• Title/Summary/Keyword: Brain ischemia

Search Result 403, Processing Time 0.025 seconds

Hyperglycemia aggravates decrease in alpha-synuclein expression in a middle cerebral artery occlusion model

  • Kang, Ju-Bin;Kim, Dong-Kyun;Park, Dong-Ju;Shah, Murad-Ali;Kim, Myeong-Ok;Jung, Eun-Jung;Lee, Han-Shin;Koh, Phil-Ok
    • Laboraroty Animal Research
    • /
    • v.34 no.4
    • /
    • pp.195-202
    • /
    • 2018
  • Hyperglycemia is one of the major risk factors for stroke. Hyperglycemia can lead to a more extensive infarct volume, aggravate neuronal damage after cerebral ischemia. ${\alpha}$-Synuclein is especially abundant in neuronal tissue, where it underlies the etiopathology of several neurodegenerative diseases. This study investigated whether hyperglycemic conditions regulate the expression of ${\alpha}$-synuclein in middle cerebral artery occlusion (MCAO)-induced cerebral ischemic injury. Male Sprague-Dawley rats were treated with streptozotocin (40 mg/kg) via intraperitoneal injection to induce hyperglycemic conditions. MCAO were performed four weeks after streptozotocin injection to induce focal cerebral ischemia, and cerebral cortex tissues were obtained 24 hours after MCAO. We confirmed that MCAO induced neurological functional deficits and cerebral infarction, and these changes were more extensive in diabetic animals compared to non-diabetic animals. Moreover, we identified a decrease in ${\alpha}$-synuclein after MCAO injury. Diabetic animals showed a more serious decrease in ${\alpha}$-synuclein than non-diabetic animals. Western blot and reverse-transcription PCR analyses confirmed more extensive decreases in ${\alpha}$-synuclein expression in MCAO-injured animals with diabetic condition than these of non-diabetic animals. It is accepted that ${\alpha}$-synuclein modulates neuronal cell death and exerts a neuroprotective effect. Thus, the results of this study suggest that hyperglycemic conditions cause more serious brain damage in ischemic brain injuries by decreasing ${\alpha}$-synuclein expression.

Taurine transport at the blood-brain barrier in spontaneously hypertensive rats and normotensive rats

  • Lim, Ji-Hyoun;Kang, Young-Sook
    • Proceedings of the Korean Society of Applied Pharmacology
    • /
    • 1998.11a
    • /
    • pp.154-155
    • /
    • 1998
  • Taurine, 2-aminoethanesulfonic acid is widely distributed in animal tissues and has a variety of biological activities. A recent worldwide study demonstrated beneficial effects of taurine on aging and age-associated disorders. In general, taurine levels in the brain decrease when an animal is subjected to pathologic conditions such as ischemia-anoxia and seizure. But taurine levels tend to increase in the brain in hypertention. In the present study, the blood-brain barrier BBB) transport of [$^3$H]taurine was compared between spontaneously hypertensive rats (SHR) and normotensive Sprague-Dawley rats (SD) using Internal artery carotid perfusion (ICAP) at a rate of 4$m\ell$/min for 10, 15 and 30 second. Calculated V$\_$D/, volume of distribution in brain, and PS, the permeability surface area product of [$^3$H]taurine through the BBB in SHR was a little lower than that in SD. PS for 15s is more higher than that of other seconds in both of them. It could be followed by taurine efflux back into blood after 15s. We also obtained pharmacokinetic parameters using intravenous injection of plasma volume marker, [$\^$14/C]sucrose and [$^3$H] taurine. PS value of [$^3$H]taurine in SHR (16.1 ${\pm}$ 2.9 ${\times}$ 10$\^$-3/ $m\ell$/min/g) was significantly higher than that in SD (7.4 ${\pm}$ 0.8 ${\times}$ 10$\^$-3/ $m\ell$/min/g). There is also significant difference for %ID/g in brain between SHR (0.195 ${\pm}$ 0.031) and SD (0.058 ${\pm}$ 0.003).

  • PDF

Acid-sensing ion channels (ASICs): therapeutic targets for neurological diseases and their regulation

  • Kweon, Hae-Jin;Suh, Byung-Chang
    • BMB Reports
    • /
    • v.46 no.6
    • /
    • pp.295-304
    • /
    • 2013
  • Extracellular acidification occurs not only in pathological conditions such as inflammation and brain ischemia, but also in normal physiological conditions such as synaptic transmission. Acid-sensing ion channels (ASICs) can detect a broad range of physiological pH changes during pathological and synaptic cellular activities. ASICs are voltage-independent, proton-gated cation channels widely expressed throughout the central and peripheral nervous system. Activation of ASICs is involved in pain perception, synaptic plasticity, learning and memory, fear, ischemic neuronal injury, seizure termination, neuronal degeneration, and mechanosensation. Therefore, ASICs emerge as potential therapeutic targets for manipulating pain and neurological diseases. The activity of these channels can be regulated by many factors such as lactate, $Zn^{2+}$, and Phe-Met-Arg-Phe amide (FMRFamide)-like neuropeptides by interacting with the channel's large extracellular loop. ASICs are also modulated by G protein-coupled receptors such as CB1 cannabinoid receptors and 5-$HT_2$. This review focuses on the physiological roles of ASICs and the molecular mechanisms by which these channels are regulated.

Matrix Metalloproteinases, New Insights into the Understanding of Neurodegenerative Disorders

  • Kim, Yoon-Seong;Joh, Tong-H.
    • Biomolecules & Therapeutics
    • /
    • v.20 no.2
    • /
    • pp.133-143
    • /
    • 2012
  • Matrix metalloproteinases (MMPs) are a subfamily of zinc-dependent proteases that are re-sponsible for degradation and remodeling of extracellular matrix proteins. The activity of MMPs is tightly regulated at several levels including cleavage of prodomain, allosteric activation, com-partmentalization and complex formation with tissue inhibitor of metalloproteinases (TIMPs). In the central nervous system (CNS), MMPs play a wide variety of roles ranging from brain devel-opment, synaptic plasticity and repair after injury to the pathogenesis of various brain disorders. Following general discussion on the domain structure and the regulation of activity of MMPs, we emphasize their implication in various brain disorder conditions such as Alzheimer's disease, multiple sclerosis, ischemia/reperfusion and Parkinson's disease. We further highlight accumu-lating evidence that MMPs might be the culprit in Parkinson's disease (PD). Among them, MMP-3 appears to be involved in a range of pathogenesis processes in PD including neuroinflamma-tion, apoptosis and degradation of ${\alpha}$-synuclein and DJ-1. MMP inhibitors could represent poten-tial novel therapeutic strategies for treatments of neurodegenerative diseases.

The Effects of Nerve Growth Factor Expression of Central Nerve System by Environmental Enrichment and Peripheral Nerve Electrical Stimulation in Brain Ischemia Model Rats (뇌졸중 유발 백서모델에서 환경강화와 말초신경전기자극이 중추신경계의 신경성장인자에 미치는 영향)

  • Kim, Sa-Youl;Kim, Eun-Jung;Kim, Gye-Yeop
    • The Journal of Korean Physical Therapy
    • /
    • v.19 no.4
    • /
    • pp.33-41
    • /
    • 2007
  • Purpose: To investigate environmental enrichment and nerve stimulation follows in application times with the change of BDNF & Trk-B receptor in the motor cortex and spinal cord. Methods: Experimental groups were divided into the five groups. Group I: normal control group, Group II: experiment control group, Group III: sciatic never electrical stimulation after MCAO, Group IV: application of only environmental enrichment after MCAO, Group V: never electrical stimulation with environmental enrichment after MCAO. Histologic observation and coronal sections were processed individually in goat polyclonal antibody phosphorylated BDNF and rabbit polyclonal antibody Trk-B receptor. Results: In immunohistochemistric response of BDNF and Trk-B, group II were showed that lower response effect at postischemic 1 days, 3 days, and 7 days. Group V were showed that increase response effect at postischemic 3 days, 7 days and 14 days. Specially showed that the most response effect at postischemic 14 days. In neurobehavioral assessment, group V were significantly difference from other groups on between-subject effects. Conclusion: The above results suggest that combined environmental enrichment with peripheral nerve electrical stimulation in focal ischemic brain injury were more improved that the change of BDNF & Trk-B receptor expression than non treatment.

  • PDF

Role of ginseng in the neurovascular unit of neuroinflammatory diseases focused on the blood-brain barrier

  • Kim, Minsu;Mok, Hyejung;Yeo, Woon-Seok;Ahn, Joong-Hoon;Choi, Yoon Kyung
    • Journal of Ginseng Research
    • /
    • v.45 no.5
    • /
    • pp.599-609
    • /
    • 2021
  • Ginseng has long been considered as an herbal medicine. Recent data suggest that ginseng has antiinflammatory properties and can improve learning- and memory-related function in the central nervous system (CNS) following the development of CNS neuroinflammatory diseases such as Alzheimer's disease, cerebral ischemia, and other neurological disorders. In this review, we discuss the role of ginseng in the neurovascular unit, which is composed of endothelial cells surrounded by astrocytes, pericytes, microglia, neural stem cells, oligodendrocytes, and neurons, especially their blood-brain barrier maintenance, anti-inflammatory effects and regenerative functions. In addition, cell-cell communication enhanced by ginseng may be attributed to regeneration via induction of neurogenesis and angiogenesis in CNS diseases. Thus, ginseng may have therapeutic potential to exert cognitive improvement in neuroinflammatory diseases such as stroke, traumatic brain injury, multiple sclerosis, Parkinson's disease, and Alzheimer's disease.

Ischemic Stroke in a Patient with Heterozygote Fabry's Disease (보인자 파브리병 환자에서 발생한 허혈뇌졸중)

  • Yang, Sun Woo;Choi, Jay Chol;Kim, Hyun Woo;Jeong, Jin-Ho;Hyun, Chang-Lim;Koh, Myeong Ju
    • Journal of the Korean neurological association
    • /
    • v.36 no.4
    • /
    • pp.341-344
    • /
    • 2018
  • It is uncommon for Fabry's disease (FD) patient to present with an isolated ischemic stroke without other typical symptoms or signs of FD. A 48-year-old woman presented with recurrent limb weakness and her brain magnetic resonance imaging revealed multiple ischemic brain lesions. Ten years ago, the patient had been diagnosed with heterozygote FD by the genetic test, but she had not shown any typical symptoms or sign of FD so far. Isolated organ involvement could occur in heterozygote FD.

Hypoxia-inducible factor: role in cell survival in superoxide dismutase overexpressing mice after neonatal hypoxia-ischemia

  • Jeon, Ga Won;Sheldon, R. Ann;Ferriero, Donna M.
    • Clinical and Experimental Pediatrics
    • /
    • v.62 no.12
    • /
    • pp.444-449
    • /
    • 2019
  • Background: Sixty percent of infants with severe neonatal hypoxic-ischemic encephalopathy die, while most survivors have permanent disabilities. Treatment for neonatal hypoxic-ischemic encephalopathy is limited to therapeutic hypothermia, but it does not offer complete protection. Here, we investigated whether hypoxia-inducible factor (HIF) promotes cell survival and suggested neuroprotective strategies. Purpose: HIF-1α deficient mice have increased brain injury after neonatal hypoxia-ischemia (HI), and the role of HIF-2α in HI is not well characterized. Copper-zinc superoxide dismutase (SOD)1 overexpression is not beneficial in neonatal HI. The expression of HIF-1α and HIF-2α was measured in SOD1 overexpressing mice and compared to wild-type littermates to see if alteration in expression explains this lack of benefit. Methods: On postnatal day 9, C57Bl/6 mice were subjected to HI, and protein expression was measured by western blotting in the ipsilateral cortex of wild-type and SOD1 overexpressing mice to quantify HIF-1α and HIF-2α. Spectrin expression was also measured to characterize the mechanism of cell death. Results: HIF-1α protein expression did not significantly change after HI injury in the SOD1 overexpressing or wild-type mouse cortex. However, HIF-2α protein expression increased 30 minutes after HI injury in the wild-type and SOD1 overexpressing mouse cortex and decreased to baseline value at 24 hours after HI injury. Spectrin 145/150 expression did not significantly change after HI injury in the SOD1 overexpressing or wild-type mouse cortex. However, spectrin 120 expression increased in both wild-type and SOD1 overexpressing mouse at 4 hours after HI, which decreased by 24 hours, indicating a greater role of apoptotic cell death. Conclusion: HIF-1α and HIF-2α may promote cell survival in neonatal HI in a cell-specific and regional fashion. Our findings suggest that early HIF-2α upregulation precedes apoptotic cell death and limits necrotic cell death. However, the influence of SOD was not clarified; it remains an intriguing factor in neonatal HI.

Cilostazol Promotes the Migration of Brain Microvascular Endothelial Cells (Cilostazol에 의한 뇌혈관내피세포의 세포이동 증진 효과연구)

  • Lee, Sae-Won;Park, Jung Hwa;Shin, Hwa Kyoung
    • Journal of Life Science
    • /
    • v.26 no.12
    • /
    • pp.1367-1375
    • /
    • 2016
  • Cilostazol is known to be a selective inhibitor of phosphodiesterase III and is generally used to treat stroke. Our previous findings showed that cilostazol enhanced capillary density through angiogenesis after focal cerebral ischemia. Angiogenesis is an important physiological process for promoting revascularization to overcome tissue ischemia. It is a multistep process consisting of endothelial cell proliferation, migration, and tubular structure formation. Here, we examined the modulatory effect of cilostazol at each step of the angiogenic mechanism by using human brain microvascular endothelial cells (HBMECs). We found that cilostazol increased the migration of HBMECs in a dose-dependent manner. However, it did not enhance HBMEC proliferation and capillary-like tube formation. We used a cDNA microarray to analyze the mechanisms of cilostazol in cell migration. We picked five candidate genes that were potentially related to cell migration, and we confirmed the gene expression levels by real-time PCR. The genes phosphoserine aminotransferase 1 (PSAT1) and CCAAT/enhancer binding protein ${\beta}$ ($C/EBP{\beta}$) were up-regulated. The genes tissue factor pathway inhibitor 2 (TFPI2), retinoic acid receptor responder 1 (RARRES1), and RARRES3 were down-regulated. Our observations suggest that cilostazol can promote angiogenesis by promoting endothelial migration. Understanding the cilostazol-modulated regulatory mechanisms in brain endothelial cells may help stimulate blood vessel formation for the treatment of ischemic diseases.

A study of the Mori Radicis Cortex pre-treatment on transient ischemic brain injury in mice (상백피(桑白皮) 메탄올 추출물 전처치가 일과성 허혈에 의한 생쥐의 뇌 손상에 미치는 영향)

  • Chung, Byung-Woo;Lim, Jae-Yu;Lee, Se-Eun;Lee, Byoungho;Lim, Sehyun;Lim, Chiyeon;Cho, Suin
    • The Korea Journal of Herbology
    • /
    • v.32 no.1
    • /
    • pp.25-31
    • /
    • 2017
  • Objectives : Mori Radicis Cortex (MRC), the root epidermis of Morus alba L., has been traditionally used to treat lung-related diseases in Korean Medicine. The common of MRC is Mulberry bark Morus bark, and it's pharmaceutical properties and taste are known as sweet and cold, and it promotes urination and reduce edema by reducing heat from the lungs and soothe asthma. In the present study, anti-apoptotic mechanism of MRC in middle cerebral artery occlusion (MCAO) model in mice. Methods : Two-hundred grams of MRC was extracted with methanol at room temperature for 5 days, and this was repeated one time. After filtration, the methanol was removed using vacuum evaporator, then stored at $-20^{\circ}C$ until use. C57BL/6 male mice were housed in an environment with controlled humidity, temperature, and light cycle. In order to determine beneficial effects of MRC on ischemia induced brain damage, infarct volume, neurological deficit scores, activities of several apoptosis-related proteins such as caspase-8, -9, Bcl-xL in MCAO-induced brains of mice were analyzed. Mice in MRC-treated groups were orally administered 30, 100, or 300 mg/kg of body weight for three consecutive days before commencing the MCAO procedure. Results : Pre-treatment of MRC significantly reduced infarct volume in MCAO subjected mice applied with 300 mg/kg of MRC methanol extract, and MRC effectively inhibited Bcl-xL reduction and caspase-9 activation caused by MCAO-induced brain damage. Conclusions : MRC showed neuro-protective effects by regulating apoptosis-related protein signals, and it can be a potential candidate for the therapy of ischemia-induced brain damage.