• Title/Summary/Keyword: Neuronal Damage

Search Result 355, Processing Time 0.022 seconds

Application of Near-Infrared Spectroscopy in Neurological Disorders: Especially in Orthostatic Intolerance (신경계 질환에서 근적외선분광분석법의 적용: 기립불내증을 중심으로)

  • Kim, Yoo Hwan;Paik, Seung-ho;Phillips V, Zephaniah;Seok, Hung Youl;Jeon, Nam-Joon;Kim, Beop-Min;Kim, Byung-Jo
    • Journal of the Korean neurological association
    • /
    • v.35 no.1
    • /
    • pp.8-15
    • /
    • 2017
  • Near-infrared spectroscopy (NIRS), a noninvasive optical method, utilizes the characteristic absorption spectra of hemoglobin in the near-infrared range to provide information on cerebral hemodynamic changes in various clinical situations. NIRS monitoring have been used mainly to detect reduced perfusion of the brain during orthostatic stress for three common forms of orthostatic intolerance (OI); orthostatic hypotension, neurally mediated syncope, and postural orthostatic tachycardia syndrome. Autonomic function testing is an important diagnostic test to assess their autonomic nervous systems for patients with symptom of OI. However, these techniques cannot measure dynamic changes in cerebral blood flow. There are many experimentations about study of NIRS to reveal the pathophysiology of patients with OI. Research using NIRS in other neurologic diseases (stroke, epilepsy and migraine) are ongoing. NIRS have been experimentally used in all stages of stroke and may complement the established diagnostic and monitoring tools. NIRS also provide pathophysiological approach during rehabilitation and secondary prevention of stroke. The hemodynamic response to seizure has long been a topic for discussion in association with the neuronal damage resulting from convulsion. One critical issue when unpredictable events are to be detected is how continuous NIRS data are analyzed. Besides, NIRS studies targeting pathophysiological aspects of migraine may contribute to a deeper understanding of mechanisms relating to aura of migraine. NIRS monitoring may play an important role to trend regional hemodynamic distribution of flow in real time and also highlights the pathophysiology and management of not only patients with OI symptoms but also those with various neurologic diseases.

Neuroprotective effects of hesperetin on H2O2-induced damage in neuroblastoma SH-SY5Y cells

  • Ha-Rin Moon;Jung-Mi Yun
    • Nutrition Research and Practice
    • /
    • v.17 no.5
    • /
    • pp.899-916
    • /
    • 2023
  • BACKGROUND/OBJECTIVES: Oxidative stress is a fundamental neurodegenerative disease trigger that damages and decimates nerve cells. Neurodegenerative diseases are chronic central nervous system disorders that progress and result from neuronal degradation and loss. Recent studies have extensively focused on neurodegenerative disease treatment and prevention using dietary compounds. Heseperetin is an aglycone hesperidin form with various physiological activities, such as anti-inflammation, antioxidant, and antitumor. However, few studies have considered hesperetin's neuroprotective effects and mechanisms; thus, our study investigated this in hydrogen peroxide (H2O2)-treated SH-SY5Y cells. MATERIALS/METHODS: SH-SY5Y cells were treated with H2O2 (400 µM) in hesperetin absence or presence (10-40 µM) for 24 h. Three-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assays detected cell viability, and 4',6-diamidino-2-phenylindole staining allowed us to observe nuclear morphology changes such as chromatin condensation and apoptotic nuclei. Reactive oxygen species (ROS) detection assays measured intracellular ROS production; Griess reaction assays assessed nitric oxide (NO) production. Western blotting and quantitative polymerase chain reactions quantified corresponding mRNA and proteins. RESULTS: Subsequent experiments utilized various non-toxic hesperetin concentrations, establishing that hesperetin notably decreased intracellular ROS and NO production in H2O2-treated SH-SY5Y cells (P < 0.05). Furthermore, hesperetin inhibited H2O2-induced inflammation-related gene expression, including interluekin-6, tumor necrosis factor-α, and nuclear factor kappa B (NF-κB) p65 activation. In addition, hesperetin inhibited NF-κB translocation into H2O2-treated SH-SY5Y cell nuclei and suppressed mitogen-activated protein kinase protein expression, an essential apoptotic cell death regulator. Various apoptosis hallmarks, including shrinkage and nuclear condensation in H2O2-treated cells, were suppressed dose-dependently. Additionally, hesperetin treatment down-regulated Bax/Bcl-2 expression ratios and activated AMP-activated protein kinase-mammalian target of rapamycin autophagy pathways. CONCLUSION: These results substantiate that hesperetin activates autophagy and inhibits apoptosis and inflammation. Hesperetin is a potentially potent dietary agent that reduces neurodegenerative disease onset, progression, and prevention.

Neuroprotective effects of resveratrol via anti-apoptosis on hypoxic-ischemic brain injury in neonatal rats (신생 백서의 저 산소 허혈 뇌손상에서 항세포사멸사를 통한 resveratrol의 신경보호 효과)

  • Shin, Jin Young;Seo, Min Ae;Choi, Eun Jin;Kim, Jin Kyung;Seo, Eok Su;Lee, Jun Hwa;Chung, Hai Lee;Kim, Woo Taek
    • Clinical and Experimental Pediatrics
    • /
    • v.51 no.10
    • /
    • pp.1102-1111
    • /
    • 2008
  • Purpose : Resveratrol, extracted from red wine and grapes, has an anti-cancer effect, an antiinflammatory effect, and an antioxidative effect mainly in heart disease and also has neuroprotective effects in the adult animal model. No studies for neuroprotective effects during the neonatal periods have been reported. Therefore, we studied the neuroprotective effect of resveratrol on hypoxic-ischemic brain damage in neonatal rats via anti-apoptosis. Methods : Embryonic cortical neuronal cell culture of rat brain was performed using pregnant Sprague-Dawley (SD) rats at 18 days of gestation (E18) for the in vitro approach. We injured the cells with hypoxia and administered resveratrol (1, 10, and $30{\mu}g/mL$) to the cells at 30 minutes before hypoxic insults. In addition, unilateral carotid artery ligation with hypoxia was induced in 7-day-old neonatal rats for the in vivo approach. We injected resveratrol (30 mg/kg) intraperitoneally into animal models. Real-time PCR and Western blotting were performed to identify the neuroprotective effects of resveratrol through anti-apoptosis. Results : In the in vitro approach of hypoxia, the expression of Bax, caspase-3, and the ratio of Bax/Bcl-2, indicators of the level of apoptosis, were significantly increased in the hypoxia group compared to the normoxia group. In the case of the resveratrol-treated group, expression was significantly decreased compared to the hypoxia group. And the results in the in vivo approach were the same as in the in vitro approach. Conclusion : The present study demonstrates that resveratrol plays neuroprotective role in hypoxic-ischemic brain damage during neonatal periods through the mechanism of anti-apoptosis.

Manganese and Iron Interaction: a Mechanism of Manganese-Induced Parkinsonism

  • Zheng, Wei
    • Proceedings of the Korea Environmental Mutagen Society Conference
    • /
    • 2003.10a
    • /
    • pp.34-63
    • /
    • 2003
  • Occupational and environmental exposure to manganese continue to represent a realistic public health problem in both developed and developing countries. Increased utility of MMT as a replacement for lead in gasoline creates a new source of environmental exposure to manganese. It is, therefore, imperative that further attention be directed at molecular neurotoxicology of manganese. A Need for a more complete understanding of manganese functions both in health and disease, and for a better defined role of manganese in iron metabolism is well substantiated. The in-depth studies in this area should provide novel information on the potential public health risk associated with manganese exposure. It will also explore novel mechanism(s) of manganese-induced neurotoxicity from the angle of Mn-Fe interaction at both systemic and cellular levels. More importantly, the result of these studies will offer clues to the etiology of IPD and its associated abnormal iron and energy metabolism. To achieve these goals, however, a number of outstanding questions remain to be resolved. First, one must understand what species of manganese in the biological matrices plays critical role in the induction of neurotoxicity, Mn(II) or Mn(III)? In our own studies with aconitase, Cpx-I, and Cpx-II, manganese was added to the buffers as the divalent salt, i.e., $MnCl_2$. While it is quite reasonable to suggest that the effect on aconitase and/or Cpx-I activites was associated with the divalent species of manganese, the experimental design does not preclude the possibility that a manganese species of higher oxidation state, such as Mn(III), is required for the induction of these effects. The ionic radius of Mn(III) is 65 ppm, which is similar to the ionic size to Fe(III) (65 ppm at the high spin state) in aconitase (Nieboer and Fletcher, 1996; Sneed et al., 1953). Thus it is plausible that the higher oxidation state of manganese optimally fits into the geometric space of aconitase, serving as the active species in this enzymatic reaction. In the current literature, most of the studies on manganese toxicity have used Mn(II) as $MnCl_2$ rather than Mn(III). The obvious advantage of Mn(II) is its good water solubility, which allows effortless preparation in either in vivo or in vitro investigation, whereas almost all of the Mn(III) salt products on the comparison between two valent manganese species nearly infeasible. Thus a more intimate collaboration with physiochemists to develop a better way to study Mn(III) species in biological matrices is pressingly needed. Second, In spite of the special affinity of manganese for mitochondria and its similar chemical properties to iron, there is a sound reason to postulate that manganese may act as an iron surrogate in certain iron-requiring enzymes. It is, therefore, imperative to design the physiochemical studies to determine whether manganese can indeed exchange with iron in proteins, and to understand how manganese interacts with tertiary structure of proteins. The studies on binding properties (such as affinity constant, dissociation parameter, etc.) of manganese and iron to key enzymes associated with iron and energy regulation would add additional information to our knowledge of Mn-Fe neurotoxicity. Third, manganese exposure, either in vivo or in vitro, promotes cellular overload of iron. It is still unclear, however, how exactly manganese interacts with cellular iron regulatory processes and what is the mechanism underlying this cellular iron overload. As discussed above, the binding of IRP-I to TfR mRNA leads to the expression of TfR, thereby increasing cellular iron uptake. The sequence encoding TfR mRNA, in particular IRE fragments, has been well-documented in literature. It is therefore possible to use molecular technique to elaborate whether manganese cytotoxicity influences the mRNA expression of iron regulatory proteins and how manganese exposure alters the binding activity of IPRs to TfR mRNA. Finally, the current manganese investigation has largely focused on the issues ranging from disposition/toxicity study to the characterization of clinical symptoms. Much less has been done regarding the risk assessment of environmenta/occupational exposure. One of the unsolved, pressing puzzles is the lack of reliable biomarker(s) for manganese-induced neurologic lesions in long-term, low-level exposure situation. Lack of such a diagnostic means renders it impossible to assess the human health risk and long-term social impact associated with potentially elevated manganese in environment. The biochemical interaction between manganese and iron, particularly the ensuing subtle changes of certain relevant proteins, provides the opportunity to identify and develop such a specific biomarker for manganese-induced neuronal damage. By learning the molecular mechanism of cytotoxicity, one will be able to find a better way for prediction and treatment of manganese-initiated neurodegenerative diseases.

  • PDF

Sequential 1H MR Spectroscopy(MRS) Studies of Kaolin-Induced Hydrocephalic Cat Brain (Kaolin 유발 고양이 수두증 모델에서 양자 자기공명 분광상의 경시적 변화)

  • Kim, Myung Jin;Hwang, Sung Kyoo;Hwang, Jeong Hyun;Chang, Yongmin;Kim, Yong Sun;Kim, Seung Lae
    • Journal of Korean Neurosurgical Society
    • /
    • v.29 no.11
    • /
    • pp.1421-1428
    • /
    • 2000
  • Objectives : The aim of this study is to evaluate the sequential metabolic changes in experimental hydrocephalus and the clinical applicability to the diagnosis and prognosis of hydrocephalus using proton MR spectroscopy. Methods : Hydrocephalus was experimentally induced in 30 cats(2-3kg body weight) by injecting 1ml of sterile kaolin suspension(250mg/ml) into the cisterna magna. Proton MRS was performed with a 1.5 T MRI/MRS unit (Vision Plus, Siemens) at pre-treatment and at 1, 3, 7, 14, 21, and 28 days after the kaolin injection. PRESS(TR/TE=1500/270msec) technique was employed. The major metabolites which include N-acetyl aspartate (NAA), creatine(Cr), choline(Cho), and lactate(Lac) were quantitatively analyzed and the relative concentrations ratios were evaluated. Multislice $T_2$-weighted images were also obtained using fast spin echo sequence(TR/TE= 2500/96msec) to monitor the morphologic changes along with progression of hydrocephalus. Results : Hydrocephalus was successfully induced in all 30 cats. Twenty five cats died within 3 days and one at the end of the second week. In all animals, the NAA/Cr ratios initially decreased during the acute stage. In 4 surviving cats, the NAA/Cr ratios initially decreased during the acute stage(<14 days) and then gradually increased to the prekaolin level as follows : pre-kaolin($1.49{\pm}0.04$), day 1($1.11{\pm}0.07$), day 7($1.17{\pm}0.04$), day 14($1.40{\pm}0.03$), day 21 ($1.46{\pm}0.06$), day 28($1.43{\pm}0.03$). These levels were relatively well correlated with the symptomatologic improvement. Lactate peak, which reflects the evidence of ischemia, did not appear throughout the entire period except in one case which expired at the end of the second week. Conclusions : The NAA/Cr ratio of the sequential proton MRS in kaolin-induced hydrocephalic cats reflects a metabolic aspect of the hydrocephalus at each stage. A decreased NAA level at the early stage is from both neuronal and axonal damage which may provide diagnostic information in the acute stage of hydrocephalus. In addition, the initial fall of NAA/Cr ratio and recovery in the late stage, when no lactate peak emerges, may suggest that the main insult of the parenchyma is not to the neuron itself but to the axon, which may be related to a good prognosis. However, emergence of the lactate peak and unrecoverable NAA/Cr at the end of the acute phase may be a poor prognostic factor. In the chronic stage, recovery of NAA/Cr ratio may provide a diagnostic clue for the differentiation between hydrocephalus and cortical atrophy.

  • PDF