146, Santiago, Chile
| | - Carolyne Lespay-Rebolledo
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Andrea Tapia-Bustos
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Emmanuel Casanova-Ortiz
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Paola Morales
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
- Faculty of Sciences, University of Chile, Santiago, Chile
| | | | - Diego Bustamante
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Bruce K. Cassels
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
17
|
Sánchez-Illana Á, Núñez-Ramiro A, Cernada M, Parra-Llorca A, Valverde E, Blanco D, Moral-Pumarega MT, Cabañas F, Boix H, Pavon A, Chaffanel M, Benavente-Fernández I, Tofe I, Loureiro B, Fernández-Lorenzo JR, Fernández-Colomer B, García-Robles A, Kuligowski J, Vento M. Evolution of Energy Related Metabolites in Plasma from Newborns with Hypoxic-Ischemic Encephalopathy during Hypothermia Treatment. Sci Rep 2017; 7:17039. [PMID: 29213095 PMCID: PMC5719006 DOI: 10.1038/s41598-017-17202-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/22/2017] [Indexed: 12/17/2022] Open
Abstract
Therapeutic hypothermia (TH) initiated within 6 h from birth is the most effective therapeutic approach for moderate to severe hypoxic-ischemic encephalopathy (HIE). However, underlying mechanisms and effects on the human metabolism are not yet fully understood. This work aims at studying the evolution of several energy related key metabolites in newborns with HIE undergoing TH employing gas chromatography - mass spectrometry. The method was validated following stringent FDA requirements and applied to 194 samples from a subgroup of newborns with HIE (N = 61) enrolled in a multicenter clinical trial (HYPOTOP) for the determination of lactate, pyruvate, ketone bodies and several Krebs cycle metabolites at different sampling time points. The analysis of plasma samples from newborns with HIE revealed a decrease of lactate, pyruvate and β-hydroxybutyrate concentrations, whereas rising malate concentrations were observed. In healthy control newborns (N = 19) significantly lower levels of pyruvate and lactate were found in comparison to age-matched newborns with HIE undergoing TH, whereas acetoacetate and β-hydroxybutyrate levels were clearly increased. Access to a validated analytical method and a controlled cohort of newborns with HIE undergoing hypothermia treatment for the first time allowed the in-depth study of the evolution of key metabolites of metabolic junctions in this special population.
Collapse
Affiliation(s)
- Ángel Sánchez-Illana
- Neonatal Research Unit, Health Research Institute La Fe, Valencia, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Antonio Núñez-Ramiro
- Neonatal Research Unit, Health Research Institute La Fe, Valencia, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - María Cernada
- Neonatal Research Unit, Health Research Institute La Fe, Valencia, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Anna Parra-Llorca
- Neonatal Research Unit, Health Research Institute La Fe, Valencia, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Eva Valverde
- Hospital Universitario La Paz, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Dorotea Blanco
- Hospital Universitario Gregorio Marañón, C/O'Donnell 48-50, 28009, Madrid, Spain
| | | | - Fernando Cabañas
- Hospital Universitario Quirónsalud Madrid, Calle Diego de Velazquez s/n, 28223, Madrid, Spain
| | - Hector Boix
- Hospital Universitario Vall d'Hebron, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Antonio Pavon
- Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot, s/n, 41013, Sevilla, Spain
| | - Mercedes Chaffanel
- Hospital Materno Infantil Carlos Haya, Avda. Arroyo de los Angeles s/n, 29011, Málaga, Spain
| | | | - Inés Tofe
- Hospital Universitario Reina Sofía, Avda. Menendez Pidal s/n, 14004, Córdoba, Spain
| | - Begoña Loureiro
- Hospital Universitario Cruces, Plaza Cruces s/n, 48903, Barakaldo, Vizcaya, Spain
| | | | | | - Ana García-Robles
- Neonatal Research Unit, Health Research Institute La Fe, Valencia, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Julia Kuligowski
- Neonatal Research Unit, Health Research Institute La Fe, Valencia, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain.
| | - Máximo Vento
- Neonatal Research Unit, Health Research Institute La Fe, Valencia, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain.
- Division of Neonatology, University & Polytechnic Hospital La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain.
| |
Collapse
|
18
|
Movsas TZ, Weiner RL, Greenberg MB, Holtzman DM, Galindo R. Pretreatment with Human Chorionic Gonadotropin Protects the Neonatal Brain against the Effects of Hypoxic-Ischemic Injury. Front Pediatr 2017; 5:232. [PMID: 29164084 PMCID: PMC5675846 DOI: 10.3389/fped.2017.00232] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 10/16/2017] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Though the human fetus is exposed to placentally derived human chorionic gonadotropin (hCG) throughout gestation, the role of hCG on the fetal brain is unknown. Review of the available literature appears to indicate that groups of women with higher mean levels of hCG during pregnancy tend to have offspring with lower cerebral palsy (CP) risk. Given that newborn cerebral injury often precedes the development of CP, we aimed to determine whether hCG may protect against the neurodegenerative effects of neonatal brain injury. METHODS We utilized the Rice-Vannucci model of neonatal cerebral hypoxia-ischemia (HI) in postnatal day 7 mice to examine whether intraperitoneal administration of hCG 15-18 h prior, 1 h after or immediately following HI decrease brain tissue loss 7 days after injury. We next studied whether hCG has pro-survival and trophic properties in neurons by exposing immature cortical and hippocampal neurons to hCG in vitro and examining neurite sprouting and neuronal survival prior and after glutamate receptor-mediated excitotoxic injury. RESULTS We found that intraperitoneal injection of hCG 15 h prior to HI, but not at or 1 h after HI induction, resulted in a significant decrease in hippocampal and striatal tissue loss 7 days following brain injury. Furthermore, hCG reduced N-methyl-d-aspartate (NMDA)-mediated neuronal excitotoxicity in vitro when neurons were continuously exposed to this hormone for 10 days or when given at the time and following neuronal injury. In addition, continuous in vitro administration of hCG for 6-9 days increased neurite sprouting and basal neuronal survival as assessed by at least a 1-fold increase in MAP2 immunoreactivity and a 2.5-fold increase in NeuN + immunoreactivity. CONCLUSION Our findings suggest that hCG can decrease HI-associated immature neural degeneration. The mechanism of action for this neuroprotective effect may partly involve inhibition of NMDA-dependent excitotoxic injury. This study supports the hypothesis that hCG during pregnancy has the potential for protecting the developing brain against HI, an important CP risk factor.
Collapse
Affiliation(s)
- Tammy Z. Movsas
- Zietchick Research Institute, Plymouth, MI, United States
- Department of Pediatrics and Human Development, Michigan State University College of Human Medicine, East Lansing, MI, United States
| | - Rebecca L. Weiner
- Department of Neurology, Hope Center for Neurological Disorders, Washington University, St. Louis, MO, United States
| | - M. Banks Greenberg
- Department of Neurology, Hope Center for Neurological Disorders, Washington University, St. Louis, MO, United States
| | - David M. Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Washington University, St. Louis, MO, United States
| | - Rafael Galindo
- Department of Neurology, Hope Center for Neurological Disorders, Washington University, St. Louis, MO, United States
| |
Collapse
|
19
|
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: 213] [Impact Index Per Article: 30.4] [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
|
20
|
Glucose and Intermediary Metabolism and Astrocyte–Neuron Interactions Following Neonatal Hypoxia–Ischemia in Rat. Neurochem Res 2016; 42:115-132. [DOI: 10.1007/s11064-016-2149-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 11/27/2022]
|
21
|
Rumajogee P, Bregman T, Miller SP, Yager JY, Fehlings MG. Rodent Hypoxia-Ischemia Models for Cerebral Palsy Research: A Systematic Review. Front Neurol 2016; 7:57. [PMID: 27199883 PMCID: PMC4843764 DOI: 10.3389/fneur.2016.00057] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 04/03/2016] [Indexed: 12/28/2022] Open
Abstract
Cerebral palsy (CP) is a complex multifactorial disorder, affecting approximately 2.5-3/1000 live term births, and up to 22/1000 prematurely born babies. CP results from injury to the developing brain incurred before, during, or after birth. The most common form of this condition, spastic CP, is primarily associated with injury to the cerebral cortex and subcortical white matter as well as the deep gray matter. The major etiological factors of spastic CP are hypoxia/ischemia (HI), occurring during the last third of pregnancy and around birth age. In addition, inflammation has been found to be an important factor contributing to brain injury, especially in term infants. Other factors, including genetics, are gaining importance. The classic Rice-Vannucci HI model (in which 7-day-old rat pups undergo unilateral ligation of the common carotid artery followed by exposure to 8% oxygen hypoxic air) is a model of neonatal stroke that has greatly contributed to CP research. In this model, brain damage resembles that observed in severe CP cases. This model, and its numerous adaptations, allows one to finely tune the injury parameters to mimic, and therefore study, many of the pathophysiological processes and conditions observed in human patients. Investigators can recreate the HI and inflammation, which cause brain damage and subsequent motor and cognitive deficits. This model further enables the examination of potential approaches to achieve neural repair and regeneration. In the present review, we compare and discuss the advantages, limitations, and the translational value for CP research of HI models of perinatal brain injury.
Collapse
Affiliation(s)
- Prakasham Rumajogee
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network , Toronto, ON , Canada
| | - Tatiana Bregman
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network , Toronto, ON , Canada
| | - Steven P Miller
- Department of Pediatrics, Hospital for Sick Children , Toronto, ON , Canada
| | - Jerome Y Yager
- Division of Pediatric Neurosciences, Stollery Children's Hospital, University of Alberta , Edmonton, AB , Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada; Division of Neurosurgery, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
22
|
Al Balushi A, Guilbault MP, Wintermark P. Secondary Increase of Lactate Levels in Asphyxiated Newborns during Hypothermia Treatment: Reflect of Suboptimal Hemodynamics (A Case Series and Review of the Literature). AJP Rep 2016; 6:e48-58. [PMID: 26929870 PMCID: PMC4737629 DOI: 10.1055/s-0035-1565921] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 09/02/2015] [Indexed: 12/02/2022] Open
Abstract
Objective To evaluate whether a secondary increase of serum lactate levels in asphyxiated newborns during hypothermia treatment may reflect suboptimal dynamics. Methods-Retrospective case series and review of the literature. We present the clinical course of four asphyxiated newborns treated with hypothermia who presented with hypotension requiring inotropic support, and who displayed a secondary increase of serum lactate levels during hypothermia treatment. Serial serum lactate levels are correlated with blood pressure and inotropic support within the first 96 hours of life. Results Lactate levels initially decreased in the four patients. However, each of them started to present lower blood pressure, and lactate levels started to increase again. Inotropic support was started to raise blood pressure. The introduction of an epinephrine drip consistently worsened the increase of lactate levels in these newborns, whereas dopamine and dobutamine enabled the clearance of lactate in addition to raising the blood pressure. Rewarming was associated with hemodynamics perturbations (a decrease of blood pressure and/or an increase of lactate levels) in the three newborns who survived. Conclusions Lactate levels during the first 4 days of life should be followed as a potential marker for suboptimal hemodynamic status in term asphyxiated newborns treated with hypothermia, for whom the maintenance of homeostasis during hypothermia treatment is of utmost importance to alleviate brain injury.
Collapse
Affiliation(s)
- Asim Al Balushi
- Department of Pediatrics, Montreal Children's Hospital, McGill University, Montreal, Canada
| | - Marie-Pier Guilbault
- Department of Pediatrics, Montreal Children's Hospital, McGill University, Montreal, Canada
| | - Pia Wintermark
- Division of Newborn Medicine, Department of Pediatrics, Montreal Children's Hospital, McGill University, Montreal, Canada
| |
Collapse
|
23
|
Cai L, Stevenson J, Peng C, Xin R, Rastogi R, Liu K, Geng X, Gao Z, Ji X, Rafols JA, Ji Z, Ding Y. Adjuvant therapies using normobaric oxygen with hypothermia or ethanol for reducing hyperglycolysis in thromboembolic cerebral ischemia. Neuroscience 2016; 318:45-57. [PMID: 26794589 DOI: 10.1016/j.neuroscience.2016.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/25/2015] [Accepted: 01/06/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE Normobaric oxygen (NBO), ethanol (EtOH), and therapeutic hypothermia (TH) delivered alone or in combination have neuroprotective properties after acute stroke. We used an autologous thromboembolic rat stroke model to assess the additive effects of these treatments for reducing the deleterious effects of hyperglycolysis post-stroke in which reperfusion is induced with recombinant tissue plasminogen activator (rt-PA). METHODS Sprague-Dawley rats were subjected to middle cerebral artery (MCA) occlusion with an autologous embolus. One hour after occlusion, rt-PA was administered alone or with NBO (60%), EtOH (1.0 g/kg), TH (33 °C), either singly or in combination. Infarct volume and neurological deficit were assessed at 24h after rt-PA-induced reperfusion with or without other treatments. The extent of hyperglycolysis, as determined by cerebral glucose and lactate levels was evaluated at 3 and 24h after rt-PA administration. At the same time points, expressions of glucose transporter 1 (Glut1), glucose transporter 3 (Glut3), phosphofructokinase1 (PFK-1), and lactate dehydrogenase were (LDH) measured by Western blotting. RESULTS Following rt-PA in rats with thromboembolic stroke, NBO combined with TH or EtOH most effectively decreased infarct volume and neurological deficit. As compared to rt-PA alone, EtOH or TH but not NBO monotherapies significantly reduced post-stroke hyperglycolysis. The increased utilization of glucose and production of lactate post-stroke was prevented most effectively when NBO was combined with either EtOH or TH after reperfusion with rt-PA, as shown by the significantly decreased Glut1, Glut3, PFK-1, and LDH levels. CONCLUSIONS In a rat thromboembolic stroke model, both EtOH and TH used individually offer neuroprotection after the administration of rt-PA. While NBO monotherapy does not appear to be effective, it significantly potentiates the efficacy of EtOH and TH. The similar neuroprotection and underlying mechanisms pertaining to the attenuation of hyperglycolysis provided by EtOH or TH in combination with NBO suggest a possibility of substituting EtOH for TH. Thus a combination of NBO and EtOH, which are widely available and easily used, could become a novel and effective neuroprotective strategy in the clinical setting.
Collapse
Affiliation(s)
- L Cai
- China-America Institute of Neuroscience, Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - J Stevenson
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - C Peng
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - R Xin
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA; Department of Radiology, Luhe Hospital, Capital Medical University, Beijing, China
| | - R Rastogi
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - K Liu
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - X Geng
- China-America Institute of Neuroscience, Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Z Gao
- Cerebral Vascular Diseases Research Institute, Capital Medical University, Beijing, China
| | - X Ji
- Cerebral Vascular Diseases Research Institute, Capital Medical University, Beijing, China
| | - J A Rafols
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Z Ji
- China-America Institute of Neuroscience, Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China.
| | - Y Ding
- China-America Institute of Neuroscience, Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
24
|
Ahmad AS, Satriotomo I, Fazal J, Nadeau SE, Doré S. Considerations for the Optimization of Induced White Matter Injury Preclinical Models. Front Neurol 2015; 6:172. [PMID: 26322013 PMCID: PMC4532913 DOI: 10.3389/fneur.2015.00172] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/20/2015] [Indexed: 11/13/2022] Open
Abstract
White matter (WM) injury in relation to acute neurologic conditions, especially stroke, has remained obscure until recently. Current advances in imaging technologies in the field of stroke have confirmed that WM injury plays an important role in the prognosis of stroke and suggest that WM protection is essential for functional recovery and post-stroke rehabilitation. However, due to the lack of a reproducible animal model of WM injury, the pathophysiology and mechanisms of this injury are not well studied. Moreover, producing selective WM injury in animals, especially in rodents, has proven to be challenging. Problems associated with inducing selective WM ischemic injury in the rodent derive from differences in the architecture of the brain, most particularly, the ratio of WM to gray matter in rodents compared to humans, the agents used to induce the injury, and the location of the injury. Aging, gender differences, and comorbidities further add to this complexity. This review provides a brief account of the techniques commonly used to induce general WM injury in animal models (stroke and non-stroke related) and highlights relevance, optimization issues, and translational potentials associated with this particular form of injury.
Collapse
Affiliation(s)
- Abdullah Shafique Ahmad
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA
| | - Irawan Satriotomo
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA
| | - Jawad Fazal
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA
| | - Stephen E Nadeau
- Research Service, Brain Rehabilitation Research Center, Malcom Randall Veterans Affairs Medical Center , Gainesville, FL , USA ; Department of Neurology, University of Florida , Gainesville, FL , USA
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida , Gainesville, FL , USA ; Research Service, Brain Rehabilitation Research Center, Malcom Randall Veterans Affairs Medical Center , Gainesville, FL , USA ; Department of Neurology, University of Florida , Gainesville, FL , USA ; Department of Neuroscience, University of Florida , Gainesville, FL , USA ; Department of Neurology, University of Florida , Gainesville, FL , USA ; Department of Pharmaceutics, University of Florida , Gainesville, FL , USA ; Department of Psychology, University of Florida , Gainesville, FL , USA ; Department of Psychiatry, University of Florida , Gainesville, FL , USA
| |
Collapse
|
25
|
Abe T, Suzuki M, Sasabe J, Takahashi S, Unekawa M, Mashima K, Iizumi T, Hamase K, Konno R, Aiso S, Suzuki N. Cellular origin and regulation of D- and L-serine in in vitro and in vivo models of cerebral ischemia. J Cereb Blood Flow Metab 2014; 34:1928-35. [PMID: 25294127 PMCID: PMC4269747 DOI: 10.1038/jcbfm.2014.164] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 08/19/2014] [Accepted: 08/31/2014] [Indexed: 01/24/2023]
Abstract
D-Serine is known to be essential for the activation of the N-methyl-D-aspartate (NMDA) receptor in the excitation of glutamatergic neurons, which have critical roles in long-term potentiation and memory formation. D-Serine is also thought to be involved in NMDA receptor-mediated neurotoxicity. The deletion of serine racemase (SRR), which synthesizes D-serine from L-serine, was recently reported to improve ischemic damage in mouse middle cerebral artery occlusion model. However, the cell type in which this phenomenon originates and the regulatory mechanism for D-/L-serine remain elusive. The D-/L-serine content in ischemic brain increased until 20 hours after recanalization and then leveled off gradually. The results of in vitro experiments using cultured cells suggested that D-serine is derived from neurons, while L-serine seems to be released from astroglia. Immunohistochemistry studies of brain tissue after cerebral ischemia showed that SRR is expressed in neurons, and 3-phosphoglycerate dehydrogenase (3-PGDH), which synthesizes L-serine from 3-phosphoglycerate, is located in astrocytes, supporting the results of the in vitro experiments. A western blot analysis showed that neither SRR nor 3-PGDH was upregulated after cerebral ischemia. Therefore, the increase in D-/L-serine was not related to an increase in SRR or 3-PGDH, but to an increase in the substrates of SRR and 3-PGDH.
Collapse
Affiliation(s)
- Takato Abe
- Department of Neurology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Masataka Suzuki
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Jumpei Sasabe
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shinichi Takahashi
- Department of Neurology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Miyuki Unekawa
- Department of Neurology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kyoko Mashima
- Department of Neurology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Takuya Iizumi
- Department of Neurology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kenji Hamase
- Graduate School of Pharmaceutical Sciences Kyushu University, Higashi-ku, Fukuoka, Japan
| | - Ryuichi Konno
- Department of Pharmacological Sciences, International University of Health and Welfare, Ohtawara, Tochigi, Japan
| | - Sadakazu Aiso
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Norihiro Suzuki
- Department of Neurology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
26
|
13C NMR metabolomic evaluation of immediate and delayed mild hypothermia in cerebrocortical slices after oxygen-glucose deprivation. Anesthesiology 2013; 119:1120-36. [PMID: 23748856 DOI: 10.1097/aln.0b013e31829c2d90] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Mild brain hypothermia (32°-34°C) after human neonatal asphyxia improves neurodevelopmental outcomes. Astrocytes but not neurons have pyruvate carboxylase and an acetate uptake transporter. C nuclear magnetic resonance spectroscopy of rodent brain extracts after administering [1-C]glucose and [1,2-C]acetate can distinguish metabolic differences between glia and neurons, and tricarboxylic acid cycle entry via pyruvate dehydrogenase and pyruvate carboxylase. METHODS Neonatal rat cerebrocortical slices receiving a C-acetate/glucose mixture underwent a 45-min asphyxia simulation via oxygen-glucose-deprivation followed by 6 h of recovery. Protocols in three groups of N=3 experiments were identical except for temperature management. The three temperature groups were: normothermia (37°C), hypothermia (32°C for 3.75 h beginning at oxygen--glucose deprivation start), and delayed hypothermia (32°C for 3.75 h, beginning 15 min after oxygen-glucose deprivation start). Multivariate analysis of nuclear magnetic resonance metabolite quantifications included principal component analyses and the L1-penalized regularized regression algorithm known as the least absolute shrinkage and selection operator. RESULTS The most significant metabolite difference (P<0.0056) was [2-C]glutamine's higher final/control ratio for the hypothermia group (1.75±0.12) compared with ratios for the delayed (1.12±0.12) and normothermia group (0.94±0.06), implying a higher pyruvate carboxylase/pyruvate dehydrogenase ratio for glutamine formation. Least Absolute Shrinkage and Selection Operator found the most important metabolites associated with adenosine triphosphate preservation: [3,4-C]glutamate-produced via pyruvate dehydrogenase entry, [2-C]taurine-an important osmolyte and antioxidant, and phosphocreatine. Final principal component analyses scores plots suggested separate cluster formation for the hypothermia group, but with insufficient data for statistical significance. CONCLUSIONS Starting mild hypothermia simultaneously with oxygen-glucose deprivation, compared with delayed starting or no hypothermia, has higher pyruvate carboxylase throughput, suggesting that better glial integrity is one important neuroprotection mechanism of earlier hypothermia.
Collapse
|
27
|
The potential roles of 18F-FDG-PET in management of acute stroke patients. BIOMED RESEARCH INTERNATIONAL 2013; 2013:634598. [PMID: 23762852 PMCID: PMC3671294 DOI: 10.1155/2013/634598] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 04/14/2013] [Indexed: 01/17/2023]
Abstract
Extensive efforts have recently been devoted to developing noninvasive imaging tools capable of delineating brain tissue viability (penumbra) during acute ischemic stroke. These efforts could have profound clinical implications for identifying patients who may benefit from tPA beyond the currently approved therapeutic time window and/or patients undergoing neuroendovascular treatments. To date, the DWI/PWI MRI and perfusion CT have received the most attention for identifying ischemic penumbra. However, their routine use in clinical settings remains limited. Preclinical and clinical PET studies with [18F]-fluoro-2-deoxy-D-glucose (18F-FDG) have consistently revealed a decreased 18F-FDG uptake in regions of presumed ischemic core. More importantly, an elevated 18F-FDG uptake in the peri-ischemic regions has been reported, potentially reflecting viable tissues. To this end, this paper provides a comprehensive review of the literature on the utilization of 14C-2-DG and 18F-FDG-PET in experimental as well as human stroke studies. Possible cellular mechanisms and physiological underpinnings attributed to the reported temporal and spatial uptake patterns of 18F-FDG are addressed. Given the wide availability of 18F-FDG in routine clinical settings, 18F-FDG PET may serve as an alternative, non-invasive tool to MRI and CT for the management of acute stroke patients.
Collapse
|
28
|
Kochanski R, Peng C, Higashida T, Geng X, Hüttemann M, Guthikonda M, Ding Y. Neuroprotection conferred by post-ischemia ethanol therapy in experimental stroke: an inhibitory effect on hyperglycolysis and NADPH oxidase activation. J Neurochem 2013; 126:113-21. [PMID: 23350720 DOI: 10.1111/jnc.12169] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/09/2013] [Accepted: 01/24/2013] [Indexed: 01/04/2023]
Abstract
Ethanol provides neuroprotection following ischemia/reperfusion. This study assessed ethanol's effect on hyperglycolysis and NADPH oxidase (NOX) activation. Adult, male Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 2 h. Three sets of experiments were conducted to determine ethanol's effect on (i) conferring neuroprotection by measuring infarct volume and neurological deficits 24 h post reperfusion; (ii) cerebral glucose metabolism and lactic acidosis by measuring brain and blood glucose concentrations and protein expression of glucose transporter 1 and 3 (GLUT1, GLUT3), phosphofructokinase (PFK), as well as lactic acidosis by measuring lactate dehydrogenase (LDH), and lactate; and (iii) nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) activation by detecting enzymatic activity and subunit expression at 3 h after reperfusion. When administered upon reperfusion, ethanol (1.5 g/kg) reduced infarct volume by 40% (p < 0.01) and neurological deficits by 48% at 24 h post reperfusion while reducing (p < 0.01) elevations in glycolytic protein expression and lactate levels during early reperfusion (3 h). Ethanol increased the reductions in cerebral glucose concentration at 3 h post reperfusion by 64% (p < 0.01) while enhancing (p < 0.01) post stroke blood glucose concentration, suggesting a reduced cellular glucose uptake and utilization. Ethanol decreased (p < 0.01) stroke-induced NOX activation by reducing enzymatic activity and gp91(phox) expression by 45% and 38%, respectively. Post-ischemia ethanol treatment exerts neuroprotection through attenuation of hyperglycolysis and associated NOX activation. Because of the lack of associated hypoglycemia and selectivity toward decreasing cerebral metabolism, further investigation of ethanol's use as a post-stroke therapy, especially in the context of hyperglycemia, seems warranted.
Collapse
Affiliation(s)
- Ryan Kochanski
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Bennet L, Davidson JO, Koome M, Gunn AJ. Glucocorticoids and preterm hypoxic-ischemic brain injury: the good and the bad. J Pregnancy 2012; 2012:751694. [PMID: 22970371 PMCID: PMC3431094 DOI: 10.1155/2012/751694] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 07/13/2012] [Indexed: 12/20/2022] Open
Abstract
Fetuses at risk of premature delivery are now routinely exposed to maternal treatment with synthetic glucocorticoids. In randomized clinical trials, these substantially reduce acute neonatal systemic morbidity, and mortality, after premature birth and reduce intraventricular hemorrhage. However, the overall neurodevelopmental impact is surprisingly unclear; worryingly, postnatal glucocorticoids are consistently associated with impaired brain development. We review the clinical and experimental evidence on how glucocorticoids may affect the developing brain and highlight the need for systematic research.
Collapse
Affiliation(s)
- Laura Bennet
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland 1142, New Zealand.
| | | | | | | |
Collapse
|
30
|
Kendall GS, Hristova M, Zbarsky V, Clements A, Peebles DM, Robertson NJ, Raivich G. Distribution of pH changes in mouse neonatal hypoxic-ischaemic insult. Dev Neurosci 2012; 33:505-18. [PMID: 22343485 DOI: 10.1159/000333850] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 09/26/2011] [Indexed: 11/19/2022] Open
Abstract
We assessed the distribution in brain pH after neonatal hypoxic-ischaemic insult and its correlation with local injury. Postnatal day 7 mice were injected with neutral red and underwent left carotid occlusion and exposure to 8% oxygen. Images captured from the cut surface of snap-frozen brain were used to calculate the pH from the blue-green absorbance ratios. Carotid occlusion alone had no effect, but combined with hypoxia caused rapid, biphasic pH decline, with the first plateau at 15-30 min, and the second at 60-90 min. The ipsilateral dorsal cortex, hippocampus, striatum and thalamus were most affected. Contralateral pH initially showed only 30% of the ipsilateral decline, becoming more acidotic with increasing duration. Systemic blood analysis revealed, compared with hypoxia alone, that combined insult caused a 63% decrease in blood glucose (1.3 ± 0.2 mM), a 2-fold increase in circulating lactate (17.7 ± 2.9 mM), a reduction in CO(2) to 1.9 ± 0.1 kPa and a drop in pH (7.26 ± 0.06). Re-oxygenation resulted in the normalisation of systemic changes, as well as a global alkaline rebound in brain pH at 4-6 h. A topographic comparison of brain injury showed only a partial correlation with pH changes, with the severest injury occurring in the ipsilateral hippocampus and sparing acidic parts of the contralateral cortex.
Collapse
Affiliation(s)
- Giles S Kendall
- Centre for Perinatal Brain Protection and Repair, Department of Obstetrics and Gynaecology, University College London, London, UK
| | | | | | | | | | | | | |
Collapse
|
31
|
Yang ZJ, Ni X, Carter EL, Kibler K, Martin LJ, Koehler RC. Neuroprotective effect of acid-sensing ion channel inhibitor psalmotoxin-1 after hypoxia-ischemia in newborn piglet striatum. Neurobiol Dis 2011; 43:446-54. [PMID: 21558004 DOI: 10.1016/j.nbd.2011.04.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 04/18/2011] [Accepted: 04/22/2011] [Indexed: 11/29/2022] Open
Abstract
Na+,Ca2+-permeable acid-sensing ion channel 1a (ASIC1a) is involved in the pathophysiologic process of adult focal brain ischemia. However, little is known about its role in the pathogenesis of global cerebral ischemia or newborn hypoxia-ischemia (H-I). Here, using a newborn piglet model of asphyxia-induced cardiac arrest, we investigated the effect of ASIC1a-specific blocker psalmotoxin-1 on neuronal injury. During asphyxia and the first 30min of recovery, brain tissue pH fell below 7.0, the approximate activation pH of ASIC1a. Psalmotoxin-1 injection at 20min before hypoxia, but not at 20min of recovery, partially protected the striatonigral and striatopallidal neurons in putamen. Psalmotoxin-1 pretreatment largely attenuated the increased protein kinase A-dependent phosphorylation of DARPP-32 and N-methyl-d-aspartate (NMDA) receptor NR1 subunit and decreased nitrative and oxidative damage to proteins at 3h of recovery. Pretreatment with NMDA receptor antagonist MK-801 also provided partial neuroprotection in putamen, and combined pretreatment with psalmotoxin-1 and MK-801 yielded additive neuroprotection. These results indicate that ASIC1a activation contributes to neuronal death in newborn putamen after H-I through mechanisms that may involve protein kinase A-dependent phosphorylation of NMDA receptor and nitrative and oxidative stress.
Collapse
Affiliation(s)
- Zeng-Jin Yang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
Perioperative fluid management in paediatrics has been the subject of many controversies in recent years, but fluid management in the neonatal period has not been considered in most reviews and guidelines. The literature regarding neonatal fluid management mainly appears in the paediatric textbooks and few recent data are available, except for resuscitation and fluid loading during shock and major surgery. In the context of anaesthesia, many neonates requiring surgery within the first month of life have organ malformation and/or dysfunction. This article aims at reviewing basic physiological considerations important for neonatal fluid management and mainly focusses on fluid maintenance and replacement during surgery.
Collapse
Affiliation(s)
- Isabelle Murat
- Department of Anesthesia, Hôpital d'Enfants Armand Trousseau, 26 avenue du Dr. Arnold Netter, 75571 Paris, Cedex 12, France.
| | | | | | | |
Collapse
|
33
|
Abstract
Neonatal encephalopathy affects 2 to 5 of every 1000 live births and represents a major cause of mortality and long-term morbidity in affected infants. Hypoxic ischemic encephalopathy (HIE) is the major cause of encephalopathy in the neonatal period. Until recently, management of a newborn with encephalopathy has consisted largely of supportive care to restore and maintain cerebral perfusion, provide adequate gas exchange and treat seizure activity. Recent randomized controlled trials have shown that mild therapeutic hypothermia (cooling) initiated within 6 h of birth reduces death and disability in these infants. Cooling can be accomplished through whole-body cooling or selective head cooling. Meta-analysis of these trials suggests that for every six or seven infants with moderate to severe HIE who are treated with mild hypothermia, there will be one fewer infant who dies or has significant neurodevelopmental disability. In response to this evidence, major policy makers and guideline developers have recommended that cooling therapy be offered to infants with moderate to severe HIE. The dissemination of this new therapy will require improved identification of infants with HIE and regional commitment to allow these infants to be cared for in a timely manner.
Collapse
|
34
|
Van Herreweghe F, Festjens N, Declercq W, Vandenabeele P. Tumor necrosis factor-mediated cell death: to break or to burst, that's the question. Cell Mol Life Sci 2010; 67:1567-79. [PMID: 20198502 PMCID: PMC11115929 DOI: 10.1007/s00018-010-0283-0] [Citation(s) in RCA: 159] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 01/20/2010] [Indexed: 02/06/2023]
Abstract
In this review, we discuss the signal-transduction pathways of three major cellular responses induced by tumor necrosis factor (TNF): cell survival through NF-kappaB activation, apoptosis, and necrosis. Recruitment and activation of caspases plays a crucial role in the initiation and execution of TNF-induced apoptosis. However, experimental inhibition of caspases reveals an alternative cell death pathway, namely necrosis, also called necroptosis, suggesting that caspases actively suppress the latter outcome. TNF-induced necrotic cell death crucially depends on the kinase activity of receptor interacting protein serine-threonine kinase 1 (RIP1) and RIP3. It was recently demonstrated that ubiquitination of RIP1 determines whether it will function as a pro-survival or pro-cell death molecule. Deeper insight into the mechanisms that control the molecular switches between cell survival and cell death will help us to understand why TNF can exert so many different biological functions in the etiology and pathogenesis of human diseases.
Collapse
Affiliation(s)
- Franky Van Herreweghe
- Unit For Molecular Signalling and Cell Death, Department for Molecular Biomedical Research, VIB, Technologiepark 927, 9052 Ghent (Zwijnaarde), Belgium
- Unit for Molecular Signalling and Cell Death, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Ghent, Belgium
| | - Nele Festjens
- Unit for Molecular Glycobiology, Department for Molecular Biomedical Research, VIB, Ghent University, Technologiepark 927, 9052 Ghent, Belgium
- Laboratory for Protein Biochemistry and Biomolecular Engineering, Department of Biochemistry and Microbiology, Ghent University, Ledeganckstraat 35, 9052 Ghent, Belgium
| | - Wim Declercq
- Unit For Molecular Signalling and Cell Death, Department for Molecular Biomedical Research, VIB, Technologiepark 927, 9052 Ghent (Zwijnaarde), Belgium
- Unit for Molecular Signalling and Cell Death, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Ghent, Belgium
| | - Peter Vandenabeele
- Unit For Molecular Signalling and Cell Death, Department for Molecular Biomedical Research, VIB, Technologiepark 927, 9052 Ghent (Zwijnaarde), Belgium
- Unit for Molecular Signalling and Cell Death, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Ghent, Belgium
| |
Collapse
|
35
|
|
36
|
Abstract
Hypoxic-ischemic encephalopathy (HIE) is one of the most important complications found in the newborn period. It is the result of a deprivation of oxygen and glucose to the neural tissue, which may be the result of either hypoxemia or ischemia. Experimental animal research and clinical observations in humans have noted that the pattern of injury occurs in 2 phases. The first phase is a primary energy failure related to the insult, and then a second energy failure occurs some hours later. The combined effects of cellular energy failure, acidosis, glutamate release, intracellular accumulation of calcium, lipid peroxidation, and nitric oxide neurotoxicity destroy essential components of the cell, culminating in cell death. The clinical presentation depends on the severity, timing, and duration of the insult, with symptoms typically evolving over approximately 72 hours. Hypothermia strategies are aimed at targeting this narrow window of opportunity to ameliorate the brain injury.
Collapse
|
37
|
Abstract
We are entering an era in which hypothermia will be used in combination with other novel neuroprotective interventions. The targeting of multiple sites in the cascade leading to brain injury may prove to be a more effective treatment strategy after hypoxic-ischemic encephalopathy in newborn infants than hypothermia alone.
Collapse
Affiliation(s)
- Rakesh Sahni
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| | | |
Collapse
|
38
|
Gibbs ME, Rodricks CL, Hutchinson DS, Summers RJ, Miller SL. Importance of adrenergic receptors in prenatally induced cognitive impairment in the domestic chick. Int J Dev Neurosci 2008; 27:27-35. [PMID: 19027843 DOI: 10.1016/j.ijdevneu.2008.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2008] [Revised: 10/16/2008] [Accepted: 10/29/2008] [Indexed: 11/30/2022] Open
Abstract
In the domestic chick, mild hypoxia (24h of 14% oxygen) at two stages of embryonic development results in post-hatch memory deficiencies tested using a discriminated bead avoidance task. The nature of the memory loss depends on the gestational age at which the hypoxia occurs. Hypoxia on embryonic day 10 (E10) of a 21 day incubation results in chicks with no short-term memory 10 min after training, whereas hypoxia on day 14 (E14) results in chicks with good labile memory 30 min after training but no consolidation of memory into permanent storage (120 min). Hypoxia at E14 is associated with increased plasma levels of noradrenaline and therefore we suggest that altered catecholamine exposure within the brain contributes to cognitive problems by modifying the responsiveness of brain beta-adrenoceptors. In ovo administration of noradrenaline, or the beta(2)-adrenoceptor agonist formoterol, at E14 had the same effect on memory consolidation as hypoxia. Following hypoxia at E14, memory could be rescued after training by central injection of a beta(3)-adrenoceptor agonist, but not by a beta(2)-adrenoceptor agonist. The differences in the responsiveness of memory processing to beta(2)-adrenoceptor agonists suggests alterations to the receptors or downstream of the receptor activation. However, both types of beta-adrenoceptor agonists rescued memory in E10 treated chicks implying that at this age hypoxia does not affect the receptors. In summary, hypoxia or increased levels of stress hormones during incubation alters beta-adrenoceptor responsiveness; the outcome of the insult depends upon the cellular developmental processes at a given embryonic stage.
Collapse
Affiliation(s)
- Marie E Gibbs
- Department of Anatomy & Developmental Biology, Monash University, Wellington Road, Clayton, 3800 Victoria, Australia.
| | | | | | | | | |
Collapse
|
39
|
Haiju Z, Suyuan H, Xiufang F, Lu Y, Sun R. The combined detection of umbilical cord nucleated red blood cells and lactate: early prediction of neonatal hypoxic ischemic encephalopathy. J Perinat Med 2008; 36:240-7. [PMID: 18576934 DOI: 10.1515/jpm.2008.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To establish a simple and quick method that could be used to predict the occurrence of hypoxic ischemic encephalopathy (HIE) as early as possible by investigating the variations of nucleated red blood cells per 100 white blood cell (NRBC/100 WBC) counts and lactate levels in cord blood. METHODS In 46 cases of acute fetal distress (AFD) and 54 cases of chronic fetal distress (CFD) neonates we measured the percentage (NRBCs/100 WBC) and lactate in the umbilical blood. RESULTS Both lactate levels and NRBC/100WBC counts were higher in CFD and AFD groups than in controls (both P<0.01). The numbers of NRBC/100 WBC and the values of lactate in moderate-severe HIE group were higher than in mild-HIE group (P=0.002 and P=0.042, respectively). The combined sensitivity and specificity was 94% and 96% at 15NRBC/100WBC and 4.25 mmol/L level by combined detecting NRBC and lactate to predict HIE. Three infants (including 1 death and 2 survivors) had the highest levels of NRBC/100WBC and lactate in cord blood, and the 2 survivors had the lowest mental development index (MDI) and psychomotor development index (PDI). CONCLUSIONS Combined detection with NRBC/100WBC and lactate allows early prediction of development and severity of HIE. The levels of these parameters are related to the neurodevelopment outcome of HIE infants.
Collapse
Affiliation(s)
- Zhang Haiju
- Department of Pediatrics, Qilu Hospital, Shandong University, China
| | | | | | | | | |
Collapse
|
40
|
Yang Z, Levison SW. Perinatal hypoxic/ischemic brain injury induces persistent production of striatal neurons from subventricular zone progenitors. Dev Neurosci 2007; 29:331-40. [PMID: 17762201 DOI: 10.1159/000105474] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2006] [Accepted: 01/02/2007] [Indexed: 11/19/2022] Open
Abstract
Ischemia-induced production of new striatal neurons in young and adult rodents has been studied. However, it is unclear whether neonatal hypoxic/ischemic (H/I) brain injury-induced neuronogenesis in the striatum is transient or sustained, nor has it been established whether these new neurons arise from progenitors within the striatum or from precursors residing in the adjacent subventricular zone. Here, we report that from 2 weeks to 5 months after H/I there are more doublecortin-positive (Dcx+) cells and Dcx+/NeuN+ cells in the damaged striatum compared to the contralateral striatum. After the S-phase marker 5-bromo-2'-deoxyuridine (BrdU) was injected at both short and long intervals (2 days and 2 months) after H/I to label newly born cells, more BrdU+/Dcx+ and BrdU+/NeuN+ cells were observed in the ipsilateral striatum compared to the contralateral striatum. Retroviral fate-mapping studies demonstrated that these newly born striatal neurons are generated from precursors within the subventricular zone. Altogether, these observations indicate the neonatal brain initiates a prolonged regenerative response from the precursors of the subventricular zone (SVZ) that results in persistent production of new striatal neurons.
Collapse
Affiliation(s)
- Zhengang Yang
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China
| | | |
Collapse
|
41
|
|