51
|
Granulocyte-colony stimulating factor in combination with stem cell factor confers greater neuroprotection after hypoxic-ischemic brain damage in the neonatal rats than a solitary treatment. Transl Stroke Res 2012; 4:171-8. [PMID: 23565130 DOI: 10.1007/s12975-012-0225-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Neonatal hypoxia-ischemia (HI) is a devastating condition resulting in neuronal cell death and often culminates in neurological deficits. Granulocyte-colony stimulating factor (G-CSF) has been shown to have neuroprotective activity via inhibition of apoptosis and inflammation in various stroke models. Stem cell factor (SCF) regulates hematopoietic stem cells in the bone marrow and has been reported to have neuroprotective properties in an experimental ischemic stroke model. In this study we aim to determine the protective effects of G-CSF in combination with SCF treatment after experimental HI. Seven-day old Sprague-Dawley rats were subjected to unilateral carotid artery ligation followed by 2.5 hours of hypoxia. Animals were randomly assigned to five groups: Sham (n=8), Vehicle (n=8), HI with G-CSF treatment (n=9), HI with SCF treatment (n=9) and HI with G-CSF+SCF treatment (coadministration group; n=10). G-CSF (50 µg/kg), SCF (50 µg/kg) and G-CSF+SCF (50 µg/kg) were administered intraperitoneally 1 hour post HI followed by daily injection for 4 consecutive days (five total injections). Animals were euthanized 14 days after HI for neurological testing. Additionally assessment of brain, heart, liver, spleen and kidney atrophy was performed. Both G-CSF and G-CSF+SCF treatments improved body growth and decreased brain atrophy at 14 days post HI. No significant differences were found in the peripheral organ weights between groups. Finally, the G-CSF+SCF coadministration group showed significant improvement in neurological function. Our data suggest that administration of G-CSF in combination with SCF not only prevented brain atrophy but also significantly improved neurological function.
Collapse
|
52
|
Wasielewski B, Jensen A, Roth-Härer A, Dermietzel R, Meier C. Neuroglial activation and Cx43 expression are reduced upon transplantation of human umbilical cord blood cells after perinatal hypoxic-ischemic injury. Brain Res 2012; 1487:39-53. [PMID: 22796290 DOI: 10.1016/j.brainres.2012.05.066] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2012] [Accepted: 05/31/2012] [Indexed: 12/15/2022]
Abstract
Glial cells play a crucial role in the pathomechanism of perinatal hypoxic-ischemic brain injury (HI) and are involved in the maintenance of a chronic state of inflammation that causes delayed neuronal damage. Activation of astrocytes is one factor prolonging brain damage and contributing to the formation of a glial scar that limits neuronal plasticity. In this context, the major astrocytic gap junction protein Connexin 43 (Cx43) has been ascribed various functions including regulation of astrocytic migration and proliferation. Here, we investigate glial responses like microglia/macrophages and astrocytic activation in a rat model of neonatal HI and characterize changes of these parameters upon transplantation of human umbilical cord blood cells (hUCB). As an alleviation of motor function in lesioned rats has previously been described in transplanted animals, we analyze the putative correlation between motor function and glial activation over time. The lesion-induced impairment of motor function, assessed by forelimb use bias, muscle strength and distal spasticity, was alleviated upon transplantation of hUCB short and long term. HI induced an acute inflammatory reaction with activation of microglia/macrophages and reactive astrogliosis associated with perilesional upregulation of Cx43 that slowly declined during the chronic post-ischemic phase. hUCB transplantation accelerated the regression of inflammatory events, narrowed the perilesional astrocytic wall and led to a downregulation of the investigated astrocytic proteins. Thus, in the immature brain, hUCB may indirectly reduce secondary cell death upon hypoxia-ischemia and facilitate post-ischemic plasticity through the attenuation of reactive gliosis. This article is part of a Special Issue entitled Electrical Synapses.
Collapse
Affiliation(s)
- Bianca Wasielewski
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, D-44801 Bochum, Germany
| | | | | | | | | |
Collapse
|
53
|
Wang X, Guo S, Lu S, Zhou J, Li J, Xia S. Ultrasound-induced release of GDNF from lipid coated microbubbles injected into striatum reduces hypoxic-ischemic injury in neonatal rats. Brain Res Bull 2012; 88:495-500. [PMID: 22579834 DOI: 10.1016/j.brainresbull.2012.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 05/01/2012] [Accepted: 05/02/2012] [Indexed: 12/11/2022]
Abstract
Previous studies showed that inflammation and apoptosis were involved in the pathogenesis of hypoxic-ischemic brain injury. The immature brain is particularly vulnerable to damage. Intracerebral injection of glial cell line-derived neurotrophic factor (GDNF) has been shown to reduce the injury induced by hypoxia-ischemia (HI). In this study, the neuroprotective effect of intracerebral ultrasound-induced dissolution of lipid-coated GDNF microbubbles was investigated in a neonatal rat model of hypoxic-ischemic brain injury. Hypoxic-ischemic injury was induced in 7-day-old rats in the present study. The rats with hypoxia-ischemia received intracerebral injections of GDNF-containing microbubbles (0.5 mg/kg). They then received low frequency ultrasound stimulation (20 kHz, 2 h intervals for a total of 24 h and each time lasted for 1 min) to induce release of GDNF into the right striatum. We found that low frequency ultrasound stimulation can induce lipid-coated GDNF microbubbles to release GDNF. Ultrasound-induced dissolution of lipid-coated GDNF microbubbles treatment reduced infarction volume and improved neurological outcomes in neonatal rats. In the meanwhile, the microbubbles attenuated the production of inducible nitric oxide synthase, nitric oxide and tumor necrosis factor-alpha, as well as the activation of caspase-3 in insulted side of brain in neonatal rats. These data demonstrated that ultrasound-induced dissolution of lipid-coated GDNF microbubbles treatment can provide a neuroprotective effect against hypoxia-ischemia in neonatal rats.
Collapse
Affiliation(s)
- Xiaoying Wang
- Department of Ultrasound, Xuzhou Medical College Affiliated Hospital, Xuzhou, China.
| | | | | | | | | | | |
Collapse
|
54
|
Rodent neonatal germinal matrix hemorrhage mimics the human brain injury, neurological consequences, and post-hemorrhagic hydrocephalus. Exp Neurol 2012; 236:69-78. [PMID: 22524990 DOI: 10.1016/j.expneurol.2012.04.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 04/03/2012] [Accepted: 04/07/2012] [Indexed: 01/24/2023]
Abstract
Germinal matrix hemorrhage (GMH) is the most common neurological disease of premature newborns. GMH causes neurological sequelae such as cerebral palsy, post-hemorrhagic hydrocephalus, and mental retardation. Despite this, there is no standardized animal model of spontaneous GMH using newborn rats to depict the condition. We asked whether stereotactic injection of collagenase type VII (0.3 U) into the ganglionic eminence of neonatal rats would reproduce the acute brain injury, gliosis, hydrocephalus, periventricular leukomalacia, and attendant neurological consequences found in humans. To test this hypothesis, we used our neonatal rat model of collagenase-induced GMH in P7 pups, and found that the levels of free-radical adducts (nitrotyrosine and 4-hyroxynonenal), proliferation (mammalian target of rapamycin), inflammation (COX-2), blood components (hemoglobin and thrombin), and gliosis (vitronectin and GFAP) were higher in the forebrain of GMH pups, than in controls. Neurobehavioral testing showed that pups with GMH had developmental delay, and the juvenile animals had significant cognitive and motor disability, suggesting clinical relevance of the model. There was also evidence of white-matter reduction, ventricular dilation, and brain atrophy in the GMH animals. This study highlights an instructive animal model of the neurological consequences after germinal matrix hemorrhage, with evidence of brain injuries that can be used to evaluate strategies in the prevention and treatment of post-hemorrhagic complications.
Collapse
|
55
|
Paes-Branco D, Abreu-Villaça Y, Manhães AC, Filgueiras CC. Unilateral hemispherectomy at adulthood asymmetrically affects motor performance of male Swiss mice. Exp Brain Res 2012; 218:465-76. [PMID: 22367398 DOI: 10.1007/s00221-012-3034-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 02/06/2012] [Indexed: 01/23/2023]
Abstract
Evidence exists indicating that cerebral lateralization is a fundamental feature of all vertebrates. In humans, a series of studies demonstrated that the left hemisphere plays a major role in controlling movement. No such asymmetries have been identified in rodents, in spite of the fact that these animals have been frequently used in studies assessing motor behavior. In this regard, here, we used unilateral hemispherectomy to study the relative importance of each hemisphere in controlling movement. Adult Swiss mice were submitted to right unilateral hemispherectomy (RH), left unilateral hemispherectomy (LH) or sham surgery. Fifteen days after surgery, motor performance was assessed in the accelerating rotarod test and in the foot-fault test (in which performance depends on skilled limb use) and in the elevated body swing test (in which performance depends on trunk movements). The surgical removal of the right hemisphere caused a more pronounced impairment in performance than the removal of the left hemisphere both in the rotarod and in the foot-fault tests. In the rotarod, the RH group presented smaller latencies to fall than both LH and sham groups. In the foot-fault test, while both the sham and the LH groups showed no differences between left and right hind limbs, the RH group showed significantly worse performance with the left hind limb than with the right one. The elevated body swing test revealed a similar impairment in the two hemispherectomized groups. Our data suggest a major role of the right hemisphere in controlling skilled limb movements in mice.
Collapse
Affiliation(s)
- Danielle Paes-Branco
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Avenida Professor Manoel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | | | | | | |
Collapse
|
56
|
Sanches EF, Arteni NS, Spindler C, Moysés F, Siqueira IR, Perry ML, Netto CA. Effects of pre- and postnatal protein malnutrition in hypoxic-ischemic rats. Brain Res 2011; 1438:85-92. [PMID: 22244305 DOI: 10.1016/j.brainres.2011.12.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 12/09/2011] [Accepted: 12/13/2011] [Indexed: 11/17/2022]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HI) is a major cause of nervous system damage and neurological morbidity. Perinatal malnutrition affects morphological, biochemical and behavioral aspects of neural development, including pathophysiological cascades of cell death triggered by ischemic events, so modifying resulting brain damage. Female Wistar rats were subjected to protein restriction during pregnancy and lactation (control group: 25% soybean protein; malnourished group: 7%). Seven days after delivery (PND7), their offspring were submitted to unilateral cerebral HI; rats were then tested for sensorimotor (PND7 and PND60) and memory (PND60) functions. Offspring of malnourished mothers showed marked reduction in body weight starting in lactation and persisting during the entire period of observation. There was a greater sensorimotor deficit after HI in malnourished (M) animals, in righting reflex and in home bedding task, indicating an interaction between diet and hypoxia-ischemia. At PND60, HI rats showed impaired performance when compared to controls in training and test sessions of rota-rod task, however there was no effect of malnutrition per se. In the open field, nourished HI (HI-N) presented an increase in crossings number; this effect was not present in HI-M group. Surprisingly, HI-M rats presented a better performance in inhibitory avoidance task and a smaller hemispheric brain damage as compared to HI-N animals. Our data points to a possible metabolic adaptation in hypoxic-ischemic animals receiving protein malnutrition during pregnancy and lactation; apparently we observed a neuroprotective effect of diet, possibly decreasing the brain energy demand, under a hypoxic-ischemic situation.
Collapse
Affiliation(s)
- Eduardo Farias Sanches
- Programa de Pós-Graduação em Neurociências, ICBS, Universidade Federal do Rio Grande do Sul, Brazil.
| | | | | | | | | | | | | |
Collapse
|
57
|
Karalis F, Soubasi V, Georgiou T, Nakas CT, Simeonidou C, Guiba-Tziampiri O, Spandou E. Resveratrol ameliorates hypoxia/ischemia-induced behavioral deficits and brain injury in the neonatal rat brain. Brain Res 2011; 1425:98-110. [DOI: 10.1016/j.brainres.2011.09.044] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 09/20/2011] [Accepted: 09/22/2011] [Indexed: 10/17/2022]
|
58
|
Idrus NM, McGough NN, Spinetta MJ, Thomas JD, Riley EP. The effects of a single memantine treatment on behavioral alterations associated with binge alcohol exposure in neonatal rats. Neurotoxicol Teratol 2011; 33:444-50. [PMID: 21565269 PMCID: PMC3144286 DOI: 10.1016/j.ntt.2011.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 04/19/2011] [Accepted: 04/22/2011] [Indexed: 01/09/2023]
Abstract
BACKGROUND The third trimester in human fetal development represents a critical time of brain maturation referred to as the "brain growth spurt". This period occurs in rats postnatally, and exposure to ethanol during this time can increase the risk of impairments on a variety of cognitive and motor tasks. It has been proposed that one potential mechanism for the teratogenic effects of ethanol is NMDA receptor-mediated excitotoxicity during periods of ethanol withdrawal. In neonatal rats, antagonism of NMDA receptors during ethanol withdrawal, with drugs such as MK-801 and eliprodil, has been shown to mitigate some of the behavioral deficits induced by developmental ethanol exposure. The current study examined whether memantine, an NMDA receptor antagonist and a drug used clinically in Alzheimer's patients, would attenuate impairments associated with binge ethanol exposure in neonatal rats. METHODS On postnatal day 6, rats were exposed to 6 g/kg ethanol via intubation with controls receiving an isocaloric maltose dextrin solution. Twenty-one hours following the ethanol binge, rats received intraperitoneal injections of memantine at 0, 10, 15, or 20 mg/kg. Ethanol's teratogenic effects were assessed using multiple behavioral tasks: open field activity, parallel bars and spatial discrimination reversal learning. RESULTS Ethanol-treated rats were overactive in the open field and were impaired on both reversal learning and motor performance. Administration of 15 or 20 mg/kg memantine during withdrawal significantly attenuated ethanol's adverse effects on motor coordination, but did not significantly alter activity levels or improve the spatial learning deficits associated with neonatal alcohol exposure. CONCLUSION These results indicate that a single memantine administration during ethanol withdrawal can mitigate motor impairments but not spatial learning impairments or overactivity observed following a binge ethanol exposure during development in the rat.
Collapse
Affiliation(s)
- Nirelia M. Idrus
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, 6330 Alvarado Ct., Ste 100, San Diego, CA 92120, USA
| | - Nancy N.H. McGough
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, 6330 Alvarado Ct., Ste 100, San Diego, CA 92120, USA
| | - Michael J. Spinetta
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, 6330 Alvarado Ct., Ste 100, San Diego, CA 92120, USA
| | - Jennifer D. Thomas
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, 6330 Alvarado Ct., Ste 100, San Diego, CA 92120, USA
| | - Edward P. Riley
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, 6330 Alvarado Ct., Ste 100, San Diego, CA 92120, USA
| |
Collapse
|
59
|
Woodworth KN, Palmateer J, Swide J, Grafe MR. Short- and long-term behavioral effects of exposure to 21%, 40% and 100% oxygen after perinatal hypoxia-ischemia in the rat. Int J Dev Neurosci 2011; 29:629-38. [PMID: 21600973 DOI: 10.1016/j.ijdevneu.2011.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 04/21/2011] [Accepted: 05/04/2011] [Indexed: 01/22/2023] Open
Abstract
Until recently, supplementation with 100% oxygen was standard therapy for newborns who required resuscitation at birth or suffered later hypoxic-ischemic events. Exposure to high concentrations of oxygen, however, may worsen oxidative stress induced by ischemic injury. In this study we investigated the short- and long-term behavioral outcomes in rats that had undergone hypoxic-ischemic brain injury on postnatal day 7, followed by 2h exposure to 21%, 40%, or 100% oxygen, compared to normal controls. There were no differences in the development of walking, head lifting and righting reflexes from postnatal days 9 to 15. Cliff avoidance showed some abnormal responses in the H21 animals. From postnatal days 28 to 56, three tests of sensorimotor coordination were performed weekly: ledged tapered beam, cylinder, and bilateral tactile stimulation. The ledged tapered beam test without prior training of animals was sensitive to injury, but did not distinguish between treatment groups. The cylinder test showed a greater use of the unimpaired limb in female 21% and 40% oxygen groups compared to controls. Performance in both cylinder and the beam tests showed a correlation with the degree of brain injury. The bilateral tactile stimulation test showed that the male 21% oxygen groups had worse sensory asymmetry than male 40% or 100% oxygen groups, but was not statistically significantly different from controls. We thus found a minor benefit to post-hypoxia-ischemic treatment with 100% and 40% oxygen compared to 21% in one test of early motor skills. Our results for long-term sensorimotor behavior, however, showed conflicting results, however, as males treated with 40% or 100% oxygen had less sensory asymmetry (better performance) in the bilateral tactile stimulation test than males treated with 21% oxygen, while females had impaired motor performance in the cylinder test with both 21% and 40% oxygen.
Collapse
Affiliation(s)
- K Nina Woodworth
- Department of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, L113 Portland, OR 97239-3098, United States
| | | | | | | |
Collapse
|
60
|
Liu Y, Xue F, Liu G, Shi X, Liu Y, Liu W, Luo X, Sun X, Kang Z. Helium preconditioning attenuates hypoxia/ischemia-induced injury in the developing brain. Brain Res 2011; 1376:122-9. [DOI: 10.1016/j.brainres.2010.12.068] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 12/18/2010] [Accepted: 12/21/2010] [Indexed: 01/13/2023]
|
61
|
Song J, Chen L, Gu Y. Effect of ipsilateral C7 nerve root transfer on restoration of rat upper trunk muscle and nerve function after brachial plexus root avulsion. Orthopedics 2010; 33:886. [PMID: 21162507 DOI: 10.3928/01477447-20101021-12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The effects of ipsilateral cervical nerve root transfer on the restoration of the rat upper trunk muscle and nerve brachial plexus root avulsion were studied. After simulated root avulsion of the upper trunk brachial plexus, 120 rats were randomly divided into 4 groups: (A) ipsilateral C7 root transfer group; (B) Oberlin group; (C) phrenic nerve group; and (D) no axillary nerve restoration group. At 3, 6, and 12 weeks postoperatively, Ochiai score, Barth feet overreaching test, Terzis grooming test, and indices of neurotization were determined in 10 rats from each group. Twelve weeks postoperatively, nearly all the behavioral, neuroelectrophysiological, and histological outcomes of the axillary nerve and deltoid muscle and some of the indices of musculocutaneous nerve and biceps brachii function in the ipsilateral C7 group were superior to those in the other 3 groups. No significant difference was found between the ipsilateral C7 group and the other 3 groups in recovery rate of wet biceps muscle weight. No significant difference was found between the ipsilateral C7 group and the Oberlin group in the recovery of the axillary nerve compound muscle action potential and biceps brachii cell size. No significant difference was found between the ipsilateral C7 group and the phrenic nerve and no axillary nerve restoration groups in amplitude recovery rate of musculocutaneous nerve compound muscle action potential. No significant difference was found between the ipsilateral C7 and the Oberlin groups in the early recovery of musculocutaneous nerve compound muscle action potential, but recovery was significantly better in the ipsilateral C7 group at 12 weeks. Ipsilateral C7 root transfer can improve the quality of restoration of muscle and nerve function in the rat upper trunk after brachial plexus root avulsion.
Collapse
Affiliation(s)
- Jie Song
- Department of Hand Surgery, Shanghai Huashan Hospital, Shanghai, China
| | | | | |
Collapse
|
62
|
Rosenkranz K, Kumbruch S, Lebermann K, Marschner K, Jensen A, Dermietzel R, Meier C. The chemokine SDF-1/CXCL12 contributes to the 'homing' of umbilical cord blood cells to a hypoxic-ischemic lesion in the rat brain. J Neurosci Res 2010; 88:1223-33. [PMID: 19937807 DOI: 10.1002/jnr.22292] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Previous studies have shown that transplanted human umbilical cord blood (hUCB)-derived mononuclear cells exert therapeutic effects in various animal models of CNS impairments, including those of perinatal hypoxic-ischemic brain injury. However, the mechanisms of how transplanted cells exert their beneficial effects on the damaged tissue are still unclear. As detection of hUCB cells at the lesion site coincides with the therapeutic effects observed in our model, we investigated the role of the chemokine stromal derived factor (SDF)-1 (CXCL12) as a possible candidate for chemotaxis-mediated 'homing' of transplanted hUCB cells to a hypoxic-ischemic lesion in the perinatal rat brain. Following the hypoxic-ischemic insult expression of SDF-1 significantly increased in lesioned brain hemispheres and was mainly associated with astrocytes. Transplanted hUCB cells expressing the SDF-1 receptor CXCR4 migrated to the lesion site within one day. Inhibition of SDF-1 by application of neutralizing antibodies in vivo resulted in a significantly reduced number of hUCB cells at the lesioned area. The increase in glial SDF-1 expression shortly after induction of the lesion and hUCB cells expressing the corresponding receptor makes SDF-1 a potential chemotactic factor for hUCB cell migration. The reduction of hUCB cells present at the lesion site upon functional inhibition of SDF-1 strengthens the view that the SDF-1/CXCR4 axis is of major importance for cell 'homing'.
Collapse
Affiliation(s)
- Katja Rosenkranz
- Department of Neuroanatomy and Mol. Brain Research, Ruhr-University, Bochum, Germany
| | | | | | | | | | | | | |
Collapse
|
63
|
Zhou Y, Lekic T, Fathali N, Ostrowski RP, Martin RD, Tang J, Zhang JH. Isoflurane posttreatment reduces neonatal hypoxic-ischemic brain injury in rats by the sphingosine-1-phosphate/phosphatidylinositol-3-kinase/Akt pathway. Stroke 2010; 41:1521-7. [PMID: 20508187 DOI: 10.1161/strokeaha.110.583757] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND PURPOSE Isoflurane, administered before or during cerebral ischemia, has been shown to exhibit neuroprotection in animal models of ischemic stroke. However, the underlying mechanism remains to be elucidated. In the present study, we determined whether isoflurane posttreatment provides neuroprotection after neonatal hypoxia-ischemia (HI) in rats and evaluated the role of the sphingosine-1-phosphate/phosphatidylinositol-3-kinase/Akt pathway in this volatile anesthetic-mediated neuroprotection. METHODS HI was induced in postnatal day 10 (P10) rat pups by unilateral carotid ligation and 2 hours of hypoxia. For treatment, 2% isoflurane was administered immediately after HI for 1 hour. As pharmacological interventions, the sphingosine-1-phosphate antagonist VPC23019, phosphatidylinositol-3-kinase inhibitor wortmannin, or opioid antagonist naloxone was administered before HI. Isoflurane posttreatment was evaluated for effects on infarct volume at 48 hours after HI and brain atrophy and neurological outcomes at 4 weeks after HI. The expression of phosphorylated Akt and cleaved caspase-3 was determined by Western blotting and immunofluorescence analysis. RESULTS Isoflurane posttreatment significantly reduced infarct volume at 48 hours after HI. VPC23019 or wortmannin abrogated the neuroprotective effect of isoflurane, whereas naloxone did not inhibit the isoflurane-induced neuroprotection. Isoflurane posttreatment significantly preserved phosphorylated Akt expression and decreased cleaved caspase-3 levels. These effects were reversed by VPC23019 and wortmannin, respectively. Isoflurane also confers long-term neuroprotective effects against brain atrophy and neurological deficits at 4 weeks after HI. CONCLUSIONS Isoflurane posttreatment provides lasting neuroprotection against hypoxic-ischemic brain injury in neonatal rats. Activation of the sphingosine-1-phosphate/phosphatidylinositol-3-kinase/Akt pathway may play a key role in isoflurane posttreatment-induced neuroprotection.
Collapse
Affiliation(s)
- Yilin Zhou
- Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, Calif 92354, USA
| | | | | | | | | | | | | |
Collapse
|
64
|
Fathali N, Lekic T, Zhang JH, Tang J. Long-term evaluation of granulocyte-colony stimulating factor on hypoxic-ischemic brain damage in infant rats. Intensive Care Med 2010; 36:1602-8. [PMID: 20461500 DOI: 10.1007/s00134-010-1913-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 03/26/2010] [Indexed: 11/24/2022]
Abstract
PURPOSE Hypoxia-ischemia (HI), as a major cause of fetal brain damage, has long-lasting neurological implications. Therefore, therapeutic interventions that attenuate the neuropathological outcome of HI while also improving the neurofunctional outcome are of paramount clinical importance. The aim of this study was to investigate the long-term functional and protective actions of granulocyte-colony stimulating factor (G-CSF) treatment in an experimental model of cerebral HI. METHODS Postnatal day-7 Sprague-Dawley rats were subjected to HI surgery, which entailed ligation of the right common carotid artery followed by 2 h of hypoxia (8% O(2)). Treatment consisted of subcutaneous injection of G-CSF at 1 h after hypoxia followed by an additional one injection per day for 5 days (6 total injections) or for 10 days (11 total injections). Animals were euthanized 5 weeks post-insult for extensive evaluation of neurological deficits and assessment of brain, spleen, heart, and liver damage. RESULTS G-CSF treatment promoted somatic growth and prevented brain atrophy and underdevelopment of the heart. Moreover, reflexes, limb placing, muscle strength, motor coordination, short-term memory, and exploratory behavior were all significantly improved by both G-CSF dosing regimens. CONCLUSIONS Long-term neuroprotection afforded by G-CSF in both morphological and functional parameters after a hypoxic-ischemic event in the neonate provides a rationale for exploring clinical translation.
Collapse
Affiliation(s)
- Nancy Fathali
- Department of Human Pathology and Anatomy, Loma Linda University, Loma Linda, CA, USA
| | | | | | | |
Collapse
|
65
|
Cyclooxygenase-2 inhibition provides lasting protection against neonatal hypoxic-ischemic brain injury. Crit Care Med 2010; 38:572-8. [PMID: 20029340 DOI: 10.1097/ccm.0b013e3181cb1158] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate whether inhibition of cyclooxygenase-2, a critical component of the inflammatory pathway, is neuroprotective in a neonatal rat model of cerebral hypoxia-ischemia. The development of brain inflammation largely contributes to neonatal brain injury that may lead to a lifetime of neurologic deficits. DESIGN Laboratory investigation. SETTING University research laboratory. SUBJECTS Postnatal day ten Sprague-Dawley rats. INTERVENTIONS Neonatal hypoxia-ischemia was induced by ligation of the right common carotid artery followed by 2 hrs of hypoxia (8% oxygen). The pups in treatment groups were administered 10 mg/kg (low dose) or 30 mg/kg (high dose) of a known selective cyclooxygenase-2 inhibitor (NS398). Animals were euthanized at three time points: 72 hrs, 2 wks, or 6 wks. Inflammation outcomes were assessed at 72 hrs; brain damage was assessed at 2 wks and 6 wks along with other organs (heart, spleen). Detailed neurobehavioral examination was performed at 6 wks. MEASUREMENTS AND MAIN RESULTS Pharmacologic inhibition of cyclooxygenase-2 markedly increased survivability within the first 72 hrs compared with untreated rats (100% vs. 72%). Low- and high-dose NS398 significantly attenuated the loss of brain and body weights observed after hypoxia-ischemia. Neurobehavioral outcomes were significantly improved in some parameters with low-dose treatment, whereas high-dose treatment consistently improved all neurologic deficits. Immunohistochemical results showed a marked decrease in macrophage, microglial, and neutrophil abundance in ipsilateral hemisphere of the NS398-treated group along with a reduction in interleukin-6 expression. CONCLUSIONS Selective cyclooxygenase-2 inhibition protected neonatal rats against death, progression of brain injury, growth retardation, and neurobehavioral deficits after a hypoxic-ischemic insult.
Collapse
|
66
|
Lateralized and sex-dependent behavioral and morphological effects of unilateral neonatal cerebral hypoxia-ischemia in the rat. Behav Brain Res 2010; 210:92-8. [PMID: 20156487 DOI: 10.1016/j.bbr.2010.02.015] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2009] [Revised: 02/03/2010] [Accepted: 02/07/2010] [Indexed: 10/19/2022]
Abstract
Neonatal cerebral hypoxia-ischemia (HI) is an important cause of neurological deficits. The Levine-Rice model of unilateral HI is a useful experimental tool, but the resulting brain damage is mainly restricted to one hemisphere. Since the rat presents morphological and biochemical asymmetries between brain hemispheres, behavioral outcome from this model is probably dependent on which hemisphere is damaged. We here investigated the effects of sex and lesioned hemisphere on the outcome of open field, plus maze, inhibitory avoidance and water maze tasks in adult rats previously submitted to neonatal unilateral HI. Females were more active than males in some of studied parameters and males presented better spatial learning. Hypoxia-ischemia caused spatial deficits independently of sex or damaged hemisphere. Right-HI increased locomotion only in males and caused working memory in females and on aversive learning in both males and females. Morphological analysis showed that right-HI animals presented greater reduction of ipsilateral striatum area, with females being more affected. Interestingly, males showed greater hippocampal volume. These results show that task performance and cerebral damage extension are lateralized and sex-dependent, and that the right hemisphere, irrespective of sex, is more vulnerable to neonatal cerebral hypoxia-ischemia.
Collapse
|
67
|
Zheng XR, Zhang SS, Yang YJ, Yin F, Wang X, Zhong L, Yu XH. Adenoviral vector-mediated transduction of VEGF improves neural functional recovery after hypoxia-ischemic brain damage in neonatal rats. Brain Res Bull 2009; 81:372-7. [PMID: 19945513 DOI: 10.1016/j.brainresbull.2009.11.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2009] [Revised: 11/22/2009] [Accepted: 11/22/2009] [Indexed: 01/17/2023]
Abstract
Previous studies have showed that vascular endothelial growth factor (VEGF) displayed neurotrophic and neuroprotective activities. To examine whether target delivery of VEGF gene directly into brain may prevent ischemic brain damage, the VEGF expression adenoviral vectors, AVHP.VEGF-with 476bp of the human preproendothelin-1 (ppET-1) promoter and 35bp of the hypoxia-reponse element (HRE) driving VEGF expression and CMV.VEGF were transferred into hypoxic-induced ischemic (HI) rat brains. Seven-day-old rats that were underwent left carotid ligation followed by 2h of hypoxic stress (8% O(2) at 37 degrees C) were received VEGF adenoviral vectors or buffer (PBS) injection 3 days after HI. The body weight, VEGF expression, neuronal apoptosis, cerebral morphology and brain functional assays were performed between 7 and 28 days after HI. There were remarkable increases in the body weight and VEGF protein expression, and decrease in the number of TUNEL-positive cells in the VEGF vector groups as compared with PBS group. The VEGF vector groups also had better brain functional performs than PBS group. The better performs by the animals that received VEGF vectors may be directly linked to the inhibitory effect of VEGF on neuronal apoptosis because the animals had less neural loss in the cortex and hippocampal CA1 region as compared with PBS group. Overall, these results indicated that over-expression of VEGF in the brain exerted a neuroprotective effect and promoted neural functional recovery in neonatal rats after hypoxic-ischemic brain damage, suggesting that in vivo target VEGF gene transfer to brain may be a promising approch for the treatment of such implications.
Collapse
Affiliation(s)
- Xiang-Rong Zheng
- Department of Pediatrics, XiangYa Hospital, Central South University, Changsha 410008, Hunan, China.
| | | | | | | | | | | | | |
Collapse
|
68
|
Liu C, Lin N, Wu B, Qiu Y. Neuroprotective effect of memantine combined with topiramate in hypoxic-ischemic brain injury. Brain Res 2009; 1282:173-82. [PMID: 19501064 DOI: 10.1016/j.brainres.2009.05.071] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2009] [Revised: 05/19/2009] [Accepted: 05/20/2009] [Indexed: 02/05/2023]
Abstract
Glutamate receptor-mediated neurotoxicity is a major mechanism contributing to hypoxic-ischemic brain injury (HIBI). Memantine is a safe non-competitive NMDA receptor blocker characterized by its low affinity and fast unblocking kinetics. Topiramate is an AMPA/KA receptor blocker and use-dependent sodium channel blocker with several other neuroprotective actions and little neurotoxicity. We hypothesized that the coadministration of memantine and topiramate would be highly effective to attenuate HIBI in neonatal rats. Seven-day-old Sprague-Dawley rat pups were subjected to right common carotid artery ligation and hypoxia for 2 h, and then were randomly and blindly assigned to one of four groups: vehicle, memantine, topiramate and combination group. Brain injury was evaluated by gross damage and weight deficit of the right hemisphere at 22d after hypoxic-ischemia (HI) and by neurofunctional assessment (foot-fault test) at 21d post-HI. Acute neuronal injury was also evaluated by microscopic damage grading at 72 h post-HI. Results showed the combination of memantine and topiramate improved both pathological outcome and performance significantly. The drug-induced apoptotic neurodegeneration was assessed by TUNEL staining at 48 h post-HI and the result showed no elevated apoptosis in all observed areas. The result of the experiment indicates the combination therapy is safe and highly effective to reduce brain damage after HIBI.
Collapse
Affiliation(s)
- Chunhua Liu
- Department of Pediatrics, The First Affiliated Hospital of Shantou University Medical College, 515000, Shantou, China
| | | | | | | |
Collapse
|
69
|
Rodent neonatal bilateral carotid artery occlusion with hypoxia mimics human hypoxic-ischemic injury. J Cereb Blood Flow Metab 2009; 29:1305-16. [PMID: 19436315 DOI: 10.1038/jcbfm.2009.56] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We report a new clinically relevant model of neonatal hypoxic-ischemic injury in a 10-day-old rat pup. Bilateral carotid artery occlusion and 8% hypoxia (1 to 15 mins, BCAO-H) was induced with varying degrees of injury (mild, moderate, severe), which was quantified using magnetic resonance imaging including diffusion-weighted and T2-weighted imaging at 24 h and 21/28 days. We developed a magnetic resonance imaging-based rat pup severity score and compared 3D ischemic injury volumes/rat pup severity score with histology and behavioral testing. At 24 h, hypoxic-ischemic injury was observed in 17/27 animals; long-term survival was 81%. Magnetic resonance imaging lesion volumes did not correlate with hypoxia duration but correlated with rat pup severity score, which was used to classify animals into mild (n=21), moderate (n=6), and severe (n=10) groups with average brain lesion volumes of 0.9%, 33.2%, and 56.3%, respectively. Histology confirmed lesion location and histologic scoring correlated with the rat pup severity score. We also found excellent correlation between injury severity and multiple behavioral tasks. Bilateral carotid artery occlusion and hypoxia in the P10 rat pup is an excellent model of neonatal hypoxic-ischemic injury because it induces diffuse global injury similar to the term infant. This model can produce graded injury severity, similar to that seen in human neonates, but manipulation with hypoxia duration is unpredictable.
Collapse
|
70
|
Chan KC, Khong P, Lau H, Cheung P, Wu EX. Late measures of microstructural alterations in severe neonatal hypoxic–ischemic encephalopathy by MR diffusion tensor imaging. Int J Dev Neurosci 2009; 27:607-15. [DOI: 10.1016/j.ijdevneu.2009.05.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 05/07/2009] [Accepted: 05/22/2009] [Indexed: 01/02/2023] Open
Affiliation(s)
- Kevin C. Chan
- Laboratory of Biomedical Imaging and Signal ProcessingThe University of Hong KongPokfulamHong Kong SARChina
- Department of Electrical and Electronic EngineeringThe University of Hong KongPokfulamHong Kong SARChina
| | - Pek‐lan Khong
- Department of Diagnostic RadiologyThe University of Hong KongPokfulamHong Kong SARChina
| | - Ho‐fai Lau
- Laboratory of Biomedical Imaging and Signal ProcessingThe University of Hong KongPokfulamHong Kong SARChina
- Department of Electrical and Electronic EngineeringThe University of Hong KongPokfulamHong Kong SARChina
| | - Pik‐to Cheung
- Department of PediatricsThe University of Hong KongPokfulamHong Kong SARChina
| | - Ed X. Wu
- Laboratory of Biomedical Imaging and Signal ProcessingThe University of Hong KongPokfulamHong Kong SARChina
- Department of Electrical and Electronic EngineeringThe University of Hong KongPokfulamHong Kong SARChina
- Department of AnatomyThe University of Hong KongPokfulamHong Kong SARChina
| |
Collapse
|
71
|
Zhou Y, Fathali N, Lekic T, Tang J, Zhang JH. Glibenclamide improves neurological function in neonatal hypoxia-ischemia in rats. Brain Res 2009; 1270:131-9. [PMID: 19306849 DOI: 10.1016/j.brainres.2009.03.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 03/03/2009] [Accepted: 03/05/2009] [Indexed: 11/26/2022]
Abstract
Recent studies demonstrated that sulfonylurea receptor 1 (SUR 1) regulated nonselective cation channel, the NC(Ca-ATP) channel, is involved in brain injury in rodent models of stroke. Block of SUR 1 with sulfonylurea such as glibenclamide has been shown to be highly effective in reducing cerebral edema, infarct volume and mortality in adult rat models of ischemic stroke. In this study, we tested glibenclamide in both severe and moderate models of neonatal hypoxia-ischemia (HI) in postnatal day 10 Sprague-Dawley rat pups. A total of 150 pups were used in the present study. Pups were subjected to unilateral carotid artery ligation followed by 2.5 or 2 h of hypoxia in the severe and moderate HI models, respectively. In the severe HI model, glibenclamide, administered immediately after HI and on postoperative Day 1, was not effective in attenuating short-term effects (brain edema and infarct volume) or long-term effects (brain weight and neurological function) of neonatal HI. In the moderate HI model, when injected immediately after HI and on postoperative Day 1, glibenclamide at 0.01 mg/kg improved several neurological parameters at 3 weeks after HI. We conclude that glibenclamide provided some long-term neuroprotective effect after neonatal HI.
Collapse
Affiliation(s)
- Yilin Zhou
- Department of Anesthesiology, Loma Linda University, Loma Linda, California 92354, USA
| | | | | | | | | |
Collapse
|
72
|
Leonardo CC, Hall AA, Collier LA, Gottschall PE, Pennypacker KR. Inhibition of gelatinase activity reduces neural injury in an ex vivo model of hypoxia-ischemia. Neuroscience 2009; 160:755-66. [PMID: 19272421 DOI: 10.1016/j.neuroscience.2009.02.080] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 02/25/2009] [Accepted: 02/27/2009] [Indexed: 11/15/2022]
Abstract
Perinatal hypoxia-ischemia (H-I) often manifests as cognitive and/or motor disturbances that appear early in development. Growing evidence indicates that neuroinflammation may exacerbate H-I injury. Resident microglia release proinflammatory cytokines and proteases in response to ischemia. Matrix metalloproteinases (MMPs), in particular, activate cytokines and degrade basement membrane proteins. These actions ultimately permit entry of peripheral leukocytes into the CNS neuropil, enhancing neuroinflammation and cell death. Currently, the relative contributions of resident and peripheral immune cells to ischemic brain injury are unclear. The present study employed an ex vivo model of H-I through oxygen glucose deprivation (OGD) to identify the cellular localization of MMP-9 in organotypic hippocampal slices from rat, and to determine whether inhibiting gelatin-degrading MMPs affords neuroprotection in the absence of peripheral immune cells. Immunohistochemistry revealed ubiquitous neuronal MMP-9 expression in both normoxic and hypoxic slices. Increased MMP-9 expression was detected in CD11b-positive microglia after 48 h exposure to OGD relative to normoxic controls. Consistent with these data, in situ zymography showed increased gelatinolytic activity after OGD. Gelatin-cleaved fluorescence localized to astrocytic processes and somata of various cellular morphologies. Treatment with either the MMP inhibitor AG3340 (prinomastat) or minocycline dampened OGD-induced gelatinolytic activity and neural injury, as measured by Fluoro-Jade staining, relative to vehicle controls. These results show that resident microglia, in the absence of peripheral immune cells, were sufficient to enhance neural injury after OGD in the organotypic hippocampal slice. Additionally, these effects were associated with upregulation or secretion of MMP-9, and were blocked after treatment with either the gelatinase-selective compound AG3340 or the anti-inflammatory compound minocycline. These data, coupled with the effectiveness of these compounds previously shown in vivo, support the selective targeting of gelatin-degrading MMPs and activated microglia as potential therapeutic approaches to combat neonatal H-I injury.
Collapse
Affiliation(s)
- C C Leonardo
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, College of Medicine, University of South Florida, 12901 Bruce B. Downs Boulevard, Tampa, FL 33612, USA
| | | | | | | | | |
Collapse
|
73
|
Animal models of perinatal hypoxic-ischemic brain damage. Pediatr Neurol 2009; 40:156-67. [PMID: 19218028 DOI: 10.1016/j.pediatrneurol.2008.10.025] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 10/02/2008] [Accepted: 10/06/2008] [Indexed: 12/22/2022]
Abstract
Animal models are often presumably the first step in determining mechanisms underlying disease, and the approach and effectiveness of therapeutic interventions. Perinatal brain damage, however, evolves over months of gestation, during the rapid maturation of the fetal and newborn brain. Despite marked advances in our understanding of these processes and technologic advances providing an improved window on the timing and duration of injury, neonatal brain injury remains a "moving target" regarding our ability to "mimic" its processes in an animal model. Moreover, interfering with normal processes of development as part of a therapeutic intervention may do "more harm than good." Hence, controversy continues over which animal model can reflect human disease states. Numerous models have provided information regarding the pathophysiology of brain damage in term and preterm infants. Our challenges consist of identifying infants at greatest risk for permanent injury, identifying the timing of injury, and adapting therapies that provide more benefit than harm. A combination of appropriately suitable animal models to conduct these studies will bring us closer to understanding human perinatal damage and the means to treat it.
Collapse
|
74
|
Sasaoka N, Kawaguchi M, Kawaraguchi Y, Nakamura M, Konishi N, Patel H, Patel PM, Furuya H. Isoflurane exerts a short-term but not a long-term preconditioning effect in neonatal rats exposed to a hypoxic-ischaemic neuronal injury. Acta Anaesthesiol Scand 2009; 53:46-54. [PMID: 19032558 DOI: 10.1111/j.1399-6576.2008.01822.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Isoflurane has been shown to induce tolerance against ischaemic injury in adult rodents. Although the delayed preconditioning effect of isoflurane has been demonstrated in neonatal rat pups, the acute preconditioning effects of isoflurane remained undetermined. The present study was therefore conducted to evaluate the acute preconditioning efficacy of isoflurane in neonatal rats subjected to a hypoxic-ischaemic (HI) injury. METHODS Post-natal day 7 pups were exposed to 1 or 2% isoflurane in oxygen for either 30, 60 or 90 min. Fifteen minutes after isoflurane exposure, the pups were subjected to an HI injury induced by left common carotid artery ligation and exposure to 8% oxygen for 2 h. Pups not exposed to isoflurane or not subjected to HI served as controls. Histopathologic injury to the cortex and hippocampus was evaluated 7 and 49 days after HI. RESULTS Isoflurane 2% exposure for 60 or 90 min before HI induced tolerance in the hippocampus and the number of normal neurons in the CA1 sector 7 days after HI was significantly greater than in non-preconditioned animals. This protective efficacy of isoflurane preconditioning was not observed 49 days after HI. CONCLUSIONS Exposure of 2% isoflurane for at least 60 min is required to induce tolerance against HI injury in rat pups. However, this neuroprotective efficacy results in only transient neuroprotection.
Collapse
Affiliation(s)
- N Sasaoka
- Department of Anesthesiology, Nara Medical University, Nara, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Neuroprotective effects of hydrogen saline in neonatal hypoxia-ischemia rat model. Brain Res 2008; 1256:129-37. [PMID: 19063869 DOI: 10.1016/j.brainres.2008.11.048] [Citation(s) in RCA: 184] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 11/10/2008] [Accepted: 11/10/2008] [Indexed: 11/24/2022]
Abstract
Cerebral hypoxia-ischemia (HI) represents a major cause of brain damage in the term newborn. This study aimed to examine the short and long-term neuroprotective effect of hydrogen saline (H(2) saline) using an established neonatal HI rat pup model. Seven-day-old rat pups were subjected to left common carotid artery ligation and then 90 min hypoxia (8% oxygen at 37 degrees C). H(2) saturated saline was administered by peritoneal injection (5 ml/kg) immediately and again at 8 h after HI insult. At 24 h after HI, the pups were decapitated and brain morphological injury was assessed by 2,3,5-triphenyltetrazolium chloride (TTC), Nissl, and TUNEL staining. Acute cell death, inflammation and oxidative stress were evaluated at 24 h by studying caspase-3 activity, MDA measurement as well as Iba-1 immunochemistry in the brain. At 5 weeks after HI, spontaneous activity test and Morris water maze test were conducted. We observed that H(2) saline treatment reduced the caspase activity, MDA, Iba-1 levels, the infarct ratio, and improved the long-term neurological and neurobehavioral functions. H(2) saline has potentials in the clinical treatment of HI and other ischemia-related cerebral diseases.
Collapse
|
76
|
Tuor UI, Meng S, Qiao M, Webster NB, Crowley SM, Dyck RH, Tomanek B. Differential progression of magnetization transfer imaging changes depending on severity of cerebral hypoxic-ischemic injury. J Cereb Blood Flow Metab 2008; 28:1613-23. [PMID: 18506197 DOI: 10.1038/jcbfm.2008.49] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We hypothesized that magnetic resonance magnetization transfer (MT) imaging would be sensitive for detecting cerebral ischemic injury in white matter of neonatal brain. We compared the progression of changes in T(2) and the MT ratio (MTR) after cerebral hypoxic-ischemic insults of differing severity in neonatal rats. Magnetization transfer imaging parameters were first optimized, and then MTR and T(2) maps were acquired at various times after a mild (rather selective white matter) or substantial insult produced by unilateral cerebral hypoxia-ischemia. Depending on insult severity, time after insult, and region (e.g., subcortical white matter or cortex), cerebral hypoxia-ischemia produced reductions in MTR and an increase in T(2). The exception was acutely at 1 to 5 h at which time points MTR was reduced ipsilaterally in white matter, whereas T(2) was not affected significantly. Progression of imaging changes differed in rats grouped according to whether gross damage was present after chronic recovery. Behavioral changes were generally associated with chronic reductions in MTR and gross brain damage. Magnetization transfer imaging was capable of early detection of hypoxic-ischemic injury and particularly sensitive for identifying the progression of cerebral injury in white matter. Magnetization transfer ratio has potential for assisting with early diagnosis and treatment assessment for infants affected by perinatal hypoxia-ischemia.
Collapse
Affiliation(s)
- Ursula I Tuor
- MR Technology, Institute for Biodiagnostics (West), Calgary, Alberta, Canada.
| | | | | | | | | | | | | |
Collapse
|
77
|
Leonardo CC, Eakin AK, Ajmo JM, Collier LA, Pennypacker KR, Strongin AY, Gottschall PE. Delayed administration of a matrix metalloproteinase inhibitor limits progressive brain injury after hypoxia-ischemia in the neonatal rat. J Neuroinflammation 2008; 5:34. [PMID: 18694515 PMCID: PMC2527306 DOI: 10.1186/1742-2094-5-34] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Accepted: 08/11/2008] [Indexed: 12/01/2022] Open
Abstract
Background Hypoxia-ischemia (H-I) can produce widespread neurodegeneration and deep cerebral white matter injury in the neonate. Resident microglia and invading leukocytes promote lesion progression by releasing reactive oxygen species, proteases and other pro-inflammatory mediators. After injury, expression of the gelatin-degrading matrix metalloproteinases (MMPs), MMP-2 and MMP-9, are thought to result in the proteolysis of extracellular matrix (ECM), activation of cytokines/chemokines, and the loss of vascular integrity. Thus, therapies targeting ECM degradation and progressive neuroinflammation may be beneficial in reducing H-I – induced neuropathy. Minocycline has MMP-inhibitory properties and is both anti-inflammatory and neuroprotective. AG3340 (prinomastat) is an MMP inhibitor with high selectivity for the gelatinases. The purpose of this study was to determine whether these compounds could limit H-I – induced injury when administered at a delayed time point. Methods Sprague-Dawley rats were exposed to H-I at postnatal day 7 (P7), consisting of unilateral carotid artery ligation followed by 90 min exposure to 8% O2. Minocycline, AG3340, or vehicle were administered once daily for 6 days, beginning 24 hours after insult. Animals were sacrificed at P14 for neurohistological assessments. Immunohistochemistry was performed to determine the degree of reactive astrogliosis and immune cell activation/recruitment. Neural injury was detected using the Fluoro-Jade stain, a marker that identifies degenerating cells. Results CD11b and glial fibrillary acidic protein (GFAP) immunopositive cells increased in ipsilateral cortex after treatment with vehicle alone, demonstrating microglia/macrophage recruitment and reactive astrogliosis, respectively. Fluoro-Jade staining was markedly increased throughout the fronto-parietal cortex, striatum and hippocampus. Treatment with minocycline or AG3340 inhibited microglia/macrophage recruitment, attenuated astrogliosis and reduced Fluoro-Jade staining when compared to vehicle alone. Conclusion The selective gelatinase inhibitor AG3340 showed equal efficacy in reducing neural injury and dampening neuroinflammation when compared to the anti-inflammatory compound minocycline. Thus, MMP-2 and MMP-9 may be viable therapeutic targets to treat neonatal brain injury.
Collapse
Affiliation(s)
- Christopher C Leonardo
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | | | | | | | | | | | | |
Collapse
|
78
|
Wang P, Wang WP, Sun-Zhang, Wang HX, Yan-Lou, Fan YH. Impaired spatial learning related with decreased expression of calcium/calmodulin-dependent protein kinase IIalpha and cAMP-response element binding protein in the pentylenetetrazol-kindled rats. Brain Res 2008; 1238:108-17. [PMID: 18710651 DOI: 10.1016/j.brainres.2008.07.103] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 07/12/2008] [Accepted: 07/21/2008] [Indexed: 11/25/2022]
Abstract
Although its effect on cognitive functions has been one of the hot topics in the present neuroscience research, the mechanism of epilepsy related cognitive impairment is not clearly revealed. Intracellular Ca(2+) plays an important role in regulating many cellular functions including learning and memory, this experiment was therefore conducted, in which, we observed the behaviors of chronic epileptic rats kindled by pentylenetetrazol (PTZ) through Morris water maze (MWM), examined the concentration of intracellular free calcium ([Ca(2+)](i)) with flow cytometry, and tested the expression of calcium/calmodulin-dependent protein kinase IIalpha (CaMKII(alpha)) and cAMP-response element binding protein (CREB) in hippocampus of those rats using western blot and reverse transcription-polymerase chain reaction (RT-PCR). The results demonstrate impaired ability of spatial learning and memory, increased concentration of [Ca(2+)](i), decreased expression levels of total CaMKII(alpha), phosphorylated CaMKII(alpha) (P-CaMKII(alpha)) and phosphorylated CREB (P-CREB) and decreased levels of CaMKII(alpha) mRNA and CREB mRNA of the epileptic rats compared with the normal control rats. Moreover, Nimodipine, an inhibitor of voltage-dependent L-type Ca(2+) channels (VDCCs), reduced the Racine's stage, improved the ability of spatial learning and memory, reversed the effect of Ca(2+) influx and expression levels of CaMKII(alpha) and CREB of the epileptic rats. We concluded that Ca(2+) influx, CaMKII(alpha) and CREB expression levels in hippocampus of chronic epileptic rats may be related with their impaired spatial learning and memory.
Collapse
Affiliation(s)
- Pei Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| | | | | | | | | | | |
Collapse
|
79
|
Jones NM, Kardashyan L, Callaway JK, Lee EM, Beart PM. Long-term functional and protective actions of preconditioning with hypoxia, cobalt chloride, and desferrioxamine against hypoxic-ischemic injury in neonatal rats. Pediatr Res 2008; 63:620-4. [PMID: 18317402 DOI: 10.1203/pdr.0b013e31816d9117] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Preconditioning with hypoxia and hypoxia-mimetic compounds cobalt chloride (CoCl2) and desferrioxamine (DFX) protects against hypoxic-ischemic (HI) injury in neonatal rat brain. We examined long-term functional and protective actions of preconditioning induced by hypoxia, CoCl(2) and DFX in a neonatal rat model of HI. Postnatal day six rat pups were exposed to preconditioning with hypoxia (8% oxygen) or injections of CoCl(2), DFX or saline vehicle and 24 h later rats underwent HI or sham surgery. Behavioral tests were performed and at the conclusion of experiments, brains removed for morphologic analyses. HI resulted in a large unilateral lesion in the ipsilateral hemisphere compared with sham control rats. All preconditioning treatments significantly reduced the total lesion volume. Behavioral deficits were observed in HI rats compared with sham controls. The reduction in forelimb grasping strength in HI rats was attenuated by preconditioning with hypoxia, CoCl(2) and DFX. HI increased the number of foot faults in a grid-walking test and resulted in forelimb asymmetry in the cylinder test. Only preconditioning with hypoxia reversed all three functional deficits after HI. These findings indicate that preconditioning, especially when induced by hypoxia, has the potential to minimize the morphologic and functional effects of neonatal HI injury.
Collapse
Affiliation(s)
- Nicole M Jones
- Brain Injury and Repair Group, Howard Florey Institute, University of Melbourne, VIC 3010 Melbourne, Victoria, Australia.
| | | | | | | | | |
Collapse
|
80
|
Wang XL, Zhao YS, Yang YJ, Xie M, Yu XH. Therapeutic window of hyperbaric oxygen therapy for hypoxic-ischemic brain damage in newborn rats. Brain Res 2008; 1222:87-94. [PMID: 18582850 DOI: 10.1016/j.brainres.2008.05.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Revised: 04/24/2008] [Accepted: 05/05/2008] [Indexed: 01/08/2023]
Abstract
Previous studies showed that hyperbaric oxygen (HBO) promoted cell proliferation in hypoxic-ischemic (HI) neonate rats. Neural stem cells (NSC) existed in the brain lifelong and can be activated. This study was undertaken to assess whether HBO treatment promoted the proliferation of NSC and repaired the brain damage regardless of when it is started, thus to explore the therapeutic window of HBO treatment. Seven-day-old Sprague-Dawley rats underwent left carotid ligation followed by 2 h of hypoxic stress (8% O(2) at 37 degrees C). Hyperbaric oxygen therapy was administered 3, 6, 12, 24, and 72 h after HI. 5-bromo-2'-deoxyurindine and 5-bromo-2'-deoxyuridine/nestin were detected by immunofluorescence and nestin was examined by western blot analysis 10 days after HI. T-maze forced alternation, the foot-fault test, and the radial arm maze were conducted at P 22 days (14 days after HI), P 30 days, and P 34 days. Thereafter, cerebral morphology was examined by Nissl-staining 28 days after HI. There were remarkable increases in the proliferation of neural stem cells in the HBO-treated group, 3, 6, 12, and 24 h after HI, as compared with the HIBD group. The HBO-treated group, 3, 6, and 12 h after HI, performed better in the behavioral test and had less neural loss in the hippocampal CA1 region as compared with the HIBD group. The therapeutic window for effective HBO treatment could be delayed up to 12 h after HIBD, while the effect decreased 24 h after HI.
Collapse
Affiliation(s)
- Xiao-Li Wang
- Division of Neonatology, Department of Pediatrics, Xiang Ya Hospital, Central South University, 87 Xiang Ya Road, Changsha, PR China
| | | | | | | | | |
Collapse
|
81
|
Abstract
Disorders of the maternal-placental-fetal unit often results in fetal brain injury, which in turn results in one of the highest burdens of disease, because of the lifelong consequences and cost to society. Investigating hypoxia-ischemia in the perinatal period requires the factoring of timing of the insult, determination of end-points, taking into account the innate development, plasticity, and enhanced recovery. Prenatal hypoxia-ischemia is believed to account for a majority of cerebral palsy cases. We have modeled sustained and repetitive hypoxia-ischemia in the pregnant rabbit in utero to mimic the insults of abruptio placenta and labor, respectively. Rabbits have many advantages over other animal species; principally, their motor development is in the perinatal period, akin to humans. Sustained hypoxia-ischemia at 70% (E22) and 79% (E25) caused stillbirths and multiple deficits in the postnatal survivors. The deficits included impairment in multiple tests of spontaneous locomotion, reflex motor activity, motor responses to olfactory stimuli, and the coordination of suck and swallow. Hypertonia was observed in the E22 and E25 survivors and persisted for at least 11 days. Noninvasive imaging using MRI suggests that white matter injury in the internal capsule could explain some of the hypertonia. Further investigation is underway in other vulnerable regions such as the basal ganglia, thalamus and brain stem, and development of other noninvasive determinants of motor deficits. For the first time critical mechanistic pathways can be tested in a clinically relevant animal model of cerebral palsy.
Collapse
Affiliation(s)
- Matthew Derrick
- Department of Pediatrics, Northwestern University and Evanston Northwestern Healthcare, Evanston, IL 60201, USA
| | | | | | | |
Collapse
|
82
|
Friess SH, Ichord RN, Owens K, Ralston J, Rizol R, Overall KL, Smith C, Helfaer MA, Margulies SS. Neurobehavioral functional deficits following closed head injury in the neonatal pig. Exp Neurol 2007; 204:234-43. [PMID: 17174304 PMCID: PMC1892165 DOI: 10.1016/j.expneurol.2006.10.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2006] [Revised: 09/21/2006] [Accepted: 10/30/2006] [Indexed: 01/22/2023]
Abstract
Neurobehavioral deficits in higher cortical systems have not been described previously in a large animal model of diffuse brain injury. Anesthetized 3-5 day old piglets were subjected to either mild (142 rad/s) or moderate (188 rad/s) rapid non-impact axial rotations of the head. Multiple domains of cortical function were evaluated 5 times during the 12 day post-injury period using tests of neurobehavioral function devised for piglets. There were no observed differences in neurobehavioral outcomes between mild injury pigs (N=8) and instrumented shams (N=4). Moderately injured piglets (N=7) had significantly lower interest in exploring their environment and had higher failure rates in visual-based problem solving compared to instrumented shams (N=5) on days 1 and 4 after injury. Neurobehavioral functional deficits correlated with neuropathologic damage in the neonatal pigs after inertial head injury. Injured axons detected by immunohistochemistry (beta-APP) were absent in mild injury and sham piglets, but were observed in moderately injured piglet brains. In summary, we have developed a quantitative battery of neurobehavioral functional assessments for large animals that correlate with neuropathologic axonal damage and may have wide applications in the fields of cardiac resuscitation, stroke, and hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Stuart H Friess
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Feng Y, Liu YM, Leblanc MH, Bhatt AJ, Rhodes PG. Grape seed extract given three hours after injury suppresses lipid peroxidation and reduces hypoxic-ischemic brain injury in neonatal rats. Pediatr Res 2007; 61:295-300. [PMID: 17314686 DOI: 10.1203/pdr.0b013e318030c92d] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We have reported that pretreatment with grape seed extract (GSE), a potent antioxidant, is neuroprotective. This study examined whether treatment after injury with GSE is protective. Seven-day-old rat pups had the right carotid artery ligated, and then 2.5 h of 8% oxygen. GSE (50 mg/kg) or vehicle was administered by i.p. initial injection at 5 min to 5 h after reoxygenation, with an additional three doses within 26 h after injury. Brain damage was evaluated by weight deficit of the right hemisphere at 22 d after hypoxia. Treatment at 3 h after reoxygenation reduced brain weight loss from 21.0 +/- 3.3% in vehicle-treated pups (n = 31) to 11.4 +/- 2.8% in treated pups (n = 31, p < 0.05). GSE lowered body temperature, but reduced brain injury even when body temperature was controlled. GSE reduced neurofunctional abnormalities caused by the hypoxia-ischemia (HI). GSE reduced a HI induced increase in 8-isoprostaglandin F2alpha (8-isoPGF2alpha) and reduced an HI-induced increase in the proapoptotic protein c-jun in the brain cortex. GSE up to 3 h after reoxygenation reduces brain injury in rat pups, probably by suppressing lipid peroxidation and the proapoptotic protein c-jun.
Collapse
Affiliation(s)
- Yangzheng Feng
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA.
| | | | | | | | | |
Collapse
|
84
|
Drobyshevsky A, Derrick M, Wyrwicz AM, Ji X, Englof I, Ullman LM, Zelaya ME, Northington FJ, Tan S. White matter injury correlates with hypertonia in an animal model of cerebral palsy. J Cereb Blood Flow Metab 2007; 27:270-81. [PMID: 16736047 DOI: 10.1038/sj.jcbfm.9600333] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hypertonia and postural deficits are observed in cerebral palsy and similar abnormalities are observed in postnatal rabbits after antenatal hypoxia-ischemia. To explain why some kits become hypertonic, we hypothesized that white matter injury was responsible for the hypertonia. We compared newborn kits at postnatal day 1 (P1) with and without hypertonia after in vivo global fetal hypoxia-ischemia in pregnant rabbits at 70% gestation. The aim was to examine white matter injury by diffusion tensor magnetic resonance imaging indices, including fractional anisotropy (FA). At P1, FA and area of white matter were significantly lower in corpus callosum, internal capsule, and corona radiata of the hypertonic kits (n=32) than that of controls (n=19) while nonhypertonic kits (n=20) were not different from controls. The decrease in FA correlated with decrease in area only in hypertonia. A threshold of FA combined with area identified only hypertonic kits. A reduction in volume and loss of phosphorylated neurofilaments in corpus callosum and internal capsule were observed on immunostaining. Concomitant hypertonia with ventriculomegaly resulted in a further decrease of FA from P1 to P5 while those without ventriculomegaly had a similar increase of FA as controls. Thus, hypertonia is associated with white matter injury, and a population of hypertonia can be identified by magnetic resonance imaging variables. The white matter injury manifests as a decrease in the number and density of fiber tracts causing the decrease in FA and volume. Furthermore, the dynamic response of FA may be a good indicator of the plasticity and repair of the postnatal developing brain.
Collapse
Affiliation(s)
- Alexander Drobyshevsky
- Department of Pediatrics, Evanston Northwestern Healthcare and Northwestern University, Evanston, Illinois 60201, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Skoff RP, Bessert D, Barks JDE, Silverstein FS. Plasticity of neurons and glia following neonatal hypoxic-ischemic brain injury in rats. Neurochem Res 2006; 32:331-42. [PMID: 17103331 DOI: 10.1007/s11064-006-9188-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Accepted: 09/29/2006] [Indexed: 01/19/2023]
Abstract
Periventricular white matter injury in premature infants is linked to chronic neurological dysfunction. Periventricular white matter injury is caused by many mechanisms including hypoxia-ischemia (HI). Animal models of HI in the neonatal rodent brain can replicate some important features of periventricular white matter injury. Most rodent studies have focused upon early cellular and tissue events following unilateral neonatal HI that is elicited by unilateral carotid artery ligation and followed by timed exposure to moderate hypoxia. Milder hypoxic-ischemic insults elicit preferential white matter injury. Little information is available about long-term cellular effects of unilateral HI. One month after unilateral neonatal hypoxia ischemia, we show that all the components for structural reorganization of the brain are present in moderately injured rats. These components in the injured side include extensive influx of neurites, axonal and dendritic growth cones, abundant immature synapses, and myelination of many small axons. Surprisingly, this neural recovery is often found in and adjacent to cysts that have the ultrastructural features of bone extracellular matrix. In contrast, brains with severe hypoxia ischemia one month after injury still undergo massive neuronal degeneration. While massive destruction of neurons and glia are striking events shortly after brain HI, neural cells re-express their intrinsic properties and attempt an anatomical recovery long after injury.
Collapse
Affiliation(s)
- Robert P Skoff
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
86
|
Northington FJ. Brief update on animal models of hypoxic-ischemic encephalopathy and neonatal stroke. ILAR J 2006; 47:32-8. [PMID: 16391429 DOI: 10.1093/ilar.47.1.32] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The discovery of safe and effective therapies for perinatal hypoxia ischemia (HI) and stroke remains an unmet goal of neonatal-perinatal medicine. Because of the many developmental and functional differences between the neonatal brain and the adult brain, the ability to extrapolate adult data to the neonatal condition is very limited. For this reason, it is incumbent on scientists in the field of neonatal brain injury to address the questions of therapeutic efficacy of an array of potential therapies in a developmentally appropriate model. Toward that end, a number of new models of neonatal HI and stroke have been introduced recently. Additionally, some of the established models have been adapted to different species and different ages, giving scientists a greater choice of models for the study of neonatal HI and stroke. Many of these models are now also being used for functional and behavioral testing, an absolute necessity for preclinical therapeutic trials. This review focuses primarily on the newly developed models, recent adaptations to established models, and the studies of functional outcome that have been published since 2000.
Collapse
Affiliation(s)
- Frances J Northington
- Department of Pediatrics, Eudowood Neonatal Pulmonary Division, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
87
|
Golan H, Huleihel M. The effect of prenatal hypoxia on brain development: short- and long-term consequences demonstrated in rodent models. Dev Sci 2006; 9:338-49. [PMID: 16764607 DOI: 10.1111/j.1467-7687.2006.00498.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Hypoxia (H) and hypoxia-ischemia (HI) are major causes of foetal brain damage with long-lasting behavioral implications. The effect of hypoxia has been widely studied in human and a variety of animal models. In the present review, we summarize the latest studies testing the behavioral outcomes following prenatal hypoxia/hypoxia-ischemia in rodent models. Delayed development of sensory and motor reflexes during the first postnatal month of rodent life was observed by various groups. Impairment of motor function, learning and memory was evident in the adult animals. Activation of the signaling leading to cell death was detected as early as three hours following H/HI. An increase in the counts of apoptotic cells appeared approximately three days after the insult and peaked about seven days later. Around 14-20 days following the H/HI, the amount of cell death observed in the tissue returned to its basal levels and cell loss was apparent in the brain tissue. The study of the molecular mechanism leading to brain damage in animal models following prenatal hypoxia adds valuable insight to our knowledge of the central events that account for the morphological and functional outcomes. This understanding provides the starting point for the development and improvement of efficient treatment and intervention strategies.
Collapse
Affiliation(s)
- Hava Golan
- Department of Developmental Molecular Genetics and Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | | |
Collapse
|
88
|
Meier C, Middelanis J, Wasielewski B, Neuhoff S, Roth-Haerer A, Gantert M, Dinse HR, Dermietzel R, Jensen A. Spastic paresis after perinatal brain damage in rats is reduced by human cord blood mononuclear cells. Pediatr Res 2006; 59:244-9. [PMID: 16439586 DOI: 10.1203/01.pdr.0000197309.08852.f5] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Brain damage around birth may cause lifelong neurodevelopmental deficits. We examined the therapeutic potential of human umbilical cord blood-derived mononuclear cells containing multipotent stem cells to facilitate motor recovery after cerebral hypoxic-ischemic damage in neonatal rats. Left carotid artery ligation followed by 8% O(2) inhalation for 80 min was performed on postnatal d 7, succeeded by intraperitoneal transplantation of human umbilical cord blood-derived mononuclear cells on postnatal d 8 in a sham-controlled design. Histologic and immunohistochemical analysis on postnatal d 21 revealed that neonates developed severe cerebral damage after the hypoxic-ischemic insult. These animals also suffered from contralateral spastic paresis, as evidenced by their locomotor behavior. After transplantation of human umbilical cord blood-derived mononuclear cells, spastic paresis was largely alleviated, resulting in a normal walking behavior. This "therapeutic" effect was accompanied by the fact that mononuclear cells had entered the brain and were incorporated around the lesion without obvious signs of transdifferentiation. This study demonstrates that intraperitoneal transplantation of human umbilical cord blood-derived mononuclear cells in a rat model of perinatal brain damage leads to both incorporation of these cells in the lesioned brain area and to an alleviation of the neurologic effects of cerebral palsy as assessed by footprint and walking pattern analysis.
Collapse
Affiliation(s)
- Carola Meier
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Northington FJ, Graham EM, Martin LJ. Apoptosis in perinatal hypoxic-ischemic brain injury: how important is it and should it be inhibited? ACTA ACUST UNITED AC 2005; 50:244-57. [PMID: 16216332 DOI: 10.1016/j.brainresrev.2005.07.003] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2005] [Revised: 07/12/2005] [Accepted: 07/14/2005] [Indexed: 01/21/2023]
Abstract
The discovery of safe and effective therapies for perinatal hypoxia-ischemia (HI) and stroke remains an unmet goal of perinatal medicine. Hypothermia and antioxidants such as allopurinol are currently under investigation as treatments for neonatal HI. Drugs targeting apoptotic mechanisms are currently being studied in adult diseases such as cancer, stroke, and trauma and have been proposed as potential therapies for perinatal HI and stroke. Before developing antiapoptosis therapies for perinatal brain injury, we must determine whether this form of cell death plays an important role in these injuries and if the inhibition of these pathways promotes more benefit than harm. This review summarizes current evidence for apoptotic mechanisms in perinatal brain injury and addresses issues pertinent to the development of antiapoptosis therapies for perinatal HI and stroke.
Collapse
Affiliation(s)
- Frances J Northington
- Department of Pediatrics, Eudowood Neonatal Pulmonary Division, Dept. of Pediatrics, CMSC 6-104, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA.
| | | | | |
Collapse
|
90
|
Feng Y, Liu YM, Fratkins JD, LeBlanc MH. Grape seed extract suppresses lipid peroxidation and reduces hypoxic ischemic brain injury in neonatal rats. Brain Res Bull 2005; 66:120-7. [PMID: 15982528 DOI: 10.1016/j.brainresbull.2005.04.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2005] [Revised: 03/22/2005] [Accepted: 04/11/2005] [Indexed: 11/17/2022]
Abstract
Oxygen radicals play a crucial role in brain injury. Grape seed extract is a potent anti-oxidant. Does grape seed extract reduce brain injury in the rat pup? Seven-day-old rat pups had the right carotid arteries permanently ligated followed by 2.5 h of hypoxia (8% oxygen). Grape seed extract, 50 mg/kg, or vehicle was administered by i.p. 5 min prior to hypoxia and 4 h after reoxygenation and twice daily for 1 day. Brain damage was evaluated by weight deficit of the right hemisphere at 22 days following hypoxia and by histopathology. Grape seed extract reduced brain weight loss from 20.0+/-4.4% S.E.M. in vehicle pups (n=21) to 3.1+/-1.6% in treated pups (n=20, P<0.01). Grape seed extract improved the histopathologic brain score in cortex, hippocampus and thalamus (P<0.05 versus vehicle). Concentrations of brain 8-isoprostaglandin F2alpha and thiobarbituric acid reacting substances significantly increased due to hypoxic ischemia. Grape seed extract reduced this increase. Treatment with grape seed extract suppresses lipid peroxidation and reduces hypoxic ischemic brain injury in neonatal rat.
Collapse
Affiliation(s)
- Yangzheng Feng
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | | | | | | |
Collapse
|
91
|
Spandou E, Papadopoulou Z, Soubasi V, Karkavelas G, Simeonidou C, Pazaiti A, Guiba-Tziampiri O. Erythropoietin prevents long-term sensorimotor deficits and brain injury following neonatal hypoxia-ischemia in rats. Brain Res 2005; 1045:22-30. [PMID: 15910759 DOI: 10.1016/j.brainres.2005.03.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Revised: 02/27/2005] [Accepted: 03/01/2005] [Indexed: 11/29/2022]
Abstract
Perinatal asphyxia accounts for behavioral dysfunctions that often manifest as sensorimotor, learning or memory disabilities throughout development and into maturity. Erythropoietin (Epo) has been shown to exert neuroprotective effects in different models of brain injury including experimental models of perinatal asphyxia. However, the effect of Epo on functional abilities following cerebral hypoxia-ischemia (HI) in neonatal rats is not known. The aim of the present study is to investigate the effect of Epo on sensorimotor deficits and brain injury induced by hypoxia-ischemia. Seven-day-old rats underwent unilateral, permanent carotid artery ligation followed by 1 h of hypoxia. Epo was administered as a single dose immediately after the hypoxic insult (2000 U/kg). The neuroprotective effect of Epo was evaluated at postnatal day 42 by using a battery of behavioral tests and histological analysis. The results of the present study suggest that Epo treatment immediately after HI insult significantly facilitated recovery of sensorimotor function. Consistently, histopathological evaluation demonstrated that Epo significantly attenuated brain injury and preserved the integrity of cerebral cortex. These findings indicate that long-term neuroprotective effect of Epo on neonatal HI-induced brain injury might be associated with the preservation of sensorimotor functions.
Collapse
Affiliation(s)
- Evangelia Spandou
- Department of Physiology and Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | | | | | | | | | | | | |
Collapse
|
92
|
Lubics A, Reglodi D, Tamás A, Kiss P, Szalai M, Szalontay L, Lengvári I. Neurological reflexes and early motor behavior in rats subjected to neonatal hypoxic-ischemic injury. Behav Brain Res 2005; 157:157-65. [PMID: 15617782 DOI: 10.1016/j.bbr.2004.06.019] [Citation(s) in RCA: 182] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Revised: 06/18/2004] [Accepted: 06/23/2004] [Indexed: 11/19/2022]
Abstract
Severe perinatal hypoxia-ischemia is an important cause of brain injury in both full-term and premature newborns, with a high risk of future behavioral and neurological deficits. The most commonly used animal model of neonatal hypoxia-ischemia is the unilateral ligation of the common carotid artery followed by exposure to hypoxia in 7-day-old rats. In spite of the wide use of this model, lot of contradictions and discrepancies exist between the results obtained by different laboratories regarding behavioral deficits and there are no data regarding the possible delay of the appearance of neurological reflexes and the time-course of reflex performances following neonatal hypoxic-ischemic injury in rats. In the present study we showed that neonatal hypoxia-ischemia retarded the development of somatic growth and several neurological reflexes (ear twitch, grasping, gait and negative geotaxis). Hypoxic animals also displayed retarded performance in righting, geotaxis and gait reflexes. Although hypoxic pups performed worse in most tests for motor coordination, they reached normal levels by 5 weeks of age except in the footfault test. In the open-field, hypoxic animals were generally more active, except at 3 weeks, when activity of normal pups increased enormously as well. Brain areas were significantly reduced in hypoxic animals, but no close correlation was found with behavioral deficits.
Collapse
Affiliation(s)
- Andrea Lubics
- Department of Anatomy, Neurohumoral Regulations Research Group of the Hungarian Academy of Sciences, Pécs University, Szigeti u 12, 7624 Pecs, Hungary.
| | | | | | | | | | | | | |
Collapse
|
93
|
Vicente E, Boer M, Netto C, Fochesatto C, Dalmaz C, Rodrigues Siqueira I, Gonçalves CA. Hippocampal antioxidant system in neonates from methylmercury-intoxicated rats. Neurotoxicol Teratol 2005; 26:817-23. [PMID: 15451045 DOI: 10.1016/j.ntt.2004.08.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2004] [Revised: 08/05/2004] [Accepted: 08/05/2004] [Indexed: 11/25/2022]
Abstract
Methylmercury (MeHg) is a well-known environmental pollutant toxic to the nervous tissue, particularly during development. We recently described transitory hippocampal changes in neonate rats prenatally exposed to MeHg. In this study, we evaluate oxidative stress in the hippocampus on the 1st and 30th postnatal days. Motor behavior (open-field, foot-fault and strength tests) of these animals also was studied after the 30th postnatal day. Female Wistar rats were injected with MeHg (5 mg/Hg/day) on the 12th, 13th and 14th gestational days. Biochemical parameters measured for oxidative stress were levels of the antioxidant enzymes superoxide dismutase (SOD), glutathione peroxidase (GPx) and catalase (CAT). Total antioxidant reactivity (TAR) and protein oxidation (contents of tryptophan and tyrosine) were also recorded. Our results showed low activities of antioxidant enzymes in the MeHg group at birth. SOD activity remained reduced on the 30th postnatal day. Moreover, a decrease of TAR and protein oxidation was observed only at 30 days of age. No changes were observed in the motor behavior of these animals. Although mercury content in hippocampus is present at undetectable levels at 30 days of age, we observed more persistent changes in oxidative balance. Our data confirm that mercury induces oxidative stress in hippocampus and that this alteration, particularly SOD activity, remained altered even when mercury was no longer present.
Collapse
Affiliation(s)
- Evelin Vicente
- Programa de Neurociências, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | | | | | | |
Collapse
|
94
|
Feng Y, Fratkins JD, LeBlanc MH. Estrogen attenuates hypoxic-ischemic brain injury in neonatal rats. Eur J Pharmacol 2004; 507:77-86. [PMID: 15659297 DOI: 10.1016/j.ejphar.2004.11.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2004] [Accepted: 11/12/2004] [Indexed: 11/27/2022]
Abstract
Estrogen is neuroprotective in adult animals. We wished to determine if estrogen protects against brain injury in the newborn. Four-day-old rat pups were treated with subcutaneously implanted pellets containing 0.05 mg (2.4 microg/day) of 17beta-estradiol or vehicle, designed to release the estrogen over 21 days. At 7 days old the pups had the right carotid artery ligated followed by 2.5 h of 8% oxygen. Brain damage was evaluated by weight deficit of the right hemisphere at 22 days following hypoxia. Estradiol treatments reduced brain weight loss from -17.4+/-2.8% S.E.M. in the vehicle group (n=32) to -9.3+/-2.7% in the treated group (n=32, P<0.05). Brain cortex thiobarbituric acid reacting substances and caspase activities were assessed 24 h after reoxygenation. Estradiol significantly reduced a hypoxia-induced increase in brain thiobarbituric acid reactive substances (P<0.05). Levels of caspase-3, -8 and -9 activity increased due to hypoxia-ischemia. Estradiol had no effect on caspase activity. Estradiol reduced brain injury in the neonatal rat.
Collapse
Affiliation(s)
- Yangzheng Feng
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, USA
| | | | | |
Collapse
|
95
|
Mishima K, Ikeda T, Aoo N, Takai N, Takahashi S, Egashira N, Ikenoue T, Iwasaki K, Fujiwara M. Hypoxia-ischemic insult in neonatal rats induced slowly progressive brain damage related to memory impairment. Neurosci Lett 2004; 376:194-9. [PMID: 15721220 DOI: 10.1016/j.neulet.2004.11.055] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2004] [Revised: 11/18/2004] [Accepted: 11/18/2004] [Indexed: 11/26/2022]
Abstract
The present study was designed to determine potential associations between the brain damage induced by hypoxic-ischemic (HI) insult and spatial learning impairment in an eight-arm radial maze task. We first determined the pathological outcomes after 2, 5, 9, and 17 weeks of recovery following the HI insult. The results show that the brain damage progressed from 2 up to 17 weeks of recovery. To clarify the time course of the brain damage changes, we investigated the histological changes of the same individual with magnetic resonance imaging (MRI) after 5, 9, and 57 weeks of recovery following the HI insult. The MRI changes were similar to the histological changes, and the brain damages were exacerbated in the contralateral hemisphere after 57 weeks of recovery following the HI insult. To investigate whether alteration in brain function was correlated with MRI and histological changes, the rats were made to find their way through an eight-arm radial maze was performed at either 7th or 16th weeks of recovery. According to the results, the spatial learning impairments of rats in the maze starting at 16 weeks of recovery were more severe than those at 7 weeks of recovery, indicating that the impairments were progressive and depended on the degree of brain damage. The results of the present study are the first demonstration that the evolutional and specific brain damage following the HI insult is slowly and progressively exacerbated to the contralateral hemisphere and rats who experience the HI are at risk for showing a late impairment of brain function.
Collapse
Affiliation(s)
- Kenichi Mishima
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma 8-19-1, Fukuoka city, Fukuoka 814-0180 Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Ten VS, Wu EX, Tang H, Bradley-Moore M, Fedarau MV, Ratner VI, Stark RI, Gingrich JA, Pinsky DJ. Late Measures of Brain Injury After Neonatal Hypoxia–Ischemia in Mice. Stroke 2004; 35:2183-8. [PMID: 15272130 DOI: 10.1161/01.str.0000137768.25203.df] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE This work was undertaken to determine to what degree long-term neurofunctional outcome of neonatal hypoxic-ischemic (HI) brain injury in mice correlates with anatomical extent of cerebral damage assessed by magnetic resonance imaging (MRI) and histopathology. METHODS On postnatal day 7, mice were subjected to HI. At 7 to 9 weeks after HI neurofunctional outcome was assessed by water-maze, rota-rod, and open-field test performance, followed by cerebral MRI and histopathology evaluation. RESULTS At 10 weeks after HI, MRI revealed ipsilateral brain atrophy alone or with porencephalic cyst formation and contralateral ventriculomegaly. Adult HI-affected mice, especially those that developed a porencephalic cyst, demonstrated significant neurofunctional deficit compared with age-matched naïve mice. HI-affected mice with ipsilateral cerebral atrophy but without porencephaly demonstrated no or an intermediate level of neurofunctional deficit. Neurobehavioral assessment of mice subjected to HI insult revealed a strong correlation between degree of brain injury and functional neurohandicap. CONCLUSIONS This is the first study to demonstrate that long-term neurofunctional outcome in mice after a neonatal HI correlates tightly with anatomical pattern/extent of cerebral damage, defined by MRI and histopathology.
Collapse
Affiliation(s)
- Vadim S Ten
- Department of Pediatrics, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Pabello NG, Tracy SJ, Keller RW. Protective effects of brief intra- and delayed postischemic hypothermia in a transient focal ischemia model in the neonatal rat. Brain Res 2004; 995:29-38. [PMID: 14644468 DOI: 10.1016/j.brainres.2003.09.065] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hypothermia provides neuroprotection in virtually all animal models of ischemia, including adult stroke models and the neonatal hypoxic-ischemic (HI) model. In these studies, brief periods of hypothermia are examined in a neonatal model employing transient focal ischemia in a 7-day-old rat pup. Pups underwent permanent middle cerebral artery (MCA) occlusion coupled with a temporary (1 h) occlusion of the ipsilateral common carotid artery (CCA). This study included five treatment groups: (1) normothermic (Normo)-brain temperature was maintained at 37 degrees C; (2) intraischemic hypothermia (IntraH)-28 degrees C during the 1-h ischemic period only; (3) postischemic hypothermia (PostH)-28 degrees C for the second hour of reperfusion only; (4) late-onset postischemic hypothermia (LPostH) cooled to 28 degrees C for the fifth and sixth hours of reperfusion only; and (5) Shams. After various times (3 days-6 weeks), the lesion was assessed using 2,3,5-triphenyltetrazolium chloride (TTC) or hematoxylin and eosin (H&E) stains. Intraischemic hypothermia resulted in significant protection in terms of survival, lesion size, and histology. Postischemic hypothermia was not effective in reducing lesion size early after ischemia, but significantly reduced the eventual long-term damage (2-6 weeks). Late-onset postischemic hypothermia did not reduce infarct volume. Therefore, both intraischemic and postischemic hypothermia provided neuroprotection in the neonatal rat, but with different effects on the degenerative time course. While there were no observable differences in simple behaviors or growth, all hypothermic conditions significantly reduced mortality rates. While the protection resulting from intraischemic hypothermia is similar to what is observed in other models, the degree of long-term ischemic protection observed after 1 h of postischemic hypothermia was remarkable and distinct from what has been observed in other adult or neonatal models.
Collapse
Affiliation(s)
- Nina G Pabello
- Center for Neuropharmacology and Neuroscience, Albany Medical College MC-136, 47 New Scotland Avenue, Albany, NY 12208, USA
| | | | | |
Collapse
|
98
|
Abstract
Controversy continues over which animal model to use as a reflection of human disease states. With respect to perinatal brain disorders, scientists must contend with a disease in evolution. In that regard, the perinatal brain is at risk during a time of extremely rapid development and maturation, involving processes that are required for normal growth. Interfering with these processes, as part of therapeutic intervention must be efficacious and safe. To date, numerous models have provided tremendous information regarding the pathophysiology of brain damage to term and preterm infants. Our challenges will continue to be in identifying those infants at greatest risk for permanent injury, and adapting therapies that provide more benefit than harm. Using animal models to conduct these studies will bring us closer to that goal.
Collapse
Affiliation(s)
- Jerome Y Yager
- Division of Pediatric Neurology, Department of Pediatrics and Child Health, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
99
|
Feng Y, Fratkins JD, LeBlanc MH. Treatment with tamoxifen reduces hypoxic–ischemic brain injury in neonatal rats. Eur J Pharmacol 2004; 484:65-74. [PMID: 14729383 DOI: 10.1016/j.ejphar.2003.10.048] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Tamoxifen, an estrogen receptor modulator, is neuroprotective in adult rats. Does tamoxifen reduce brain injury in the rat pup? Seven-day-old rat pups had the right carotid artery permanently ligated followed by 2.5 h of hypoxia (8% oxygen). Tamoxifen (10 mg/kg) or vehicle was given i.p. 5 min prior to hypoxia, or 5 min after reoxygenation, with a second dose given 6 h after the first. Brain damage was evaluated by weight deficit of the right hemisphere 22 days following hypoxia and gross and microscopic morphology. Tamoxifen pre-treatment reduced brain weight loss from 21.5+/-4.0% in vehicle pups (n=27) to 2.6+/-2.5% in the treated pups (n=22, P<0.05). Treatment 5 min after reoxygenation reduced brain weight loss from 27.5+/-4.0% in vehicle pups (n=42) to 12.0+/-3.9% in the treated pups (n=30, P<0.05). Tamoxifen reduces brain injury in the neonatal rat.
Collapse
Affiliation(s)
- Yangzheng Feng
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, 39216-4505, Jackson, MS, USA.
| | | | | |
Collapse
|
100
|
Ten VS, Bradley-Moore M, Gingrich JA, Stark RI, Pinsky DJ. Brain injury and neurofunctional deficit in neonatal mice with hypoxic-ischemic encephalopathy. Behav Brain Res 2003; 145:209-19. [PMID: 14529818 DOI: 10.1016/s0166-4328(03)00146-3] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Birth asphyxia accounts for the majority of developmental motor and cognitive deficits. Studies were undertaken to develop a reproducible murine model of perinatal hypoxic-ischemic encephalopathy (HIE) which would permit both anatomic and neurofunctional quantification of injury. Short-term neurofunctional outcomes consisted of three developmental reflexes (righting, cliff aversion and geotaxis) assessed in 7-day-old mouse pups 24 h after unilateral carotid artery ligation followed by inhalation of 8% oxygen. Cerebral infarct volume was dependent on duration of hypoxia, being approximately 2.5-fold greater with longer (60 min) versus shorter (30 min) hypoxia exposure (P=0.001). All three sensorimotor neonatal reflexes assessed at 24 h after HIE injury correlated significantly with long-term neurofunction evaluated using a water-maze test of navigational learning and memory assessed 8 weeks later in the same animals. Cerebral atrophy, a delayed consequence of HIE in this model, also correlated strongly with water-maze performance (r=0.86, P=0.002). These data demonstrate for the first time that murine neonatal sensorimotor reflex performance can be rigorously quantified in the acute phase of perinatal HIE and has strong predictive value not only for anatomic extent of cerebral injury, but also for long-term neurofunctional outcome.
Collapse
Affiliation(s)
- Vadim S Ten
- Department of Pediatrics, Columbia University, 3959 Broadway, BHS-12, Room 115, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|