Dissertations / Theses on the topic 'Hypoxic ischaemic brain injury'
Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles
Consult the top 44 dissertations / theses for your research on the topic 'Hypoxic ischaemic brain injury.'
Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.
You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.
Browse dissertations / theses on a wide variety of disciplines and organise your bibliography correctly.
Peden, Carol J. "Proton and phosphorus spectroscopy of hypoxic, ischaemic and haemorrhagic perinatal brain injury." Thesis, University of Edinburgh, 1996. http://hdl.handle.net/1842/21460.
Full textPennell, Craig Edward. "The role of lactate measurement in the prediction of fetal hypoxic-ischaemic brain injury during labour." University of Western Australia. School of Women's and Infants' Health, 2004. http://theses.library.uwa.edu.au/adt-WU2003.0037.
Full textWilkinson, Dominic James Clifford. "Ethical issues in the use of magnetic resonance imaging of the brain in newborn infants with hypoxic-ischaemic encephalopathy : neuroimaging and decision-making for brain injured newborns." Thesis, University of Oxford, 2010. http://ora.ox.ac.uk/objects/uuid:d61e4318-3568-4310-bf92-c7d70f2cb3da.
Full textFairlie, John. "Neuronal and microvascular adaptions to hypoxic/ischaemic injury in animal models." Thesis, University of Newcastle Upon Tyne, 2005. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.427287.
Full textFlynn, Liam Martin Clint. "Physiological responses to brain tissue hypoxia and blood flow after acute brain injury." Thesis, University of Edinburgh, 2018. http://hdl.handle.net/1842/31268.
Full textRutherford, Mary. "Magnetic resonance imaging of hypoxic-ischaemic brain lesions in the term infant." Thesis, University of Bristol, 1998. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.262817.
Full textLim, Ta. "The role of hypoxia-inducible factor-1α in xenon preconditioning versus hypoxic-ischaemic organ injury." Thesis, Imperial College London, 2008. http://hdl.handle.net/10044/1/7663.
Full textWhiteley, Tara. "Mitochondria and secondary ischaemic neural injury after head trauma." Thesis, University of Newcastle Upon Tyne, 1999. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.285401.
Full textRocha, Ferreira E. "The role of infection/inflammation, the TNF family of cytokines and myeloid cells in perinatal hypoxia-ischaemia brain injury." Thesis, University College London (University of London), 2014. http://discovery.ucl.ac.uk/1447553/.
Full textCrooks, Suzanne. "Long-term neuropsychological and psychosocial outcomes of hypoxic-ischemic brain injury." Thesis, Queen's University Belfast, 2014. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.676279.
Full textFu, Wai. "In silico prediction of cis-regulatory elements of genes involved in hypoxic-ischaemic insult." Click to view the E-thesis via HKUTO, 2006. http://sunzi.lib.hku.hk/hkuto/record/B36986896.
Full textDobrucki, Wawrzyniec L. "Nitrogen and Oxygen Radicals in Ischemic and Hypoxic Injury of the Brain." Ohio University / OhioLINK, 2003. http://rave.ohiolink.edu/etdc/view?acc_num=ohiou1057171850.
Full textFu, Wai, and 符慧. "In silico prediction of cis-regulatory elements of genes involved in hypoxic-ischaemic insult." Thesis, The University of Hong Kong (Pokfulam, Hong Kong), 2006. http://hub.hku.hk/bib/B36986896.
Full textWiderøe, Marius. "Magnetic Resonance Imaging of Hypoxic-Ischemic Brain Injury Development in the Newborn Rat." Doctoral thesis, Norges teknisk-naturvitenskapelige universitet, Institutt for laboratoriemedisin, barne- og kvinnesykdommer, 2012. http://urn.kb.se/resolve?urn=urn:nbn:no:ntnu:diva-17210.
Full textManderino, Lisa M. "Cognitive Functioning Under Hypoxic Stress in Individuals with History of Mild Traumatic Brain Injury." Kent State University / OhioLINK, 2020. http://rave.ohiolink.edu/etdc/view?acc_num=kent1591713552152285.
Full textLeonardo, Christopher C. "The Role of Extracellular Matrix and Matrix-Degrading Proteases in Neonatal Hypoxic-Ischemic Injury." [Tampa, Fla] : University of South Florida, 2008. http://purl.fcla.edu/usf/dc/et/SFE0002587.
Full textMathur, Manik. "The Blood-Brain Barrier integrity and function in ischaemic injury and multiple sclerosis with role of hypothermia." Thesis, University of Nottingham, 2016. http://eprints.nottingham.ac.uk/37404/.
Full textRanasinghe, Himani Sumudumalee. "Mechanisms underlying hypoxic ischemic injury to the developing brain: The significance of matrix metalloproteinase 2 and 9." Thesis, University of Auckland, 2009. http://hdl.handle.net/2292/4962.
Full textChristophidis, Larissa Joy. "The roles of growth hormone and prolactin in the brain during development and recovery from hypoxic-ischemic injury." Thesis, University of Auckland, 2011. http://hdl.handle.net/2292/6781.
Full textKendall, G. "The role of endotoxin, the TNF family of cytokines and intracellular pH in perinatal hypoxic-ischemic brain injury." Thesis, University College London (University of London), 2010. http://discovery.ucl.ac.uk/19212/.
Full textGregg, Robert. "Functional outcomes of hypoxic brain injury : a systematic review : the influence of childhood trauma and coping on the psychosis phenotype in the general population." Thesis, Queen's University Belfast, 2015. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.695346.
Full textXu, Kui. "The Central Nervous System Aspects of Cardiac Arrest and Resuscitation in a Rat Model of Global Ischemia." Case Western Reserve University School of Graduate Studies / OhioLINK, 2010. http://rave.ohiolink.edu/etdc/view?acc_num=case1270689501.
Full textBattin, Malcolm Richard. "Selective cerebral hypothermia for term infants following hypoxic ischaemic injury." 2008. http://hdl.handle.net/2292/5545.
Full textWhole document restricted until 11/2014, but available by request, use the feedback form to request access.
Lai, Pei-Chun, and 賴佩君. "Ceftriaxone attenuates hypoxic-ischemic brain injury in neonatal rat." Thesis, 2012. http://ndltd.ncl.edu.tw/handle/20603848098666369351.
Full text慈濟大學
藥理暨毒理學碩士班/博士班
100
Perinatal brain injury is the leading cause of subsequent neurological disability in both term and preterm baby. Glutamate excitotoxicity is one of the major factors involved in perinatal hypoxic-ischemic encephalopathy (HIE). Glutamate transporter GLT1, expressed mainly in mature astrocytes, is the major glutamate transporter in the brain. HIE induced excessive glutamate release which is not reuptaked by immature astrocytes may induce neuronal damage. Compounds that enhance the expression of GLT1 may exert neuroprotective effect in HIE. In this study, we used a neonatal rat model of HIE by unilateral ligation of carotid artery and subsequent exposure to 8% oxygen for 2 hrs on postnatal day 7 (P7) rats. Neonatal rats were administered three dosages of an antibiotic, ceftriaxone, 48 hrs prior to experimental HIE. Our results showed that pre-treatment with 200 mg/kg ceftriaxone significantly reduced the brain injury scores and apoptotic cells in the hippocampus, restored myelination in the external capsule of P14 rats. Pre-treatment with 200 mg/kg ceftriaxone improved the hypoxia-ischemia induced learning and memory deficit of P23-24 rats. GLT1 expression was observed in the cortical neurons of ceftriaxone treated rats. These results suggest that pre-treatment of infants at risk for HIE with ceftriaxone may reduce subsequent brain injury.
Sizonenko, Stéphane Vladimir. "Hypoxic-ischemic injury in the developing brain: pathogenesis and neuroprotection." 2002. http://hdl.handle.net/2292/3212.
Full textChang, Kang-Fan, and 張綱凡. "LPS preconditioning mediates neuroprotection against hypoxic-ischemic injury in the neonatal rat brain." Thesis, 2004. http://ndltd.ncl.edu.tw/handle/91278671965730347711.
Full text國立成功大學
微生物及免疫學研究所
92
Perinatal hypoxic-ischemic (HI) brain injury is a major cause of neonatal mortality and long-term disability such as mental retardation, cerebral palsy, learning disability, and seizures. There are still no effective therapies against neonatal HI brain injury to date. A sublethal stress before a lethal injury can reduce the neuronal death, a phenomenon called “preconditioning”. Elucidating the underlying mechanisms of preconditioning may provide potential neuroprotective therapy for neonatal HI encephalopathy. We first established a lipopolysaccharide (LPS) preconditioning model in neonatal rats by pretreating rat pups with a low dose of LPS 24 hours before HI injury on postnatal day 7. We found that the degree of brain injury of LPS-preconditioned rats were significantly less than that of NS-pretreated rats. The behavioral performance measured by Morris water maze of LPS-preconditioned rats was also significantly better than that of NS-pretreated rats. The expression levels of apoptosis markers, such as cleavaged form of caspase-3, caspase-9, poly (ADP-ribose) polymerase (PARP), and the production of reactive oxygen species (ROS) in the cortex were significantly lower in the LPS-preconditioned rats compared to NS-pretreated rats at 24 hours after HI injury. Among the mitogen-activated protein kinases (MAPK) family, LPS preconditioning could increase the phosphorylation levels of extracellular signal-related kinase (ERK) instead of p38. However, inhibition of MAP kinase kinase (MEK) or p38 did not affect the neuroprotection induced by LPS preconditioning. We also observed that LPS could up-regulate ROS production up to 24 hours after LPS injection. However, ROS scavenger, N-(2-mercaptopropionyl) glycine (N-2-MPG), could not abolish the LPS induced neuroprotection. In contrast, toll-like receptor 4 (TLR4) was involved in the LPS-induced neuroprotection since LPS preconditioning could not be induced in the C3H/HeJ mice, a TLR4 deficient mice. In conclusion, we established a LPS preconditioning model in immature rat brains, and the neuroprotection mechanisms in this model involved the down-regulation of ROS production and the reduction of apoptosis. The LPS-preconditioning mechanisms in the neonatal rat brain remain to be elucidated.
Yi-ChingHsu and 許宜菁. "The mechanism of neurovascular damage in neonatal rat with hypoxic-ischemic brain injury." Thesis, 2014. http://ndltd.ncl.edu.tw/handle/89meyn.
Full text國立成功大學
基礎醫學研究所
102
Brain cells are extremely sensitive to oxygen deprivation. Some brain cells actually start dying just under five minutes after their oxygen supply is cut. As a result, brain hypoxia can kill brain cells and rapidly cause severe brain damage. This is an emergency, and the sooner medical attention is given and the oxygen supply restored, the lower the chances of severe brain damage or death. Neonatal hypoxic-ischemic (HI) stress can lead to HI encephalopathy. There is still no effective drug against neonatal HI brain injury. Most neuroprotective agents have not benefitted patients with stroke. Because researchers have reported that neurovascular units can be the targets of hypoxic stress and that both neurons and microvessels respond equally rapidly to the insult, it has been hypothesized that vascular protection would be more effective than neuroprotection against HI brain damage. To clarify the function of neurovascular unit in neonatal HI brain injury, a major cause of neonatal mortality and long-term disability, we used 7-day-old rat pups with an animal model of HI injury: one side carotid artery was permanently ligated and each rat pup was subjected to systemic hypoxia (8% O for 2 h) to induce ipsilateral cerebral HI injury. This study investigated whether neurovascular unit damage is an early event in HI neonatal brain damage and how nitric oxide contributes to HI-induced brain injury. The data show that IgG leakage and microvascular change were observed with transmission electron microscopy (TEM) as early as 1 h after HI insult. Nitrotyrosine was overexpressed immediately after reoxygenation. The hypothesis that “microvascular damage occurs soon after hypoxic-ischemia: neuronal nitric oxide synthase (nNOS) is activated and contributes to brain injury” was tested. Treating the rat pups with 7-nitroindazole (7-NI), an nNOS inhibitor, and aminoguanidine (AG), an inducible NOS (iNOS) inhibitor, before hypoxia provided complete and partial neuroprotection, respectively. I also use ischemic preconditioning (IP) as a complete brain protection model for comparison. Pretreatment with 7-NI and IP protected cerebral blood flow (CBF) from hypoxia-induced hypoperfusion that reduced the brain infarct area. In summary, nNOS-mediated vascular damage is an early event caused by hypoxic-ischemia and that vascular protection might be better than neuroprotection. However, how the IP is related to endothelial cells requires additional investigation.
Wu, Hsin-Chieh, and 吳欣潔. "The Effect of Neonatal Obesity on Hypoxic-ischemic Brain Injury in Rat Pups." Thesis, 2007. http://ndltd.ncl.edu.tw/handle/70012301530373508973.
Full text國立成功大學
分子醫學研究所
95
Perinatal hypoxic–ischemic (HI) brain damage remains a major cause of acute mortality and chronic neurologic morbidity in children. Obesity is a growing problem in modern society. Evidence has shown that obese adult persons suffer a higher risk of stroke and have worse prognosis than non-obese adults. Here, using a well-established HI brain injury model in neonatal rats, we first tested the hypothesis that the over-fed (OF) obese rat pups had a higher degree of HI brain injury compared with the normal-fed (NF) rat pups. The OF group, defined by reducing the litter size to 6 pups from postnatal day 1 (P1), had a significantly higher body-weight (P<0.001), excess fat deposition in the subcutaneous and perirenal space (P<0.001) and bore hyperglycemia (P<0.01) at P7 compared with the NF group defined by keeping the litter size of 12 pups. The OF-HI group had a higher mortality rate than the NF-HI group during HI. In addition, the OF-HI survivors showed a significantly poorer learning behavior performance assessed by water maze task and higher degree of brain damage measured by pathology at adulthood. Twenty-four hours after HI, more brain damage (Nissl stain) and more TUNEL postive cells were observed in the OF-HI brain than in the NF-HI brain. Caspase-3 activation also significantly increased in cerebral cortex in the OF-HI group compared with the NF-HI group. Immunofluorescence imaging showed that the OF-HI group had increased number of injured cells that exhibited pyknotic morphology and cells that showed nuclear translocalization of apoptotic inducing factor in the cortex and hippocampus than the NF-HI group. These results suggest that mitochondria-dependent apoptosis is involved in the obesity-aggravated HI neuronal death in the neonatal rat brain. Thus, we examined whether the over-nutrient status in the neonatal period would cause endoplasmic reticulum (ER) stress and hyperactivation of c-Jun N-terminal kinase (JNK). Our results showed that there was no difference in the ER chaperon protein Grp78 expression between the two groups. In contrast, the OF pups had higher levels of activated JNKs in the cortex than the NF group before and after HI. Furthermore, the OF-HI group had increased phosphorylation at Ser65 site of cell death molecular BIMEL, and the activation of stress-activated protein kinase p38 was not detected after HI in the two groups. Intracerebroventricular administration of JNK inhibitor SP600125 was operated to prove the causal relationship of JNK hyper-activation in neonatal obesity-aggravated HI brain injury. In the OF-HI group, JNK inhibition reduced the mortality during HI and BIMEL phosphorylation after HI, and also provided significant neuroprotection at pathological level. In conclusion, our study shows that obesity in the neonatal period could aggravate HI neuronal death and JNK activation plays a significant role in obesity-aggravated HI neuronal death in neonatal rats.
Scheepens, Arjan. "Studies on the GH/IGF axis in the infant rat brain following hypoxic ischemic injury." 1999. http://hdl.handle.net/2292/3200.
Full textMing-CheLee and 李明哲. "Inhibition of HDAC activity by sodium butyrate attenuates hypoxic-ischemic injury in the neonatal brain." Thesis, 2010. http://ndltd.ncl.edu.tw/handle/95704696191172209196.
Full text國立成功大學
分子醫學研究所
98
Background: Hypoxic-ischemia (HI) encephalopathy is a major cause of neonatal mortality and subsequent neurodevelopmental disability in the surviving infants, however, there is still no effective treatment. Recent studies have showed that increased histone acetylation by inhibitors of histone deacetylase (HDAC) protects against neurodegenerative disorders and acute cerebral ischemia in adult animals. The neuroprotective effect of HDAC inhibitors involves transcription activation through increasing histone acetylation of prosurvival genes. Hypothesis: This study was to test the hypotheses that increased histone acetylation by a HDAC inhibitor- sodium butyrate (SB): 1) provides neuroprotection against HI, and 2) increases neuroplasticity after HI in the neonatal rat brain through chromatin remodeling and upregulating protective factors. Materials and methods: In the pre-treatment part, we intraperitoneally injected SB (1.2 and 1.8 g/kg) or vehicle (normal saline) daily in SD rat pups from postnatal (P) day 1 to P7. On P7, rats were subjected to HI induced by permanent ligation of unilateral carotid artery followed by 2 hours of 8% O2 hypoxia at 37°C. Histone acetylation, apoptosis and inflammatory marker activity were investigated by western blotting before and after HI. Behavior test was performed by Morris water maze ,and inhibitory avoidance test from P42 to P49, and brain injury was investigated by pathology on P49. In the post-treatment part, we treated the pups with SB or vehicle daily after HI from P14 to P42. Behavior test measured by Morris water maze test and inhibitory avoidance test was done from P42 to P49 and brain injury by pathology on P49. Results: In the pretreatment part, compared with vehicle, SB pretreatment (1.8g/kg) significantly reduced HI injury in the cortex (P<0.001), striatum (P<0.05) and hippocampus (P<0.01) and improved learning and memory performance (P <0.05). SB pretreatment (1.2g/kg) didn’t show protective effect in pathology and learning and memory performance, compared with vehicle group. Western blotting analysis showed that SB (1.8g/kg) increased acetylation levels of histone H3 and H4, and down-regulated apoptotic (caspase-3, PARP) and inflammatory (COX-2, p53) markers after HI. SB also increased Bcl-xL mRNA expression (3.2 fold) and heat shock protein 70 (HSP70) mRNA and protein levels. We also found that SB upregulated acetylation levels of Sp1, a transcription factor of HSP70. In the post-HI treatment part, SB post-treatment did not provide protective effect at pathology and behavioral levels. Conclusion: SB treatment before HI provides neuroprotection in the neonatal rat brain in association with down-regulating apoptotic and inflammatory responses and up-regulating HSP70 possibly through Sp1 acetylation. In contrast, SB post-treatment after HI does not upregulate neuroplasticity.
Lee, Hsueh-Te, and 李學德. "The neuroprotective mechanisms of carotid artery ligation-induced preconditioning against neonatal hypoxic-ischemia brain injury." Thesis, 2006. http://ndltd.ncl.edu.tw/handle/72625919712700630778.
Full text國立成功大學
基礎醫學研究所
94
Perinatal hypoxic-ischemic brain injury is a major cause of permanent neurological dysfunction in children. An approach to study the treatment of neonatal hypoxic-ischemic encephalopathy that allows for neuroprotection is to investigate the states of tolerance to hypoxic-ischemia. Twenty-four-hour carotid-artery-ligation preconditioning established by delaying the onset of hypoxia for 24 h after permanent unilateral carotid ligation in neonatal rats markedly diminished the cerebral injury. Although much has been learned about the ischemic preconditioning mechanisms in adult rats, the signaling mechanisms of this 24-h-carotid-artery-ligation preconditioning in neonatal rats remain unknown. We demonstrated that carotid artery ligation 24 h before hypoxia on postnatal day 7 (P7) rat pups provided complete neuroprotection, while artery ligation 6 h produced intermediate benefit, at behavioral and pathological levels compared to ligation 1 h before hypoxia. We first showed that the 24-h-carotid-artery-ligation preconditioning was associated with a robust and sustained activation of cAMP response element-binding protein (CREB), a transcription factor that acts as a key mediator of stimulus-induced nuclear responses underlying learning and memory, survival, and synaptic plasticity of the nervous system. Intracerebroventricular infusions of antisense CREB oligodeoxynucleotides significantly reduced the 24-h-carotid-artery ligation-induced neuroprotection by decreasing CREB expression. Pharmacological activation of the cAMP-CREB signaling with rolipram 24 h prehypoxia protected rat pups at behavioral and pathological levels by sustained increased CREB phosphorylation. These findings suggest that CREB activation provides important mechanism for potential pharmacological treatment against neonatal hypoxic-ischemic brain injury. The upstream signaling mechanisms leading to CREB activation, however, in this 24 h carotid-artery ligation preconditioning of neonatal rat brain remained unknown. We next found that vascular endothelial growth factor (VEGF)-A and VEGF receptor-2 (VEGFR-2) instead of VEGFR-1 were expressed in vessels and neurons of the P7 rat brain. Increased angiogenesis and upregulated expression of VEGF-A and VEGFR-2, but not VEGFR-1, was also found in vessels and neurons in the ipsilateral cerebral cortex 24 h after carotid artery ligation. A blockade of VEGF-A or VEGFR-2, instead of VEGFR-1, by antisense oligodeoxynucleotides decreased VEGFR-2 and pCREB expression and abolished the neuroprotective effect of carotid artery ligation preconditioning. In contrast, VEGF-A treatment or selective activation of VEGFR-2 before hypoxic-ischemia selectively upregulated VEGFR-2 and pCREB expression and provided neuroprotection against neonatal hypoxic-ischemic brain injury. Furthermore, selective activation of VEGFR-2 but not VEGFR-1 after hypoxic-ischemia also significantly protected P7 rat pups against hypoxic-ischemic brain injury. Further in vitro oxygen-glucose deprivation (OGD) study confirmed that VEGFR-2 and CREB activation was required for VEGF-A-induced neuroprotection against oxygen glucose deprivation neuronal death in differentiated H19-7 cells. Taken together, these in vivo and in vitro evidences suggest that VEGF-A/VEGFR-2 signaling leading to CREB activation is an important event in neuroprotection against hypoxic-ischemic injury in the neonatal brain. Pharmacological activation of VEGFR-2 might be an important strategy for the treatment of neonatal hypoxic-ischemic brain injury.
Ming-YiHuang and 黃名儀. "Endothelial IRS-1 aggravates neurovascular damage after neonatal hypoxic-ischemic brain injury through increasing neuroinflammation." Thesis, 2015. http://ndltd.ncl.edu.tw/handle/nppwu2.
Full text國立成功大學
臨床醫學研究所
103
Perinatal hypoxic-ischemia (HI) is a major cause of neonatal mortality and long-term neurological morbidity among survivors. HI injury not only causes neuron death but also damages endothelial cells within neurovascular unit. Maintaining the integrity of whole neurovascular unit is required for proper brain function. Disruption of neurovascular unit, especially cerebral microvascular endothelial cells, leads to BBB perturbation and causes vasogenic cerebral edema and secondary neuronal damage, which eventually exacerbates long-term disability. Thus, there is a new idea to develop means to protect the vasculature to improve HI outcome. Our previous study found that dietary restriction (DR) reduced neurovascular damage after HI and conferred long-term protection in the neonatal brain through insulin receptor substrate-1 (IRS-1)-Akt pathway. Moreover, IRS-1 over-expression protected against oxygen-glucose deprivation (OGD) cell death mainly in the endothelial cells. Therefore, an endothelium specific IRS-1 transgenic rat was created to delineate the role of IRS-1 in vascular endothelial cells against HI in the neonatal brain. We found that over-expressed IRS-1 in the vascular endothelial cells worsened the HI infarct volume with decreased tight junction proteins expression, and increased matrix metalloprotein 9 (MMP 9) production and BBB damage. Since upregulating adhesion molecules of endothelium increases peripheral leukocytes infiltration in to the damaged brain tissue and subsequently worsen the brain injury, we further investigated the expression of adhesion molecules, such as intracellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) in our transgenic rats. We found endothelial IRS-1 transgenic pups had upregulated VCAM-1 expression as early as 3 hours and ICAM-1 expression at 24 hours after HI. It associated with significantly increased microglia activation and neutrophils infiltration at 24 hours after HI. The in vitro experiments confirmed that IRS-1 over-expression in human brain microvascular endothelial cells could increase VCAM-1 and ICAM-1 expression before and after oxygen-glucose deprivation (OGD). Blockage of VCAM-1 and ICAM-1 using anti-VCAM-1 and anti-ICAM-1 antibody in our transgenic rats decreased neuronal apoptosis, and attenuated MMP 9 production, microglia activation and neutrophils infiltration after HI. In conclusion, IRS-1 specific over-expression in the vascular endothelial cells enhanced VCAM-1 and ICMA-1 expression, and exaggerated neuroinflammation and neurovascular damage after HI in the neonatal brain.
Lin, Wan-Ying, and 林宛瑩. "Anti-apoptotic mechanism of ischemic preconditioning against hypoxic-ischemic injury in the neonatal rat brain." Thesis, 2005. http://ndltd.ncl.edu.tw/handle/7j466r.
Full text國立成功大學
分子醫學研究所
93
Neonatal hypoxic-ischemic (HI) brain injury is a major cause of neonatal mortality and long-term disability. Currently, there are still no effective therapies against neonatal HI brain injury. A sublethal stress, such as a brief episode of ischemia, before a lethal injury may reduce neuronal death against subsequent lethal injury; a phenomenon called “preconditioning”. Elucidating the underlying mechanisms of preconditioning may provide potential neuroprotective therapy for neonatal HI brain injury. Permanent ligation of unilateral carotid artery followed by systemic hypoxia (8% O2 for 2 h) could induce ipsilateral cerebral HI injury in 7-day-old rat pups. To test whether the ischemia preconditioning (IP) could be established in neonatal brain in a time-dependent manner, rat pups were subjected to HI at 22 h (IP-22h group), 6 h (IP-6h group) or 2 h (IP-2h group) after reversible unilateral carotid artery ligation for 2 h. The outcome was measured by behavior assessment (P35) and pathology (P40) (percentage of brain weight reduction) and behavior (Morris water maze) on P35-P40. Compared to the No-IP group, all the three IP groups had significantly neuroprotective effect at morphological and behavioral levels. Among the three IP groups, the IP-6h group had the worst behavioral performance and also showed more TUNEL positive cells in the cortex and hippocampus. Twenty-four hours after HI, the reactive oxygen species (ROS) production and the expression levels of the cleavaged form of pro-apoptotic markers, such as caspase-3, caspase-8, caspase-9, poly (ADP-ribose) polymerase, and apoptosis inducing factor, were significantly lower in the IP-22h group compared to the no-IP group. In addition, the release of cytochrome c and Smac from mitochondria to cytosol after HI was also significantly reduced in the IP-22h group. In contrast, the expression of anti-apoptotic markers, such as cellular inhibitor of apoptosis-1(cIAP-1) and BCL2, but not X-linked inhibitor of apoptosis, were significantly higher in the IP-22h group. Increased expression of cIAP-1 but not activated caspase-3 was also found in the IP-22h group during preconditioning phase. Our study suggests that but both rapid and delayed phase of IP can be established in the immature brain, suggesting the unique plasticity in the development brain. In addition, IP-mediated neuroprotective mechanisms in the immature brain involve not only the inhibition of apoptosis (caspase-dependent and caspase-independent pathways, and intrinsic and extrinsic pathways) but also increase expression of anti-apoptotic markers, such as cIAP-1. Further work will be extended to determine the role of cIAP-1 in IP and elucidate its upstream signaling during IP in the immature rat brain.
Chien-HangTseng and 曾千航. "Lipopolysaccharide Sensitizes Hypoxic-ischemia Injury in the Immature Brain through Tumor Necrosis Factor Receptor 1 Signaling." Thesis, 2011. http://ndltd.ncl.edu.tw/handle/85537877104177878639.
Full textHsiang-YinLin and 林相吟. "The protective mechanism of low-dose lipopolysaccharide preconditioning against hypoxic-iscjemic brain injury in neonatal rats." Thesis, 2010. http://ndltd.ncl.edu.tw/handle/21367016492112758777.
Full text國立成功大學
基礎醫學研究所
98
Hypoxic-ischemic encephalopathy (HIE) in newborn is a common and important neonatal disease. It always resulted in learning disability, mental retardation, cerebral palsy and even fetal death and there is no effective prevention or therapy for HIE. Based on the concept of preconditioning, which is a sub-lethal insult providing tissue or organs resistance to subsequent lethal insult, we established a low-dose lipopolysaccharide (LPS, 0.05mg/Kg) preconditioning against hypoxic-ischemic (HI) brain damage on both behavioral (learning and memory ability) and pathological levels (Brain weight reduction) in the neonatal rats. Delineating the underlying protective mechanisms of LPS preconditioning is the aim of this study. We found that LPS preconditioning-derived eNOS up-regulation and PI3K/Akt activation in cortical neuron and cerebral vessels are required for the neuroprotection. And the PI3k/Akt activation is responsible for the eNOS up-regulation. In vitro study showed that the expression level of eNOS is associated with the susceptibility to HI in neuronal cell and vascular endothelial cell. Low-dose LPS preconditioning-mediated PI3K/Akt-eNOS is suggested to contribute to neuro-vascular protection in the immature brain. It implicated that eNOS up-regulation is a potential approach for clinical prevention of HIE. We also got insight into how low-dose LPS preconditioning influences the pathogenetic cascades of HI brain injury including ROS accumulation, neuroinflammation, apoptotic cell death and apoptotic signaling cascades. Our findings revealed that low-dose LPS preconditioning reduced HI-induced inflammation, apoptosis and oxidative stress in the neonatal HI brain. And LPS preconditioning-mediated neuroprotective effects were correlated with LPS preconditioning-mediated GSK-3 beta inhibition after HI. Inhibition of GSK-3 beta by pretreatment of GSK-3 beta inhibitor ( lithium chloride) 30 minutes before HI can mimic LPS preconditioning-induced neuroprotective effects including anti-apoptotic and anti-inflammatory effects in the neonatal HI brain. It implicated that GSK-3 beta activation contributes to the pathogenesis of neonatal HI brain injury and inhibition of GSK-3 beta may be required for the acquisition of LPS preconditioning. Taken all together, we have demonstrated the protective mechanisms underlying low-dose LPS preconditioning against HI brain injury in neonatal rats. And it may be helpful for designing potential clinical therapy or prevention for HIE.
Yi-LunChiang and 江宜倫. "Shear stress and erythromycin stimulations in endothelial cells for treatment of neonatal hypoxic-ischemia brain injury." Thesis, 2017. http://ndltd.ncl.edu.tw/handle/s85d6f.
Full text國立成功大學
細胞生物與解剖學研究所
105
Stem cells can differentiate to endothelial lineage as a source for tissue engineering of vessels. However, the rarity of cell source and low differentiation efficiency are the main limitation for stem cell therapy in vessel regeneration. Our previously study demonstrated a successful endothelial cell (EC) differentiation by integration of biomechanical force and chemical factors, but the mechanism is still unknown. Furthermore, we build up a cell therapy of brain hypoxic-ischemia (HI) model by using human umbilical vein endothelial cells (HUVECs). The differentiated cells will apply to this animal model in the future. By static HUVECs transplantation in brain HI model, brain injury and cell death were decreased. Laminar shear stress (LSS)-treated HUVECs group in animal model had not significantly repair function than static HUVECs. Previously research indicates that CXCR4, a chemoattractant receptor, is reduced by LSS. One paper showed that erythromycin (EM) increased CXCR4 expression. Therefore, we combined LSS with EM to stimulate HUVECs. We found that EM treatment after LSS stimulation of HUVEC transplantation was useful for rescuing brain injury. In this study, we built up successful cell therapy for brain injury model. Furthermore, repair function of ECs can be enlarged by increasing EM-induced CXCR4 expression.
Dunlop, Kate. "Neuroprotective Effects of a Novel Apple Peel Extract AF4 in a Mouse Model of Hypoxic-Ischemic Brain Injury." 2011. http://hdl.handle.net/10222/13997.
Full textDunlop, Kate Elizabeth. "Neuroprotective Effects of a Novel Apple Peel Extract AF4 in a Mouse Model of Hypoxic-Ischemic Brain Injury." 2011. http://hdl.handle.net/10222/15722.
Full textGuan, Jian. "Insulin-like growth factor-1 after hypoxic-ischemic brain injury: effects and modes of action on neuronal survival." 1996. http://hdl.handle.net/2292/3116.
Full textSukha, Neelam. "The developmental motor outcomes of infants with hypoxic ischaemic encephalopathy II and III between the ages of 12-14 months at Chris Hani Baragwanath academic hospital." Thesis, 2013.
Find full textThis study determined outcomes for motor developmental delay in infants, 12-14 months, diagnosed with HIE II and III, at Chris Hani Baragwanath Academic Hospital. Twenty nine infants diagnosed with HIE II and nine infants diagnosed with HIE III were assessed using the Peabody Development Motor Scale- 2, at their corrected age. Demographic, antenatal and perinatal factors similar to those in other studies were found for this sample. Infants with HIE III had significantly more developmental delay (p=0.01) than infants with HIE II. Fifty two percent of infants with HIE II had no delay while a 100% of infants with HIE III presented with disability. A greater percentage of infants had delay in fine motor skills. Infants with severe and moderate disabilities were receiving intervention whereas those mild disabilities were often missed in screening clinics. It is vital to ensure these infants are assessed and followed up to remediate difficulties as soon as they arise.
FitriHandayani and 蘇涵亭. "Protection of neurovascular structure from hypoxic-ischemic brain injury by different progenitor cells derived from adipose-derived stem cell." Thesis, 2014. http://ndltd.ncl.edu.tw/handle/ykhwz8.
Full text國立成功大學
細胞生物與解剖學研究所
102
Adipose-derived stem cells (ASCs) show promise for regenerative medicine researches and able to differentiate into either endothelial or neuronal lineages. The current study aimed to investigate the therapeutic potential of human adipose-derived stem cell for hypoxic-ischemic (HI) brain injury. The HI brain injury was created by right common carotid artery ligation and then exposure to hypoxia (8% O2) for 2h. The human ASCs were differentiated into NPCs and EPCs by culturing ASCs in the chitosan and shear stress microenvironments. The rat pups were divided into 5 groups to receive different treatments by using intra-peritoneum injection, including Naïve, PBS, hASCs, EPCs, NPCs, and combination of EPCs and NPCs (E+N). All rat pups were sacrificed at 7th day after HI. The infracted area and ratio of cell apoptosis were determined by TTC, and Nissl staining. Our results showed significant reduction of cerebral infarction, increase of neurons, and decrease of cell apoptosis after injected with therapeutic cells. Therapeutic effects were analyzed with immunofluorescence staining to detect neurons, astrocytes, and endothelial cells. The quantified results showed that GFAP positive cells on the ischemic side of cortex were lower in therapeutic cell treatment as compared with PBS group. The branch point of vessel and NeuN positive were enhanced after cell therapies for 7 days in HI brain injury. The positive cells with human chromatin staining confirmed the engraftment of transplanted cells homing to the damaged brain. Combination between EPC and NPC involve signaling in Nrp1 signal in EPCs to promote the angiogenesis. Therefore, the progenitor cells from hASCs may benefit as a treatment of HI brain injury.
Lan-WanWang and 王藍浣. "The Effects of Hypoxic-Ischemia and Inflammation on White Matter Injury in the Immature Brain — Clinical and Experimental Approaches." Thesis, 2012. http://ndltd.ncl.edu.tw/handle/98c2d2.
Full text國立成功大學
臨床醫學研究所
101
Very low-birth-weight (VLBW, 〈 1500 g) premature infants have high risk of neurodevelopmental impairment, and cerebral palsy (CP) is the major disability in very preterm survivors. Cerebral white matter injury is the principal brain injury leading to CP in VLBW infants. Hypoxic-ischemia (HI) and inflammation/infection are the two major pathogenic factors of white matter injury and CP. From both experimental and clinical approaches, we examined whether HI and inflammation/infection have joint effects on the risk of CP in very preterm infants, and white matter injury in the immature brain. Objective: Clinical Study: To examine the joint effects of HI and infectious events in the perinatal and neonatal periods on CP risk in VLBW preterm infants. Experimental Study: To investigate whether low-dose lipopolysaccharide (LPS) sensitizes HI-induced white matter injury in the immature brain by selectively up-regulating neuroinflammation and blood-brain barrier (BBB) damage in the white matter; and if so, whether c-Jun N-terminal kinases (JNK) signaling is the shared pathway linking neuroinflammation, BBB leakage and oligodendroglial apoptosis in the white matter injury. Study Design: Clinical Study: From 1995 to 2005, prospective registry of 6318 VLBW preterm infants admitted to Taiwan hospitals was conducted. CP was diagnosed at corrected age 24 months. The cumulative effects of HI and infectious events during the perinatal and neonatal periods on CP risk were analyzed. Experimental Study: Postpartum (P) day 2 rat pups received LPS (0.05 mg/kg) (LPS+HI) or normal saline (NS+HI) followed by 90-minute HI. LPS and NS group were the pups that had LPS or NS only. Immunohistochemistry and immunoblotting were used to determine microglia activation, tumor necrosis factor-alpha (TNF-α), BBB damage, cleaved caspase-3, JNK and phospho-JNK (p-JNK), myelin basic protein (MBP), and glial fibrillary acidic protein (GFAP) expression. Immunofluorescence was performed to determine the cellular distribution of p-JNK. Pharmacological and genetic approaches were used to inhibit JNK activity. Results: Clinical Study: Of the 3946 infants who had completed 24-month neuromotor examinations, 466 (11.8%) had CP, 2238 (56.7%) normal outcomes, and 1242 (31.5%) minor or other impairment. CP group had significantly higher incidences of HI events in the perinatal and neonatal periods, and sepsis in the neonatal period than normal group. Three HI events, including birth cardiopulmonary resuscitation, chronic lung disease and severe apnea/bradycardia, were the most significant predictors for CP. Relative to CP risk for infants with neither HI nor sepsis, the CP odds increased 1.91-fold, 2.22-fold and 5.07-fold for infants with birth cardiopulmonary resuscitation, chronic lung disease, and severe apnea/bradycardia, respectively; while the combination with sepsis increased the odds by 3.05-fold, 3.10-fold and 7.11-fold, respectively. Using the three HI events plus sepsis, CP rates were 10.3%, 18.5%, 28.0%, 41.3% and 55.6% for infants with none, one, two, three and four events, respectively. Experimental Study: Myelin basic protein immunohistochemistry on P11 showed white matter injury in LPS+HI group, but not in NS+HI, LPS and NS groups. In contrast, no gray matter injury was found in the four groups. In the white matter, increases of activated microglia, TNF-α expression, BBB leakage and cleaved caspase-3-positive oligodendrocyte progenitors were much more prominent in LPS+HI group than in the other three groups 24 hours post-insult. Immunoblotting and immunohistochemical analyses showed early and sustained JNK activation in the white matter at 6 and 24 hours post-insult. Immunofluorescence demonstrated up-regulation of p-JNK in activated microglia, vascular endothelial cells and oligodendrocyte progenitors, and also showed perivascular aggregation of p-JNK-positive cells around the vessels 24 hours post-insult. JNK inhibition by AS601245 or by antisense oligodeoxynucleotides significantly reduced microglial activation, TNF-α immunoreactivity, IgG extravasation, and cleaved caspase 3 in the endothelial cells and oligodendrocyte progenitors, and also attenuated perivascular aggregation of p-JNK-positive cells 24 hours post-insult. The AS601245 or JNK antisense oligodeoxynucleotide group had significantly increased MBP and decreased GFAP expression in the white matter on P11 than the vehicle or scrambled oligodeoxynucleotides group. Conclusions: We concluded that HI and inflammation/infection have joint effects on CP risk in very preterm infants, and on the pathogenesis of white matter injury in the immature brain. Clinically, HI events and sepsis across the perinatal and neonatal periods exert cumulative effects on CP risk in VLBW premature infants. Experimentally, low-dose LPS sensitizes HI-induced white matter injury in the immature brain by selectively up-regulating neuroinflammation and BBB damage in the white matter, and JNK signaling is the shared pathway linking neuroinflammation, BBB leakage and oligodendroglial apoptosis in the white matter injury. The clinical and experimental studies may help reduce significant risk factors and develop effective therapeutic strategies against white matter injury to improve neurodevelopmental outcomes of very preterm infants.
Lin, Chien-min, and 林乾閔. "The basic and clinical researches of Chinese and Western medicine in the prevention and treatment of severe brain injury with hypoxic change." Thesis, 2012. http://ndltd.ncl.edu.tw/handle/17631660336843753740.
Full textDostálová, Veronika. "Vliv cerebrálního hypoxického poškození na kognitivní funkce a psychosociální faktory." Doctoral thesis, 2019. http://www.nusl.cz/ntk/nusl-405841.
Full text