1
|
Di Martino E, Rayasam A, Vexler ZS. Brain Maturation as a Fundamental Factor in Immune-Neurovascular Interactions in Stroke. Transl Stroke Res 2024; 15:69-86. [PMID: 36705821 PMCID: PMC10796425 DOI: 10.1007/s12975-022-01111-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 01/28/2023]
Abstract
Injuries in the developing brain cause significant long-term neurological deficits. Emerging clinical and preclinical data have demonstrated that the pathophysiology of neonatal and childhood stroke share similar mechanisms that regulate brain damage, but also have distinct molecular signatures and cellular pathways. The focus of this review is on two different diseases-neonatal and childhood stroke-with emphasis on similarities and distinctions identified thus far in rodent models of these diseases. This includes the susceptibility of distinct cell types to brain injury with particular emphasis on the role of resident and peripheral immune populations in modulating stroke outcome. Furthermore, we discuss some of the most recent and relevant findings in relation to the immune-neurovascular crosstalk and how the influence of inflammatory mediators is dependent on specific brain maturation stages. Finally, we comment on the current state of treatments geared toward inducing neuroprotection and promoting brain repair after injury and highlight that future prophylactic and therapeutic strategies for stroke should be age-specific and consider gender differences in order to achieve optimal translational success.
Collapse
Affiliation(s)
- Elena Di Martino
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Aditya Rayasam
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
2
|
Barks JD, Liu Y, Dopp IA, Silverstein FS. Azithromycin reduces inflammation-amplified hypoxic-ischemic brain injury in neonatal rats. Pediatr Res 2022; 92:415-423. [PMID: 34625655 PMCID: PMC8989723 DOI: 10.1038/s41390-021-01747-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/07/2021] [Accepted: 06/16/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Systemic inflammation amplifies neonatal hypoxic-ischemic (HI) brain injury. Azithromycin (AZ), an antibiotic with anti-inflammatory properties, improves sensorimotor function and reduces tissue damage after neonatal rat HI brain injury. The objective of this study was to determine if AZ is neuroprotective in two neonatal rat models of inflammation-amplified HI brain injury. DESIGN/METHODS Seven-day-old (P7) rats received injections of toll-like receptor agonists lipopolysaccharide (LPS) or Pam3Cys-Ser-(Lys)4 (PAM) prior to right carotid ligation followed by 50 min (LPS + HI) or 60 min (PAM + HI) in 8% oxygen. Outcomes included contralateral forelimb function (forepaw placing; grip strength), survival, %Intact right hemisphere (brain damage), and a composite score incorporating these measures. We compared postnatal day 35 outcomes in controls and groups treated with three or five AZ doses. Then, we compared P21 outcomes when the first (of five) AZ doses were administered 1, 2, or 4 h after HI. RESULTS In both LPS + HI and PAM + HI models, AZ improved sensorimotor function, survival, brain tissue preservation, and composite scores. Benefits increased with five- vs. three-dose AZ and declined with longer initiation delay. CONCLUSIONS Perinatal systemic infection is a common comorbidity of neonatal asphyxia brain injury and contributes to adverse outcomes. These data support further evaluation of AZ as a candidate treatment for neonatal neuroprotection. IMPACT AZ treatment decreases sensorimotor impairment and severity of brain injury, and improves survival, after inflammation-amplified HI brain injury, and this can be achieved even with a 2 h delay in initiation. This neuroprotective benefit is seen in models of inflammation priming by both Gram-negative and Gram-positive infections. This extends our previous findings that AZ treatment is neuroprotective after HI brain injury in neonatal rats.
Collapse
Affiliation(s)
- John D.E. Barks
- Department of Pediatrics, University of Michigan Medical School, The University of Michigan, Ann Arbor, MI
| | - Yiqing Liu
- Department of Pediatrics, University of Michigan Medical School, The University of Michigan, Ann Arbor, MI
| | - Ian A. Dopp
- Department of Pediatrics, University of Michigan Medical School, The University of Michigan, Ann Arbor, MI
| | - Faye S. Silverstein
- Department of Pediatrics, University of Michigan Medical School, The University of Michigan, Ann Arbor, MI,Department of Neurology, University of Michigan Medical School, The University of Michigan, Ann Arbor, MI
| |
Collapse
|
3
|
Yazdani A, Howidi B, Shi MZ, Tugarinov N, Khoja Z, Wintermark P. Sildenafil improves hippocampal brain injuries and restores neuronal development after neonatal hypoxia-ischemia in male rat pups. Sci Rep 2021; 11:22046. [PMID: 34764335 PMCID: PMC8586032 DOI: 10.1038/s41598-021-01097-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/08/2021] [Indexed: 02/07/2023] Open
Abstract
The hippocampus is a fundamental structure of the brain that plays an important role in neurodevelopment and is very sensitive to hypoxia-ischemia (HI). The purpose of this study was to investigate the effects of sildenafil on neonatal hippocampal brain injuries resulting from HI, and on neuronal development in this context. HI was induced in male Long-Evans rat pups at postnatal day 10 (P10) by a left common carotid ligation followed by a 2-h exposure to 8% oxygen. Rat pups were randomized to vehicle or sildenafil given orally twice daily for 7 days starting 12 h after HI. Hematoxylin and eosin staining was performed at P30 to measure the surface of the hippocampus; immunohistochemistry was performed to stain neurons, oligodendrocytes, and glial cells in the hippocampus. Western blots of the hippocampus were performed at P12, P17, and P30 to study the expression of neuronal markers and mTOR pathway. HI caused significant hippocampal atrophy and a significant reduction of the number of mature neurons, and induced reactive astrocytosis and microgliosis in the hippocampus. HI increased apoptosis and caused significant dysregulation of the normal neuronal development program. Treatment with sildenafil preserved the gross morphology of the hippocampus, reverted the number of mature neurons to levels comparable to sham rats, significantly increased both the immature and mature oligodendrocytes, and significantly reduced the number of microglia and astrocytes. Sildenafil also decreased apoptosis and reestablished the normal progression of post-natal neuronal development. The PI3K/Akt/mTOR pathway, whose activity was decreased after HI in the hippocampus, and restored after sildenafil treatment, may be involved. Sildenafil may have both neuroprotective and neurorestorative properties in the neonatal hippocampus following HI.
Collapse
Affiliation(s)
- Armin Yazdani
- grid.63984.300000 0000 9064 4811Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Belal Howidi
- grid.63984.300000 0000 9064 4811Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Meng Zhu Shi
- grid.63984.300000 0000 9064 4811Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Nicol Tugarinov
- grid.63984.300000 0000 9064 4811Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Zehra Khoja
- grid.63984.300000 0000 9064 4811Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Pia Wintermark
- Research Institute of the McGill University Health Centre, Montreal, Canada. .,Division of Newborn Medicine, Department of Pediatrics, Montreal Children's Hospital, 1001 boul. Décarie, Site Glen Block E, EM0.3244, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
4
|
Faillot M, Chaillet A, Palfi S, Senova S. Rodent models used in preclinical studies of deep brain stimulation to rescue memory deficits. Neurosci Biobehav Rev 2021; 130:410-432. [PMID: 34437937 DOI: 10.1016/j.neubiorev.2021.08.012] [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: 02/08/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
Deep brain stimulation paradigms might be used to treat memory disorders in patients with stroke or traumatic brain injury. However, proof of concept studies in animal models are needed before clinical translation. We propose here a comprehensive review of rodent models for Traumatic Brain Injury and Stroke. We systematically review the histological, behavioral and electrophysiological features of each model and identify those that are the most relevant for translational research.
Collapse
Affiliation(s)
- Matthieu Faillot
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Antoine Chaillet
- Laboratoire des Signaux et Systèmes (L2S-UMR8506) - CentraleSupélec, Université Paris Saclay, Institut Universitaire de France, France
| | - Stéphane Palfi
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Suhan Senova
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France.
| |
Collapse
|
5
|
Huang A, Jia L. Crocin enhances hypothermia therapy in hypoxic ischemia-induced brain injury in mice. Acta Neurol Belg 2021; 121:429-436. [PMID: 31367946 DOI: 10.1007/s13760-019-01198-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a serious medical situation at labor which leads to severe brain damage. Hypothermia therapy is the standard treatment for infants with HIE, but the efficacy is limited. Combination treatments are considered to enhance the efficacy of hypothermia. Crocin is an extract from saffron which has anti-inflammatory, anti-oxidant, and neuroprotective properties. The present study sought to investigate whether crocin could act as a combined treatment with hypothermia in a mouse model of HIE. C57BL/6J mice at post-natal day 7 were subjected to left common carotid artery ligation, followed by treatment of crocin (10 mg/kg) and hypothermia, either alone or in combination. Brain edema and tissue infarct were measured to evaluate brain damage. Mediators involved in inflammatory response and oxidative stress were measured. Neurological severity score test was performed to evaluate the functional outcome. Results show that crocin treatment alone could reduce inflammation and brain damage after hypoxia-ischemia. Combined treatment of crocin and hypothermia exerted enhanced therapeutic effect compared with single treatment, resulting in significantly less brain damage, reduced inflammatory and oxidative responses, and improved functional outcome. Together, these data suggest that crocin plays a beneficial effect in the mouse model of HIE. It could also enhance the neuroprotective effect of hypothermia and might be considered as a combination therapeutic treatment with hypothermia in HIE.
Collapse
|
6
|
Laštůvka Z, Borbélyová V, Janišová K, Otáhal J, Mysliveček J, Riljak V. Neonatal hypoxic-ischemic brain injury leads to sex-specific deficits in rearing and climbing in adult mice. Physiol Res 2020; 69:S499-S512. [PMID: 33476172 DOI: 10.33549/physiolres.934604] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The study examined the morphological and long-term behavioral impacts of neonatal hypoxic-ischemic brain injury in a mouse model. We investigated the modification of different behavioral domains, such as spontaneous climbing, which represents fine motor skills. We also focused on sex-dependent differences during hypoxic-ischemic encephalopathy. The Rice-Vannucci model of hypoxia-ischemia was used, adjusted and adapted to 7-day-old C57BL/6NTac mice. The effects of induced hypoxia and ischemia were also studied separately. At postnatal day 60, mice underwent behavioral testing using the LABORAS apparatus. The perfusion for histological evaluation was performed one day after the behavioral analyses. In groups with separately induced hypoxia or ischemia, the observed alterations in behavior were not accompanied by morphological changes in the cortex or hippocampal formation. Female mice naturally climbed significantly more and hypoxic females reared less than hypoxic males (p<0.05). Male mice postnatally exposed to hypoxia-ischemia exhibited significantly lower vertical activity and higher horizontal activity (p<0.05). Mild hypoxic damage may not be morphologically detectable but may induce substantial behavioral changes in adult mice. There were significant differences between horizontal and vertical activity in reaction to hypoxia-ischemia. Our study indicates that the importance of behavioral testing is irreplaceable and may be reflected in neonatal medicine.
Collapse
Affiliation(s)
- Z Laštůvka
- Institute of Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
7
|
Emili M, Guidi S, Uguagliati B, Giacomini A, Bartesaghi R, Stagni F. Treatment with the flavonoid 7,8-Dihydroxyflavone: a promising strategy for a constellation of body and brain disorders. Crit Rev Food Sci Nutr 2020; 62:13-50. [DOI: 10.1080/10408398.2020.1810625] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| |
Collapse
|
8
|
Hamdy N, Eide S, Sun HS, Feng ZP. Animal models for neonatal brain injury induced by hypoxic ischemic conditions in rodents. Exp Neurol 2020; 334:113457. [PMID: 32889009 DOI: 10.1016/j.expneurol.2020.113457] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023]
Abstract
Neonatal hypoxia-ischemia and resulting encephalopathies are of significant concern. Intrapartum asphyxia is a leading cause of neonatal death globally. Among surviving infants, there remains a high incidence of hypoxic-ischemic encephalopathy due to neonatal hypoxic-ischemic brain injury, manifesting as mild conditions including attention deficit hyperactivity disorder, and debilitating disorders such as cerebral palsy. Various animal models of neonatal hypoxic brain injury have been implemented to explore cellular and molecular mechanisms, assess the potential of novel therapeutic strategies, and characterize the functional and behavioural correlates of injury. Each of the animal models has individual advantages and limitations. The present review looks at several widely-used and alternative rodent models of neonatal hypoxia and hypoxia-ischemia; it highlights their strengths and limitations, and their potential for continued and improved use.
Collapse
Affiliation(s)
- Nancy Hamdy
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sarah Eide
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
9
|
Wood T, Nance E. Disease-directed engineering for physiology-driven treatment interventions in neurological disorders. APL Bioeng 2019; 3:040901. [PMID: 31673672 PMCID: PMC6811362 DOI: 10.1063/1.5117299] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023] Open
Abstract
Neurological disease is killing us. While there have long been attempts to develop therapies for both acute and chronic neurological diseases, no current treatments are curative. Additionally, therapeutic development for neurological disease takes 15 years and often costs several billion dollars. More than 96% of these therapies will fail in late stage clinical trials. Engineering novel treatment interventions for neurological disease can improve outcomes and quality of life for millions; however, therapeutics should be designed with the underlying physiology and pathology in mind. In this perspective, we aim to unpack the importance of, and need to understand, the physiology of neurological disease. We first dive into the normal physiological considerations that should guide experimental design, and then assess the pathophysiological factors of acute and chronic neurological disease that should direct treatment design. We provide an analysis of a nanobased therapeutic intervention that proved successful in translation due to incorporation of physiology at all stages of the research process. We also provide an opinion on the importance of keeping a high-level view to designing and administering treatment interventions. Finally, we close with an implementation strategy for applying a disease-directed engineering approach. Our assessment encourages embracing the complexity of neurological disease, as well as increasing efforts to provide system-level thinking in our development of therapeutics for neurological disease.
Collapse
|
10
|
Halis H, Bitiktaş S, Baştuğ O, Tan B, Kavraal Ş, Güneş T, Süer C. Differential Effects of Pentoxifylline on Learning and Memory Impairment Induced by Hypoxic-ischemic Brain Injury in Rats. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2019; 17:388-399. [PMID: 31352705 PMCID: PMC6705102 DOI: 10.9758/cpn.2019.17.3.388] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 01/10/2023]
Abstract
Objective Hypoxic-ischemic (HI) brain injury in the human perinatal period often leads to significant long-term neurobehavioral dysfunction in the cognitive and sensory-motor domains. Using a neonatal HI injury model (unilateral carotid ligation followed by hypoxia) in postnatal day seven rats, the present study investigated the long-term effects of HI and potential behavioral protective effect of pentoxifylline. Methods Seven-day-old rats underwent right carotid ligation, followed by hypoxia (FiO2 = 0.08). Rats received pentoxifylline immediately after and again 2 hours after hypoxia (two doses, 60‒100 mg/kg/dose), or serum physiologic. Another set of seven-day-old rats was included to sham group exposed to surgical stress but not ligated. These rats were tested for spatial learning and memory on the simple place task in the Morris water maze from postnatal days 77 to 85. Results HI rats displayed significant tissue loss in the right hippocampus, as well as severe spatial memory deficits. Low-dose treatment with pentoxifylline resulted in significant protection against both HI-induced hippocampus tissue losses and spatial memory impairments. Beneficial effects are, however, negated if pentoxifylline is administered at high dose. Conclusion These findings indicate that unilateral HI brain injury in a neonatal rodent model is associated with cognitive deficits, and that low dose pentoxifylline treatment is protective against spatial memory impairment.
Collapse
Affiliation(s)
- Hülya Halis
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Soner Bitiktaş
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Osman Baştuğ
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Burak Tan
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Şehrazat Kavraal
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Tamer Güneş
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Cem Süer
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
11
|
Neurite orientation dispersion and density imaging for evaluating the severity of neonatal hypoxic-ischemic encephalopathy in rats. Magn Reson Imaging 2019; 62:214-219. [PMID: 31325487 DOI: 10.1016/j.mri.2019.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/27/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE To evaluate the utility of neurite orientation dispersion and density imaging (NODDI) for longitudinally assessing neonatal hypoxic-ischemic (HI) encephalopathy severity with 7.0 T magnetic resonance imaging. METHODS Thirteen 8-day-old Wistar rats underwent unilateral ligation of the left common carotid artery followed by mild (1 h; n = 6) or severe (2 h; n = 7) hypoxic exposure (8% O2, 34 °C). Diffusion-weighted, T2-weighted (T2W), and flow-sensitive alternating inversion recovery images were obtained with a horizontal 7.0 T scanner at 1, 24, 72, and 168 h after HI insult. The fractional anisotropy (FA), apparent diffusion coefficient (ADC), intracellular volume fraction (ICVF), isotropic volume fraction (ISO), orientation dispersion index (ODI), and cerebral blood flow (CBF) values were calculated for each group (mild and severe) at each time point (1, 24, 72, and 168 h). ICVF, ISO, and ODI were the NODDI parameters. RESULTS Left hemisphere brain damage was identified as slight hyperintensity on T2W images after 1 h in both groups. In the severe group only, the signal hyperintensity increased time-dependently over 168 h. The ADC and CBF were not significantly different between the groups within any region. The ICVF and ODI were significantly higher in the severe vs. mild group at various points between 1 and 168 h (cortex, striatum, or white matter), whereas the FA was significantly higher in the mild vs. severe group at 168 h (cortex and white matter). The ISO was higher in the severe vs. mild group at 72 h (striatum) and 168 h (all regions), while the ISO was significantly higher in the mild vs. severe group at 24 h (all regions). CONCLUSION Here, ODI, a NODDI metric, identified early differences between mild and severe HI injuries. Our findings support the potential utility of NODDI for determining neonatal HI encephalopathy severity in rats.
Collapse
|
12
|
Kim YE, Sung SI, Chang YS, Ahn SY, Sung DK, Park WS. Thrombin Preconditioning Enhances Therapeutic Efficacy of Human Wharton's Jelly-Derived Mesenchymal Stem Cells in Severe Neonatal Hypoxic Ischemic Encephalopathy. Int J Mol Sci 2019; 20:E2477. [PMID: 31137455 PMCID: PMC6566845 DOI: 10.3390/ijms20102477] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/10/2019] [Accepted: 05/17/2019] [Indexed: 02/08/2023] Open
Abstract
We investigated whether thrombin preconditioning of human Wharton's jelly-derived mesenchymal stem cells (MSCs) improves paracrine potency and thus the therapeutic efficacy of naïve MSCs against severe hypoxic ischemic encephalopathy (HIE). Thrombin preconditioning significantly enhances the neuroprotective anti-oxidative, anti-apoptotic, and anti-cytotoxic effects of naïve MSCs against oxygen-glucose deprivation (OGD) of cortical neurons in vitro. Severe HIE was induced in vivo using unilateral carotid artery ligation and hypoxia for 2 h and confirmed using brain magnetic resonance imaging (MRI) involving >40% of ipsilateral hemisphere at postnatal day (P) 7 in newborn rats. Delayed intraventricular transplantation of 1 × 105 thrombin preconditioned but not naïve MSCs at 24 h after hypothermia significantly enhanced observed anti-inflammatory, anti-astroglial, and anti-apoptotic effects and the ensuing brain infarction; behavioral tests, such as cylinder rearing and negative geotaxis tests, were conducted at P42. In summary, thrombin preconditioning of human Wharton's jelly-derived MSCs significantly boosted the neuroprotective effects of naïve MSCs against OGD in vitro by enhancing their anti-oxidative, anti-apoptotic, and anti-cytotoxic effects, and significantly attenuated the severe HIE-induced brain infarction and improved behavioral function tests in vivo by maximizing their paracrine anti-inflammatory, anti-astroglial, and anti-apoptotic effects.
Collapse
Affiliation(s)
- Young Eun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
| | - Dong Kyung Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea.
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea.
| |
Collapse
|
13
|
Breu M, Reisinger D, Tao L, Wu D, Zhang Y, Budde MD, Fatemi A, Pathak AP, Zhang J. In vivo high-resolution diffusion tensor imaging of the developing neonatal rat cortex and its relationship to glial and dendritic maturation. Brain Struct Funct 2019; 224:1815-1829. [PMID: 31011813 DOI: 10.1007/s00429-019-01878-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/11/2019] [Indexed: 12/25/2022]
Abstract
Diffusion tensor imaging (DTI) is increasingly utilized as a sensitive tool for studying brain maturation and injuries during the neonatal period. In this study, we acquired high resolution in vivo DTI data from neonatal rat brains from postnatal day 2 (P2) to P10 and correlated temporal changes in DTI derived markers with microstructural organization of glia, axons, and dendrites during this critical period of brain development. Group average images showed dramatic temporal changes in brain morphology, fractional anisotropy (FA) and mean diffusivity (MD). Most cortical regions showed a monotonous decline in FA and an initial increase in MD from P2 to P8 that declined slightly by P10. Qualitative histology revealed rapid maturation of the glial and dendritic networks in the developing cortex. In the cingulate and motor cortex, the decreases in FA over time significantly correlated with structural anisotropy values computed from histological sections stained with glial and dendritic markers. However, in the sensory and visual cortex, other factors probably contributed to the observed decreases in FA. We did not observe any significant correlations between FA and structural anisotropy computed from the axonal histological marker.
Collapse
Affiliation(s)
- Markus Breu
- Division of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Dominik Reisinger
- Division of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Liangcheng Tao
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dan Wu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yajing Zhang
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew D Budde
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ali Fatemi
- Division of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arvind P Pathak
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiangyang Zhang
- Department of Radiology, New York University School of Medicine, 660 First Avenue, Room 207, New York, NY, 10016, USA.
| |
Collapse
|
14
|
Mikrogeorgiou A, Xu D, Ferriero DM, Vannucci SJ. Assessing Cerebral Metabolism in the Immature Rodent: From Extracts to Real-Time Assessments. Dev Neurosci 2019; 40:463-474. [PMID: 30991389 DOI: 10.1159/000496921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/09/2019] [Indexed: 12/27/2022] Open
Abstract
Brain development is an energy-expensive process. Although glucose is irreplaceable, the developing brain utilizes a variety of substrates such as lactate and the ketone bodies, β-hydroxybutyrate and acetoacetate, to produce energy and synthesize the structural components necessary for cerebral maturation. When oxygen and nutrient supplies to the brain are restricted, as in neonatal hypoxia-ischemia (HI), cerebral energy metabolism undergoes alterations in substrate use to preserve the production of adenosine triphosphate. These changes have been studied by in situ biochemical methods that yielded valuable quantitative information about high-energy and glycolytic metabolites and established a temporal profile of the cerebral metabolic response to hypoxia and HI. However, these analyses relied on terminal experiments and averaging values from several animals at each time point as well as challenging requirements for accurate tissue processing.More recent methodologies have focused on in vivo longitudinal analyses in individual animals. The emerging field of metabolomics provides a new investigative tool for studying cerebral metabolism. Magnetic resonance spectroscopy (MRS) has enabled the acquisition of a snapshot of the metabolic status of the brain as quantifiable spectra of various intracellular metabolites. Proton (1H) MRS has been used extensively as an experimental and diagnostic tool of HI in the pursuit of markers of long-term neurodevelopmental outcomes. Still, the interpretation of the metabolite spectra acquired with 1H MRS has proven challenging, due to discrepancies among studies, regarding calculations and timing of measurements. As a result, the predictive utility of such studies is not clear. 13C MRS is methodologically more challenging, but it provides a unique window on living tissue metabolism via measurements of the incorporation of 13C label from substrates into brain metabolites and the localized determination of various metabolic fluxes. The newly developed hyperpolarized 13C MRS is an exciting method for assessing cerebral metabolism in vivo, that bears the advantages of conventional 13C MRS but with a huge gain in signal intensity and much shorter acquisition times. The first part of this review article provides a brief description of the findings of biochemical and imaging methods over the years as well as a discussion of their associated strengths and pitfalls. The second part summarizes the current knowledge on cerebral metabolism during development and HI brain injury.
Collapse
Affiliation(s)
- Alkisti Mikrogeorgiou
- Department of Pediatrics, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Duan Xu
- Department of Radiology and Biomedical Imaging, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Donna M Ferriero
- Department of Pediatrics, University of California San Francisco School of Medicine, San Francisco, California, USA.,Department of Neurology, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Susan J Vannucci
- Department of Pediatrics and Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA,
| |
Collapse
|
15
|
Short-, Mid-, and Long-Term Effect of Granulocyte Colony-Stimulating Factor/Stem Cell Factor and Fms-Related Tyrosine Kinase 3 Ligand Evaluated in an In Vivo Model of Hypoxic-Hyperoxic Ischemic Neonatal Brain Injury. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5935279. [PMID: 31001556 PMCID: PMC6436372 DOI: 10.1155/2019/5935279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/01/2019] [Accepted: 02/10/2019] [Indexed: 01/01/2023]
Abstract
Hematopoietic growth factors are considered to bear neuroprotective potential. We have previously shown that delayed treatment with granulocyte colony-stimulating factor (G-CSF)/stem cell factor (SCF) and Fms-related tyrosine kinase 3 ligand (FL) ameliorates excitotoxic neonatal brain injury. The effect of these substances in combined-stressor neonatal brain injury models more closely mimicking clinical conditions has not been investigated. The aim of this study was to assess the short-, mid-, and long-term neuroprotective potential of G-CSF/SCF and FL in a neonatal model of hypoxic-hyperoxic ischemic brain injury. Five-day-old (P5) CD-1 mice were subjected to unilateral common carotid artery ligation and subsequent alternating periods of hypoxia and hyperoxia for 65 minutes. Sixty hours after injury, pups were randomly assigned to intraperitoneal treatment with (i) G-CSF (200 μg/kg)/SCF (50 μg/kg), (ii) FL (100 μg/kg), or (iii) vehicle every 24 hours for three or five consecutive days. Histopathological and functional outcomes were evaluated on P10, P18, and P90. Baseline outcome parameters were established in sham-treated and healthy control animals. Gross brain injury did not significantly differ between treatment groups at any time point. On P10, caspase-3 activation and caspase-independent apoptosis were similar between treatment groups; cell proliferation and the number of BrdU-positive vessels did not differ on P18 or P90. Neurobehavioral assessment did not reveal significant differences between treatment groups in accelerod performance, open field behavior, or novel object recognition capacity on P90. Turning behavior was more frequently observed in G-CSF/SCF- and FL-treated animals. No sex-specific differences were detected in any outcome parameter evaluated. In hypoxic-hyperoxic ischemic neonatal brain injury, G-CSF/SCF and FL treatment does not convey neuroprotection. Prior to potential clinical use, meticulous assessment of these hematopoietic growth factors is mandated.
Collapse
|
16
|
Muntsant A, Shrivastava K, Recasens M, Giménez-Llort L. Severe Perinatal Hypoxic-Ischemic Brain Injury Induces Long-Term Sensorimotor Deficits, Anxiety-Like Behaviors and Cognitive Impairment in a Sex-, Age- and Task-Selective Manner in C57BL/6 Mice but Can Be Modulated by Neonatal Handling. Front Behav Neurosci 2019; 13:7. [PMID: 30814939 PMCID: PMC6381068 DOI: 10.3389/fnbeh.2019.00007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022] Open
Abstract
Perinatal brain injury (PBI) leads to neurological disabilities throughout life, from motor deficits, cognitive limitations to severe cerebral palsy. Yet, perinatal brain damage has limited therapeutic outcomes. Besides, the immature brain of premature children is at increased risk of hypoxic/ischemic (HI) injury, with males being more susceptible to it and less responsive to protective/therapeutical interventions. Here, we model in male and female C57BL/6 mice, the impact of neonatal HI and the protective effects of neonatal handling (NH), an early life tactile and proprioceptive sensory stimulation. From postnatal day 1 (PND1, modeling pre-term) to PND21 randomized litters received either NH or left undisturbed. HI brain damage occurred by permanent left carotid occlusion followed by hypoxia at PND7 (modeling full-term) in half of the animals. The behavioral and functional screening of the pups at weaning (PND23) and their long-term outcomes (adulthood, PND70) were evaluated in a longitudinal study, as follows: somatic development (weight), sensorimotor functions (reflexes, rods and hanger tests), exploration [activity (ACT) and open-field (OF) test], emotional and anxiety-like behaviors [corner, open-field and dark-light box (DLB) tests], learning and memory [T-maze (TM) and Morris Water-Maze (MWM)]. HI induced similar brain damage in both sexes but affected motor development, sensorimotor functions, induced hyperactivity at weaning, and anxiety-like behaviors and cognitive deficits at adulthood, in a sex- and age-dependent manner. Thus, during ontogeny, HI affected equilibrium especially in females and prehensility in males, but only reflexes at adulthood. Hyperactivity of HI males was normalized at adulthood. HI increased neophobia and other anxiety-like behaviors in males but emotionality in females. Both sexes showed worse short/long-term learning, but memory was more affected in males. Striking neuroprotective effects of NH were found, with significantly lower injury scores, mostly in HI males. At the functional level, NH reversed the impaired reflex responses and improved memory performances in hippocampal-dependent spatial-learning tasks, especially in males. Finally, neuropathological correlates referred to atrophy, neuronal densities and cellularity in the affected areas [hippocampal-CA, caudate/putamen, thalamus, neocortex and corpus callosum (CC)] point out distinct neuronal substrates underlying the sex- and age- functional impacts of these risk/protection interventions on sensorimotor, behavioral and cognitive outcomes from ontogeny to adulthood.
Collapse
Affiliation(s)
- Aida Muntsant
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kalpana Shrivastava
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology & Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mireia Recasens
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology & Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lydia Giménez-Llort
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
17
|
Huun MU, Garberg H, Løberg EM, Escobar J, Martinez-Orgado J, Saugstad OD, Solberg R. DHA and therapeutic hypothermia in a short-term follow-up piglet model of hypoxia-ischemia: Effects on H+MRS biomarkers. PLoS One 2018; 13:e0201895. [PMID: 30086156 PMCID: PMC6080779 DOI: 10.1371/journal.pone.0201895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/24/2018] [Indexed: 12/22/2022] Open
Abstract
Background Therapeutic hypothermia has become the standard of care for newborns with hypoxic-ischemic encephalopathy in high and middle income countries. Docosahexaenoic acid (DHA) has neuroprotective properties of reducing excitotoxicity, neuroinflammation and apoptosis in rodent models. We aim to study whether post hypoxic administration of i.v. DHA will reduce H+MRS biomarkers and gene expression of inflammation and apoptosis both with and without hypothermia in a large animal model. Methods Fifty-five piglets were randomized to severe global hypoxia (N = 48) or not (Sham, N = 7). Hypoxic piglets were further randomized by factorial design: Vehicle (VEH), DHA, VEH + Hypothermia (HT), or DHA + HT. 5 mg/kg DHA was given intravenously 210 min after end of hypoxia. Two-way ANOVA analyses were performed with DHA and hypothermia as main effects. Results Cortical lactate/N-acetylaspartate (Lac/NAA) was significantly reduced in DHA + HT compared to HT. DHA had significant main effects on increasing N-acetylaspartate and glutathione in hippocampus. Therapeutic hypothermia significantly reduced the Lac/NAA ratio and protein expression of IL-1β and TNFα in hippocampus and reduced Troponin T in serum. Neuropathology showed significant differences between sham and hypoxia, but no differences between intervention groups. Conclusion DHA and therapeutic hypothermia significantly improve specific H+MRS biomarkers in this short-term follow up model of hypoxia-ischemia. Longer recovery periods are needed to evaluate whether DHA can offer translational neuroprotection.
Collapse
Affiliation(s)
- Marianne Ullestad Huun
- Department of Pediatric Research, Women and Children's Division and Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- University of Oslo, Oslo, Norway
- * E-mail:
| | - Håvard Garberg
- Department of Pediatric Research, Women and Children's Division and Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Else Marit Løberg
- Department of Pathology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Javier Escobar
- Department of Pediatric Research, Women and Children's Division and Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Neonatal Research Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | | | - Ola Didrik Saugstad
- Department of Pediatric Research, Women and Children's Division and Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- University of Oslo, Oslo, Norway
| | - Rønnaug Solberg
- Department of Pediatric Research, Women and Children's Division and Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Pediatrics, Vestfold Hospital Trust, Tønsberg, Norway
| |
Collapse
|
18
|
Neuroprotective Effects of Bioactive Compounds and MAPK Pathway Modulation in "Ischemia"-Stressed PC12 Pheochromocytoma Cells. Brain Sci 2018; 8:brainsci8020032. [PMID: 29419806 PMCID: PMC5836051 DOI: 10.3390/brainsci8020032] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/24/2018] [Accepted: 02/02/2018] [Indexed: 02/08/2023] Open
Abstract
This review surveys the efforts taken to investigate in vitro neuroprotective features of synthetic compounds and cell-released growth factors on PC12 clonal cell line temporarily deprived of oxygen and glucose followed by reoxygenation (OGD/R). These cells have been used previously to mimic some of the properties of in vivo brain ischemia-reperfusion-injury (IRI) and have been instrumental in identifying common mechanisms such as calcium overload, redox potential, lipid peroxidation and MAPKs modulation. In addition, they were useful for establishing the role of certain membrane penetrable cocktails of antioxidants as well as potential growth factors which may act in neuroprotection. Pharmacological mechanisms of neuroprotection addressing modulation of the MAPK cascade and increased redox potential by natural products, drugs and growth factors secreted by stem cells, in either undifferentiated or nerve growth factor-differentiated PC12 cells exposed to ischemic conditions are discussed for future prospects in neuroprotection studies.
Collapse
|
19
|
Li B, Concepcion K, Meng X, Zhang L. Brain-immune interactions in perinatal hypoxic-ischemic brain injury. Prog Neurobiol 2017; 159:50-68. [PMID: 29111451 PMCID: PMC5831511 DOI: 10.1016/j.pneurobio.2017.10.006] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/26/2017] [Indexed: 01/07/2023]
Abstract
Perinatal hypoxia-ischemia remains the primary cause of acute neonatal brain injury, leading to a high mortality rate and long-term neurological deficits, such as behavioral, social, attentional, cognitive and functional motor deficits. An ever-increasing body of evidence shows that the immune response to acute cerebral hypoxia-ischemia is a major contributor to the pathophysiology of neonatal brain injury. Hypoxia-ischemia provokes an intravascular inflammatory cascade that is further augmented by the activation of resident immune cells and the cerebral infiltration of peripheral immune cells response to cellular damages in the brain parenchyma. This prolonged and/or inappropriate neuroinflammation leads to secondary brain tissue injury. Yet, the long-term effects of immune activation, especially the adaptive immune response, on the hypoxic-ischemic brain still remain unclear. The focus of this review is to summarize recent advances in the understanding of post-hypoxic-ischemic neuroinflammation triggered by the innate and adaptive immune responses and to discuss how these mechanisms modulate the brain vulnerability to injury. A greater understanding of the reciprocal interactions between the hypoxic-ischemic brain and the immune system will open new avenues for potential immunomodulatory therapy in the treatment of neonatal brain injury.
Collapse
Affiliation(s)
- Bo Li
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Katherine Concepcion
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Xianmei Meng
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
20
|
Adstamongkonkul D, Hess DC. Ischemic Conditioning and neonatal hypoxic ischemic encephalopathy: a literature review. CONDITIONING MEDICINE 2017; 1:9-16. [PMID: 30215057 PMCID: PMC6131706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Hypoxic Ischemic Encephalopathy (HIE) is the result of severe anoxic brain injury during the neonatal period and causes life-long morbidity and premature mortality. Currently, therapeutic hypothermia immediately after birth is the standard of care for clinically relevant HIE. However, therapeutic hypothermia alone does not provide complete neuroprotection and there is an urgent need for adjunctive therapies. Ischemic conditioning is an adaptive process of endogenous protection in which small doses of sub-lethal ischemia can provide a protection against a lethal ischemic event. Remote Ischemic Post-conditioning (RIPC), a form of ischemic conditioning, is highly translatable for HIE diagnosed immediately after birth as the conditioned ischemic stimulus is applied at the limb after the lethal ischemic episode. A number of studies in neonatal rats have demonstrated that RIPC is effective at reducing injury in focal cerebral ischemia models and improves neurological outcomes. In this review, we focus on the available data on HIE and its current treatment, models in HIE studies, ischemic conditioning/RIPC and its mechanism. We discuss in particular the effect of RIPC on neonatal brain with HIE. We postulate that combining RIPC with standard therapeutic hypothermia can be an attractive therapeutic approach for HIE.
Collapse
Affiliation(s)
- Dusit Adstamongkonkul
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA
| | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA
| |
Collapse
|
21
|
Sukhanova IA, Sebentsova EA, Levitskaya NG. The acute and delayed effects of perinatal hypoxic brain damage in children and in model experiments with rodents. NEUROCHEM J+ 2016. [DOI: 10.1134/s1819712416040127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
22
|
Lingam I, Avdic-Belltheus A, Robertson NJ. Using animal models to improve care of neonatal encephalopathy. Arch Dis Child Educ Pract Ed 2016; 101:271-6. [PMID: 27147551 DOI: 10.1136/archdischild-2015-309927] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 04/13/2016] [Indexed: 11/04/2022]
Affiliation(s)
- Ingran Lingam
- Institute for Women's Health, University College London, London, UK
| | | | | |
Collapse
|
23
|
Yazdani A, Khoja Z, Johnstone A, Dale L, Rampakakis E, Wintermark P. Sildenafil Improves Brain Injury Recovery following Term Neonatal Hypoxia-Ischemia in Male Rat Pups. Dev Neurosci 2016; 38:251-263. [DOI: 10.1159/000448327] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/08/2016] [Indexed: 11/19/2022] Open
Abstract
Term asphyxiated newborns remain at risk of developing brain injury despite available neuropreventive therapies such as hypothermia. Neurorestorative treatments may be an alternative. This study investigated the effect of sildenafil on brain injury induced by neonatal hypoxia-ischemia (HI) at term-equivalent age. Neonatal HI was induced in male Long-Evans rat pups at postnatal day 10 (P10) by left common carotid ligation followed by a 2-hour exposure to 8% oxygen; sham-operated rat pups served as the control. Both groups were randomized to oral sildenafil or vehicle twice daily for 7 consecutive days. Gait analysis was performed on P27. At P30, the rats were sacrificed, and their brains were extracted. The surfaces of both hemispheres were measured on hematoxylin and eosin-stained brain sections. Mature neurons and endothelial cells were quantified near the infarct boundary zone using immunohistochemistry. HI caused significant gait impairment and a reduction in the size of the left hemisphere. Treatment with sildenafil led to an improvement in the neurological deficits as measured by gait analysis, as well as an improvement in the size of the left hemisphere. Sildenafil, especially at higher doses, also caused a significant increase in the number of neurons near the infarct boundary zone. In conclusion, sildenafil administered after neonatal HI may improve brain injury recovery by promoting neuronal populations.
Collapse
|
24
|
Kartal Ö, Aydınöz S, Kartal AT, Kelestemur T, Caglayan AB, Beker MC, Karademir F, Süleymanoğlu S, Kul M, Yulug B, Kilic E. Time dependent impact of perinatal hypoxia on growth hormone, insulin-like growth factor 1 and insulin-like growth factor binding protein-3. Metab Brain Dis 2016; 31:827-35. [PMID: 26943480 DOI: 10.1007/s11011-016-9816-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/29/2016] [Indexed: 12/25/2022]
Abstract
Hypoxic-ischemia (HI) is a widely used animal model to mimic the preterm or perinatal sublethal hypoxia, including hypoxic-ischemic encephalopathy. It causes diffuse neurodegeneration in the brain and results in mental retardation, hyperactivity, cerebral palsy, epilepsy and neuroendocrine disturbances. Herein, we examined acute and subacute correlations between neuronal degeneration and serum growth factor changes, including growth hormone (GH), insulin-like growth factor 1 (IGF-1) and insulin-like growth factor binding protein-3 (IGFBP-3) after hypoxic-ischemia (HI) in neonatal rats. In the acute phase of hypoxia, brain volume was increased significantly as compared with control animals, which was associated with reduced GH and IGF-1 secretions. Reduced neuronal survival and increased DNA fragmentation were also noticed in these animals. However, in the subacute phase of hypoxia, neuronal survival and brain volume were significantly decreased, accompanied by increased apoptotic cell death in the hippocampus and cortex. Serum GH, IGF-1, and IGFBP-3 levels were significantly reduced in the subacute phase of HI. Significant retardation in the brain and body development were noted in the subacute phase of hypoxia. Here, we provide evidence that serum levels of growth-hormone and factors were decreased in the acute and subacute phase of hypoxia, which was associated with increased DNA fragmentation and decreased neuronal survival.
Collapse
Affiliation(s)
- Ömer Kartal
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler cad. 19, TR-34810, Istanbul, Turkey
- Department of Pediatrics, Gulhane Military Medical Academy, Haydarpasa Teaching Hospital, Istanbul, Turkey
| | - Seçil Aydınöz
- Department of Pediatrics, Gulhane Military Medical Academy, Haydarpasa Teaching Hospital, Istanbul, Turkey
| | - Ayşe Tuğba Kartal
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler cad. 19, TR-34810, Istanbul, Turkey
- Department of Pediatrics, Marmaris State Hospital, Mugla, Turkey
| | - Taha Kelestemur
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler cad. 19, TR-34810, Istanbul, Turkey
| | - Ahmet Burak Caglayan
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler cad. 19, TR-34810, Istanbul, Turkey
| | - Mustafa Caglar Beker
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler cad. 19, TR-34810, Istanbul, Turkey
| | - Ferhan Karademir
- Department of Pediatrics, Gulhane Military Medical Academy, Haydarpasa Teaching Hospital, Istanbul, Turkey
| | - Selami Süleymanoğlu
- Department of Pediatrics, Gulhane Military Medical Academy, Haydarpasa Teaching Hospital, Istanbul, Turkey
| | - Mustafa Kul
- Department of Pediatrics, Gulhane Military Medical Academy, Haydarpasa Teaching Hospital, Istanbul, Turkey
| | - Burak Yulug
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler cad. 19, TR-34810, Istanbul, Turkey
| | - Ertugrul Kilic
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Ekinciler cad. 19, TR-34810, Istanbul, Turkey.
| |
Collapse
|
25
|
Rumajogee P, Bregman T, Miller SP, Yager JY, Fehlings MG. Rodent Hypoxia-Ischemia Models for Cerebral Palsy Research: A Systematic Review. Front Neurol 2016; 7:57. [PMID: 27199883 PMCID: PMC4843764 DOI: 10.3389/fneur.2016.00057] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 04/03/2016] [Indexed: 12/28/2022] Open
Abstract
Cerebral palsy (CP) is a complex multifactorial disorder, affecting approximately 2.5-3/1000 live term births, and up to 22/1000 prematurely born babies. CP results from injury to the developing brain incurred before, during, or after birth. The most common form of this condition, spastic CP, is primarily associated with injury to the cerebral cortex and subcortical white matter as well as the deep gray matter. The major etiological factors of spastic CP are hypoxia/ischemia (HI), occurring during the last third of pregnancy and around birth age. In addition, inflammation has been found to be an important factor contributing to brain injury, especially in term infants. Other factors, including genetics, are gaining importance. The classic Rice-Vannucci HI model (in which 7-day-old rat pups undergo unilateral ligation of the common carotid artery followed by exposure to 8% oxygen hypoxic air) is a model of neonatal stroke that has greatly contributed to CP research. In this model, brain damage resembles that observed in severe CP cases. This model, and its numerous adaptations, allows one to finely tune the injury parameters to mimic, and therefore study, many of the pathophysiological processes and conditions observed in human patients. Investigators can recreate the HI and inflammation, which cause brain damage and subsequent motor and cognitive deficits. This model further enables the examination of potential approaches to achieve neural repair and regeneration. In the present review, we compare and discuss the advantages, limitations, and the translational value for CP research of HI models of perinatal brain injury.
Collapse
Affiliation(s)
- Prakasham Rumajogee
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network , Toronto, ON , Canada
| | - Tatiana Bregman
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network , Toronto, ON , Canada
| | - Steven P Miller
- Department of Pediatrics, Hospital for Sick Children , Toronto, ON , Canada
| | - Jerome Y Yager
- Division of Pediatric Neurosciences, Stollery Children's Hospital, University of Alberta , Edmonton, AB , Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada; Division of Neurosurgery, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
26
|
Neuroprotection as a Potential Therapeutic Perspective in Neurodegenerative Diseases: Focus on Antiepileptic Drugs. Neurochem Res 2015; 41:340-52. [PMID: 26721507 DOI: 10.1007/s11064-015-1809-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/10/2015] [Accepted: 12/14/2015] [Indexed: 02/07/2023]
Abstract
Neuroprotection is conceived as one of the potential tool to prevent or slow neuronal death and hence a therapeutic hope to treat neurodegenerative diseases, like Parkinson's and Alzheimer's diseases. Increase of oxidative stress, mitochondrial dysfunction, excitotoxicity, inflammatory changes, iron accumulation, and protein aggregation have been identified as main causes of neuronal death and adopted as targets to test experimentally the putative neuroprotective effects of various classes of drugs. Among these agents, antiepileptic drugs (AEDs), both the old and the newer generations, have shown to exert protective effects in different experimental models. Their mechanism of action is mediated mainly by modulating the activity of sodium, calcium and potassium channels as well as the glutamatergic and GABAergic (gamma-aminobutyric acid) synapses. Neurological pathologies in which a neuroprotective action of AEDs has been demonstrated in specific experimental models include: cerebral ischemia, Parkinson's disease, and Alzheimer's disease. Although the whole of experimental data indicating that neuroprotection can be achieved is remarkable and encouraging, no firm data have been produced in humans so far and, at the present time, neuroprotection still remains a challenge for the future.
Collapse
|
27
|
Moretti R, Zanin A, Pansiot J, Spiri D, Manganozzi L, Kratzer I, Favero G, Vasiljevic A, Rinaldi VE, Pic I, Massano D, D'Agostino I, Baburamani A, La Rocca MA, Rodella LF, Rezzani R, Ek J, Strazielle N, Ghersi-Egea JF, Gressens P, Titomanlio L. Melatonin reduces excitotoxic blood-brain barrier breakdown in neonatal rats. Neuroscience 2015; 311:382-97. [PMID: 26542996 DOI: 10.1016/j.neuroscience.2015.10.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/14/2015] [Accepted: 10/23/2015] [Indexed: 12/30/2022]
Abstract
The blood-brain barrier (BBB) is a complex structure that protects the central nervous system from peripheral insults. Understanding the molecular basis of BBB function and dysfunction holds significant potential for future strategies to prevent and treat neurological damage. The aim of our study was (1) to investigate BBB alterations following excitotoxicity and (2) to test the protective properties of melatonin. Ibotenate, a glutamate analog, was injected intracerebrally in postnatal day 5 (P5) rat pups to mimic excitotoxic injury. Animals were than randomly divided into two groups, one receiving intraperitoneal (i.p.) melatonin injections (5mg/kg), and the other phosphate buffer saline (PBS) injections. Pups were sacrificed 2, 4 and 18 h after ibotenate injection. We determined lesion size at 5 days by histology, the location and organization of tight junction (TJ) proteins by immunohistochemical studies, and BBB leakage by dextran extravasation. Expression levels of BBB genes (TJs, efflux transporters and detoxification enzymes) were determined in the cortex and choroid plexus by quantitative PCR. Dextran extravasation was seen 2h after the insult, suggesting a rapid BBB breakdown that was resolved by 4h. Extravasation was significantly reduced in melatonin-treated pups. Gene expression and immunohistochemical assays showed dynamic BBB modifications during the first 4h, partially prevented by melatonin. Lesion-size measurements confirmed white matter neuroprotection by melatonin. Our study is the first to evaluate BBB structure and function at a very early time point following excitotoxicity in neonates. Melatonin neuroprotects by preventing TJ modifications and BBB disruption at this early phase, before its previously demonstrated anti-inflammatory, antioxidant and axonal regrowth-promoting effects.
Collapse
Affiliation(s)
- R Moretti
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris, France; Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France; Università degli studi di Udine, 33100 Udine, Italy
| | - A Zanin
- Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France
| | - J Pansiot
- Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France
| | - D Spiri
- Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France
| | - L Manganozzi
- Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France
| | - I Kratzer
- Lyon Neurosciences Research Center, Inserm U1028, CNRS UMR5292 - Lyon University, Lyon, France
| | - G Favero
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - A Vasiljevic
- Lyon Neurosciences Research Center, Inserm U1028, CNRS UMR5292 - Lyon University, Lyon, France
| | - V E Rinaldi
- Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France
| | - I Pic
- Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France
| | - D Massano
- Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France
| | - I D'Agostino
- Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France
| | - A Baburamani
- Perinatal Center, Dept Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - M A La Rocca
- Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France
| | - L F Rodella
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - R Rezzani
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - J Ek
- Perinatal Center, Dept Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - N Strazielle
- Lyon Neurosciences Research Center, Inserm U1028, CNRS UMR5292 - Lyon University, Lyon, France; Brain-i, Lyon, France
| | - J-F Ghersi-Egea
- Lyon Neurosciences Research Center, Inserm U1028, CNRS UMR5292 - Lyon University, Lyon, France
| | - P Gressens
- Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, St. Thomas' Hospital, London, United Kingdom
| | - L Titomanlio
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris, France; Inserm, U1141, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; PremUP, Paris, France.
| |
Collapse
|
28
|
Dixon BJ, Reis C, Ho WM, Tang J, Zhang JH. Neuroprotective Strategies after Neonatal Hypoxic Ischemic Encephalopathy. Int J Mol Sci 2015; 16:22368-401. [PMID: 26389893 PMCID: PMC4613313 DOI: 10.3390/ijms160922368] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/31/2015] [Accepted: 09/06/2015] [Indexed: 12/21/2022] Open
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) is a devastating disease that primarily causes neuronal and white matter injury and is among the leading cause of death among infants. Currently there are no well-established treatments; thus, it is important to understand the pathophysiology of the disease and elucidate complications that are creating a gap between basic science and clinical translation. In the development of neuroprotective strategies and translation of experimental results in HIE, there are many limitations and challenges to master based on an appropriate study design, drug delivery properties, dosage, and use in neonates. We will identify understudied targets after HIE, as well as neuroprotective molecules that bring hope to future treatments such as melatonin, topiramate, xenon, interferon-beta, stem cell transplantation. This review will also discuss some of the most recent trials being conducted in the clinical setting and evaluate what directions are needed in the future.
Collapse
Affiliation(s)
- Brandon J Dixon
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | - Cesar Reis
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Wing Mann Ho
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Medical University Innsbruck, Tyrol 6020, Austria.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|
29
|
Empie K, Rangarajan V, Juul SE. Is the ferret a suitable species for studying perinatal brain injury? Int J Dev Neurosci 2015; 45:2-10. [PMID: 26102988 PMCID: PMC4793918 DOI: 10.1016/j.ijdevneu.2015.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/09/2015] [Accepted: 06/01/2015] [Indexed: 11/26/2022] Open
Abstract
Ferret brain architecture, composition, and development are similar to humans. Postnatal ferret brain development is comparable to that of premature infants. Ferrets have potential to model preterm and term neonatal brain injury. Ferrets may fulfill the need for an intermediate model species of neurodevelopment. Many opportunities exist to expand the use of ferrets as research subjects.
Complications of prematurity often disrupt normal brain development and/or cause direct damage to the developing brain, resulting in poor neurodevelopmental outcomes. Physiologically relevant animal models of perinatal brain injury can advance our understanding of these influences and thereby provide opportunities to develop therapies and improve long-term outcomes. While there are advantages to currently available small animal models, there are also significant drawbacks that have limited translation of research findings to humans. Large animal models such as newborn pig, sheep and nonhuman primates have complex brain development more similar to humans, but these animals are expensive, and developmental testing of sheep and piglets is limited. Ferrets (Mustela putorius furo) are born lissencephalic and undergo postnatal cortical folding to form complex gyrencephalic brains. This review examines whether ferrets might provide a novel intermediate animal model of neonatal brain disease that has the benefit of a gyrified, altricial brain in a small animal. It summarizes attributes of ferret brain growth and development that make it an appealing animal in which to model perinatal brain injury. We postulate that because of their innate characteristics, ferrets have great potential in neonatal neurodevelopmental studies.
Collapse
Affiliation(s)
- Kristen Empie
- Department of Neonatology, University of Washington, Seattle, USA
| | | | - Sandra E Juul
- Department of Neonatology, University of Washington, Seattle, USA.
| |
Collapse
|
30
|
Shaikh H, Lechpammer M, Jensen FE, Warfield SK, Hansen AH, Kosaras B, Shevell M, Wintermark P. Increased Brain Perfusion Persists over the First Month of Life in Term Asphyxiated Newborns Treated with Hypothermia: Does it Reflect Activated Angiogenesis? Transl Stroke Res 2015; 6:224-33. [PMID: 25620793 DOI: 10.1007/s12975-015-0387-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 01/06/2015] [Accepted: 01/13/2015] [Indexed: 12/23/2022]
Abstract
Many asphyxiated newborns still develop brain injury despite hypothermia therapy. The development of brain injury in these newborns has been related partly to brain perfusion abnormalities. The purposes of this study were to assess brain hyperperfusion over the first month of life in term asphyxiated newborns and to search for some histopathological clues indicating whether this hyperperfusion may be related to activated angiogenesis following asphyxia. In this prospective cohort study, regional cerebral blood flow was measured in term asphyxiated newborns treated with hypothermia around day 10 of life and around 1 month of life using magnetic resonance imaging (MRI) and arterial spin labeling. A total of 32 MRI scans were obtained from 24 term newborns. Asphyxiated newborns treated with hypothermia displayed an increased cerebral blood flow in the injured brain areas around day 10 of life and up to 1 month of life. In addition, we looked at the histopathological clues in a human asphyxiated newborn and in a rat model of neonatal encephalopathy. Vascular endothelial growth factor (VEGF) was expressed in the injured brain of an asphyxiated newborn treated with hypothermia in the first days of life and of rat pups 24-48 h after the hypoxic-ischemic event, and the endothelial cell count increased in the injured cortex of the pups 7 and 11 days after hypoxia-ischemia. Our data showed that the hyperperfusion measured by imaging persisted in the injured areas up to 1 month of life and that angiogenesis was activated in the injured brain of asphyxiated newborns.
Collapse
Affiliation(s)
- Henna Shaikh
- Department of Pediatrics, McGill University, Montreal, QC, Canada
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Shi RY, Zhu SH, Li V, Gibson SB, Xu XS, Kong JM. BNIP3 interacting with LC3 triggers excessive mitophagy in delayed neuronal death in stroke. CNS Neurosci Ther 2014; 20:1045-55. [PMID: 25230377 DOI: 10.1111/cns.12325] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/14/2014] [Accepted: 08/15/2014] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION A basal level of mitophagy is essential in mitochondrial quality control in physiological conditions, while excessive mitophagy contributes to cell death in a number of diseases including ischemic stroke. Signals regulating this process remain unknown. BNIP3, a pro-apoptotic BH3-only protein, has been implicated as a regulator of mitophagy. AIMS Both in vivo and in vitro models of stroke, as well as BNIP3 wild-type and knock out mice were used in this study. RESULTS We show that BNIP3 and its homologue BNIP3L (NIX) are highly expressed in a "delayed" manner and contribute to delayed neuronal loss following stroke. Deficiency in BNIP3 significantly decreases both neuronal mitophagy and apoptosis but increases nonselective autophagy following ischemic/hypoxic insults. The mitochondria-localized BNIP3 interacts with the autophagosome-localized LC3, suggesting that BNIP3, similar to NIX, functions as a LC3-binding receptor on mitochondria. Although NIX expression is upregulated when BNIP3 is silenced, up-regulation of NIX cannot functionally compensate for the loss of BNIP3 in activating excessive mitophagy. CONCLUSIONS NIX primarily regulates basal level of mitophagy in physiological conditions, whereas BNIP3 exclusively activates excessive mitophagy leading to cell death.
Collapse
Affiliation(s)
- Ruo-Yang Shi
- Department of Human Anatomy and Cell Science, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | | | | | | | | | | |
Collapse
|
32
|
Ginet V, Pittet MP, Rummel C, Osterheld MC, Meuli R, Clarke PGH, Puyal J, Truttmann AC. Dying neurons in thalamus of asphyxiated term newborns and rats are autophagic. Ann Neurol 2014; 76:695-711. [PMID: 25146903 DOI: 10.1002/ana.24257] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 08/19/2014] [Accepted: 08/19/2014] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Neonatal hypoxic-ischemic encephalopathy (HIE) still carries a high burden by its mortality and long-term neurological morbidity in survivors. Apart from hypothermia, there is no acknowledged therapy for HIE, reflecting the lack of mechanistic understanding of its pathophysiology. (Macro)autophagy, a physiological intracellular process of lysosomal degradation, has been proposed to be excessively activated in excitotoxic conditions such as HIE. The present study examines whether neuronal autophagy in the thalamus of asphyxiated human newborns or P7 rats is enhanced and related to neuronal death processes. METHODS Neuronal autophagy and cell death were evaluated in the thalamus (frequently injured in severe HIE) of both human newborns who died after severe HIE (n = 5) and P7 hypoxic-ischemic rats (Rice-Vannuci model). Autophagic (LC3, p62), lysosomal (LAMP1, cathepsins), and cell death (TUNEL, caspase-3) markers were studied by immunohistochemistry in human and rat brain sections, and by additional methods in rats (immunoblotting, histochemistry, and electron microscopy). RESULTS Following severe perinatal asphyxia in both humans and rats, thalamic neurons displayed up to 10-fold (p < 0.001) higher numbers of autophagosomes and lysosomes, implying an enhanced autophagic flux. The highly autophagic neurons presented strong features of apoptosis. These findings were confirmed and elucidated in more detail in rats. INTERPRETATION These results show for the first time that autophagy is enhanced in severe HIE in dying thalamic neurons of human newborns, as in rats. Experimental neuroprotective strategies targeting autophagy could thus be a promising lead to follow for the development of future therapeutic approaches.
Collapse
Affiliation(s)
- Vanessa Ginet
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Albertsson AM, Bi D, Duan L, Zhang X, Leavenworth JW, Qiao L, Zhu C, Cardell S, Cantor H, Hagberg H, Mallard C, Wang X. The immune response after hypoxia-ischemia in a mouse model of preterm brain injury. J Neuroinflammation 2014; 11:153. [PMID: 25187205 PMCID: PMC4172879 DOI: 10.1186/s12974-014-0153-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/14/2014] [Indexed: 01/04/2023] Open
Abstract
Background Preterm brain injury consists primarily of periventricular leukomalacia accompanied by elements of gray-matter injury, and these injuries are associated with cerebral palsy and cognitive impairments. Inflammation is believed to be an important contributing factor to these injuries. The aim of this study was to examine the immune response in a postnatal day (PND) 5 mouse model of preterm brain injury induced by hypoxia-ischemia (HI) that is characterized by focal white and gray-matter injury. Methods C57Bl/6 mice at PND 5 were subjected to unilateral HI induced by left carotid artery ligation and subsequent exposure to 10% O2 for 50 minutes, 70 minutes, or 80 minutes. At seven days post-HI, the white/gray-matter injury was examined. The immune responses in the brain after HI were examined at different time points after HI using RT-PCR and immunohistochemical staining. Results HI for 70 minutes in PND 5 mice induced local white-matter injury with focal cortical injury and hippocampal atrophy, features that are similar to those seen in preterm brain injury in human infants. HI for 50 minutes resulted in a small percentage of animals being injured, and HI for 80 minutes produced extensive infarction in multiple brain areas. Various immune responses, including changes in transcription factors and cytokines that are associated with a T-helper (Th)1/Th17-type response, an increased number of CD4+ T-cells, and elevated levels of triggering receptor expressed on myeloid cells 2 (TREM-2) and its adaptor protein DNAX activation protein of 12 kDa (DAP12) were observed using the HI 70 minute preterm brain injury model. Conclusions We have established a reproducible model of HI in PND 5 mice that produces consistent local white/gray-matter brain damage that is relevant to preterm brain injury in human infants. This model provides a useful tool for studying preterm brain injury. Both innate and adaptive immune responses are observed after HI, and these show a strong pro-inflammatory Th1/Th17-type bias. Such findings provide a critical foundation for future studies on the mechanism of preterm brain injury and suggest that blocking the Th1/Th17-type immune response might provide neuroprotection after preterm brain injury.
Collapse
|
34
|
Gonzales-Portillo GS, Reyes S, Aguirre D, Pabon MM, Borlongan CV. Stem cell therapy for neonatal hypoxic-ischemic encephalopathy. Front Neurol 2014; 5:147. [PMID: 25161645 PMCID: PMC4130306 DOI: 10.3389/fneur.2014.00147] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 07/22/2014] [Indexed: 11/27/2022] Open
Abstract
Treatments for neonatal hypoxic-ischemic encephalopathy (HIE) have been limited. The aim of this paper is to offer translational research guidance on stem cell therapy for neonatal HIE by examining clinically relevant animal models, practical stem cell sources, safety and efficacy of endpoint assays, as well as a general understanding of modes of action of this cellular therapy. In order to do so, we discuss the clinical manifestations of HIE, highlighting its overlapping pathologies with stroke and providing insights on the potential of cell therapy currently investigated in stroke, for HIE. To this end, we draw guidance from recommendations outlined in stem cell therapeutics as an emerging paradigm for stroke or STEPS, which have been recently modified to Baby STEPS to cater for the “neonatal” symptoms of HIE. These guidelines recognized that neonatal HIE exhibit distinct disease symptoms from adult stroke in need of an innovative translational approach that facilitates the entry of cell therapy in the clinic. Finally, new information about recent clinical trials and insights into combination therapy are provided with the vision that stem cell therapy may benefit from available treatments, such as hypothermia, already being tested in children diagnosed with HIE.
Collapse
Affiliation(s)
| | - Stephanny Reyes
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| | - Daniela Aguirre
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| | - Mibel M Pabon
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| |
Collapse
|
35
|
Kesavan K, Ezell T, Bierman A, Nunes AR, Northington FJ, Tankersley CG, Gauda EB. Breathing and temperature control disrupted by morphine and stabilized by clonidine in neonatal rats. Respir Physiol Neurobiol 2014; 201:93-100. [PMID: 25008573 DOI: 10.1016/j.resp.2014.06.015] [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] [Received: 12/18/2013] [Revised: 06/09/2014] [Accepted: 06/29/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Sedative-analgesics are often given to newborn infants and are known to affect many components of the autonomic nervous system. While morphine is most frequently used, α-2 adrenergic receptor agonists are being increasingly used in this population. Alpha-2 adrenergic receptors agonists also have anti-shivering properties which may make it a desirable drug to give to infants undergoing therapeutic hypothermia. The aim of this study was to systematically compare two different classes of sedative-analgesics, morphine, a μ-opioid receptor agonist, and clonidine an α-2 adrenergic receptor agonist on breathing, metabolism and core body temperature (CBT) in neonatal rodents. METHODS Breathing parameters, oxygen consumption (VO2) and carbon dioxide production (VCO2), were measured prior to, 10 and 90 min after intraperitoneal (IP) administration of morphine (2, 10 or 20 mg/kg), clonidine (40, 200 or 400 μg/kg), or saline in Sprague-Dawley rat pups at postnatal day 7 (p7) while continuously monitoring CBT. RESULTS Morphine reduced the respiratory rate, VO2 and VCO2 greater than clonidine at all dosages used (p<0.05, morphine vs. clonidine, for all metabolic and respiratory parameters). Furthermore, morphine induced prolonged respiratory pauses, which were not observed in animals treated with clonidine or saline. Morphine caused hypothermia which was dose dependent, while clonidine stabilized CBT in comparison to saline treated animals (p<0.0001). CONCLUSION In the newborn rat, morphine causes profound respiratory depression and hypothermia while clonidine causes minimal respiratory depression and stabilizes CBT. All together, we suggest that clonidine promotes autonomic stability and may be a desirable agent to use in infants being treated with therapeutic hypothermia.
Collapse
Affiliation(s)
- Kalpashri Kesavan
- Pediatrics, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Tarrah Ezell
- Pediatrics, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Alexis Bierman
- Environmental Health Sciences, School of Public Health of Johns Hopkins, Baltimore, MD, United States
| | | | | | - Clarke G Tankersley
- Environmental Health Sciences, School of Public Health of Johns Hopkins, Baltimore, MD, United States
| | - Estelle B Gauda
- Pediatrics, Johns Hopkins Medical Institutions, Baltimore, MD, United States.
| |
Collapse
|
36
|
Teo L, Bourne JA. A reproducible and translatable model of focal ischemia in the visual cortex of infant and adult marmoset monkeys. Brain Pathol 2014; 24:459-74. [PMID: 25469561 DOI: 10.1111/bpa.12129] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/21/2014] [Indexed: 12/12/2022] Open
Abstract
Models of ischemic brain injury in the nonhuman primate (NHP) are advantageous for investigating mechanisms of central nervous system (CNS) injuries and testing of new therapeutic strategies. However, issues of reproducibility and survivability persist in NHP models of CNS injuries. Furthermore, there are currently no pediatric NHP models of ischemic brain injury. Therefore, we have developed a NHP model of cortical focal ischemia that is highly reproducible throughout life to enable better understanding of downstream consequences of injury. Posterior cerebral arterial occlusion was induced through intracortical injections of endothelin-1 in adult (n = 5) and neonatal (n = 3) marmosets, followed by magnetic resonance imaging (MRI), histology and immunohistochemistry. MRI revealed tissue hyperintensity at the lesion site at 1-7 days followed by isointensity at 14-21 days. Peripheral macrophage and serum albumin infiltration was detected at 1 day, persisting at 21 days. The proportional loss of total V1 as a result of infarction was consistent in adults and neonates. Minor hemorrhagic transformation was detected at 21 days at the lesion core, while neovascularization was detected in neonates, but not in adults. We have developed a highly reproducible and survivable model of focal ischemia in the adult and neonatal marmoset primary visual cortex, demonstrating similar downstream anatomical and cellular pathology to those observed in post-ischemic humans.
Collapse
Affiliation(s)
- Leon Teo
- Australian Regenerative Medicine Institute, Monash University, Clayton, Vic., Australia
| | | |
Collapse
|
37
|
Kilicdag H, Daglioglu YK, Erdogan S, Zorludemir S. Effects of caffeine on neuronal apoptosis in neonatal hypoxic-ischemic brain injury. J Matern Fetal Neonatal Med 2014; 27:1470-5. [PMID: 24392823 DOI: 10.3109/14767058.2013.878694] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Hypoxia-ischemia (HI) in rat pups leads to strong activation of apoptosis, and apoptosis contributes significantly to cerebral damage in the perinatal period. Caffeine displays a broad array of actions on the brain. The aim of this study was to investigate the effects of caffeine on neuronal apoptosis in a hypoxic-ischemic neonatal model. METHODS Twenty-four seven-day-old Wistar rat pups were subjected to right common carotid artery ligation and hypoxia for 2 h. Sham group (n = 8) had a median neck incision, but the rats were not subjected to ligation or hypoxia. The pups were treated with 20 mg/kg/day caffeine citrate (n = 8) or saline (n = 8) immediately before HI and at 0, 24, 48 and 72 h post-hypoxia. Neuronal apoptosis was evaluated by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) and caspase-3 in the hippocampus and parietal cortex of both hemispheres. RESULTS The numbers of apoptotic cells in the hippocampus and parietal cortex were significantly higher in the saline group than they were in the sham group (p < 0.0001). The number of apoptotic cells in the hippocampus (p < 0.0001) and parietal cortex (p < 0.0001, TUNEL and p = 0.001, caspase-3) were higher in the caffeine-treated group than they were in the sham group, but the number of apoptotic cells decreased significantly in the caffeine-treated group compared with the saline group in the hippocampus (p < 0.0001, TUNEL and p = 0.001, caspase-3) and parietal cortex (p = 0.001, TUNEL and p = 0.002, caspase-3). CONCLUSIONS We show that caffeine administration in hypoxic-ischemic brain injury reduces neuronal apoptosis in the developing brain. We suggest that caffeine may be effective in reducing brain injury.
Collapse
Affiliation(s)
- Hasan Kilicdag
- Division of Neonatology, Department of Paediatrics, Acıbadem Hospital , Adana , Turkey
| | | | | | | |
Collapse
|
38
|
Brockmann MD, Kukovic M, Schönfeld M, Sedlacik J, Hanganu-Opatz IL. Hypoxia-ischemia disrupts directed interactions within neonatal prefrontal-hippocampal networks. PLoS One 2013; 8:e83074. [PMID: 24376636 PMCID: PMC3869754 DOI: 10.1371/journal.pone.0083074] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 11/07/2013] [Indexed: 02/04/2023] Open
Abstract
Due to improved survival rates and outcome of human infants experiencing a hypoxic-ischemic episode, cognitive dysfunctions have become prominent. They might result from abnormal communication within prefrontal-hippocampal networks, as synchrony and directed interactions between the prefrontal cortex and hippocampus account for mnemonic and executive performance. Here, we elucidate the structural and functional impact of hypoxic-ischemic events on developing prefrontal-hippocampal networks in an immature rat model of injury. The magnitude of infarction, cell loss and astrogliosis revealed that an early hypoxic-ischemic episode had either a severe or a mild/moderate outcome. Without affecting the gross morphology, hypoxia-ischemia with mild/moderate outcome diminished prefrontal neuronal firing and gamma network entrainment. This dysfunction resulted from decreased coupling synchrony within prefrontal-hippocampal networks and disruption of hippocampal theta drive. Thus, early hypoxia-ischemia may alter the functional maturation of neuronal networks involved in cognitive processing by disturbing the communication between the neonatal prefrontal cortex and hippocampus.
Collapse
Affiliation(s)
- Marco D. Brockmann
- Developmental Neurophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maja Kukovic
- Developmental Neurophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Schönfeld
- Department of Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Sedlacik
- Department of Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ileana L. Hanganu-Opatz
- Developmental Neurophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
39
|
Subchronic perinatal asphyxia in rats: Embryo–foetal assessment of a new model of oxidative stress during critical period of development. Food Chem Toxicol 2013; 61:233-9. [DOI: 10.1016/j.fct.2013.07.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/10/2013] [Accepted: 07/10/2013] [Indexed: 11/19/2022]
|
40
|
Aggarwal M, Burnsed J, Martin LJ, Northington FJ, Zhang J. Imaging neurodegeneration in the mouse hippocampus after neonatal hypoxia-ischemia using oscillating gradient diffusion MRI. Magn Reson Med 2013; 72:829-40. [PMID: 24123409 DOI: 10.1002/mrm.24956] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/31/2013] [Accepted: 08/22/2013] [Indexed: 12/26/2022]
Abstract
PURPOSE To investigate if frequency-dependent contrasts using oscillating gradient diffusion MRI (dMRI) can detect hypoxia-ischemia (HI) -induced neurodegeneration in the neonatal mouse hippocampus. METHODS Pulsed- and oscillating-gradient dMR images (at 50, 100, and 150 Hz) were acquired from postmortem fixed brains of mice exposed to neonatal HI using the Rice-Vanucci model. MRI data were acquired at 1, 4, and 8 days following HI, and compared with histological data from the same mice for in situ histological validation of the MRI findings. RESULTS The rate of change of apparent diffusion coefficient with gradient frequency (Δf ADC) revealed unique layer-specific contrasts in the neonatal mouse hippocampus. Δf ADC measurements were found to show a significant decrease in response to neonatal HI injury, in the pyramidal (Py) and granule (GrDG) cell layers compared with contralateral regions. The areas of reduced intensity in the Δf ADC maps corresponded to regional neurodegeneration seen with H&E and Fluoro-Jade C stainings, indicating that alterations in Δf ADC contrasts are sensitive to early microstructural changes due to HI-induced neurodegeneration in the studied regions. CONCLUSION The findings show that the frequency-dependence of ADC measurements with oscillating-gradient dMRI can provide a sensitive contrast to detect HI-induced neurodegeneration in neuronal layers of the neonatal mouse hippocampus.
Collapse
Affiliation(s)
- Manisha Aggarwal
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | |
Collapse
|
41
|
Kilicdag H, Daglıoglu K, Erdogan S, Guzel A, Sencar L, Polat S, Zorludemir S. The effect of levetiracetam on neuronal apoptosis in neonatal rat model of hypoxic ischemic brain injury. Early Hum Dev 2013; 89:355-60. [PMID: 23266150 DOI: 10.1016/j.earlhumdev.2012.12.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 11/29/2012] [Accepted: 12/01/2012] [Indexed: 12/15/2022]
Abstract
BACKGROUND Hypoxic-ischemic brain injury (HIBI) is a common cause of neonatal mortality and morbidity. The use of levetiracetam (LEV), as a potential neuroprotective in brain ischemia, receives an increasingly high attention, and it could have a crucial role in the regulation of epileptogenesis and neuroprotection. Potential effects of LEV on neuronal apoptosis in HIBI have not previously been reported in literature. OBJECTIVES The aim of this study is to evaluate the possible effects of LEV on neuronal apoptosis in neonatal rat model of HIBI. METHODS Seven-day-old Wistar rat pups were subjected to right common carotid artery ligation and hypoxia (92% nitrogen and 8% oxygen) for 2h. The pups were treated with LEV or saline after hypoxia. In sham group rats, neither ligation, nor hypoxia was performed. Neuronal apoptosis was evaluated by the terminal deoxynucleotidyl-transferase- mediated dUTP nick-end labeling (TUNEL) methods. RESULTS The counts of apoptotic cells in both hippocampus and cerebral cortex were significantly higher in the saline treatment group than in the sham group. The counts of apoptotic cells in both hippocampus and cerebral cortex were similar to those in the sham group and in the LEV treatment group. The number of apoptotic cells decreased significantly in the LEV-treated group compared with the saline group. CONCLUSIONS These results show that LEV administration after hypoxia reduces neuronal apoptosis. Thus, we propose that LEV, as an effective antiepileptic and antiapoptotic drug, may be a viable choice for the control of seizure activity in neonates with HIBI.
Collapse
Affiliation(s)
- Hasan Kilicdag
- Division of Neonatology, Department of Paediatrics, Acıbadem Hospital, Adana, Turkey.
| | | | | | | | | | | | | |
Collapse
|
42
|
Pluripotent possibilities: human umbilical cord blood cell treatment after neonatal brain injury. Pediatr Neurol 2013; 48:346-54. [PMID: 23583051 DOI: 10.1016/j.pediatrneurol.2012.10.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 10/29/2012] [Indexed: 12/14/2022]
Abstract
Perinatal hypoxic-ischemic brain injury and stroke in the developing brain remain important causes of chronic neurologic morbidity. Emerging data suggest that transplantation of umbilical cord blood-derived stem cells may have therapeutic potential for neuroregeneration and improved functional outcome. The pluripotent capacity of stem cells from the human umbilical cord blood provides simultaneous targeting of multiple neuropathologic events initiated by a hypoxic-ischemic insult. Their high regenerative potential and naïve immunologic phenotype makes them a preferable choice for transplantation. A multiplicity of transplantation protocols have been studied with a variety of brain injury models; however, only a few have been conducted on immature animals. Biological recipient characteristics, such as age and sex, appear to differentially modulate responses of the animals to the transplanted cord blood stem cells. Survival, migration, and function of the transplanted cells have also been studied and reveal insights into the mechanisms of cord blood stem cell effects. Data from preclinical studies have informed current clinical safety trials of human cord blood in neonates, and further work is needed to continue progress in this field.
Collapse
|
43
|
Shrivastava K, Llovera G, Recasens M, Chertoff M, Giménez-Llort L, Gonzalez B, Acarin L. Temporal expression of cytokines and signal transducer and activator of transcription factor 3 activation after neonatal hypoxia/ischemia in mice. Dev Neurosci 2013; 35:212-25. [PMID: 23571161 DOI: 10.1159/000348432] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 01/23/2013] [Indexed: 11/19/2022] Open
Abstract
Hypoxia/ischemia (HI) is a prevalent reason for neonatal brain injury with inflammation being an inevitable phenomenon following such injury; but there is a scarcity of data regarding the signaling pathway involved and the effector molecules. The signal transducer and activator of transcription factor 3 (STAT3) is known to modulate injury following imbalance between pro- and anti-inflammatory cytokines in peripheral and central nervous system injury making it a potential molecule for study. The current study investigates the temporal expression of interleukin (IL)-6, IL-1β, tumor necrosis factor-α, IL-1ra, IL-4, IL-10, IL-13 and phosphorylated STAT3 (pSTAT3) after carotid occlusion and hypoxia (8% O2, 55 min) in postnatal day 7 C57BL/6 mice from 3 h to 21 days after hypoxia. Protein array illustrated notable changes in cytokines expressed in both hemispheres in a time-dependent manner. The major pro-inflammatory cytokines showing immediate changes between ipsi- and contralateral hemispheres were IL-6 and IL-1β. The anti-inflammatory cytokines IL-4 and IL-13 demonstrated a delayed augmentation with no prominent differences between hemispheres, while IL-1ra showed two distinct peaks of expression spread over time. We also illustrate for the first time the spatiotemporal activation of pSTAT3 (Y705 phosphorylation) after a neonatal HI in mice brain. The main regions expressing pSTAT3 were the hippocampus and the corpus callosum. pSTAT3+ cells were mostly a subpopulation of activated astrocytes (GFAP+) and microglia/macrophages (F4/80+) seen only in the ipsilateral hemisphere at most time points studied (till 7 days after hypoxia). The highest expression of pSTAT3+ cells was observed to be around 24-48 h, where the presence of pSTAT3+ astrocytes and pSTAT3+ microglia/macrophages was seen by confocal micrographs. In conclusion, our study highlights a synchronized expression of some pro- and anti-inflammatory cytokines, especially in the long term not previously defined. It also points towards a significant role of STAT3 signaling following micro- and astrogliosis in the pathophysiology of neonatal HI-related brain injury. In the study, a shift from pro-inflammatory to anti-inflammatory cytokine profile was also noted as the injury progressed. We suggest that while designing efficient neuroprotective therapies using inflammatory molecules, the time of intervention and balance between the pro- and anti-inflammatory cytokines must be considered.
Collapse
Affiliation(s)
- K Shrivastava
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma Barcelona, Bellaterra, Spain.
| | | | | | | | | | | | | |
Collapse
|
44
|
Modèles animaux de la prématurité : mesures comportementales des effets des lésions cérébrales. ENFANCE 2013. [DOI: 10.4074/s0013754513001080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Pabon MM, Borlongan CV. ADVANCES IN THE CELL-BASED TREATMENT OF NEONATAL HYPOXIC-ISCHEMIC BRAIN INJURY. FUTURE NEUROLOGY 2013; 8:193-203. [PMID: 23565051 DOI: 10.2217/fnl.12.85] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stem cell therapy for adult stroke has reached limited clinical trials. Here, we provide translational research guidance on stem cell therapy for neonatal hypoxic-ischemic brain injury requiring a careful consideration of clinically relevant animal models, feasible stem cell sources, and validated safety and efficacy endpoint assays, as well as a general understanding of modes of action of this cellular therapy. To this end, we refer to existing translational guidelines, in particular the recommendations outlined in the consortium of academicians, industry partners and regulators called Stem cell Therapeutics as an Emerging Paradigm for Stroke or STEPS. Although the STEPS guidelines are directed at enhancing the successful outcome of cell therapy in adult stroke, we highlight overlapping pathologies between adult stroke and neonatal hypoxic-ischemic brain injury. We are, however, cognizant that the neonatal hypoxic-ischemic brain injury displays disease symptoms distinct from adult stroke in need of an innovative translational approach that facilitates the entry of cell therapy in the clinic. Finally, insights into combination therapy are provided with the vision that stem cell therapy may benefit from available treatments, such as hypothermia, already being tested in children diagnosed with hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Mibel M Pabon
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, Florida 33612 USA
| | | |
Collapse
|
46
|
Titomanlio L, Zanin A, Sachs P, Khaled J, Elmaleh M, Blanc R, Piotin M. Pediatric ischemic stroke: acute management and areas of research. J Pediatr 2013; 162:227-35.e1. [PMID: 23153863 DOI: 10.1016/j.jpeds.2012.09.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 08/08/2012] [Accepted: 09/10/2012] [Indexed: 11/28/2022]
Affiliation(s)
- Luigi Titomanlio
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris Diderot University, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
47
|
Roy V, Leroux P, Arabo A, Marret S, Gonzalez B. Modèles animaux de la prématurité : mesures comportementales des effets des lésions cérébrales. ENFANCE 2013. [DOI: 10.3917/enf1.131.0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
48
|
Leger PL, Bonnin P, Nguyen T, Renolleau S, Baud O, Charriaut-Marlangue C. Ischemic postconditioning fails to protect against neonatal cerebral stroke. PLoS One 2012; 7:e49695. [PMID: 23251348 PMCID: PMC3520965 DOI: 10.1371/journal.pone.0049695] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 10/11/2012] [Indexed: 11/18/2022] Open
Abstract
The lack of efficient neuroprotective strategies for neonatal stroke could be ascribed to pathogenic ischemic processes differentiating adults and neonates. We explored this hypothesis using a rat model of neonatal ischemia induced by permanent occlusion of the left distal middle cerebral artery combined with 50 min of occlusion of both common carotid arteries (CCA). Postconditioning was performed by repetitive brief release and occlusion (30 s, 1 and/or 5 min) of CCA after 50 min of CCA occlusion. Alternative reperfusion was generated by controlled release of the bilateral CCA occlusion. Blood-flow velocities in the left internal carotid artery were measured using color-coded pulsed Doppler ultrasound imaging. Cortical perfusion was measured using laser Doppler. Cerebrovascular vasoreactivity was evaluated after inhalation with the hypercapnic gas or inhaled nitric oxide (NO). Whatever the type of serial mechanical interruptions of blood flow at reperfusion, postconditioning did not reduce infarct volume after 72 hours. A gradual perfusion was found during early re-flow both in the left internal carotid artery and in the cortical penumbra. The absence of acute hyperemia during early CCA re-flow, and the lack of NO-dependent vasoreactivity in P7 rat brain could in part explain the inefficiency of ischemic postconditioning after ischemia-reperfusion.
Collapse
Affiliation(s)
- Pierre-Louis Leger
- Univ Paris Diderot, Sorbonne Paris Cité, INSERM, U676, Paris, France
- UPMC-Paris6, AP-HP, Hôpital Armand Trousseau, Service de Réanimation pédiatrique, Paris, France
| | - Philippe Bonnin
- Univ Paris Diderot, Sorbonne Paris Cité, AP-HP, Hôpital Lariboisière, Physiologie Clinique, Explorations-Fonctionnelles, Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, INSERM, U965, Paris, France
| | - Thao Nguyen
- Univ Paris Diderot, Sorbonne Paris Cité, INSERM, U676, Paris, France
| | - Sylvain Renolleau
- UPMC-Paris6, AP-HP, Hôpital Armand Trousseau, Service de Réanimation pédiatrique, Paris, France
| | - Olivier Baud
- Univ Paris Diderot, Sorbonne Paris Cité, INSERM, U676, Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, AP-HP, Service de Réanimation néonatale et pédiatrique, Hôpital Robert Debré, Paris, France
| | | |
Collapse
|
49
|
Brima T, Mikulecká A, Otáhal J. Impacts of perinatal induced photothrombotic stroke on sensorimotor performance in adult rats. Physiol Res 2012; 62:85-94. [PMID: 23173682 DOI: 10.33549/physiolres.932447] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Perinatal ischemic stroke is a leading cerebrovascular disorder occurring in infants around the time of birth associated with long term comorbidities including motor, cognitive and behavioral deficits. We sought to determine the impact of perinatal induced stroke on locomotion, behavior and motor function in rats. A photothrombotic model of ischemic stroke was used in rat at postnatal day 7. Presently, we induced two lesions of different extents, to assess the consequences of stroke on motor function, locomotion and possible correlations to morphological changes. Behavioral tests sensitive to sensorimotor changes were used; locomotion expressed as distance moved in the open field was monitored and histological changes were also assessed. Outcomes depicted two kinds of lesions of different shapes and sizes, relative to laser illumination. Motor performance of rats submitted to stroke was poor when compared to controls; a difference in motor performance was also noted between rats with small and large lesions. Correlations were observed between: motor performance and exposition time; volume ratio and exposition time; and in the rotarod between motor performance and volume ratio. Outcomes demonstrate that photothrombotic cerebral ischemic stroke induced in early postnatal period and tested in adulthood, indeed influenced functional performance governed by the affected brain regions.
Collapse
Affiliation(s)
- T Brima
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | |
Collapse
|
50
|
Short and long-term analysis and comparison of neurodegeneration and inflammatory cell response in the ipsilateral and contralateral hemisphere of the neonatal mouse brain after hypoxia/ischemia. Neurol Res Int 2012; 2012:781512. [PMID: 22701792 PMCID: PMC3372286 DOI: 10.1155/2012/781512] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Accepted: 02/02/2012] [Indexed: 12/21/2022] Open
Abstract
Understanding the evolution of neonatal hypoxic/ischemic is essential for novel neuroprotective approaches. We describe the neuropathology and glial/inflammatory response, from 3 hours to 100 days, after carotid occlusion and hypoxia (8% O2, 55 minutes) to the C57/BL6 P7 mouse. Massive tissue injury and atrophy in the ipsilateral (IL) hippocampus, corpus callosum, and caudate-putamen are consistently shown. Astrogliosis peaks at 14 days, but glial scar is still evident at day 100. Microgliosis peaks at 3–7 days and decreases by day 14. Both glial responses start at 3 hours in the corpus callosum and hippocampal fissure, to progressively cover the degenerating CA field. Neutrophils increase in the ventricles and hippocampal vasculature, showing also parenchymal extravasation at 7 days. Remarkably, delayed milder atrophy is also seen in the contralateral (CL) hippocampus and corpus callosum, areas showing astrogliosis and microgliosis during the first 72 hours. This detailed and long-term cellular response characterization of the ipsilateral and contralateral hemisphere after H/I may help in the design of better therapeutic strategies.
Collapse
|