1
|
Samaiya PK, Krishnamurthy S, Kumar A. Mitochondrial dysfunction in perinatal asphyxia: role in pathogenesis and potential therapeutic interventions. Mol Cell Biochem 2021; 476:4421-4434. [PMID: 34472002 DOI: 10.1007/s11010-021-04253-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/20/2021] [Indexed: 01/13/2023]
Abstract
Perinatal asphyxia (PA)-induced brain injury may present as hypoxic-ischemic encephalopathy in the neonatal period, and long-term sequelae such as spastic motor deficits, intellectual disability, seizure disorders and learning disabilities. The brain injury is secondary to both the hypoxic-ischemic event and oxygenation-reperfusion following resuscitation. Following PA, a time-dependent progression of neuronal insult takes place in terms of transition of cell death from necrosis to apoptosis. This transition is the result of time-dependent progression of pathomechanisms which involve excitotoxicity, oxidative stress, and ultimately mitochondrial dysfunction in developing brain. More precisely mitochondrial respiration is suppressed and calcium signalling is dysregulated. Consequently, Bax-dependent mitochondrial permeabilization occurs leading to release of cytochrome c and activation of caspases leading to transition of cell death in developing brain. The therapeutic window lies within this transition process. At present, therapeutic hypothermia (TH) is the only clinical treatment available for treating moderate as well as severe asphyxia in new-born as it attenuates secondary loss of high-energy phosphates (ATP) (Solevåg et al. in Free Radic Biol Med 142:113-122, 2019; Gunn et al. in Pediatr Res 81:202-209, 2017), improving both short- and long-term outcomes. Mitoprotective therapies can offer a new avenue of intervention alone or in combination with therapeutic hypothermia for babies with birth asphyxia. This review will explore these mitochondrial pathways, and finally will summarize past and current efforts in targeting these pathways after PA, as a means of identifying new avenues of therapeutic intervention.
Collapse
Affiliation(s)
- Puneet K Samaiya
- Department of Pharmacy, Shri G.S. Institute of Technology and Science, Indore, MP, 452003, India.
| | - Sairam Krishnamurthy
- Neurotherapeutics Lab, Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Department of Pediatrics, Institute of Medical Sciences, BHU, Varanasi, UP, India
| |
Collapse
|
2
|
Davidson JO, Dhillon SK, Wassink G, Zhou KQ, Bennet L, Gunn AJ. Endogenous neuroprotection after perinatal hypoxia-ischaemia: the resilient developing brain. J R Soc N Z 2018. [DOI: 10.1080/03036758.2018.1529685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Joanne O. Davidson
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Simerdeep K. Dhillon
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Guido Wassink
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Kelly Q. Zhou
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Alistair J. Gunn
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
3
|
Thornton C, Leaw B, Mallard C, Nair S, Jinnai M, Hagberg H. Cell Death in the Developing Brain after Hypoxia-Ischemia. Front Cell Neurosci 2017; 11:248. [PMID: 28878624 PMCID: PMC5572386 DOI: 10.3389/fncel.2017.00248] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/07/2017] [Indexed: 01/11/2023] Open
Abstract
Perinatal insults such as hypoxia–ischemia induces secondary brain injury. In order to develop the next generation of neuroprotective therapies, we urgently need to understand the underlying molecular mechanisms leading to cell death. The cell death mechanisms have been shown to be quite different in the developing brain compared to that in the adult. The aim of this review is update on what cell death mechanisms that are operating particularly in the setting of the developing CNS. In response to mild stress stimuli a number of compensatory mechanisms will be activated, most often leading to cell survival. Moderate-to-severe insults trigger regulated cell death. Depending on several factors such as the metabolic situation, cell type, nature of the stress stimulus, and which intracellular organelle(s) are affected, the cell undergoes apoptosis (caspase activation) triggered by BAX dependent mitochondrial permeabilzation, necroptosis (mixed lineage kinase domain-like activation), necrosis (via opening of the mitochondrial permeability transition pore), autophagic cell death (autophagy/Na+, K+-ATPase), or parthanatos (poly(ADP-ribose) polymerase 1, apoptosis-inducing factor). Severe insults cause accidental cell death that cannot be modulated genetically or by pharmacologic means. However, accidental cell death leads to the release of factors (damage-associated molecular patterns) that initiate systemic effects, as well as inflammation and (regulated) secondary brain injury in neighboring tissue. Furthermore, if one mode of cell death is inhibited, another route may step in at least in a scenario when upstream damaging factors predominate over protective responses. The provision of alternative routes through which the cell undergoes death has to be taken into account in the hunt for novel brain protective strategies.
Collapse
Affiliation(s)
- Claire Thornton
- Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' HospitalLondon, United Kingdom
| | - Bryan Leaw
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia
| | - Carina Mallard
- Department of Physiology, Perinatal Center, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Syam Nair
- Department of Physiology, Perinatal Center, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Masako Jinnai
- Department of Physiology, Perinatal Center, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Henrik Hagberg
- Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' HospitalLondon, United Kingdom.,Department of Clinical Sciences and Physiology and Neuroscience, Perinatal Center, Sahlgrenska Academy, Gothenburg UniversityGothenburg, Sweden
| |
Collapse
|
4
|
Fineschi V, Viola RV, La Russa R, Santurro A, Frati P. A Controversial Medicolegal Issue: Timing the Onset of Perinatal Hypoxic-Ischemic Brain Injury. Mediators Inflamm 2017; 2017:6024959. [PMID: 28883688 PMCID: PMC5572618 DOI: 10.1155/2017/6024959] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/18/2017] [Indexed: 12/11/2022] Open
Abstract
Perinatal hypoxic-ischemic brain injury, as a result of chronic, subacute, and acute insults, represents the pathological consequence of fetal distress and birth or perinatal asphyxia, that is, "nonreassuring fetal status." Hypoxic-ischemic injury (HII) is typically characterized by an early phase of damage, followed by a delayed inflammatory local response, in an apoptosis-necrosis continuum. In the early phase, the cytotoxic edema and eventual acute lysis take place; with reperfusion, additional damage should be assigned to excitotoxicity and oxidative stress. Finally, a later phase involves all the inflammatory activity and long-term neural tissue repairing and remodeling. In this model mechanism, loss of mitochondrial function is supposed to be the hallmark of secondary injury progression, and autophagy which is lysosome-mediated play a role in enhancing brain injury. Early-induced molecules driven by hypoxia, as chaperonins HSPs and ORP150, besides common markers for inflammatory responses, have predictive value in timing the onset of neonatal HII; on the other hand, clinical biomarkers for HII diagnosis, as CK-BB, LDH, S-100beta, and NSE, could be useful to predict outcomes.
Collapse
Affiliation(s)
- Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| | - Rocco Valerio Viola
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
| | - Raffaele La Russa
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| | - Alessandro Santurro
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
| | - Paola Frati
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| |
Collapse
|
5
|
Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal Hypoxia Ischaemia: Mechanisms, Models, and Therapeutic Challenges. Front Cell Neurosci 2017; 11:78. [PMID: 28533743 PMCID: PMC5420571 DOI: 10.3389/fncel.2017.00078] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Neonatal hypoxia-ischaemia (HI) is the most common cause of death and disability in human neonates, and is often associated with persistent motor, sensory, and cognitive impairment. Improved intensive care technology has increased survival without preventing neurological disorder, increasing morbidity throughout the adult population. Early preventative or neuroprotective interventions have the potential to rescue brain development in neonates, yet only one therapeutic intervention is currently licensed for use in developed countries. Recent investigations of the transient cortical layer known as subplate, especially regarding subplate's secretory role, opens up a novel set of potential molecular modulators of neonatal HI injury. This review examines the biological mechanisms of human neonatal HI, discusses evidence for the relevance of subplate-secreted molecules to this condition, and evaluates available animal models. Neuroserpin, a neuronally released neuroprotective factor, is discussed as a case study for developing new potential pharmacological interventions for use post-ischaemic injury.
Collapse
Affiliation(s)
- Lancelot J. Millar
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Lei Shi
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan UniversityGuangzhou, China
| | | | - Zoltán Molnár
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| |
Collapse
|
6
|
Aksoy T, Richardson BS, Han VK, Gagnon R. Apoptosis in the Ovine Fetal Brain Following Placental Embolization and Intermittent Umbilical Cord Occlusion. Reprod Sci 2015; 23:249-56. [PMID: 26346442 DOI: 10.1177/1933719115602774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The purpose of this study was to compare the regional distribution of apoptotic cells in the near term ovine fetal brain caused by prolonged moderate hypoxia, as seen in placental insufficiency, and intermittent severe hypoxia, as seen in umbilical cord compression, which may then contribute to adverse neurodevelopment in the postnatal life. We hypothesized that apoptosis in the fetal brain will be increased in response to both prolonged moderate hypoxia and intermittent severe hypoxia. Twenty-one near term (126-127 days) sheep were divided into 3 groups: control (CON; n = 7), placental embolization (EMB; n = 7), and umbilical cord occlusion (UCO; n = 8). The EMB group had microsphere injections into the umbilical arterial circulation until the oxygen content was at 50% of baseline value. The UCO group had complete cord occlusion for 2 minutes every hour, 6 times a day for 2 consecutive days. At 4 pm on day 2, the animals were euthanized; fetal brains were fixed and prepared for apoptosis staining using the terminal uridine deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay method. In the cerebellar white matter, there was a 3-fold increase in the number of TUNEL positive cells per 1000 cells in both EMB and UCO animals as compared to CON (P = .017). There was also a significant increase in the frontal cortical grey matter (layers 1-3) in EMB animals as compared to CON (P = .014). As such, apoptosis in the near term fetal sheep brain is altered with both sustained moderate hypoxia and intermittent severe hypoxia in the latter part of pregnancy, with potential for long-term neurological sequelae.
Collapse
Affiliation(s)
- Tuba Aksoy
- Departments of Obstetrics and Gynecology, Physiology, and Pediatrics, The Lawson Health Research Institute, Western University, London, Ontario, Canada
| | - Bryan S Richardson
- Departments of Obstetrics and Gynecology, Physiology, and Pediatrics, The Lawson Health Research Institute, Western University, London, Ontario, Canada
| | - Victor K Han
- Departments of Obstetrics and Gynecology, Physiology, and Pediatrics, The Lawson Health Research Institute, Western University, London, Ontario, Canada
| | - Robert Gagnon
- Department of Obstetrics and Gynecology, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Seehase M, Jennekens W, Zwanenburg A, Andriessen P, Collins JJ, Kuypers E, Zimmermann LJ, Vles JS, Gavilanes AW, Kramer BW. Propofol administration to the maternal-fetal unit improved fetal EEG and influenced cerebral apoptotic pathway in preterm lambs suffering from severe asphyxia. Mol Cell Pediatr 2015; 2:4. [PMID: 26542294 PMCID: PMC4530565 DOI: 10.1186/s40348-015-0016-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 02/24/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Term and near-term infants are at high risk of developing brain injury and life-long disability if they have suffered from severe perinatal asphyxia. We hypothesized that propofol administration to the maternal-fetal unit can diminish cerebral injury in term and near-term infant fetuses in states of progressive severe asphyxia. METHODS Forty-four late preterm lambs underwent total umbilical cord occlusion (UCO) or sham treatment in utero. UCO resulted in global asphyxia and cardiac arrest. After emergency cesarean section under either maternal propofol or isoflurane anesthesia, the fetuses were resuscitated and subsequently anesthetized the same way as their mothers. RESULTS Asphyctic lambs receiving isoflurane showed a significant increase of total and low-frequency spectral power in bursts indicating seizure activity and more burst-suppression with a marked increase of interburst interval length during UCO. Asphyctic lambs receiving propofol showed less EEG changes. Propofol increased levels of anti-apoptotic B-cell lymphoma-extra large (Bcl-xL) and phosphorylated STAT-3 and reduced the release of cytochrome c from the mitochondria and the protein levels of activated cysteinyl aspartate-specific protease (caspase)-3, -9, and N-methyl-d-aspartate (NMDA) receptor. CONCLUSIONS Improvement of fetal EEG during and after severe asphyxia could be achieved by propofol treatment of the ovine maternal-fetal unit. The underlying mechanism is probably the reduction of glutamate-induced cytotoxicity by down-regulation of NMDA receptors and an inhibition of the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Matthias Seehase
- Department of Paediatrics, Maastricht University Medical Center, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands. .,School of Mental Health and Neuroscience; School of Oncology and Developmental Biology, Maastricht University, and European Graduate School of Neuroscience (EURON), P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands. .,Department of Pediatric Cardiology and Intensive Care Medicine with Neonatology, University Hospital, Georg-August-University, Robert-Koch-Str. 40, D 37099, Göttingen, Germany.
| | - Ward Jennekens
- Department of Clinical Physics, Maasstad Hospital, Postbus 9100, 3007 AC, Rotterdam, The Netherlands.
| | - Alex Zwanenburg
- Department of Paediatrics, Maastricht University Medical Center, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands. .,Department of Biomedical Technology, Maastricht University, Faculty of Health, Medicine and Life Sciences, School of Cardiovascular Diseases, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands.
| | - Peter Andriessen
- Department of Paediatrics, Maastricht University Medical Center, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands. .,Department of Paediatrics, Máxima Medical Center, De Run 4600, 5504 DB, Veldhoven, The Netherlands.
| | - Jennifer Jp Collins
- Department of Paediatrics, Maastricht University Medical Center, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands. .,School of Mental Health and Neuroscience; School of Oncology and Developmental Biology, Maastricht University, and European Graduate School of Neuroscience (EURON), P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands. .,Regenerative Medicine Program, Sprott Centre for Stem Cell Research at the Ottawa Hospital Research Institute, 725 Parkdale Ave, Ottawa ON K1Y 4E9, Ontario, Canada.
| | - Elke Kuypers
- Department of Paediatrics, Maastricht University Medical Center, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands. .,School of Mental Health and Neuroscience; School of Oncology and Developmental Biology, Maastricht University, and European Graduate School of Neuroscience (EURON), P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands.
| | - Luc J Zimmermann
- Department of Paediatrics, Maastricht University Medical Center, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands.
| | - Johan Sh Vles
- Department of Neurology, Maastricht University Medical Center, Faculty of Health, Medicine and Life Sciences, School of Mental Health and Neuroscience, Maastricht University, and European Graduate School of Neuroscience (EURON), P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands.
| | - Antonio Wd Gavilanes
- Department of Paediatrics, Maastricht University Medical Center, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands.
| | - Boris W Kramer
- Department of Paediatrics, Maastricht University Medical Center, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands. .,School of Mental Health and Neuroscience; School of Oncology and Developmental Biology, Maastricht University, and European Graduate School of Neuroscience (EURON), P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands.
| |
Collapse
|
8
|
Tusor N, Edwards AD. Birth asphyxia: 100 years of progress. J Pediatr 2014; 165:1081-3. [PMID: 25282068 DOI: 10.1016/j.jpeds.2014.08.051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 08/26/2014] [Indexed: 01/07/2023]
Affiliation(s)
- Nora Tusor
- Center for the Developing Brain, Department of Perinatal Imaging and Health, Division of Imaging Sciences and Biomedical Engineering, King's College, London, United Kingdom
| | - A David Edwards
- Center for the Developing Brain, Department of Perinatal Imaging and Health, Division of Imaging Sciences and Biomedical Engineering, King's College, London, United Kingdom.
| |
Collapse
|
9
|
Ozyürek H, Bayrak S, Pehlivanoğlu B, Atilla P, Balkancı ZD, Cakar N, Anlar B. Effect of transient maternal hypotension on apoptotic cell death in foetal rat brain. Balkan Med J 2014; 31:88-94. [PMID: 25207175 DOI: 10.5152/balkanmedj.2013.8313] [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: 12/07/2012] [Accepted: 07/31/2013] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Intrauterine perfusion insufficiency induced by transient maternal hypotension has been reported to be associated with foetal brain malformations. However, the effects of maternal hypotension on apoptotic processes in the foetal brain have not been investigated experimentally during the intrauterine period. AIMS The aim of this study was to investigate the effects of transient maternal hypotension on apoptotic cell death in the intrauterine foetal brain. STUDY DESIGN Animal experimentation. METHODS Three-month-old female Wistar albino rats were allocated into four groups (n=5 each). The impact of hypoxic/ischemic injury induced by transient maternal hypotension on the 15th day of pregnancy (late gestation) in rats was investigated at 48 (H17 group) or 96 hours (H19 group) after the insult. Control groups underwent the same procedure except for induction of hypotension (C17 and H17 groups). Brain sections of one randomly selected foetus from each pregnant rat were histopathologically evaluated for hypoxic/ischemic injury in the metencephalon, diencephalon, and telencephalon by terminal transferase-mediated dUTP nick end labelling and active cysteine-dependent aspartate-directed protease-3 (caspase-3) positivity for cell death. RESULTS The number of terminal transferase-mediated dUTP nick end labelling (+) cells in all the areas examined was comparable in both hypotension and control groups. The H17 group had active caspase-3 (+) cells in the metencephalon and telencephalon, sparing diencephalon, whereas the C19 and H19 groups had active caspase-3 (+) cells in all three regions. The number of active caspase-3 (+) cells in the telencephalon in the H19 group was higher compared with the metencephalon and diencephalon and compared with H17 group (p<0.05). CONCLUSION Our results suggest that prenatal hypoxic/ischemic injury triggers apoptotic mechanisms. Therefore, blockade of apoptotic pathways, considering the time pattern of the insult, may constitute a potential neuroprotective approach for the detrimental effects of prenatal hypoperfusion.
Collapse
Affiliation(s)
- Hamit Ozyürek
- Pediatric Neurology Unit, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Sibel Bayrak
- Department of Physiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Bilge Pehlivanoğlu
- Department of Physiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Pergin Atilla
- Department of Histology and Embryology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | | | - Nur Cakar
- Department of Histology and Embryology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Banu Anlar
- Pediatric Neurology Unit, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
10
|
Wassink G, Gunn ER, Drury PP, Bennet L, Gunn AJ. The mechanisms and treatment of asphyxial encephalopathy. Front Neurosci 2014; 8:40. [PMID: 24578682 PMCID: PMC3936504 DOI: 10.3389/fnins.2014.00040] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 02/12/2014] [Indexed: 11/13/2022] Open
Abstract
Acute post-asphyxial encephalopathy occurring around the time of birth remains a major cause of death and disability. The recent seminal insight that allows active neuroprotective treatment is that even after profound asphyxia (the “primary” phase), many brain cells show initial recovery from the insult during a short “latent” phase, typically lasting approximately 6 h, only to die hours to days later after a “secondary” deterioration characterized by seizures, cytotoxic edema, and progressive failure of cerebral oxidative metabolism. Although many of these secondary processes are potentially injurious, they appear to be primarily epiphenomena of the “execution” phase of cell death. Animal and human studies designed around this conceptual framework have shown that moderate cerebral hypothermia initiated as early as possible but before the onset of secondary deterioration, and continued for a sufficient duration to allow the secondary deterioration to resolve, has been associated with potent, long-lasting neuroprotection. Recent clinical trials show that while therapeutic hypothermia significantly reduces morbidity and mortality, many babies still die or survive with disabilities. The challenge for the future is to find ways of improving the effectiveness of treatment. In this review, we will dissect the known mechanisms of hypoxic-ischemic brain injury in relation to the known effects of hypothermic neuroprotection.
Collapse
Affiliation(s)
- Guido Wassink
- Fetal Physiology and Neuroscience Team, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland Auckland, New Zealand
| | - Eleanor R Gunn
- Fetal Physiology and Neuroscience Team, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland Auckland, New Zealand
| | - Paul P Drury
- Fetal Physiology and Neuroscience Team, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland Auckland, New Zealand
| | - Laura Bennet
- Fetal Physiology and Neuroscience Team, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland Auckland, New Zealand
| | - Alistair J Gunn
- Fetal Physiology and Neuroscience Team, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland Auckland, New Zealand
| |
Collapse
|
11
|
Bhalala US, Koehler RC, Kannan S. Neuroinflammation and neuroimmune dysregulation after acute hypoxic-ischemic injury of developing brain. Front Pediatr 2014; 2:144. [PMID: 25642419 PMCID: PMC4294124 DOI: 10.3389/fped.2014.00144] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/22/2014] [Indexed: 12/15/2022] Open
Abstract
Hypoxic-ischemic (HI) injury to developing brain results from birth asphyxia in neonates and from cardiac arrest in infants and children. It is associated with varying degrees of neurologic sequelae, depending upon the severity and length of HI. Global HI triggers a series of cellular and biochemical pathways that lead to neuronal injury. One of the key cellular pathways of neuronal injury is inflammation. The inflammatory cascade comprises activation and migration of microglia - the so-called "brain macrophages," infiltration of peripheral macrophages into the brain, and release of cytotoxic and proinflammatory cytokines. In this article, we review the inflammatory and immune mechanisms of secondary neuronal injury after global HI injury to developing brain. Specifically, we highlight the current literature on microglial activation in relation to neuronal injury, proinflammatory and anti-inflammatory/restorative pathways, the role of peripheral immune cells, and the potential use of immunomodulators as neuroprotective compounds.
Collapse
Affiliation(s)
- Utpal S Bhalala
- Department of Anesthesiology, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Department of Critical Care Medicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Raymond C Koehler
- Department of Anesthesiology, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Department of Critical Care Medicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Sujatha Kannan
- Department of Anesthesiology, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Department of Critical Care Medicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| |
Collapse
|
12
|
Abstract
Hypoxia-ischemia is a leading cause of morbidity and mortality in the perinatal period with an incidence of 1/4000 live births. Biochemical events such as energy failure, membrane depolarization, brain edema, an increase of neurotransmitter release and inhibition of uptake, an increase of intracellular Ca(2+), production of oxygen-free radicals, lipid peroxidation, and a decrease of blood flow are triggered by hypoxia-ischemia and may lead to brain dysfunction and neuronal death. These abnormalities can result in mental impairments, seizures, and permanent motor deficits, such as cerebral palsy. The physical and emotional strain that is placed on the children affected and their families is enormous. The care that these individuals need is not only confined to childhood, but rather extends throughout their entire life span, so it is very important to understand the pathophysiology that follows a hypoxic-ischemic insult. This review will highlight many of the mechanisms that lead to neuronal death and include the emerging area of white matter injury as well as the role of inflammation and will provide a summary of therapeutic strategies. Hypothermia and oxygen will also be discussed as treatments that currently lack a specific target in the hypoxic/ischemic cascade.
Collapse
Affiliation(s)
- John W Calvert
- Departments of Neurosurgery and Molecular and Cellular Physiology, Loma Linda University Medical Center, 11234 Anderson Street, Loma Linda, CA 92354, USA
| | | |
Collapse
|
13
|
Volpe JJ. Neonatal encephalopathy: an inadequate term for hypoxic-ischemic encephalopathy. Ann Neurol 2012; 72:156-66. [PMID: 22926849 DOI: 10.1002/ana.23647] [Citation(s) in RCA: 230] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This Point of View article addresses neonatal encephalopathy (NE) presumably caused by hypoxia-ischemia and the terminology currently in wide use for this disorder. The nonspecific term NE is commonly utilized for those infants with the clinical and imaging characteristics of neonatal hypoxic-ischemic encephalopathy (HIE). Multiple magnetic resonance imaging studies of term infants with the clinical setting of presumed hypoxia-ischemia near the time of delivery have delineated a topography of lesions highly correlated with that defined by human neuropathology and by animal models, including primate models, of hypoxia-ischemia. These imaging findings, coupled with clinical features consistent with perinatal hypoxic-ischemic insult(s), warrant the specific designation of neonatal HIE.
Collapse
Affiliation(s)
- Joseph J Volpe
- Department of Neurology, Harvard Medical School, Children's Hospital Boston, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Li T, Xu F, Cheng X, Guo X, Ji L, Zhang Z, Wang X, Blomgren K, Simbruner G, Zhu C. Systemic hypothermia induced within 10 hours after birth improved neurological outcome in newborns with hypoxic-ischemic encephalopathy. Hosp Pract (1995) 2012; 37:147-52. [PMID: 20877184 DOI: 10.3810/hp.2009.12.269] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVE To evaluate the efficacy of systemic hypothermia when applied within 10 hours after birth to neonates with hypoxic-ischemic encephalopathy (HIE). STUDY DESIGN Ninety-three term infants with moderate-to-severe HIE were randomly assigned to either systemic hypothermia (n = 46) or conventional treatment (n = 47). Hypothermia was induced within 10 hours after birth, decreasing rectal temperature to 33.5°C for 72 hours, followed by slow rewarming to 36.5°C. Neurodevelopmental outcome was assessed at 18 months old. The primary outcome was death or moderate-to-severe disability. RESULTS Outcome data were available for 82 infants. Death or moderate-to-severe disability occurred in 21 of 44 infants (47.7%) in the control group and in 7 of 38 infants (18.4%) in the hypothermia group (P = 0.01) at 18 months. The primary outcome was not different whether hypothermia was started within 6 hours or 6 to 10 hours after birth. Subgroup analysis suggested that systemic hypothermia improved long-term outcome only in infants with moderate HIE (P = 0.009), but not in those with severe HIE. No severe hypothermia-related adverse events were observed. CONCLUSION Systemic hypothermia reduced the risk of disability in infants with moderate HIE, in accordance with earlier studies. Hypothermia was induced within 6 hours in most infants, but delaying the onset to 6 to 10 hours after birth did not negatively affect primary outcome. Further studies with a large number of patients are needed to confirm that delayed cooling is equally effective.
Collapse
Affiliation(s)
- Tongchuan Li
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Filippi L, Fiorini P, Daniotti M, Catarzi S, Savelli S, Fonda C, Bartalena L, Boldrini A, Giampietri M, Scaramuzzo R, Papoff P, Del Balzo F, Spalice A, la Marca G, Malvagia S, Della Bona ML, Donzelli G, Tinelli F, Cioni G, Pisano T, Falchi M, Guerrini R. Safety and efficacy of topiramate in neonates with hypoxic ischemic encephalopathy treated with hypothermia (NeoNATI). BMC Pediatr 2012; 12:144. [PMID: 22950861 PMCID: PMC3478965 DOI: 10.1186/1471-2431-12-144] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 08/31/2012] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Despite progresses in neonatal care, the mortality and the incidence of neuro-motor disability after perinatal asphyxia have failed to show substantial improvements. In countries with a high level of perinatal care, the incidence of asphyxia responsible for moderate or severe encephalopathy is still 2-3 per 1000 term newborns. Recent trials have demonstrated that moderate hypothermia, started within 6 hours after birth and protracted for 72 hours, can significantly improve survival and reduce neurologic impairment in neonates with hypoxic-ischemic encephalopathy. It is not currently known whether neuroprotective drugs can further improve the beneficial effects of hypothermia. Topiramate has been proven to reduce brain injury in animal models of neonatal hypoxic ischemic encephalopathy. However, the association of mild hypothermia and topiramate treatment has never been studied in human newborns. The objective of this research project is to evaluate, through a multicenter randomized controlled trial, whether the efficacy of moderate hypothermia can be increased by concomitant topiramate treatment. METHODS/DESIGN Term newborns (gestational age ≥ 36 weeks and birth weight ≥ 1800 g) with precocious metabolic, clinical and electroencephalographic (EEG) signs of hypoxic-ischemic encephalopathy will be randomized, according to their EEG pattern, to receive topiramate added to standard treatment with moderate hypothermia or standard treatment alone. Topiramate will be administered at 10 mg/kg once a day for the first 3 days of life. Topiramate concentrations will be measured on serial dried blood spots. 64 participants will be recruited in the study. To evaluate the safety of topiramate administration, cardiac and respiratory parameters will be continuously monitored. Blood samplings will be performed to check renal, liver and metabolic balance. To evaluate the efficacy of topiramate, the neurologic outcome of enrolled newborns will be evaluated by serial neurologic and neuroradiologic examinations. Visual function will be evaluated by means of behavioural standardized tests. DISCUSSION This pilot study will explore the possible therapeutic role of topiramate in combination with moderate hypothermia. Any favourable results of this research might open new perspectives about the reduction of cerebral damage in asphyxiated newborns.
Collapse
Affiliation(s)
- Luca Filippi
- Neonatal Intensive Care Unit, Medical Surgical Feto-Neonatal Department, A. Meyer University Children's Hospital, Viale Pieraccini, 24, I-50139, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Molecular mechanisms of neonatal brain injury. Neurol Res Int 2012; 2012:506320. [PMID: 22363841 PMCID: PMC3272851 DOI: 10.1155/2012/506320] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 10/11/2011] [Indexed: 12/12/2022] Open
Abstract
Fetal/neonatal brain injury is an important cause of neurological disability. Hypoxia-ischemia and excitotoxicity are considered important insults, and, in spite of their acute nature, brain injury develops over a protracted time period during the primary, secondary, and tertiary phases. The concept that most of the injury develops with a delay after the insult makes it possible to provide effective neuroprotective treatment after the insult. Indeed, hypothermia applied within 6 hours after birth in neonatal encephalopathy reduces neurological disability in clinical trials. In order to develop the next generation of treatment, we need to know more about the pathophysiological mechanism during the secondary and tertiary phases of injury. We review some of the critical molecular events related to mitochondrial dysfunction and apoptosis during the secondary phase and report some recent evidence that intervention may be feasible also days-weeks after the insult.
Collapse
|
17
|
Alonso-Alconada D, Alvarez A, Hilario E. Cannabinoid as a neuroprotective strategy in perinatal hypoxic-ischemic injury. Neurosci Bull 2011; 27:275-85. [PMID: 21788999 DOI: 10.1007/s12264-011-1008-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Perinatal hypoxia-ischemia remains the single most important cause of brain injury in the newborn, leading to death or lifelong sequelae. Because of the fact that there is still no specific treatment for perinatal brain lesions due to the complexity of neonatal hypoxic-ischemic pathophysiology, the search of new neuroprotective therapies is of great interest. In this regard, therapeutic possibilities of the endocannabinoid system have grown lately. The endocannabinoid system modulates a wide range of physiological processes in mammals and has demonstrated neuroprotective effects in different paradigms of acute brain injury, acting as a natural neuroprotectant. Concerning perinatal asphyxia, the neuroprotective role of this endogenous system is emerging these years. The present review mainly focused on the current knowledge of the cannabinoids as a new neuroprotective strategy against perinatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Daniel Alonso-Alconada
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, Leioa, Vizcaya, Spain
| | | | | |
Collapse
|
18
|
Chauvier D, Renolleau S, Holifanjaniaina S, Ankri S, Bezault M, Schwendimann L, Rousset C, Casimir R, Hoebeke J, Smirnova M, Debret G, Trichet AP, Carlsson Y, Wang X, Bernard E, Hébert M, Rauzier JM, Matecki S, Lacampagne A, Rustin P, Mariani J, Hagberg H, Gressens P, Charriaut-Marlangue C, Jacotot E. Targeting neonatal ischemic brain injury with a pentapeptide-based irreversible caspase inhibitor. Cell Death Dis 2011; 2:e203. [PMID: 21881605 PMCID: PMC3186905 DOI: 10.1038/cddis.2011.87] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Brain protection of the newborn remains a challenging priority and represents a totally unmet medical need. Pharmacological inhibition of caspases appears as a promising strategy for neuroprotection. In a translational perspective, we have developed a pentapeptide-based group II caspase inhibitor, TRP601/ORPHA133563, which reaches the brain, and inhibits caspases activation, mitochondrial release of cytochrome c, and apoptosis in vivo. Single administration of TRP601 protects newborn rodent brain against excitotoxicity, hypoxia-ischemia, and perinatal arterial stroke with a 6-h therapeutic time window, and has no adverse effects on physiological parameters. Safety pharmacology investigations, and toxicology studies in rodent and canine neonates, suggest that TRP601 is a lead compound for further drug development to treat ischemic brain damage in human newborns.
Collapse
Affiliation(s)
- D Chauvier
- Theraptosis Research Laboratory, Theraptosis SA, Pasteur BioTop, Institut Pasteur, Paris 75015, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
There is now compelling clinical evidence that prolonged, moderate cerebral hypothermia initiated within a few hours after severe hypoxia-ischemia and continued until resolution of the acute phase of delayed cell death can reduce subsequent neuronal loss and improve behavioral recovery in term infants and adults after cardiac arrest. Perhaps surprisingly, the specific mechanisms of hypothermic neuroprotection remain unclear, at least in part because hypothermia suppresses a broad range of potential injurious factors. In the present review we critically examine proposed mechanisms in relation to the known window of opportunity for effective protection with hypothermia. Better knowledge of the mechanisms of hypothermia is critical to help guide the rational development of future combination treatments to augment neuroprotection with hypothermia, and to identify those most likely to benefit from it.
Collapse
|
20
|
The discovery of hypothermic neural rescue therapy for perinatal hypoxic-ischemic encephalopathy. Semin Pediatr Neurol 2009; 16:200-6. [PMID: 19945654 DOI: 10.1016/j.spen.2009.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The development of the concepts of delayed post-ischaemic neuronal death and neural rescue brought about a search for clinical treatments to reduce brain damage after birth asphyxia. Cooling had long been an unproven empyrical therapy, and a 20 year programme of careful laboratory and clinical research has proved that hypothermia reduces neurological damage in infants suffering perinatal asphyxial encephalopathy.
Collapse
|
21
|
Zhu C, Kang W, Xu F, Cheng X, Zhang Z, Jia L, Ji L, Guo X, Xiong H, Simbruner G, Blomgren K, Wang X. Erythropoietin improved neurologic outcomes in newborns with hypoxic-ischemic encephalopathy. Pediatrics 2009; 124:e218-26. [PMID: 19651565 DOI: 10.1542/peds.2008-3553] [Citation(s) in RCA: 240] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE The purpose of this study was to evaluate the efficacy and safety of erythropoietin in neonatal hypoxic-ischemic encephalopathy (HIE), by using a randomized, prospective study design. METHODS A total of 167 term infants with moderate/severe HIE were assigned randomly to receive either erythropoietin (N = 83) or conventional treatment (N = 84). Recombinant human erythropoietin, at either 300 U/kg (N = 52) or 500 U/kg (N = 31), was administered every other day for 2 weeks, starting <48 hours after birth. The primary outcome was death or disability. Neurodevelopmental outcomes were assessed at 18 months of age. RESULTS Complete outcome data were available for 153 infants. Nine patients dropped out during treatment, and 5 patients were lost to follow-up monitoring. Death or moderate/severe disability occurred for 35 (43.8%) of 80 infants in the control group and 18 (24.6%) of 73 infants in the erythropoietin group (P = .017) at 18 months. The primary outcomes were not different between the 2 erythropoietin doses. Subgroup analyses indicated that erythropoietin improved long-term outcomes only for infants with moderate HIE (P = .001) and not those with severe HIE (P = .227). No negative hematopoietic side effects were observed. CONCLUSION Repeated, low-dose, recombinant human erythropoietin treatment reduced the risk of disability for infants with moderate HIE, without apparent side effects.
Collapse
Affiliation(s)
- Changlian Zhu
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Robertson CL, Scafidi S, McKenna MC, Fiskum G. Mitochondrial mechanisms of cell death and neuroprotection in pediatric ischemic and traumatic brain injury. Exp Neurol 2009; 218:371-80. [PMID: 19427308 DOI: 10.1016/j.expneurol.2009.04.030] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2009] [Revised: 04/23/2009] [Accepted: 04/28/2009] [Indexed: 11/30/2022]
Abstract
There are several forms of acute pediatric brain injury, including neonatal asphyxia, pediatric cardiac arrest with global ischemia, and head trauma, that result in devastating, lifelong neurologic impairment. The only clinical intervention that appears neuroprotective is hypothermia initiated soon after the initial injury. Evidence indicates that oxidative stress, mitochondrial dysfunction, and impaired cerebral energy metabolism contribute to the brain cell death that is responsible for much of the poor neurologic outcome from these events. Recent results obtained from both in vitro and animal models of neuronal death in the immature brain point toward several molecular mechanisms that are either induced or promoted by oxidative modification of macromolecules, including consumption of cytosolic and mitochondrial NAD(+) by poly-ADP ribose polymerase, opening of the mitochondrial inner membrane permeability transition pore, and inactivation of key, rate-limiting metabolic enzymes, e.g., the pyruvate dehydrogenase complex. In addition, the relative abundance of pro-apoptotic proteins in immature brains and neurons, and particularly within their mitochondria, predisposes these cells to the intrinsic, mitochondrial pathway of apoptosis, mediated by Bax- or Bak-triggered release of proteins into the cytosol through the mitochondrial outer membrane. Based on these pathways of cell dysfunction and death, several approaches toward neuroprotection are being investigated that show promise toward clinical translation. These strategies include minimizing oxidative stress by avoiding unnecessary hyperoxia, promoting aerobic energy metabolism by repletion of NAD(+) and by providing alternative oxidative fuels, e.g., ketone bodies, directly interfering with apoptotic pathways at the mitochondrial level, and pharmacologic induction of antioxidant and anti-inflammatory gene expression.
Collapse
Affiliation(s)
- Courtney L Robertson
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, 21201, USA
| | | | | | | |
Collapse
|
23
|
Gwak M, Park P, Kim K, Lim K, Jeong S, Baek C, Lee J. The effects of dantrolene on hypoxic-ischemic injury in the neonatal rat brain. Anesth Analg 2008; 106:227-33, table of contents. [PMID: 18165582 DOI: 10.1213/01.ane.0000287663.81050.38] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The pathophysiology of brain damage from hypoxia or ischemia has been ascribed to various mechanisms and cascades. Intracellular calcium overload and a calcium excitotoxic cascade have been implicated. It has been suggested that disturbances of endoplasmic reticulum calcium homeostasis are involved in the induction of neuronal cell injury. Two types of intracellular Ca2+-release channels, involving the ryanodyne receptor and the inositol (1,4,5)-triphosphate receptor, are essential for Ca2+ signaling in cells. Dantrolene, which is used for the treatment of malignant hyperthermia syndrome, has been reported to inhibit Ca2+ release through ryanodyne receptors from the endoplasmic reticulum into the cytosol. We designed this study to investigate the neuroprotective effects of dantrolene on hypoxic-ischemic brain damage in the neonatal rat brain. METHODS Seven-day-old Sprague-Dawley rats were assigned into two groups; control group (n = 69) and dantrolene group (n = 60). Dimethyl sulfoxide was administered intracerebroventricularly in the control group, and dantrolene in dimethyl sulfoxide was similarly administered to the dantrolene group, before hypoxic-ischemic brain injury (HII). HII was induced by the ligation of the common carotid artery under isoflurane anesthesia, followed by exposure to about 2.5 h of hypoxia (oxygen concentration was maintained at 7%-8%). 1H magnetic resonance spectroscopy was performed 1 day after HII. This noninvasive method evaluated apoptotic processes in the brain after HII. Morphologic score analyses and the calculated percentage of infarct areas after 2,3,5-triphenyltetrazolium chloride staining 14 days after HII were also used to evaluate the effects of dantrolene on HII. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL) staining was performed 1 day after HII using 24 more rats. RESULTS The lipid/creatine ratios in the right hemispheres in the dantrolene group 1 day after HII were significantly lower than those of the control group (P < 0.05). There was no significant difference between the two groups in the N-acetylaspartate/creatine ratios. The gross morphologic scores were lower in the dantrolene group than in the control group (P < 0.05), and infarct area (%) after 2,3,5-triphenyltetrazolium chloride staining was less in the dantrolene group than in the control group (P < 0.05) 14 days after HII. Further work with 24 rats showed no significant difference, however, in the number of TUNEL positive cells on the two groups. CONCLUSIONS Our results show that dantrolene, administered intracerebroventricularly before HII, had a neuroprotective effect in HII model of the neonatal rat brain.
Collapse
Affiliation(s)
- Mijeung Gwak
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, College of Medicine, Ulsan University, Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
24
|
|
25
|
Yesilirmak DC, Kumral A, Tugyan K, Cilaker S, Baskin H, Yilmaz O, Duman N, Ozkan H. Effects of activated protein C on neonatal hypoxic ischemic brain injury. Brain Res 2008; 1210:56-62. [DOI: 10.1016/j.brainres.2008.02.088] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Revised: 02/22/2008] [Accepted: 02/25/2008] [Indexed: 11/28/2022]
|
26
|
Moeller-Ehrlich K, Ludlow M, Beschorner R, Meyermann R, Rima BK, Duprex WP, Niewiesk S, Schneider-Schaulies J. Two functionally linked amino acids in the stem 2 region of measles virus haemagglutinin determine infectivity and virulence in the rodent central nervous system. J Gen Virol 2007; 88:3112-3120. [PMID: 17947537 DOI: 10.1099/vir.0.83235-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Rodent brain-adapted measles virus (MV) strains, such as CAM/RB and recombinant MVs based on the Edmonston strain containing the haemagglutinin (H) of CAM/RB, cause acute encephalitis after intracerebral infection of newborn rodents. We have demonstrated that rodent neurovirulence is modulated by two mutations at amino acid positions 195 and 200 in the H protein, one of these positions (200) being a potential glycosylation site. In order to analyse the effects of specific amino acids at these positions, we introduced a range of individual and combined mutations into the open reading frame of the H gene to generate a number of eukaryotic expression plasmids. The functionality of the mutant H proteins was assessed in transfected cells and by generating recombinant viruses. Interestingly, viruses caused acute encephalitis only if the amino acid Ser at position 200 was coupled with Gly at position 195, whereas viruses with single or combined mutations at these positions, including glycosylation at position 200, were attenuated. Neurovirulence was associated with virus spread and induction of neuronal apoptosis, whereas attenuated viruses failed to infect brain cells. Similar results were obtained by using primary brain-cell cultures. Our findings indicate that a structural alteration in the stem 2 region of the H protein at position 195 or 200 interferes with infectivity of rodent neurons, and suggest that the interaction of the viral attachment protein with cellular receptors on neurons is affected.
Collapse
Affiliation(s)
- K Moeller-Ehrlich
- Institut für Virologie und Immunbiologie, University of Würzburg, D-97078 Würzburg, Germany
| | - M Ludlow
- School of Biomedical Sciences, Centre for Cancer Research and Cell Biology, The Queen's University of Belfast, Belfast BT9 7BL, UK
| | - R Beschorner
- Institut für Hirnforschung, University of Tübingen, D-72076 Tübingen, Germany
| | - R Meyermann
- Institut für Hirnforschung, University of Tübingen, D-72076 Tübingen, Germany
| | - B K Rima
- School of Biomedical Sciences, Centre for Cancer Research and Cell Biology, The Queen's University of Belfast, Belfast BT9 7BL, UK
| | - W P Duprex
- School of Biomedical Sciences, Centre for Cancer Research and Cell Biology, The Queen's University of Belfast, Belfast BT9 7BL, UK
| | - S Niewiesk
- College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210-1093, USA
| | - J Schneider-Schaulies
- Institut für Virologie und Immunbiologie, University of Würzburg, D-97078 Würzburg, Germany
| |
Collapse
|
27
|
Moxon-Lester L, Sinclair K, Burke C, Cowin GJ, Rose SE, Colditz P. Increased cerebral lactate during hypoxia may be neuroprotective in newborn piglets with intrauterine growth restriction. Brain Res 2007; 1179:79-88. [PMID: 17936737 DOI: 10.1016/j.brainres.2007.08.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Revised: 08/08/2007] [Accepted: 08/10/2007] [Indexed: 01/07/2023]
Abstract
Intrauterine growth restriction (IUGR) can increase susceptibility to perinatal hypoxic brain injury for reasons that are unknown. Previous studies of the neonatal IUGR brain have suggested that the cerebral mitochondrial capacity is reduced but the glycolytic capacity increased relative to normal weight (NW) neonates. In view of these two factors, we hypothesized that the generation of brain lactate during a mild hypoxic insult would be greater in neonatal IUGR piglets compared to NW piglets. Brain lactate/N-acetylaspartate (NAA) ratios and apparent diffusion coefficients (ADCs) were determined by proton magnetic resonance spectroscopy and imaging of the brain before, during and after hypoxia in seven neonatal piglets with asymmetric IUGR and six NW piglets. During hypoxia, IUGR piglets had significantly higher brain lactate/NAA ratios than NW piglets (P=0.046). The lactate response in the IUGR piglets correlated inversely with apoptosis in the thalamus and frontal cortex of the brain measured 4 h post hypoxia (Pearson's r=0.86, P<0.05). Apoptosis in IUGR piglets with high brain lactate was similar to that in the NW piglets whereas IUGR piglets with low brain lactate had significantly higher apoptosis than NW piglets (P=0.019). ADCs in the high lactate IUGR piglets were significantly lower during hypoxia than in all the other piglets. This signifies increased diffusion of water into brain cells during hypoxia, possibly in response to increased intracellular osmolality caused by high intracellular lactate concentrations. These findings support previous studies showing increased susceptibility to hypoxic brain injury in IUGR neonates but suggest that increased glycolysis during hypoxia confers neuroprotection in some IUGR piglets.
Collapse
Affiliation(s)
- Leith Moxon-Lester
- Perinatal Research Centre, University of Queensland, Royal Brisbane and Women's Hospital, Level 6 Ned Hanlon Building, Brisbane, Australia.
| | | | | | | | | | | |
Collapse
|
28
|
Goñi-de-Cerio F, Alvarez A, Caballero A, Mielgo VE, Alvarez FJ, Rey-Santano MC, Gastiasoro E, Valls-i-Soler A, Bilbao J, Hilario E. Early cell death in the brain of fetal preterm lambs after hypoxic-ischemic injury. Brain Res 2007; 1151:161-71. [PMID: 17418109 DOI: 10.1016/j.brainres.2007.03.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Revised: 03/02/2007] [Accepted: 03/02/2007] [Indexed: 12/12/2022]
Abstract
The objective of the present study was to evaluate using premature fetal lambs the effect of cerebral hypoxia-ischemia induced by partial occlusion of the umbilical cord on the type of cell death which occurs in different brain regions and to ascertain some of the neural pathways which may underlie the associated pathologies. Lambs were sacrificed either immediately after a 1 h hypoxic-ischemic insult or 3 h later. Brains were fixed by perfusion and blocks of the different brain territories were processed for light microscopy (hematoxylin-eosin, Nissl staining), electron transmission microscopy and quantification of apoptosis by the TUNEL method. Other fixed brains were dissociated and labeled by nonyl acridine orange to determine mitochondrial integrity. Non-fixed brains were also used for membrane asymmetry studies, in which cell suspensions were analyzed by flow cytometry to quantify apoptosis. In both hypoxic-ischemic groups, necrotic-like neurons were observed mainly in the mesencephalon, pons, deep cerebellar nuclei and basal nuclei, whereas apoptotic cells were extensively found both in white and gray matter and were not limited to regions where necrotic neurons were present. The 3 h post-partial cord occlusion group, but not the 0 h group, showed a generalized alteration of cell membrane asymmetry and mitochondrial integrity as revealed by Annexin V/PI flow cytometry and nonyl acridine orange studies, respectively. Our results show that the apoptotic/necrotic patterns of cell death occurring early after hypoxic-ischemic injury are brain-region-specific and have distinct dynamics and suggest that therapeutic strategies aimed at rescuing cells from the effects of hypoxia/ischemia should be aimed at blocking the apoptotic components of brain damage.
Collapse
Affiliation(s)
- Felipe Goñi-de-Cerio
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, E-48940 Leioa, Vizcaya, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kim JY, Yang SH, Cha SH, Kim JY, Jang YC, Park KK, Kim JK, Chung HL, Seo ES, Kim WT. The neuroprotective effect of mycophenolic acid via anti-apoptosis in perinatal hypoxic-ischemic brain injury. KOREAN JOURNAL OF PEDIATRICS 2007. [DOI: 10.3345/kjp.2007.50.7.686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Ji Young Kim
- Department of Pediatrics, School of Medicine, Catholic University of Daegu, Daegu, S. Korea
| | - Seung Ho Yang
- Department of Pediatrics, School of Medicine, Catholic University of Daegu, Daegu, S. Korea
| | - Sun Hwa Cha
- Department of Pediatrics, School of Medicine, Catholic University of Daegu, Daegu, S. Korea
| | - Ji Yeun Kim
- Department of Neurology, School of Medicine, Catholic University of Daegu, Daegu, S. Korea
| | - Young Chae Jang
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu, S. Korea
| | - Kwan Kyu Park
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu, S. Korea
| | - Jin Kyung Kim
- Department of Pediatrics, School of Medicine, Catholic University of Daegu, Daegu, S. Korea
| | - Hai Lee Chung
- Department of Pediatrics, School of Medicine, Catholic University of Daegu, Daegu, S. Korea
| | - Eok Su Seo
- Department of Opthalmology, College of Medicine, Dongguk University, Kyungju, S. Korea
| | - Woo Taek Kim
- Department of Pediatrics, School of Medicine, Catholic University of Daegu, Daegu, S. Korea
| |
Collapse
|
30
|
Billiards SS, Pierson CR, Haynes RL, Folkerth RD, Kinney HC. Is the late preterm infant more vulnerable to gray matter injury than the term infant? Clin Perinatol 2006; 33:915-33; abstract x-xi. [PMID: 17148012 DOI: 10.1016/j.clp.2006.10.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This article addresses the issue of whether the late preterm infant is more susceptible to gray matter injury induced by hypoxia-ischemia than the term infant. Although different gray matter regions display varying patterns of neuronal injury in the face of hypoxia-ischemia during advancing gestational development, little is known about the specific patterns of injury faced by the late preterm infant. This changing pattern of neuronal vulnerability with age likely reflects developmental changes of susceptibility and protective factors essential for responding to energy deprivation at the molecular, cellular, biochemical, and vascular levels. Future research involving closer examination of the late preterm period is essential to provide a greater understanding of the neuronal vulnerability in the face of hypoxic-ischemic injury.
Collapse
Affiliation(s)
- Saraid S Billiards
- Department of Pathology, Enders Building, Room 1109, Children's Hospital Boston, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
31
|
Kumral A, Yesilirmak DC, Sonmez U, Baskin H, Tugyan K, Yilmaz O, Genc S, Gokmen N, Genc K, Duman N, Ozkan H. Neuroprotective effect of the peptides ADNF-9 and NAP on hypoxic-ischemic brain injury in neonatal rats. Brain Res 2006; 1115:169-78. [PMID: 16938277 DOI: 10.1016/j.brainres.2006.07.114] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Revised: 07/19/2006] [Accepted: 07/20/2006] [Indexed: 11/25/2022]
Abstract
Perinatal asphyxia is an important cause of neonatal mortality and subsequent serious sequelae such as motor and cognitive deficits and seizures. Recent studies have demonstrated that short peptides derived from activity-dependent neurotrophic factor (ADNF) and activity-dependent neuroprotective protein (ADNP) are neuroprotective at femtomolar concentrations. However, the effect of these peptides on the hypoxic-ischemic brain injury model is unknown. The aim of this study is to investigate the effects of the peptides ADNF-9 and NAP on neurodegeneration and cerebral nitric oxide (NO) production in a neonatal rat model of hypoxic-ischemic brain injury. Seven-day-old Wistar Albino rat pups have been used in the study (n=42). Experimental groups in the study were: sham-operated group, ADNF-9-treated hypoxia-ischemia group, NAP-treated hypoxia-ischemia group, ADNF-9+NAP-treated hypoxia-ischemia group, and vehicle-treated group. In hypoxia-ischemia groups, left common carotid artery was ligated permanently on the seventh postnatal day. Two hours after the procedure, hypoxia (92% nitrogen and 8% oxygen) was applied for 2.5 h. ADNF-9, NAP, and ADNF-9+NAP were injected (intraperitoneally; i.p.) as a single dose immediately after the hypoxia period. Brain nitrite levels, neuronal cell death, and apoptosis were evaluated in both hemispheres (carotid ligated or nonligated) 72 h after the hypoxic-ischemic insult. Histopathological evaluation demonstrated that ADNF-9 and NAP significantly diminished number of "apoptotic cells" in the hippocampal CA1, CA2, CA3, and gyrus dentatus regions in both hemispheres (ligated and nonligated). When compared with vehicle-treated group, combination treatment with ADNF-9+NAP did not significantly reduce "apoptotic cell death" in any of the hemispheres. ADNF-9 and NAP, when administered separately, significantly preserved the number of neurons CA1, CA2, CA3, and dentate gyrus regions of the hippocampus, when compared with vehicle-treated group. The density of the CA1, CA2, and dentate gyrus neurons was significantly higher when combination therapy with ADNF-9+NAP was used in the carotid ligated hemispheres. In the nonligated hemispheres, combination therapy preserved the number of neurons only in the CA1 and dentate gyrus regions. Brain nitrite levels were evaluated by Griess reagent and showed that hypoxic-ischemic injury caused a significant increase in NO production. Brain nitrite levels in ADNF-9+NAP-treated animals were not different in carotid ligated or nonligated hemispheres. The peptides ADNF-9 and NAP significantly decreased NO overproduction in the hypoxic-ischemic hemisphere, whereas no significant change appeared in hypoxia alone and also in the sham-operated group. These results suggest the beneficial neuroprotective effect of ADNF-9 and NAP in this model of neonatal hypoxic-ischemic brain injury. To our knowledge, this is the first study that demonstrates a protective effect of these peptides against hypoxia-ischemia in the developing brain.
Collapse
Affiliation(s)
- Abdullah Kumral
- Department of Pediatrics, School of Medicine, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Burke C, Sinclair K, Cowin G, Rose S, Pat B, Gobe G, Colditz P. Intrauterine growth restriction due to uteroplacental vascular insufficiency leads to increased hypoxia-induced cerebral apoptosis in newborn piglets. Brain Res 2006; 1098:19-25. [PMID: 16808906 DOI: 10.1016/j.brainres.2006.04.129] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2005] [Revised: 04/11/2006] [Accepted: 04/12/2006] [Indexed: 11/21/2022]
Abstract
Uteroplacental vascular insufficiency in humans is a common cause of intrauterine growth restriction (IUGR) and is associated with an increased incidence of perinatal asphyxia and neurodevelopmental disorders compared to normal weight newborns. Experimental models that provide an opportunity to analyze the pathogenesis of these relationships are limited. Here, we used neonatal pigs from large litters in which there were piglets of normal birth weight (for controls) and of low birth weight (for uteroplacental vascular insufficiency). Hypoxia was induced in paired littermates by reducing the fraction of inspired oxygen to 4% for 25 min. Brain tissue was collected 4 h post-hypoxia. Cerebral levels of apoptosis were quantified morphologically and verified with caspase-3 activity and TUNEL. Expression of Bcl-2, Bcl-XL and Bax proteins was investigated using immunohistochemistry. Cellular positivity for Bcl-2 was consistently higher in the non-apoptotic white matter of the hypoxic IUGR animals compared with their littermates and reached significance at P < 0.05 in several pairs of littermates. Alterations in Bax showed a trend towards higher expression in the hypoxic IUGR littermates but rarely reached significance. The IUGR piglets showed a significantly greater amount of apoptosis in response to the hypoxia than the normal weight piglets, suggesting an increased vulnerability to apoptosis in the IUGR piglets.
Collapse
Affiliation(s)
- Christopher Burke
- Centre for Perinatal Research, School of Medicine, University of Queensland, Herston, Australia.
| | | | | | | | | | | | | |
Collapse
|
33
|
Miura S, Ishida A, Nakajima W, Ohmura A, Kawamura M, Takada G. Intraventricular ascorbic acid administration decreases hypoxic-ischemic brain injury in newborn rats. Brain Res 2006; 1095:159-66. [PMID: 16725128 DOI: 10.1016/j.brainres.2006.04.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Revised: 04/03/2006] [Accepted: 04/04/2006] [Indexed: 01/29/2023]
Abstract
Neuronal cell damage following hypoxic-ischemic (HI) brain injury is partly caused by production of free radicals and reactive oxygen species (ROS). Ascorbic acid (AA) is a potent antioxidant, which scavenges various types of ROS. Some studies have shown that it is neuroprotective, however, the issue is still controversial. In this study, we examined the effect of intraventricular AA administration on immature HI brain using the Rice-Vannucci model. After unilateral carotid artery ligation under isoflurane anesthesia, 7-day-old rat pups received varying concentrations of AA (0.04, 0.2, 1 and 5 mg/kg) by intraventricular injection and were exposed to 8% oxygen for 90 min. Vehicle controls received an equal volume of phosphate saline buffer. We assessed the neuroprotective effect of AA at 7 days post-HI. The percent brain damage measured by comparing the wet weight of the ligated side of hemisphere with that of contralateral one was reduced in both 1 and 5 mg/kg groups but not in either 0.04 or 0.2 mg/kg groups compared to vehicle controls (5 mg/kg 16.0 +/- 4.3%, 1 mg/kg 10.9 +/- 5.0%, vs. controls 36.7 +/- 3.6%, P < 0.05). Macroscopic evaluation of brain injury revealed the neuroprotective effect of AA in both 1 and 5 mg/kg groups (5 mg/kg 1.1 +/- 0.4, 1 mg/kg 0.4 +/- 0.3, vs. controls 2.9 +/- 0.3, P < 0.05). Western blots of fodrin on the ligated side also showed that AA significantly suppressed 150/145-kDa bands of fodrin breakdown products, which suggested that AA suppressed activation of calpain. Neuropathological quantitative analysis of cell death revealed that 1 mg/kg of AA injection significantly reduced the number of necrotic cells in cortex, caudate putamen, thalamus and hippocampus CA1, whereas that of apoptotic cells was only reduced in cortex. These findings show that intraventricular AA injection is neuroprotective after HI in immature rats.
Collapse
Affiliation(s)
- Shinobu Miura
- Department of Pediatrics, Akita University School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.
| | | | | | | | | | | |
Collapse
|
34
|
Takizawa Y, Takashima S, Itoh M. A histopathological study of premature and mature infants with pontosubicular neuron necrosis: neuronal cell death in perinatal brain damage. Brain Res 2006; 1095:200-6. [PMID: 16712812 DOI: 10.1016/j.brainres.2006.04.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 04/07/2006] [Accepted: 04/08/2006] [Indexed: 01/28/2023]
Abstract
Perinatal hypoxic-ischemic brain damage is a major cause of neuronal and behavior deficits, in which the onset of injury can be before, at or after birth, and the effects may be delayed. Pontosubicular neuron necrosis (PSN) is one of perinatal hypoxic-ischemic brain injury and its pathological peculiarity is neuronal apoptosis. In this study, we investigated whether apoptotic cascade of PSN used a caspase-pathway or not, and whether hypoglycemia activated apoptosis or not. Sections of the pons of PSN with and without hypoglycemia were stained using terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) and immunohistochemistry for glial fibrillary acidic protein (GFAP), Bcl-2, Bcl-x and activated caspase 3. Additionally, we performed immunoblot analysis of Bcl-2, Bcl-x and activated caspase 3. TUNEL-positive cell was closely associated with the presence of karyorrhexis. Under combination of karyorrhectic and TUNEL-positive cells, number of apoptotic cells in premature brains was significantly more than in mature brains. Hypoxic-ischemic brain injury was considered to easily lead to apoptosis in premature infants. Moreover, as this pathophysiology, caspase-pathway activation contributed to neuronal death from caspase-immunoexpression analyses. PSN with hypoglycemia showed large number of apoptotic cells and higher expression of activated caspase 3. The result may be more severe with the background of hypoglycemia and prematurity complicated by hypoxia and/or ischemia.
Collapse
Affiliation(s)
- Yuji Takizawa
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | | | | |
Collapse
|
35
|
Nurmi A, Goldsteins G, Närväinen J, Pihlaja R, Ahtoniemi T, Gröhn O, Koistinaho J. Antioxidant pyrrolidine dithiocarbamate activates Akt-GSK signaling and is neuroprotective in neonatal hypoxia-ischemia. Free Radic Biol Med 2006; 40:1776-84. [PMID: 16678015 DOI: 10.1016/j.freeradbiomed.2006.01.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2005] [Revised: 01/02/2006] [Accepted: 01/10/2006] [Indexed: 12/29/2022]
Abstract
Pyrrolidine dithiocarbamate (PDTC), an antioxidant and inhibitor of transcription factor nuclear factor kappa-B (NF-kappaB), has been reported to reduce inflammation and apoptosis. Because PDTC was recently found to protect in various models of adult brain ischemia with a wide therapeutic time window, we tested the effect of PDTC in a rodent model of neonatal hypoxia-ischemia (HI) brain injury. T2-weighed magnetic resonance imaging (T2-MRI) 7 days after the insult showed that a single PDTC (50 mg/kg) injection 2.5 h after the HI reduced the mean brain infarct size by 59%. PDTC reduced the HI-induced dephosphorylation of Akt and glycogen synthase kinase-3beta (GSK-3beta), expression of cleaved caspase-3, and nuclear translocation of NF-kappaB in the neonatal brain. PDTC targeted directly neurons, as PDTC reduced hypoxia-reoxygenation-induced cell death in pure hippocampal neuronal cultures. It is suggested that in addition to the previously indicated NF-kappaB inhibition as a protective mechanism of PDTC treatment, PDTC may reduce HI-induced brain injury at least partially by acting as an antioxidant, which reduces the Akt-GSK-3beta pathway of apoptotic cell death. The clinically approved PDTC and its analogues may be beneficial after HI insults with a reasonable time window.
Collapse
Affiliation(s)
- Antti Nurmi
- Department of Neurobiology, A.I.Virtanen Institute of Molecular Sciences, University of Kuopio, Finland
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
The possibility that hypothermia during or after resuscitation from asphyxia at birth, or cardiac arrest in adults, might reduce evolving damage has tantalized clinicians for a very long time. It is now known that severe hypoxia-ischemia may not necessarily cause immediate cell death, but can precipitate a complex biochemical cascade leading to the delayed neuronal loss. Clinically and experimentally, the key phases of injury include a latent phase after reperfusion, with initial recovery of cerebral energy metabolism but EEG suppression, followed by a secondary phase characterized by accumulation of cytotoxins, seizures, cytotoxic edema, and failure of cerebral oxidative metabolism starting 6 to 15 h post insult. Although many of the secondary processes can be injurious, they appear to be primarily epiphenomena of the 'execution' phase of cell death. Studies designed around this conceptual framework have shown that moderate cerebral hypothermia initiated as early as possible before the onset of secondary deterioration, and continued for a sufficient duration in relation to the severity of the cerebral injury, has been associated with potent, long-lasting neuroprotection in both adult and perinatal species. Two large controlled trials, one of head cooling with mild hypothermia, and one of moderate whole body cooling have demonstrated that post resuscitation cooling is generally safe in intensive care, and reduces death or disability at 18 months of age after neonatal encephalopathy. These studies, however, show that only a subset of babies seemed to benefit. The challenge for the future is to find ways of improving the effectiveness of treatment.
Collapse
Affiliation(s)
- A J Gunn
- Dept of Physiology, The University of Auckland, New Zealand.
| | | |
Collapse
|
37
|
Kumral A, Genc S, Ozer E, Yilmaz O, Gokmen N, Koroglu TF, Duman N, Genc K, Ozkan H. Erythropoietin Downregulates Bax and DP5 ProApoptotic Gene Expression in Neonatal Hypoxic-Ischemic Brain Injury. Neonatology 2006; 89:205-10. [PMID: 16319448 DOI: 10.1159/000089951] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2005] [Accepted: 09/12/2005] [Indexed: 11/19/2022]
Abstract
BACKGROUND Perinatal asphyxia is an important cause of neonatal mortality and subsequent serious sequelae such as motor and cognitive deficits and seizures. The ameliorative effect of erythropoietin (Epo) on experimental hypoxic-ischemic brain injury in neonatal rats has been recently reported. Recent studies also confirm the antiapoptotic effect of Epo in a variety of in vitro and in vivo neuronal injury models including hypoxic-ischemic brain injury. However, molecular mechanisms of Epo protection and antiapoptotic effect in this model are unclear. Epo may exert its antiapoptotic effect via the differential regulation of the expression of genes involved in the apoptotic process. OBJECTIVES Thus, in the present study, we studied the effects of systemically administered Epo on antiapoptotic (bcl-2, bcl-XL), proapoptotic (bax and DP5) gene expression following hypoxic-ischemic brain injury in neonatal rats. METHODS Seven- day-old Wistar rat pups were divided into three groups: control group (n=15), saline-treated group (n=17), and Epo-treated group (n=18). Rat pups were subjected to left carotid artery occlusion followed by 2.5 h of hypoxic exposure. Epo-treated group received an intraperitoneal injection of recombinant human Epo at a dose of 1,000 units/kg, saline-treated group received an intraperitoneal injection of saline at the same volume of Epo. Forty-eight hours after hypoxia, 3 animals in each group were killed for histopathological evaluation. To detect DNA fragmentation in cell nuclei, terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling reaction was applied. Bcl-2 and bax protein expression were also analyzed with immunohistochemistry. For reverse transcriptase-polymerase chain reaction (RT-PCR) analysis, rats were sacrificed 4, 12, and 24 h after hypoxia. Bcl-2, bcl-XL, bax, and DP5 mRNA expression were analyzed by RT-PCR. RESULTS Epo significantly prevented hypoxia-ischemia-induced bax and DP5 mRNA upregulation in brain tissue. Epo did not show any effect on bcl-XL transcription altered by injury. However, Epo reversed injury-induced downregulation in bcl-2 transcription. Modulating effects of Epo on bcl-2 and bax protein expression were also revealed by immunohistochemistry. CONCLUSIONS These results suggest that Epo exerts a neuroprotective effect against hypoxic-ischemic brain injury, at least partially, via the differential regulation of the expression of genes involved in apoptotic process.
Collapse
Affiliation(s)
- Abdullah Kumral
- Department of Pediatrics, School of Medicine, Dokuz Eylul University, Inciralti, Izmir, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Perinatal hypoxic-ischaemic injury (HII) is a significant cause of neurodevelopmental impairment and disability. Studies employing 31P magnetic resonance spectroscopy to measure phosphorus metabolites in situ in the brains of newborn infants and animals have demonstrated that transient hypoxia-ischaemia leads to a delayed disruption in cerebral energy metabolism, the magnitude of which correlates with the subsequent neurodevelopmental impairment. Prominent among the biochemical features of HII is the loss of cellular ATP, resulting in increased intracellular Na+ and Ca2+, and decreased intracellular K+. These ionic imbalances, together with a breakdown in cellular defence systems following HII, can contribute to oxidative stress with a net increase in reactive oxygen species. Subsequent damage to lipids, proteins, and DNA and inactivation of key cellular enzymes leads ultimately to cell death. Although the precise mechanisms of neuronal loss are unclear, it is now clear both apoptosis and necrosis are the significant components of cell death following HII. A number of different factors influence whether a cell will undergo apoptosis or necrosis, including the stage of development, cell type, severity of mitochondrial injury and the availability of ATP for apoptotic execution. This review will focus on some pathological mechanisms of cell death in which there is a disruption to oxidative metabolism. The first sections will discuss the process of damage to oxidative metabolism, covering the data collected both from human infants and from animal models. Following sections will deal with the molecular mechanisms that may underlie cerebral energy failure and cell death in this form of brain injury, with a particular emphasis on the role of apoptosis and mitochondria.
Collapse
Affiliation(s)
- Deanna L. Taylor
- Weston Laboratory, Division of Paediatrics, Obstetrics and Gynaecology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| | - A. David Edwards
- Weston Laboratory, Division of Paediatrics, Obstetrics and Gynaecology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| | - Huseyin Mehmet
- Weston Laboratory, Division of Paediatrics, Obstetrics and Gynaecology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| |
Collapse
|
39
|
Felderhoff-Mueser U, Taylor DL, Greenwood K, Kozma M, Stibenz D, Joashi UC, Edwards AD, Mehmet H. Fas/CD95/APO-1 can function as a death receptor for neuronal cells in vitro and in vivo and is upregulated following cerebral hypoxic-ischemic injury to the developing rat brain. Brain Pathol 2006; 10:17-29. [PMID: 10668892 PMCID: PMC8098164 DOI: 10.1111/j.1750-3639.2000.tb00239.x] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Fas/CD95/Apo-1 is a cell surface receptor that transduces apoptotic death signals following activation and has been implicated in triggering apoptosis in infected or damaged cells in disease states. Apoptosis is a major mechanism of neuronal loss following hypoxic-ischemic injury to the developing brain, although the role of Fas in this process has not been studied in detail. In the present study, we have investigated the expression and function of Fas in neuronal cells in vitro and in vivo. Fas was found to be expressed in the 14 day old rat brain, with strongest expression in the cortex, hippocampus and cerebellum. Cross-linking of Fas induced neuronal apoptosis both in neuronal PC12 cells in culture and following intracerebral injection in vivo, indicating that neuronal Fas was functional as a death receptor. This death was shown to be caspase dependent in primary neuronal cultures and was blocked by the selective caspase 8 inhibitor IETD. Finally, cerebral hypoxia-ischemia resulted in a strong lateralised upregulation of Fas in the hippocampus, that peaked six to twelve hours after the insult and was greater on the side of injury. These results suggest that Fas may be involved in neuronal apoptosis following hypoxic-ischemic injury to the developing brain.
Collapse
Affiliation(s)
- U Felderhoff-Mueser
- Division of Paediatrics, Obstetrics and Gynaecology, Imperial College of Science, Technology and Medicine, Hammersmith Hospital, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Muzumdar D, Higgins MJ, Ventureyra ECG. Intrauterine penetrating direct fetal head trauma following gunshot injury: a case report and review of the literature. Childs Nerv Syst 2006; 22:398-402. [PMID: 16096718 DOI: 10.1007/s00381-005-1200-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2005] [Indexed: 11/28/2022]
Abstract
CASE REPORT An unusual case of an intrauterine penetrating head injury due to a pellet from an airgun is described. A 28-year-old pregnant woman, at term, shot herself intravaginally with a toy BB gun. Following a spontaneous precipitous vaginal delivery, the neonate presented with persistent seizure disorder, meningitis, cerebritis, and a right parietal region scalp swelling. Imaging studies revealed intracranial hemorrhage, and the metallic pellet was adjacent to the right lateral ventricle, which was removed through a parietal craniotomy. Computed tomography of the brain after 1 week demonstrated early abscess formation in the left frontal operculum and a subdural empyema in the posterior fossa. The abscesses were evacuated, and the meningitis was treated vigorously with broad-spectrum antibiotics. Although well for the past 6 years, the child demonstrates significant mental handicap and developmental delay. DISCUSSION The pathogenesis, management, and medicolegal issues pertaining to the above case are discussed, and the relevant literature is briefly reviewed.
Collapse
Affiliation(s)
- Dattatraya Muzumdar
- Division of Neurosurgery, Children's Hospital of Eastern Ontario, 401 Smyth Road, Ottawa, Ontario, K1H 8 L1, Canada
| | | | | |
Collapse
|
41
|
Castillo-Meléndez M, Yan E, Walker DW. Expression of erythropoietin and its receptor in the brain of late-gestation fetal sheep, and responses to asphyxia caused by umbilical cord occlusion. Dev Neurosci 2005; 27:220-7. [PMID: 16046857 DOI: 10.1159/000085995] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2004] [Accepted: 12/20/2004] [Indexed: 11/19/2022] Open
Abstract
Asphyxia and hypoxia are common threats faced by the fetus in utero. In late-gestation fetal sheep, asphyxia produced by umbilical cord occlusion (UCO) results in widespread lipid peroxidation and apoptosis. Adaptive mechanisms that might limit fetal brain damage include induction of the hemopoietic cytokine, erythropoietin (EPO). In unanesthetized fetal sheep, we investigated if 1 or 2 bouts of brief asphyxia (UCO for 10 min) induced EPO and EPO type I receptor (EPO-R) expressions, with the second UCO repeated 48 h after the first. Fetal brains were recovered 48 h after either sham, 1 x or 2 x UCO at 129-133 (term approximately 147) days of gestation and prepared for immunocytochemistry. In age-matched control brain, low levels of EPO and EPO-R proteins were present in oligodendrocytes (OLs), periventricular and cortical white matter (WM), with no EPO and very low EPO-R expression in neurons. After 1 x UCO, EPO and EPO-R expressions were increased in astrocytes (periventricular and cortical WM, striatum, corpus callosum), choroid plexus epithelial cells, scattered neurons in cortical layers IV-VI, hippocampal CA1 neurons, and in the molecular and granule layers of the cerebellum. After 2 x UCO, higher levels of EPO and EPO-R occurred in the periventricular and cortical WM, corpus callosum, hippocampal CA1, and in neurons of all cortical layers. Paradoxically, EPO and EPO-R were now lower in hippocampal CA1 neurons and cerebellar molecular and granule cell layers. Few OLs expressed EPO or EPO-R after 1 x or 2 x UCO. Thus, brief asphyxia induces EPO and EPO-R in fetal astrocytes, but only after repeated asphyxial insult in neurons. Whether this is a response to increased injury, or represents an adaptive response that limits further cell death and brain damage awaits further investigation.
Collapse
|
42
|
Burke C, Gobe G. Pontosubicular apoptosis (“necrosis”) in human neonates with intrauterine growth retardation and placental infarction. Virchows Arch 2005; 446:640-5. [PMID: 15838644 DOI: 10.1007/s00428-005-1251-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2005] [Accepted: 03/04/2005] [Indexed: 10/25/2022]
Abstract
In a previous study of 37 autopsied stillbirths with non-dysmorphic intrauterine growth retardation (IUGR), 26 cases were associated with placental infarction, a morphologic marker of uteroplacental insufficiency. Nine of the 26 cases with both IUGR and placental infarction, where archival tissue was available, had grey matter ischaemic lesions that were subsequently identified as "pontosubicular necrosis". This lesion is now regarded as a localized form of apoptosis. A further eight third trimester stillbirth cases with both IUGR and placental infarction were ascertained prospectively. Sixteen of these 17 cases showed pontosubicular apoptosis, identified morphologically and verified using activated caspase-3 and TUNEL. Five of the 17 cases showed apoptosis in the frontal or temporal cortex as well. In this current study, pontosubicular apoptosis was strongly associated with IUGR and placental infarction in third trimester stillborns, suggesting that uteroplacental insufficiency leading to chronic fetal hypoxaemia may cause cerebral apoptosis.
Collapse
|
43
|
Mahan VL, Ilangovan S, Cuison R, Patil J, Dockter S, Rizzo V, Ilbawi M. Does antegrade cerebral perfusion protect the brain during deep hypothermic circulatory arrest? J Pediatr Surg 2005; 40:510-5. [PMID: 15793727 DOI: 10.1016/j.jpedsurg.2004.11.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE This study compares cerebral protection using no cerebroplegia and using antegrade cerebroplegia with variable flow rates during deep hypothermic circulatory arrest (DHCA). METHODS Twenty healthy neonatal piglets (2.5-3.8 kg) underwent 60 minutes of DHCA. No cerebroplegia was used in group 1 (n = 5). Cold (16 degrees C) antegrade cerebral perfusate was administered through the innominate artery at 10 mL/kg per minute in group 2 (n = 5), at 25 mL/kg per minute in group 3 (n = 5), and at 50 mL/kg per minute in group 4 (n = 5). Venous samples for lactate, pyruvate, S-100B protein, and creatine kinase BB (CKBB) were drawn from the jugular vein before and after discontinuation of cardiopulmonary bypass--lactate at 5 minutes postbypass, pyruvate at 5 minutes postbypass, S-100B protein at 30 minutes postbypass, and CKBB at 6 hours postbypass. Piglets were killed 6 hours postbypass and their brains were harvested for histological/immunologic studies. Extent of damage was assessed using a semiquantitative score of 0 to 4 based on a validated method. RESULTS Evidence for significant apoptosis and necrosis was apparent in all groups. The mean H&E score was 2.2 for group 1, 2.3 for group 2, 2.5 for group 3, and 2.3 for group 4. The mean terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling score was 1.0 for group 1, 1.2 for group 2, 1.7 for group 3, and 0.8 for group 4. Pathological changes were not greater in the piglets that did not have antegrade cerebral perfusion. Serum lactate, pyruvate, S-100B protein, and CKBB did not distinguish between perfusion strategies. CONCLUSIONS In neonates, unmodified antegrade cerebral perfusion at flow rates of 10, 25, and 50 mL/kg per minute during DHCA does not provide additional protection of the brain as determined by histology, immunology, serum lactate, pyruvate, S-100B protein, and CKBB.
Collapse
Affiliation(s)
- Vicki L Mahan
- Department of Pediatric Cardiothoracic Surgery, Heart Institute for Children at Hope Children's Hospital, Oak Lawn, IL, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Bell JE, Becher JC, Wyatt B, Keeling JW, McIntosh N. Brain damage and axonal injury in a Scottish cohort of neonatal deaths. ACTA ACUST UNITED AC 2005; 128:1070-81. [PMID: 15705606 DOI: 10.1093/brain/awh436] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Despite the clinical and medicolegal significance attached to perinatal asphyxia, the neuropathological basis of this condition remains obscure. There are very few studies in the literature which correlate the pathological findings in neonatal brains with detailed epidemiological data, and none which are population based. In a Scotland-wide study of neonatal deaths, 70 brains have been examined. On the basis of glial and macrophage reactions, we previously identified infants with putative antepartum brain damage in this cohort and have related these reactions to signs of birth asphyxia. The present study explores the extent of neuronal/axonal injury in these infants since this is likely to be the basis for neurological deficits in surviving infants. We have also investigated these brains for beta-amyloid precursor protein (betaAPP) positivity to determine whether this is a useful marker of neuronal injury in neonates. Neuronal eosinophilia and karyorrhexes were detected in 43% and 27% of the cohort, respectively; maximally in the subiculum and ventral pons, but often present elsewhere. White matter damage was detected in 24% of cases but without classic cystic lesions of periventricular leucomalacia. betaAPP positivity was present in neuronal soma in 52% of cases and, in axons, in 27% of cases, and was seen from as early as 25-weeks gestation. Axonal bulbs were clearly delineated by betaAPP positivity and were usually located in the cerebral white matter and internal capsule, and infrequently in the brain stem. Although white matter damage and betaAPP axonal positivity were often detected in the same cases (P = 0.034), these features also occurred independently of each other. Both neuronal karyorrhexes and white matter betaAPP positivity were significantly correlated with the features of birth asphyxia, particularly a history of seizures. Immunocytochemistry for both betaAPP and glial fibrillary acidic protein proved useful in detecting neuropathological features which escaped detection on routine examination, particularly in preterm infants. The presence together of recent and older damage in individual brains suggests that there is an ongoing neuronal response to cerebral insults. We find that betaAPP is a useful marker of white matter damage in the neonatal brain. Immunopositivity for betaAPP in these circumstances is not attributable to inflicted or accidental trauma. While birth-related trauma cannot be ruled out, hypoxia/ischaemia is a likely cause in these infants. However, the exact pathogenesis of neuronal/axonal injury in the neonatal brain remains unclear.
Collapse
Affiliation(s)
- J E Bell
- Division of Pathology, University of Edinburgh, Edinburgh, UK.
| | | | | | | | | |
Collapse
|
45
|
Abstract
Parents and clinicians concerned about high-risk infants and children with motor delay or cerebral palsy seek information on cause, treatment, prognosis, and recurrence risk. Used in combination with history and examination, neuroimaging studies can improve diagnosis and management. In premature infants, cranial ultrasound is a reliable, noninvasive diagnostic modality. Nuclear magnetic resonance techniques including magnetic resonance imaging and diffusion weighted imaging can be used effectively in neonatal encephalopathies. In children with motor delay and cerebral palsy syndromes including spastic diplegia, quadriplegia, hemiplegia, and extrapyramidal movement disorders, conventional magnetic resonance imaging has become an important determinant of diagnosis and management. The aim of this article is to help clinicians select and interpret imaging studies of benefit in clinical care.
Collapse
Affiliation(s)
- Jennifer Accardo
- Johns Hopkins University School of Medicine, and the Kennedy Krieger Institute, Division of Neurology and Developmental Medicine, Baltimore, Maryland 21205, USA
| | | | | |
Collapse
|
46
|
Roelfsema V, Bennet L, George S, Wu D, Guan J, Veerman M, Gunn AJ. Window of opportunity of cerebral hypothermia for postischemic white matter injury in the near-term fetal sheep. J Cereb Blood Flow Metab 2004; 24:877-86. [PMID: 15362718 DOI: 10.1097/01.wcb.0000123904.17746.92] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Postresuscitation cerebral hypothermia is consistently neuroprotective in experimental preparations; however, its effects on white matter injury are poorly understood. Using a model of reversible cerebral ischemia in unanesthetized near-term fetal sheep, we examined the effects of cerebral hypothermia (fetal extradural temperature reduced from 39.4 +/- 0.1 degrees C to between 30 and 33 degrees C), induced at different times after reperfusion and continued for 72 hours after ischemia, on injury in the parasagittal white matter 5 days after ischemia. Cooling started within 90 minutes of reperfusion was associated with a significant increase in bioactive oligodendrocytes in the intragyral white matter compared with sham cooling (41 +/- 20 vs 18 +/- 11 per field, P < 0.05), increased myelin basic protein density and reduced expression of activated caspase-3 (14 +/- 12 vs 91 +/- 51, P < 0.05). Reactive microglia were profoundly suppressed compared with sham cooling (4 +/- 6 vs 38 +/- 18 per field, P < 0.05) with no effect on numbers of astrocytes. When cooling was delayed until 5.5 hours after reperfusion there was no significant effect on loss of oligodendrocytes (24 +/- 12 per field). In conclusion, hypothermia can effectively protect white matter after ischemia, but only if initiated early after the insult. Protection was closely associated with reduced expression of both activated caspase-3 and of reactive microglia.
Collapse
Affiliation(s)
- Vincent Roelfsema
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
47
|
Aito H, Aalto KT, Raivio KO. Adenine nucleotide metabolism and cell fate after oxidant exposure of rat cortical neurons: effects of inhibition of poly(ADP-ribose) polymerase. Brain Res 2004; 1013:117-24. [PMID: 15196974 DOI: 10.1016/j.brainres.2004.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2004] [Indexed: 12/01/2022]
Abstract
We exposed cultured neurons prelabeled with 14C-adenine to H2O2 with or without the poly(ADP-ribose) polymerase (PARP) inhibitor 3,4-Dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone (DPQ) to quantify its effects on acute ATP depletion, later ATP synthesis, cellular and nuclear morphology, extent of DNA fragmentation, and PARP cleavage. According to the extent of the acute ATP depletion, the exposures were classified as 'mild' (50 microM H2O2), 'moderate' (100-250 microM H2O2), or 'severe' (500 microM-1 mM H2O2) insults. Mild exposure had no significant effects on the parameters studied. In the 'moderately' exposed neurons, ATP depletion to 59+/-6% of control was associated with a decrease in the cell counts, apoptotic morphology, and cleavage of PARP. In this group, DPQ prevented the acute ATP (to 95+/-15% of control), preserved cell morphology, and improved cell survival. In the 'severe' group, ATP depletion to 18+/-4% was associated with necrosis and intact PARP. DPQ elevated ATP levels (to 44+/-12% of control) and post-insult ATP synthesis, improved cell counts, and altered cell morphology towards apoptosis rather than necrosis. Post-insult application of DPQ was less effective. Our results show that the extent of oxidant-induced ATP depletion and cell fate can be modified by PARP inhibition, to some extent also after the insult.
Collapse
Affiliation(s)
- Henrikka Aito
- Research Laboratory, Hospital for Children and Adolescents, University of Helsinki, Biomedicum Helsinki, V Floor, Room B524b, P.O. Box 700, FIN-00029 HUS, Helsinki, Finland.
| | | | | |
Collapse
|
48
|
|
49
|
Joly LM, Mucignat V, Mariani J, Plotkine M, Charriaut-Marlangue C. Caspase inhibition after neonatal ischemia in the rat brain. J Cereb Blood Flow Metab 2004; 24:124-31. [PMID: 14688624 DOI: 10.1097/01.wcb.0000100061.36077.5f] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Caspase-3 has been identified as a key protease in the execution of apoptosis and appears to be an important downstream event after hypoxia-ischemia in the immature brain. The efficacy of a pan-caspase inhibitor, boc-aspartyl-(Ome)-fluoromethyl-ketone (BAF), was tested in a model of unilateral focal ischemia with reperfusion in 7-day-old rats. The BAF inhibitor was given intraperitoneally 5 minutes before reperfusion via the carotid artery. This procedure reduced the activity of caspase-3 by 79% but did not induce a significant reduction in infarct volume (23.8 +/- 7.5% versus 30.1 +/- 6.4%). Animals were distributed in two populations. One population exhibited an infarct, whereas the other appeared to be fully protected. BAF-treated animals exhibiting an infarct mostly displayed necrotic cell death, whereas apoptotic nuclei were observed in untreated or vehicle-treated animals. Repeated dose of BAF (5 minutes before and 9 hours after reperfusion) did not also provide benefit after neonatal ischemia, although a general trend to reduce lesion was observed (20.5 +/- 3.7% versus 34.4 +/- 5.9%). These findings raise critical questions about the use of peptide ketone apoptotic inhibitors in improving histopathologic outcomes after neonatal stroke.
Collapse
Affiliation(s)
- Luc-Marie Joly
- UMR-CNRS 7102, Laboratoire Developpement et Vieillissement du Système Nerveux, Paris, France
| | | | | | | | | |
Collapse
|
50
|
Hargitai B, Szabó V, Cziniel M, Hajdú J, Papp Z, Szende B, Sergi C. Human brain of preterm infants after hypoxic-ischaemic injuries: no evidence of a substantial role for apoptosis by using a fine-tuned ultrasound-guided neuropathological analysis. Brain Dev 2004; 26:30-36. [PMID: 14729412 DOI: 10.1016/s0387-7604(03)00091-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Preterm birth may be associated with hypoxic-ischaemic encephalopathy (HIE) showing a well recognised number of patterns, including neuronal karyorrhexis/eosinophilia mostly at the diencephalon and brain stem and leukomalacia at the periventricular white matter. To investigate whether programmed cell death or apoptosis plays a role in HIE, we examined human brains of preterm infants. Brain tissue samples from 12 consecutive infants (24-34 weeks of gestation) were available at post-mortem examination (1998-2000) after approval of the Ethics Committee. Two tissue sections were stereologically localised after brain fixation, slice preparation, and comparison with ultrasound imaging. We studied the periventricular white matter and the corresponding cortical region in each brain. Conventional histological stains were used. In addition, apoptosis was detected using a neuronal-specific terminal deoxynucleotidyl transferase-mediated nick end-labelling (TUNEL) method (NeuroTACS). A semiquantitative evaluation was performed to compare regions close to brain lesions with injury-free areas. Neuronal apoptosis was low in both cortical and in periventricular regions. No glial apoptosis was detected. Apoptosis in neurones was, however, detected in preterm brains with bacterial or mycotic infection. These results point out to the ambiguity of the TUNEL-reactive neurons in the diseased premature infants using fine-tuned ultrasound-guided neuropathological analysis, support the probable coexistence of neuronal TUNEL-reactivity and infection, and suggest that the association between apoptosis and HIE should overall be viewed with more caution.
Collapse
Affiliation(s)
- Beáta Hargitai
- 1st Institute of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Ulloi út 26. H-1085, Hungary
| | | | | | | | | | | | | |
Collapse
|