1
|
A Case Series of 39 United States Veterans with Mild Traumatic Brain Injury Treated with Hyperbaric Oxygen Therapy. CLINICAL AND TRANSLATIONAL NEUROSCIENCE 2022. [DOI: 10.3390/ctn6030021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Importance: The Defense and Veterans Brain Injury Center reported 358,088 mild traumatic brain injury (mTBI) among U.S. service members worldwide between the years 2000 and 2020. Veterans with mTBI have higher rates of Post-Traumatic Stress Disorder (PTSD), depressive disorder, substance use disorder, anxiety disorder, and suicide than their healthy counterparts. Currently, there is no effective treatment for mTBI. Objective: To assess the efficacy of hyperbaric oxygen therapy (HBOT) as a treatment option for mTBI. Design, Setting, Participants: This is a case series of 39 U.S. Veterans diagnosed with mTBI and treated with HBOT. Of these participants, 36 were men and 3 women, and their ages ranged between 28 and 69. The treatment was administered by The 22 Project (a veteran-centered nonprofit organization) using monoplace hyperbaric chambers located in Delray Beach, Florida. Neuroimaging using Single Photon Emission Computer Tomography (SPECT) brain scans performed pre- and post-HBOT were made available for secondary analysis. Nilearn Python Library was utilized to visualize the corresponding neuroimaging data. A two-sided paired t-test in R was used to compare the pre- and post-treatment results. Intervention: A full treatment of HBOT involved 40 sessions. Each session consisted of the administration of 100% oxygen at 1.5 atmospheres for 90 min, twice a day, for 20 days, Mondays to Fridays only. Main Outcome and Measure: Perfusion in the brain’s Brodmann Areas (BA) comparing pre- and post-HBOT using NeuroGam software analysis from brain SPECT scan neuroimaging and multi-symptom self-reports. Results: A comparison between the pre- and post-HBOT brain scans showed significant improvement in the brain perfusion, and the difference was statistically significant (p < 0.001). Separately, participants reported reduced pain, improved mood, and better sleep, an outcome that translated into an average of about 46.6% improvement in the measured symptoms. Conclusions and Relevance: This series demonstrated that HBOT could be a useful treatment for mTBI in U.S. veterans. The participants in the study showed marked improvement in both brain perfusion measured on SPECT scan imaging and measured mTBI symptoms. This is the first study to use brain SPECT scans with quantitative numerical measurements to demonstrate improvement in brain perfusion in veterans with mild TBI treated with HBOT and measured mTBI symptoms. Future research studies are currently being done to validate these important findings.
Collapse
|
2
|
Zhao J, Yin L, Jiang L, Hou L, He L, Zhang C. PTEN nuclear translocation enhances neuronal injury after hypoxia-ischemia via modulation of the nuclear factor-κB signaling pathway. Aging (Albany NY) 2021; 13:16165-16177. [PMID: 34114972 PMCID: PMC8266328 DOI: 10.18632/aging.203141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/29/2021] [Indexed: 11/25/2022]
Abstract
The occurrence of hypoxia-ischemia (HI) in the developing brain is closely associated with neuronal injury and even death. However, the underlying molecular mechanism is not fully understood. This study was designed to investigate phosphatase and tensin homolog (PTEN) nuclear translocation and its possible role in rat cortical neuronal damage following oxygen-glucose deprivation (OGD) in vitro. An in vitro OGD model was established using primary cortical neurons dissected from newborn Sprague-Dawley rats to mimic HI conditions. The PTENK13R mutant plasmid, which contains a lysine-to-arginine mutation at the lysine 13 residue, was constructed. The nuclei and cytoplasm of neurons were separated. Neuronal injury following OGD was evidenced by increased lactate dehydrogenase (LDH) release and apoptotic cell counts. In addition, PTEN expression was increased and the phosphorylation of extracellular signal-regulated kinase 1/2 (p-ERK1/2) and activation of nuclear factor kappa B (NF-κB) were decreased following OGD. PTENK13R transfection prevented PTEN nuclear translocation; attenuated the effect of OGD on nuclear p-ERK1/2 and NF-κB, apoptosis, and LDH release; and increased the expression of several anti-apoptotic proteins. We conclude that PTEN nuclear translocation plays an essential role in neuronal injury following OGD via modulation of the p-ERK1/2 and NF-κB pathways. Prevention of PTEN nuclear translocation might be a candidate strategy for preventing brain injury following HI.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Linlin Yin
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Lin Jiang
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Li Hou
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Ling He
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Chunyan Zhang
- Department of Neonatology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| |
Collapse
|
3
|
Lactate Administration Reduces Brain Injury and Ameliorates Behavioral Outcomes Following Neonatal Hypoxia-Ischemia. Neuroscience 2020; 448:191-205. [PMID: 32905840 DOI: 10.1016/j.neuroscience.2020.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 01/02/2023]
Abstract
Neonatal hypoxic-ischemic encephalopathy is a major cause of mortality and disability in newborns and the only standard approach for treating this condition is therapeutic hypothermia, which shows some limitations. Thus, putative neuroprotective agents have been tested in animal models. The present study evaluated the administration of lactate, a potential energy substrate of the central nervous system (CNS) in an animal model of hypoxia-ischemia (HI), that mimics in neonatal rats the brain damage observed in human newborns. Seven-day-old (P7) male and female Wistar rats underwent permanent common right carotid occlusion combined with an exposition to a hypoxic atmosphere (8% oxygen) for 60 min. Animals were assigned to four experimental groups: HI, HI + LAC, SHAM, SHAM + LAC. Lactate was administered intraperitoneally 30 min and 2 h after hypoxia in HI + LAC and SHAM + LAC groups. HI and SHAM groups received vehicle at the same time points. The volume of brain lesion was evaluated in P9. Animals underwent behavioral assessments: negative geotaxis, righting reflex (P8 and P14), and cylinder test (P20). Lactate administration reduced the volume of brain lesion and improved behavioral parameters after HI in both sexes. Thus, lactate administration could be a neuroprotective strategy for the treatment of neonatal HI, a disorder still affecting a significant percentage of human newborns.
Collapse
|
4
|
Schuffels S, Nakada S, Wu Y, Lim YP, Chen X, Stonestreet BS. Effects of inter-alpha inhibitor proteins on brain injury after exposure of neonatal rats to severe hypoxia-ischemia. Exp Neurol 2020; 334:113442. [PMID: 32896573 DOI: 10.1016/j.expneurol.2020.113442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 12/25/2022]
Abstract
Hypoxic-ischemic (HI) brain injury is one of the most common neurological problems occurring in premature and full-term infants after perinatal complications. Hypothermia is the only treatment approved for HI encephalopathy in newborns. However, this treatment is only partially protective, cannot be used to treat premature infants, and has limited efficacy to treat severe HI encephalopathy. Inflammation contributes to the evolution of HI brain injury in neonates. Inter-alpha Inhibitor Proteins (IAIPs) are immunomodulatory proteins that have neuroprotective properties after exposure to moderate HI in neonatal rats. The objective of the current study was to determine the neuroprotective efficacy of treatment with IAIPs starting immediately after or with a delay of one hour after exposure to severe HI of 120 min duration. One hundred and forty-six 7-day-old rat pups were randomized to sham control, HI and immediate treatment with IAIPs (60 mg/kg) or placebo (PL), and sham, HI and delayed treatment with IAIPs or PL. IAIPs or PL were given at zero, 24, and 48 h after HI or 1, 24 and 48 h after HI. Total brain infarct volume was determined 72 h after exposure to HI. Treatment with IAIPs immediately after HI decreased (P < 0.05) infarct volumes by 58.0% and 44.5% in male and female neonatal rats, respectively. Delayed treatment with IAIPs after HI decreased (P < 0.05) infarct volumes by 23.7% in male, but not in female rats. We conclude that IAIPs exert neuroprotective effects even after exposure to severe HI in neonatal rats and appear to exhibit some sex-related differential effects.
Collapse
Affiliation(s)
- Stephanie Schuffels
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, United States of America
| | - Sakura Nakada
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, United States of America
| | - Yuqi Wu
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, United States of America
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI, The Alpert Medical School of Brown University, Providence, RI, United States of America; Department of Pathology and Laboratory Medicine, The Alpert Medical School of Brown University, Providence, RI, United States of America
| | - Xiaodi Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, United States of America.
| | - Barbara S Stonestreet
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, United States of America.
| |
Collapse
|
5
|
c-Mpl and TPO expression in the human central nervous system neurons inhibits neuronal apoptosis. Aging (Albany NY) 2020; 12:7397-7410. [PMID: 32341206 PMCID: PMC7202501 DOI: 10.18632/aging.103086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/13/2020] [Indexed: 12/11/2022]
Abstract
Thrombopoietin (TPO) is a growth factor for the megakaryocytic/platelet lineage. In this study, we investigated the expression of TPO and its receptor, c-Mpl, in the human central nervous system (CNS) and their roles after a neural insult. Our results demonstrate that both TPO and c-Mpl are expressed in the neurons of the human CNS. TPO was also detected in human cerebrospinal fluid. TPO was found to be neuroprotective in hypoxic-ischemic neonatal rat brain models. In these rat models, treatment with TPO reduced brain damage and improved sensorimotor functions. In addition, TPO promoted C17.2 cell proliferation through activation of the PI3K/Akt signaling pathway. Via the Bcl-2/BAX signaling pathway, TPO exerted an antiapoptotic effect by suppressing mitochondrial membrane potentials. Taken together, our results indicate that TPO is neuroprotective in the CNS.
Collapse
|
6
|
Dilek M, Orallar H, Cetinkaya A, Bozat G, Pehlivan F, Bekdas M, Kabakus N. Can Excessive Oxygen Cause Hyperactive Behavior Disorder in Preterm Children? Cognitive Effects of Hyperoxia in the Preterm Brain of Rats. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09819-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
7
|
Safety and efficacy evaluations of an adeno-associated virus variant for preparing IL10-secreting human neural stem cell-based therapeutics. Gene Ther 2019; 26:135-150. [PMID: 30692604 DOI: 10.1038/s41434-019-0057-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 12/20/2018] [Accepted: 01/07/2019] [Indexed: 12/22/2022]
Abstract
Gene therapy technologies are inevitably required to boost the therapeutic performance of cell therapies; thus, validating the efficacy of gene carriers specifically used for preparing cellular therapeutics is a prerequisite for evaluating the therapeutic capabilities of gene and cell combinatorial therapies. Herein, the efficacy of a recombinant adeno-associated virus derivative (rAAVr3.45) was examined to evaluate its potential as a gene carrier for genetically manipulating interleukin-10 (IL10)-secreting human neural stem cells (hNSCs) that can potentially treat ischemic injuries or neurological disorders. Safety issues that could arise during the virus preparation or viral infection were investigated; no replication-competent AAVs were detected in the final cell suspensions, transgene expression was mostly transient, and no severe interference on endogenous gene expression by viral infection occurred. IL10 secretion from hNSCs infected by rAAVr3.45 encoding IL10 did not alter the transcriptional profile of any gene by more than threefold, but the exogenously boosted IL10 was sufficient to provoke immunomodulatory effects in an ischemic brain injury animal model, thereby accelerating the recovery of neurological deficits and the reduction of brain infarction volume. This study presents evidence that rAAVr3.45 can be potentially used as a gene carrier to prepare stem cell therapeutics.
Collapse
|
8
|
Fabres RB, da Rosa LA, de Souza SK, Cecconello AL, Azambuja AS, Sanches EF, Ribeiro MFM, de Fraga LS. Effects of progesterone on the neonatal brain following hypoxia-ischemia. Metab Brain Dis 2018; 33:813-821. [PMID: 29363039 DOI: 10.1007/s11011-018-0193-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/17/2018] [Indexed: 11/25/2022]
Abstract
Progesterone displays a strong potential for the treatment of neonatal hypoxic-ischemic encephalopathy since it has been shown to be beneficial in the treatment of the central nervous system injuries in adult animals. Here, we evaluated the effects of the administration of progesterone (10 mg/kg) in seven-days-old male Wistar rats submitted to neonatal hypoxia-ischemia (HI). Progesterone was administered immediately before ischemia and/or 6 and 24 h after the onset of hypoxia. The body weight of the animals, the volume of brain lesion and the expression of p-Akt and procaspase-3 in the hippocampus were evaluated. All animals submitted to HI showed a reduction in the body weight. However, this reduction was more remarkable in those animals which received progesterone before surgery. Administration of progesterone was unable to reduce the volume of brain damage caused by HI. Moreover, no significant differences were observed in the expression of p-Akt and procaspase-3 in animals submitted to HI and treated with either progesterone or vehicle. In summary, progesterone did not show a neuroprotective effect on the volume of brain lesion in neonatal rats submitted to hypoxia-ischemia. Furthermore, progesterone was unable to modulate p-Akt and procaspase-3 signaling pathways, which may explain the absence of neuroprotection. On the other hand, it seems that administration of progesterone before ischemia exerts some systemic effect, leading to a remarkable reduction in the body weight.
Collapse
Affiliation(s)
- Rafael Bandeira Fabres
- Laboratory of Neurohumoral Interaction, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Laboratory of Comparative Metabolism and Endocrinology, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-903, Brazil
| | - Luciana Abreu da Rosa
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
| | - Samir Khal de Souza
- Laboratory of Comparative Metabolism and Endocrinology, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
| | - Ana Lucia Cecconello
- Laboratory of Neurohumoral Interaction, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
| | - Amanda Stapenhorst Azambuja
- Laboratory of Neurohumoral Interaction, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
| | - Eduardo Farias Sanches
- Laboratory of Cerebral Ischemia, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-000, Brazil
| | - Maria Flavia Marques Ribeiro
- Laboratory of Neurohumoral Interaction, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil
| | - Luciano Stürmer de Fraga
- Laboratory of Comparative Metabolism and Endocrinology, Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil.
- Hospital de Clínicas de Porto Alegre (HCPA), Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-903, Brazil.
| |
Collapse
|
9
|
Gurgul S, Buyukakilli B, Komur M, Okuyaz C, Balli E, Ozcan T. Does Levetiracetam Administration Prevent Cardiac Damage in Adulthood Rats Following Neonatal Hypoxia/Ischemia-Induced Brain Injury? ACTA ACUST UNITED AC 2018; 54:medicina54020012. [PMID: 30344243 PMCID: PMC6037241 DOI: 10.3390/medicina54020012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 12/12/2022]
Abstract
Cardiovascular abnormalities are widespread when a newborn is exposed to a hypoxic-ischemic injury in the neonatal period. Although the neuroprotective effects of levetiracetam (LEV) have been reported after hypoxia, the cardioprotective effects of LEV have not been documented. Therefore, we aimed to investigate whether levetiracetam (LEV) has a protective effect on cardiac-contractility and ultrastructure of heart muscle in rats exposed to hypoxia-ischemia (HI) during the neonatal period. A total of 49 seven-day-old rat pups were separated into four groups. For HI induction, a combination of right common carotid artery ligation with 8% oxygen in seven-day-old rat pups for 2 h was performed for saline, LEV100, and LEV200 groups. Just after hypoxia, LEV100 and LEV200 groups were administered with 100 mg/kg and 200 mg/kg of LEV, respectively. The arteries of rats in the control group were only detected; no ligation or hypoxia was performed. At the end of the 16th week after HI, cardiac mechanograms were recorded, and samples of tissue were explored by electronmicroscopy.While ventricular contractility in the control group was similar to LEV100, there were significant decreases in both saline and LEV200 groups (p < 0.05). Although ventricular contractile duration of the control and saline groups was found to be similar, durations in the LEV100 and LEV200 groups were significantly higher (p < 0.05). After HI, mitochondrial damage and ultrastructural deteriorative alterations in ventricles and atriums of the LEV-administered groups were significantly less severe than the saline group. The present study showed that neonatal HI caused long-term cardiac dysfunction and ultrastructural deteriorations in cardiac muscles. LEV administration just after HI might possess some protective effects against myocardial damage and contractility.
Collapse
Affiliation(s)
- Serkan Gurgul
- Department of Biophysics, Faculty of Medicine, Gaziantep University, TR-27310 Gaziantep, Turkey.
| | - Belgin Buyukakilli
- Department of Biophysics, Faculty of Medicine, Mersin University, TR-33343 Mersin, Turkey.
| | - Mustafa Komur
- Department of Child Health and Disease, Faculty of Medicine, Mersin University, TR-33343 Mersin, Turkey.
| | - Cetin Okuyaz
- Department of Child Health and Disease, Faculty of Medicine, Mersin University, TR-33343 Mersin, Turkey.
| | - Ebru Balli
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, TR-33343 Mersin, Turkey.
| | - Tuba Ozcan
- Department of Histology and Embryology, Faculty of Medicine, K. Sütcü Imam University, TR-46040 Kahramanmaraş, Turkey.
| |
Collapse
|
10
|
Li Q, Wang J, Li Y, Xu X. Neuroprotective effects of salidroside administration in a mouse model of Alzheimer's disease. Mol Med Rep 2018; 17:7287-7292. [PMID: 29568861 DOI: 10.3892/mmr.2018.8757] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 09/06/2017] [Indexed: 11/06/2022] Open
Abstract
Salidroside administration improves memory in different models of learning. However, its influence on models of Alzheimer's disease (AD) has not been widely studied. In the present study, the therapeutic effect of salidroside was investigated in an animal model of AD. APPswe/PS1ΔE9 mouse (n=20) were randomly divided into either the AD model group or the salidroside + AD model group (n=10 in each group), and C57BL/6J mouse (n=20) of identical age and genetic background were randomly divided into either the normal control (NC) group or the salidroside + NC group (n=10 in each group). The Morris water maze behavioral test was applied to all mice in order to investigate the effects of salidroside administration on learning and memory functions. The concentrations of malondialdehyde (MDA), glutathione (GSH) and nitrate in the hippocampus of the mice were determined, and hippocampal superoxide dismutase (SOD) activity was also determined. In addition, terminal deoxynucleotidyl‑transferase‑mediated dUTP nick end labeling was used to investigate the rate of neuronal apoptosis in the hippocampus. Furthermore, the concentrations of interleukin‑6 (IL‑6) and tumor necrosis factor‑α (TNF‑α) were tested for in the brain tissues of AD mice. Learning and memory functions in AD mice were revealed to improve following administration of salidroside. Furthermore, salidroside administration was revealed to decrease the concentrations of MDA and nitrate in the hippocampus, decrease the apoptotic rate of hippocampal neurons, and increase the activity of SOD and the concentration of GSH in hippocampal tissue. In addition, it was demonstrated that salidroside administration suppressed the expression levels of IL‑6 and TNF‑α. In conclusion, this study revealed that the administration of salidroside could attenuate the effects of AD‑associated memory and learning impairment in mice. Furthermore, it was demonstrated that the effects of salidroside administration on AD mice were, at least partially, via inhibition of brain oxidative/nitrosative damage, suppression of both IL‑6 and TNF‑α expression levels, and suppression of the hippocampal neuronal apoptotic rate.
Collapse
Affiliation(s)
- Qingyun Li
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| | - Jinhua Wang
- Department of Neurology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang 318000, P.R. China
| | - Yuwang Li
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| | - Xiaolin Xu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| |
Collapse
|
11
|
Chen D, Dixon BJ, Doycheva DM, Li B, Zhang Y, Hu Q, He Y, Guo Z, Nowrangi D, Flores J, Filippov V, Zhang JH, Tang J. IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic-ischemic brain injury in rats. J Neuroinflammation 2018; 15:32. [PMID: 29394934 PMCID: PMC5797348 DOI: 10.1186/s12974-018-1077-9] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/22/2018] [Indexed: 12/18/2022] Open
Abstract
Background The endoplasmic reticulum (ER) is responsible for the control of correct protein folding and protein function which is crucial for cell survival. However, under pathological conditions, such as hypoxia–ischemia (HI), there is an accumulation of unfolded proteins thereby triggering the unfolded protein response (UPR) and causing ER stress which is associated with activation of several stress sensor signaling pathways, one of them being the inositol requiring enzyme-1 alpha (IRE1α) signaling pathway. The UPR is regarded as a potential contributor to neuronal cell death and inflammation after HI. In the present study, we sought to investigate whether microRNA-17 (miR-17), a potential IRE1α ribonuclease (RNase) substrate, arbitrates downregulation of thioredoxin-interacting protein (TXNIP) and consequent NLRP3 inflammasome activation in the immature brain after HI injury and whether inhibition of IRE1α may attenuate inflammation via miR-17/TXNIP regulation. Methods Postnatal day 10 rat pups (n = 287) were subjected to unilateral carotid artery ligation followed by 2.5 h of hypoxia (8% O2). STF-083010, an IRE1α RNase inhibitor, was intranasally delivered at 1 h post-HI or followed by an additional one administration per day for 2 days. MiR-17-5p mimic or anti-miR-17-5p inhibitor was injected intracerebroventricularly at 48 h before HI. Infarct volume and body weight were used to evaluate the short-term effects while brain weight, gross and microscopic brain tissue morphologies, and neurobehavioral tests were conducted for the long-term evaluation. Western blots, immunofluorescence staining, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), and co-immunoprecipitation (Co-IP) were used for mechanism studies. Results Endogenous phosphorylated IRE1α expression was significantly increased after HI. Intranasal administration of STF-083010 alleviated brain injury and improved neurological behavior. MiR-17-5p expression was reduced after HI, and this decrease was attenuated by STF-083010 treatment. MiR-17-5p mimic administration ameliorated TXNIP expression, NLRP3 inflammasome activation, caspase-1 cleavage, and IL-1β production, as well as brain infarct volume. Conversely, anti-miR-17-5p inhibitor reversed IRE1α inhibition-induced decrease in TXNIP expression and inflammasome activation, as well as exacerbated brain injury after HI. Conclusions IRE1a-induced UPR pathway may contribute to inflammatory activation and brain injury following neonatal HI. IRE1a activation, through decay of miR-17-5p, elevated TXNIP expression to activate NLRP3 inflammasome and aggravated brain damage. Electronic supplementary material The online version of this article (10.1186/s12974-018-1077-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Di Chen
- Cerebrovascular Diseases Laboratory, Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
| | - Brandon J Dixon
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Desislava M Doycheva
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Bo Li
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Yang Zhang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Qin Hu
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Yue He
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Zongduo Guo
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Derek Nowrangi
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Jerry Flores
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Valery Filippov
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Jiping Tang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
12
|
Ding Y, Yao P, Hong T, Han Z, Zhao B, Chen W, Zhou G. Early hyperbaric oxygen effects on neuropathic pain and nitric oxide synthase isoforms in CCI rats. Oncotarget 2018; 9:7513-7521. [PMID: 29484128 PMCID: PMC5800920 DOI: 10.18632/oncotarget.23867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 12/24/2017] [Indexed: 12/20/2022] Open
Abstract
Neuropathic pain is pain caused by injury or dysfunction in the central and/or peripheral nervous system. Neuropathic pain has a high incidence with a complex mechanism, but effective treatment remains elusive. Hyperbaric oxygen (HBO) therapy has been widely used in the treatment of a variety of neurological diseases. The current study used a rat model of neuropathic pain induced by chronic constriction injury (CCI) of the sciatic nerve. We observed the effects of early use of 2.5 absolute atmosphere (ATA) HBO on neuropathic pain-related behaviors and the expression of nitric oxide synthase (NOS) isoforms in the spinal dorsal horn. In the CCI group, mechanical withdrawal threshold (MWT) was decreased, Thermal withdrawal latency (TWL) was shortened, and mRNA and protein levels of iNOS and nNOS were significantly increased compared to the sham group. MWT was increased, TWL was enhanced, and iNOS and nNOS levels were significantly decreased in the HBO group compared to the CCI group. There was no change in eNOS levels across all groups. HBO treatment at early stages can improve hyperalgesia.
Collapse
Affiliation(s)
- Yuanyuan Ding
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peng Yao
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tao Hong
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhenkai Han
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Baisong Zhao
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Weimin Chen
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guangyu Zhou
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Tan JW, Zhang F, Liu HJ, Li Z. Hyperbaric oxygen ameliorated the lesion scope and nerve function in acute spinal cord injury patients: A retrospective study. Clin Biochem 2017; 53:1-7. [PMID: 29217422 DOI: 10.1016/j.clinbiochem.2017.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/29/2017] [Accepted: 12/03/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVE This is a retrospective study to assess the therapeutic effect of hyperbaric oxygen (HBO) in early treatment of acute spinal cord injury (SCI) using magnetic resonance imaging (MRI) and electrophysiology in diagnosing. METHODS Forty acute SCI patients from Sun Yat-Sen Memorial Hospital who were assigned into HBO treatment were included during August 2013 to October 2014.The patients with adverse reactions or contraindications for HBO were assigned as controls. Both of two groups (HBO and Control) received medicine treatment with Urbason, GM-1 and mecobalamine after surgery. ASIA and the Frankel scores were used to evaluate the therapeutic effect of HBO at the 15th and 30th day after HBO treatment by using MRI and electrophysiology features. RESULTS Significant therapeutic effect of HBO treatment on acute SCI patients was observed compared with the control group (P<0.05). Comparison for ASIA and Frankel scores showed that motor and neurological functions were significantly improved in HBO group at day 15 and day 30 post treatment. MRI images showed that the grade III injury in HBO group was significant lower than the control group. In comparison with the control, the peak of somatosensory evoked potential (SEP) and motor evoked potential (MEP) amplitude increased, the latency was shortened, and the conduction velocity of sensory nerve (SCV) and motor nerve (MCV) was significantly increased in the HBO group (P<0.05). CONCLUSIONS HBO treatment has a great efficacy in acute SCI patients. HBO therapy at early stage of acute SCI is beneficiary to the recovery.
Collapse
Affiliation(s)
- Jie-Wen Tan
- Department of Rehabilitation Medicine, XinHua College, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China
| | - Fang Zhang
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China
| | - He-Jun Liu
- Department of Rehabilitation Medicine, XinHua College, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China
| | - Zhuo Li
- Department of Hyperbaric Oxygen, Beijing Chaoyang Hospital Affiliated to Capital University of Medical Science, China.
| |
Collapse
|
14
|
Sheng M, Liu P, Mao D, Ge Y, Lu H. The impact of hyperoxia on brain activity: A resting-state and task-evoked electroencephalography (EEG) study. PLoS One 2017; 12:e0176610. [PMID: 28464001 PMCID: PMC5412995 DOI: 10.1371/journal.pone.0176610] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 04/13/2017] [Indexed: 12/17/2022] Open
Abstract
A better understanding of the effect of oxygen on brain electrophysiological activity may provide a more mechanistic insight into clinical studies that use oxygen treatment in pathological conditions, as well as in studies that use oxygen to calibrate functional magnetic resonance imaging (fMRI) signals. This study applied electroencephalography (EEG) in healthy subjects and investigated how high a concentration of oxygen in inhaled air (i.e., normobaric hyperoxia) alters brain activity under resting-state and task-evoked conditions. Study 1 investigated its impact on resting EEG and revealed that hyperoxia suppressed α (8-13Hz) and β (14-35Hz) band power (by 15.6±2.3% and 14.1±3.1%, respectively), but did not change the δ (1-3Hz), θ (4-7Hz), and γ (36-75Hz) bands. Sham control experiments did not result in such changes. Study 2 reproduced these findings, and, furthermore, examined the effect of hyperoxia on visual stimulation event-related potentials (ERP). It was found that the main peaks of visual ERP, specifically N1 and P2, were both delayed during hyperoxia compared to normoxia (P = 0.04 and 0.02, respectively). In contrast, the amplitude of the peaks did not show a change. Our results suggest that hyperoxia has a pronounced effect on brain neural activity, for both resting-state and task-evoked potentials.
Collapse
Affiliation(s)
- Min Sheng
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Peiying Liu
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Deng Mao
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yulin Ge
- Department of Radiology, New York University Langone Medical Center, New York, New York, United States of America
| | - Hanzhang Lu
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
15
|
Hu Q, Manaenko A, Xu T, Guo Z, Tang J, Zhang JH. Hyperbaric oxygen therapy for traumatic brain injury: bench-to-bedside. Med Gas Res 2016; 6:102-110. [PMID: 27867476 PMCID: PMC5110132 DOI: 10.4103/2045-9912.184720] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious public health problem in the United States. Survivors of TBI are often left with significant cognitive, behavioral, and communicative disabilities. So far there is no effective treatment/intervention in the daily clinical practice for TBI patients. The protective effects of hyperbaric oxygen therapy (HBOT) have been proved in stroke; however, its efficiency in TBI remains controversial. In this review, we will summarize the results of HBOT in experimental and clinical TBI, elaborate the mechanisms, and bring out our current understanding and opinions for future studies.
Collapse
Affiliation(s)
- Qin Hu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Departments of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Anatol Manaenko
- Departments of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Ting Xu
- Departments of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Zhenni Guo
- Departments of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- Departments of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Departments of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
16
|
Chen J, Chen YH, Lv HY, Chen LT. Effect of hyperbaric oxygen on lipid peroxidation and visual development in neonatal rats with hypoxia-ischemia brain damage. Biomed Rep 2016; 5:136-140. [PMID: 27347417 DOI: 10.3892/br.2016.673] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/19/2016] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the effect of hyperbaric oxygen (HBO) on lipid peroxidation and visual development in a neonatal rat model of hypoxic-ischemic brain damage (HIBD). The rat models of HIBD were established by delayed uterus dissection and were divided randomly into two groups (10 rats each): HIBD and HBO-treated HIBD (HIBD+HBO) group. Another 20 rats that underwent sham-surgery were also divided randomly into the HBO-treated and control groups. The rats that underwent HBO treatment received HBO (0.02 MPa, 1 h/day) 24 h after the surgery and this continued for 14 days. When rats were 4 weeks old, their flash visual evoked potentials (F-VEPs) were monitored and the ultrastructures of the hippocampus were observed under transmission electron microscope. The levels of superoxide dismutase (SOD) and malonyldialdehyde (MDA) in the brain tissue homogenate were detected by xanthine oxidase and the thiobarbituric acid colorimetric method. Compared with the control group, the ultrastructures of the pyramidal neurons in the hippocampal CA3 area were distorted, the latencies of F-VEPs were prolonged (P<0.01) and the SOD activities were lower while the MDA levels were higher (P<0.01) in the HIBD group. No significant differences in ultrastructure, the latency of F-VEPs or SOD/MDA levels were identified between the HBO-treated HIBD group and the normal control group (P>0.05). HBO enhances antioxidant capacity and reduces the ultrastructural damage induced by hypoxic-ischemia, which may improve synaptic reconstruction and alleviate immature brain damage to promote the habilitation of brain function.
Collapse
Affiliation(s)
- Jing Chen
- Department of Pediatrics, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China; Department of Child Health, Xiamen Maternal and Child Health Care Hospital, Xiamen, Fujian 361003, P.R. China
| | - Yan-Hui Chen
- Department of Pediatrics, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Hong-Yan Lv
- Department of Pediatrics, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Li-Ting Chen
- Department of Pediatrics, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
17
|
Ostrowski RP, Stępień K, Pucko E, Matyja E. Hyperbaric oxygen modalities are differentially effective in distinct brain ischemia models. Med Gas Res 2016; 6:39-47. [PMID: 27826422 PMCID: PMC5075682 DOI: 10.4103/2045-9912.179344] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The effectiveness and efficacy of hyperbaric oxygen (HBO) preconditioning and post-treatment modalities have been demonstrated in experimental models of ischemic cerebrovascular diseases, including global brain ischemia, transient focal and permanent focal cerebral ischemia, and experimental neonatal hypoxia-ischemia encephalopathy. In general, early and repetitive post-treatment of HBO appears to create enhanced protection against brain ischemia whereas delayed HBO treatment after transient focal ischemia may even aggravate brain injury. This review advocates the level of injury reduction upon HBO as an important component for translational evaluation of HBO based treatment modalities. The combined preconditioning and HBO post-treatment that would provide synergistic effects is also worth considering.
Collapse
Affiliation(s)
- Robert P Ostrowski
- Department of Experimental and Clinical Neuropathology, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Stępień
- Department of Experimental and Clinical Neuropathology, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Emanuela Pucko
- Department of Experimental and Clinical Neuropathology, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Matyja
- Department of Experimental and Clinical Neuropathology, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
18
|
Romero JI, Hanschmann EM, Gellert M, Eitner S, Holubiec MI, Blanco-Calvo E, Lillig CH, Capani F. Thioredoxin 1 and glutaredoxin 2 contribute to maintain the phenotype and integrity of neurons following perinatal asphyxia. Biochim Biophys Acta Gen Subj 2015; 1850:1274-85. [PMID: 25735211 DOI: 10.1016/j.bbagen.2015.02.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 02/14/2015] [Accepted: 02/24/2015] [Indexed: 01/23/2023]
Abstract
BACKGROUND Thioredoxin (Trx) family proteins are crucial mediators of cell functions via regulation of the thiol redox state of various key proteins and the levels of the intracellular second messenger hydrogen peroxide. Their expression, localization and functions are altered in various pathologies. Here, we have analyzed the impact of Trx family proteins in neuronal development and recovery, following hypoxia/ischemia and reperfusion. METHODS We have analyzed the regulation and potential functions of Trx family proteins during hypoxia/ischemia and reoxygenation of the developing brain in both an animal and a cellular model of perinatal asphyxia. We have analyzed the distribution of 14 Trx family and related proteins in the cerebellum, striatum, and hippocampus, three areas of the rat brain that are especially susceptible to hypoxia. Using SH-SY5Y cells subjected to hypoxia and reoxygenation, we have analyzed the functions of some redoxins suggested by the animal experiment. RESULTS AND CONCLUSIONS We have described/discovered a complex, cell-type and tissue-specific expression pattern following the hypoxia/ischemia and reoxygenation. Particularly, Grx2 and Trx1 showed distinct changes during tissue recovery following hypoxia/ischemia and reoxygenation. Silencing of these proteins in SH-SY5Y cells subjected to hypoxia-reoxygenation confirmed that these proteins are required to maintain the normal neuronal phenotype. GENERAL SIGNIFICANCE These findings demonstrate the significance of redox signaling in cellular pathways. Grx2 and Trx1 contribute significantly to neuronal integrity and could be clinically relevant in neuronal damage following perinatal asphyxia and other neuronal disorders.
Collapse
Affiliation(s)
- Juan Ignacio Romero
- Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini" (ININCA), Facultad de Medicina, UBA-CONICET, Marcelo T. de Alvear 2270, C1122AAJ, Ciudad de Buenos Aires, Argentina
| | - Eva-Maria Hanschmann
- Institute for Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Ernst-Moritz-Arndt-Universität Greifswald, 17475 Greifswald, Germany
| | - Manuela Gellert
- Institute for Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Ernst-Moritz-Arndt-Universität Greifswald, 17475 Greifswald, Germany
| | - Susanne Eitner
- Institute for Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Ernst-Moritz-Arndt-Universität Greifswald, 17475 Greifswald, Germany
| | - Mariana Inés Holubiec
- Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini" (ININCA), Facultad de Medicina, UBA-CONICET, Marcelo T. de Alvear 2270, C1122AAJ, Ciudad de Buenos Aires, Argentina
| | - Eduardo Blanco-Calvo
- Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini" (ININCA), Facultad de Medicina, UBA-CONICET, Marcelo T. de Alvear 2270, C1122AAJ, Ciudad de Buenos Aires, Argentina; Facultat d'Educació, Psicologia i Treball Social Universitat de Lleida Av. de l'Estudi General, 4, 25001 Lleida, Spain
| | - Christopher Horst Lillig
- Institute for Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Ernst-Moritz-Arndt-Universität Greifswald, 17475 Greifswald, Germany
| | - Francisco Capani
- Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini" (ININCA), Facultad de Medicina, UBA-CONICET, Marcelo T. de Alvear 2270, C1122AAJ, Ciudad de Buenos Aires, Argentina; Departamento de Biología, UAJFK, C1197AAR, Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
19
|
Hu Q, Vakhmjanin A, Li B, Tang J, Zhang JH. Hyperbaric oxygen therapy fails to reduce hydrocephalus formation following subarachnoid hemorrhage in rats. Med Gas Res 2014; 4:12. [PMID: 25132956 PMCID: PMC4134116 DOI: 10.1186/2045-9912-4-12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/27/2014] [Indexed: 11/11/2022] Open
Abstract
Background & purpose Approximately 40% of hemorrhagic stroke survivors develop hydrocephalus. Hyperbaric oxygen (HBO) has been shown to be anti-inflammation following experimental stroke; however, its effect upon post-hemorrhagic hydrocephalus formation is not known. The objective of this study is to investigate whether HBO therapy can effectively reduce hydrocephalus formation and improve neurobehavioral functions in a rat model of subarachnoid hemorrhage (SAH). Method Thirty-eight male Sprague–Dawley rats (300-320 g) rats survived for 21 days from SAH by endovascular perforation or sham surgery were used. At 24 hours after SAH, HBO (3 atmospheres absolute) or normobaric oxygen (NBO) administrated for 1 hour once daily for a total of 7 days. Wire hanging and rotarod testing were conducted at 14 days after SAH, and cognitive functions were evaluated via the Morris water maze, between day 17 to day 21 after surgery. At day 21, rats were sacrificed and cerebroventricular volumes were measured histologically. Results Hydrocephalus exacerbated neurological deficits after SAH, and HBO multiple treatment tendentially improved the neurobehavioral functions. Spatial learning and memory deficits were noticed after SAH, and rats with hydrocephalus showed worse learning and memory abilities and HBO treatment showed a minor improvement. In the SAH group (room air) 4 rats showed an increased ventricular volume at day 21 after SAH-induction (n = 10). HBO or NBO therapy did not alter the occurrence of hydrocephalus after SAH, as 4 rats in each of these groups showed an increased ventricular volume (n = 10 per group). Conclusion Multiple HBO therapy does not ameliorate hydrocephalus formation in a rat model of SAH; however, HBO tendentially improved the neurological functions and spatial learning and memory abilities in rats with hydrocephalus.
Collapse
Affiliation(s)
- Qin Hu
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda CA 92354, USA
| | - Alexander Vakhmjanin
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda CA 92354, USA
| | - Bo Li
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda CA 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda CA 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda CA 92354, USA ; Department of Neurosurgery, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda CA 92354, USA
| |
Collapse
|
20
|
Kraitsy K, Uecal M, Grossauer S, Bruckmann L, Pfleger F, Ropele S, Fazekas F, Gruenbacher G, Patz S, Absenger M, Porubsky C, Smolle-Juettner F, Tezer I, Molcanyi M, Fasching U, Schaefer U. Repetitive long-term hyperbaric oxygen treatment (HBOT) administered after experimental traumatic brain injury in rats induces significant remyelination and a recovery of sensorimotor function. PLoS One 2014; 9:e97750. [PMID: 24848795 PMCID: PMC4029808 DOI: 10.1371/journal.pone.0097750] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/24/2014] [Indexed: 12/20/2022] Open
Abstract
Cells in the central nervous system rely almost exclusively on aerobic metabolism. Oxygen deprivation, such as injury-associated ischemia, results in detrimental apoptotic and necrotic cell loss. There is evidence that repetitive hyperbaric oxygen therapy (HBOT) improves outcomes in traumatic brain-injured patients. However, there are no experimental studies investigating the mechanism of repetitive long-term HBOT treatment-associated protective effects. We have therefore analysed the effect of long-term repetitive HBOT treatment on brain trauma-associated cerebral modulations using the lateral fluid percussion model for rats. Trauma-associated neurological impairment regressed significantly in the group of HBO-treated animals within three weeks post trauma. Evaluation of somatosensory-evoked potentials indicated a possible remyelination of neurons in the injured hemisphere following HBOT. This presumption was confirmed by a pronounced increase in myelin basic protein isoforms, PLP expression as well as an increase in myelin following three weeks of repetitive HBO treatment. Our results indicate that protective long-term HBOT effects following brain injury is mediated by a pronounced remyelination in the ipsilateral injured cortex as substantiated by the associated recovery of sensorimotor function.
Collapse
Affiliation(s)
- Klaus Kraitsy
- Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria
| | - Muammer Uecal
- Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria
| | - Stefan Grossauer
- Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - Lukas Bruckmann
- Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - Florentina Pfleger
- Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria
| | - Stefan Ropele
- Clinical Division of General Neurology, Medical University of Graz, Graz, Austria
| | - Franz Fazekas
- Clinical Division of General Neurology, Medical University of Graz, Graz, Austria
| | - Gerda Gruenbacher
- Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria
| | - Silke Patz
- Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria
| | - Markus Absenger
- Core Facility Microscopy, Centre for Medical Research, Medical University of Graz, Graz, Austria
| | - Christian Porubsky
- Division of Thoracic and Hyperbaric Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Freyja Smolle-Juettner
- Division of Thoracic and Hyperbaric Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Irem Tezer
- Division of Thoracic and Hyperbaric Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Marek Molcanyi
- Department of Neurosurgery, University of Cologne, Cologne, Germany
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Ulrike Fasching
- Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria
| | - Ute Schaefer
- Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria
- * E-mail:
| |
Collapse
|
21
|
Abstract
This article presents a pilot study to determine the value of hyperbaric oxygenation (HBO₂) in the acute management of neonatal hypoxia (hypoxic ischemic encephalopathy) and necrotizing enterocolitis. Neonates with hypoxic-ischemic encephalopathy and NE were treated in a Sechrist monoplace chamber. Electroencephalogram, evoked potential, ophthalmic evaluation, ultrasonograph, laboratory exams, and radiographs were obtained before and after HBO₂. Treatment protocol was 2.0 atm abs/45 minutes. Preventive myringotomies were conducted in all patients. A follow-up was done at 3 and 6 months. All patients (n = 8) were ventilator-dependent and required bag-valve-mask ventilation by a neonatologist during the treatment. All showed a resolution after HBO₂. There was also a dramatic improvement (P < .05) in hemoglobin, hematocrit, total proteins, serum sodium, triglycerides, and pH. There were favorable changes in all other studies although they did not meet statistical significance. There was a marked reduction of the morbidity and mortality. There were no adverse effects on the ophthalmologic or Central Nervous System. When used promptly, HBO₂ can modify the local and systemic inflammatory response caused by intestinal inflammation or cerebral or systemic hypoxia. It helps to preserve the marginal tissue and recover the ischemic and metabolic penumbra. This pilot study suggests that HBO₂ could be a safe and effective treatment in the acute management of neonatal necrotizing enterocolitis or hypoxic ischemic encephalopathy. There is a need for a prospective, randomized, controlled, and double-blinded study to determine the real use of HBO₂ in these cases.
Collapse
|
22
|
Drunalini Perera PN, Hu Q, Tang J, Li L, Barnhart M, Doycheva DM, Zhang JH, Tang J. Delayed remote ischemic postconditioning improves long term sensory motor deficits in a neonatal hypoxic ischemic rat model. PLoS One 2014; 9:e90258. [PMID: 24587303 PMCID: PMC3938659 DOI: 10.1371/journal.pone.0090258] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 01/28/2014] [Indexed: 11/29/2022] Open
Abstract
Objective Remote Ischemic Postconditioning (RIPC) is a promising therapeutic intervention wherein a sub-lethal ischemic insult induced in one organ (limb) improves ischemia in an organ distant to it (brain). The main objective of this study was to investigate the long-term functional effects of delayed RIPC in a neonatal hypoxia-ischemia (HI) rat model. Method 10 day old rat pups were subjected to delayed RIPC treatment and randomized into four groups: 1) Sham, 2) HI induced, 3) HI +24 hr delayed RIPC, and 4) HI +24 hr delayed RIPC with three consecutive daily treatments. Neurobehavioral tests, brain weights, gross and microscopic brain tissue morphologies, and systemic organ weights were evaluated at five weeks post surgery. Results HI induced rats performed significantly worse than sham but both groups of delayed RIPC treatment showed improvement of sensory motor functions. Furthermore, compared to the HI induced group, the delayed RIPC treatment groups showed no further detrimental changes on brain tissue, both grossly and morphologically, and no changes on the systemic organ weights. Conclusion Delayed RIPC significantly improves long term sensory motor deficits in a neonatal HI rat model. A 24 hr delayed treatment does not significantly attenuate morphological brain injury but does attenuate sensory motor deficits. Sensory motor deficits improve with both a single treatment and with three consecutive daily treatments, and the consecutive treatments are possibly being more beneficial.
Collapse
Affiliation(s)
- Pradilka N. Drunalini Perera
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Qin Hu
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Junjia Tang
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Li Li
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Margaret Barnhart
- Department of Neurosurgery Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Desislava M. Doycheva
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - John H. Zhang
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, United States of America
- Department of Neurosurgery Loma Linda University School of Medicine, Loma Linda, California, United States of America
- Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Jiping Tang
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, United States of America
- * E-mail:
| |
Collapse
|
23
|
Yu Y, Li L, Shao X, Tian F, Sun Q. Establishing a rat model of spastic cerebral palsy by targeted ethanol injection. Neural Regen Res 2013; 8:3255-62. [PMID: 25206647 PMCID: PMC4146179 DOI: 10.3969/j.issn.1673-5374.2013.34.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 11/26/2013] [Indexed: 11/26/2022] Open
Abstract
Spastic cerebral palsy is generally considered to result from cerebral cortical or pyramidal tract damage. Here, we precisely targeted the left pyramidal tract of 2-month-old Sprague-Dawley rats placed on a stereotaxic instrument under intraperitoneal anesthesia. Based on the rat brain stereotaxic map, a 1-mm hole was made 10 mm posterior to bregma and 0.8 mm left of sagittal suture. A microsyringe was inserted perpendicularly to the surface of the brain to a depth of 9.7 mm, and 15 μL of ethanol was slowly injected to establish a rat model of spastic cerebral palsy. After modeling, the rats appeared to have necrotic voids in the pyramidal tract and exhibited typical signs and symptoms of flexion spasms that lasted for a long period of time. These findings indicate that this is an effective and easy method of establishing a rat model of spastic cerebral palsy with good re-producibility. Ethanol as a chemical ablation agent specifically and thoroughly damages the pyramidal tract, and therefore, the animals display flexion spasms, which are a typical symptom of the disease.
Collapse
Affiliation(s)
- Yadong Yu
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| | - Liang Li
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| | - Xinzhong Shao
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| | - Fangtao Tian
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| | - Qinglu Sun
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| |
Collapse
|
24
|
Abstract
Hypoxia-ischemia is a leading cause of morbidity and mortality in the perinatal period with an incidence of 1/4000 live births. Biochemical events such as energy failure, membrane depolarization, brain edema, an increase of neurotransmitter release and inhibition of uptake, an increase of intracellular Ca(2+), production of oxygen-free radicals, lipid peroxidation, and a decrease of blood flow are triggered by hypoxia-ischemia and may lead to brain dysfunction and neuronal death. These abnormalities can result in mental impairments, seizures, and permanent motor deficits, such as cerebral palsy. The physical and emotional strain that is placed on the children affected and their families is enormous. The care that these individuals need is not only confined to childhood, but rather extends throughout their entire life span, so it is very important to understand the pathophysiology that follows a hypoxic-ischemic insult. This review will highlight many of the mechanisms that lead to neuronal death and include the emerging area of white matter injury as well as the role of inflammation and will provide a summary of therapeutic strategies. Hypothermia and oxygen will also be discussed as treatments that currently lack a specific target in the hypoxic/ischemic cascade.
Collapse
Affiliation(s)
- John W Calvert
- Departments of Neurosurgery and Molecular and Cellular Physiology, Loma Linda University Medical Center, 11234 Anderson Street, Loma Linda, CA 92354, USA
| | | |
Collapse
|
25
|
Abstract
Neuroprotective drugs have so far failed clinical trials, at high cost, and intravenous tissue plasminogen activator (i.v. tPA) remains the only FDA-approved acute stroke therapy. Hyperoxia, acting via multiple direct and indirect mechanisms, may be a powerful neuroprotective strategy to salvage acutely ischemic brain tissue and extend the time window for acute stroke treatment. Of the available oxygen delivery methods, hyperbaric oxygen therapy (HBO) appears to be the most potent, while even normobaric oxygen therapy (NBO) may be effective if started promptly after stroke onset. HBO has so far failed to show efficacy in three clinical trials. The failure of these trials is probably attributable to factors such as delayed time to therapy, inadequate sample size and use of excessive chamber pressures. Previous trials did not assess long-term benefit in patients with tissue reperfusion. In this modern era of stroke thrombolysis and advanced neuroimaging, oxygen therapy may have renewed significance. If applied within the first few hours after stroke onset or in patients with imaging evidence of salvageable brain tissue, oxygen therapy could be used to 'buy time' for the administration of thrombolytic or neuroprotective drugs. This article reviews the history and current rationale for using oxygen therapy in stroke, the mechanisms of action of HBO and the results of animal and human studies of hyperoxia in cerebrovascular diseases.
Collapse
Affiliation(s)
- Aneesh B Singhal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
26
|
Feng Z, Liu J, Ju R. Hyperbaric oxygen treatment promotes neural stem cell proliferation in the subventricular zone of neonatal rats with hypoxic-ischemic brain damage. Neural Regen Res 2013; 8:1220-7. [PMID: 25206416 PMCID: PMC4107609 DOI: 10.3969/j.issn.1673-5374.2013.13.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/10/2013] [Indexed: 02/05/2023] Open
Abstract
Hyperbaric oxygen therapy for the treatment of neonatal hypoxic-ischemic brain damage has been used clinically for many years, but its effectiveness remains controversial. In addition, the mechanism of this potential neuroprotective effect remains unclear. This study aimed to investigate the influence of hyperbaric oxygen on the proliferation of neural stem cells in the subventricular zone of neonatal Sprague-Dawley rats (7 days old) subjected to hypoxic-ischemic brain damage. Six hours after modeling, rats were treated with hyperbaric oxygen once daily for 7 days. Immunohistochemistry revealed that the number of 5-bromo-2'-deoxyuridine positive and nestin positive cells in the subventricular zone of neonatal rats increased at day 3 after hypoxic-ischemic brain damage and peaked at day 5. After hyperbaric oxygen treatment, the number of 5-bromo-2'-deoxyuridine positive and nestin positive cells began to increase at day 1, and was significantly higher than that in normal rats and model rats until day 21. Hematoxylin-eosin staining showed that hyperbaric oxygen treatment could attenuate pathological changes to brain tissue in neonatal rats, and reduce the number of degenerating and necrotic nerve cells. Our experimental findings indicate that hyperbaric oxygen treatment enhances the proliferation of neural stem cells in the subventricular zone of neonatal rats with hypoxic-ischemic brain damage, and has therapeutic potential for promoting neurological recovery following brain injury.
Collapse
Affiliation(s)
- Zhichun Feng
- Department of Neonatology & Neonatal Intensive Care Unit, Bayi Children's Hospital Affiliated to General Hospital of Beijing Military Command, Beijing 100700, China
| | - Jing Liu
- Department of Neonatology & Neonatal Intensive Care Unit, Bayi Children's Hospital Affiliated to General Hospital of Beijing Military Command, Beijing 100700, China
- Corresponding author: Jing Liu, Chief physician, Professor, Department of Neonatology & Neonatal Intensive Care Unit, Bayi Children's Hospital Affiliated to General Hospital of Beijing Military Command, Beijing 100700, China, (N20120314002)
| | - Rong Ju
- Department of Neonatology & Neonatal Intensive Care Unit, Bayi Children's Hospital Affiliated to General Hospital of Beijing Military Command, Beijing 100700, China
- Department of Neonatology, Chengdu Women's & Children's Central Hospital, Chengdu 610031, Sichuan Province, China
| |
Collapse
|
27
|
Zhu J, Shen W, Gao L, Gu H, Shen S, Wang Y, Wu H, Guo J. PI3K/Akt-independent negative regulation of JNK signaling by MKP-7 after cerebral ischemia in rat hippocampus. BMC Neurosci 2013; 14:1. [PMID: 23280045 PMCID: PMC3583678 DOI: 10.1186/1471-2202-14-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 12/26/2012] [Indexed: 01/26/2023] Open
Abstract
Background The inactivation of c-Jun N-terminal kinase (JNK) is associated with anti-apoptotic and anti-inflammatory effects in cerebral ischemia, which can be induced by an imbalance between upstream phosphatases and kinases. Result Mitogen-activated protein kinase phosphatase 7 (MKP-7) was upregulated significantly at 4 h of reperfusion postischemia in rat hippocampi. By administration of cycloheximide or siRNA against mitogen-activated protein kinase phosphatase 7 (MKP-7) in a rat model of ischemia/reperfusion, an obvious enhancement of JNK activity was observed in 4 h of reperfusion following ischemia, suggesting MKP-7 was involved in JNK inactivation after ischemia. The subcellular localization of MKP-7 altered after ischemia, and the inhibition of MKP-7 nuclear export by Leptomycin B up-regulated JNK activity. Although PI3K/Akt inhibition could block downregulation of JNK activity through SEK1 and MKK-7 activation, PI3K/Akt activity was not associated with the regulation of JNK by MKP-7. Conclusions MKP-7, independently of PI3K/Akt pathway, played a key role in downregulation of JNK activity after ischemia in the rat hippocampus, and the export of MKP-7 from the nucleus was involved in downregulation of cytoplasmic JNK activity in response to ischemic stimuli.
Collapse
Affiliation(s)
- JianXi Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Li Y, Xia ZL, Chen LB. HIF-1-α and survivin involved in the anti-apoptotic effect of 2ME2 after global ischemia in rats. Neurol Res 2012; 33:583-92. [PMID: 21708067 DOI: 10.1179/1743132810y.0000000013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Survivin is an anti-apoptotic gene that decreases the apoptosis by depressing the expression of caspase-3. Hypoxia-inducible factor-1-alpha (HIF-1-alpha) is a transcription factor specifically activated by hypoxia. 2-methoxyestradiol (2ME2) is an estradiol derivative and a known HIF-1-alpha inhibitor. 2ME2 decreased apoptosis by inhibiting HIF-1-alpha. The aim of the present study was to investigate if survivin is involved in the anti-apoptotic effect of 2ME2. Male adult rats were used to make the global ischemia (GI) model. Ten minutes after GI, 2ME2 was injected intraperitoneally (16 mg/kg weight). Rats were killed at 6 hours, 12 hours, 24 hours, 48 hours, 96 hours, and 7 days. GI produced a marked increase in HIF-1-alpha expressions in the hippocampus at 6 hours and peaked at 48-96 hours. The expressions of survivin and caspase-3 were increased lightly in a similar time course. These molecular changes were accompanied by massive cell loss and apoptosis in the hippocampal regions. 2ME2 treatment reduced the expression of HIF-1-alpha, increased survivin expression, and decreased the expression of caspase-3. These results indicate that survivin and HIF-1-alpha were involved in the anti-apoptotic effect of 2ME2 treated following GI. 2ME2 may decrease the HIF-1-alpha expression, up-regulate the survivin expression, inhibit the expression of caspase-3, and finally reduce apoptosis after GI.
Collapse
Affiliation(s)
- Yun Li
- Department of Physiology, Shandong University School of Medicine, Jinan, China
| | | | | |
Collapse
|
29
|
An SJ, Kim DS. Alterations in serotonin receptors and transporter immunoreactivities in the hippocampus in the rat unilateral hypoxic-induced epilepsy model. Cell Mol Neurobiol 2011; 31:1245-55. [PMID: 21681557 DOI: 10.1007/s10571-011-9726-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 06/06/2011] [Indexed: 11/26/2022]
Abstract
Unilateral hypoxic-ischemia results in the frequent occurrence of interictal spikes, and occasionally sustained ictal discharges accompanied by a reduction in paired-pulse inhibition within the non-lesioned dentate gyrus. To elucidate the roles of serotonin (5-hydroxytryptamine [5-HT]) in an epileptogenic insult, we investigated the changes in 5-HT receptors and serotonin transporter (5-HTT) immunoreactivities within the lesioned and contralateral hippocampus following unilateral hypoxic-ischemia. During epileptogenic periods following hypoxic-ischemia, both 5-HT(1A) and 5HT(1B) receptor immunoreactivities were decreased within the lesioned and the non-lesioned hippocampus. However, 5-HTT immunoreactivity was transiently increased within the hippocampus bilaterally. These findings indicate that alteration of the 5-HT system results in a "diaschisis" pattern, and may contribute to neuronal death and the development of emotional disorders in epileptic patients accompanied by psychological stress.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Epilepsy/physiopathology
- Hippocampus/physiology
- Hippocampus/physiopathology
- Humans
- Male
- Rats
- Rats, Sprague-Dawley
- Receptor, Serotonin, 5-HT1A/genetics
- Receptor, Serotonin, 5-HT1A/metabolism
- Receptor, Serotonin, 5-HT1B/genetics
- Receptor, Serotonin, 5-HT1B/metabolism
- Serotonin Plasma Membrane Transport Proteins/genetics
- Serotonin Plasma Membrane Transport Proteins/metabolism
- Stress, Psychological
Collapse
Affiliation(s)
- Sung-Jin An
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, Kangwon-Do 200-702, Republic of Korea
| | | |
Collapse
|
30
|
Fan X, Kavelaars A, Heijnen CJ, Groenendaal F, van Bel F. Pharmacological neuroprotection after perinatal hypoxic-ischemic brain injury. Curr Neuropharmacol 2011; 8:324-34. [PMID: 21629441 PMCID: PMC3080590 DOI: 10.2174/157015910793358150] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 03/31/2010] [Accepted: 04/08/2010] [Indexed: 11/22/2022] Open
Abstract
Perinatal hypoxia-ischemia (HI) is an important cause of neonatal brain injury. Recent progress in the search for neuroprotective compounds has provided us with several promising drugs to reduce perinatal HI-induced brain injury. In the early stage (first 6 hours after birth) therapies are concentrated on prevention of the production of reactive oxygen species or free radicals (xanthine-oxidase-, nitric oxide synthase-, and prostaglandin inhibition), anti-inflammatory effects (erythropoietin, melatonin, Xenon) and anti-apoptotic interventions (nuclear factor kappa B- and c-jun N-terminal kinase inhibition); in a later stage stimulation of neurotrophic properties in the neonatal brain (erythropoietin, growth factors) can be targeted to promote neuronal and oligodendrocyte regeneration. Combination of pharmacological means of treatment with moderate hypothermia, which is accepted now as a meaningful therapy, is probably the next step in clinical treatment to fight post-asphyxial brain damage. Further studies should be directed at a more rational use of therapies by determining the optimal time and dose to inhibit the different potentially destructive molecular pathways or to enhance endogenous repair while at the same time avoiding adverse effects of the drugs used.
Collapse
Affiliation(s)
- Xiyong Fan
- Department of Neonatology, University Medical Center, Utrecht, the Netherlands
| | | | | | | | | |
Collapse
|
31
|
Souvenir R, Fathali N, Ostrowski RP, Lekic T, Zhang JH, Tang J. Tissue inhibitor of matrix metalloproteinase-1 mediates erythropoietin-induced neuroprotection in hypoxia ischemia. Neurobiol Dis 2011; 44:28-37. [PMID: 21689752 DOI: 10.1016/j.nbd.2011.05.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/17/2011] [Accepted: 05/28/2011] [Indexed: 02/06/2023] Open
Abstract
Previous studies have shown that erythropoietin (EPO) is neuroprotective in both in vivo and in vitro models of hypoxia ischemia. However these studies hold limited clinical translations because the underlying mechanism remains unclear and the key molecules involved in EPO-induced neuroprotection are still to be determined. This study investigated if tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) and its upstream regulator signaling molecule Janus kinase-2 (JAK-2) are critical in EPO-induced neuroprotection. Hypoxia ischemia (HI) was modeled in-vitro by oxygen and glucose deprivation (OGD) and in-vivo by a modified version of Rice-Vannucci model of HI in 10-day-old rat pups. EPO treated cells were exposed to AG490, an inhibitor of JAK-2 or TIMP-1 neutralizing antibody for 2h with OGD. Cell death, phosphorylation of JAK-2 and signal transducers and activators of transcription protein-3 (STAT-3), TIMP-1 expression, and matrix metalloproteinase-9 (MMP-9) activity were measured and compared with normoxic group. Hypoxic ischemic animals were treated one hour following HI and evaluated 48 h after. Our data showed that EPO significantly increased cell survival, associated with increased TIMP-1 activity, phosphorylation of JAK-2 and STAT-3, and decreased MMP-9 activity in vivo and in vitro. EPO's protective effects were reversed by inhibition of JAK-2 or TIMP-1 in both models. We concluded that JAK-2, STAT-3 and TIMP-1 are key mediators of EPO-induced neuroprotection during hypoxia ischemia injury.
Collapse
Affiliation(s)
- Rhonda Souvenir
- Division of Microbiology and Molecular Genetics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | | | | | | | | | | |
Collapse
|
32
|
Mychaskiw G. Molecules, magic and forgetful fruit flies: the supernatural science of medical gas research. Med Gas Res 2011; 1:23. [PMID: 22146602 PMCID: PMC3231965 DOI: 10.1186/2045-9912-1-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 09/06/2011] [Indexed: 11/10/2022] Open
Abstract
Medical gas research often involves the study of molecules under extraphysiologic conditions, that is, conditions that do not exist in nature. This "supernatural" nature of medical gas research sometimes produces results that appear to be almost "magic" to those schooled in traditional physiology "Any sufficiently advanced technology is indistinguishable from magic". -Arthur C. Clarke
Collapse
|
33
|
Ezra N, Dang K, Heuser G. Improvement of attention span and reaction time with hyperbaric oxygen treatment in patients with toxic injury due to mold exposure. Eur J Clin Microbiol Infect Dis 2010; 30:1-6. [PMID: 20978814 PMCID: PMC2998645 DOI: 10.1007/s10096-010-0937-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 04/03/2010] [Indexed: 12/19/2022]
Abstract
It is, by now, well established that mold toxins (mycotoxins) can cause significant adverse health effects. In this study, 15 subjects who developed an attention deficit disorder (ADD) and slowing of reaction time at the time of exposure to mold toxins were identified. Deficits in attention span and reaction time were documented not only by taking a careful history, but also by performing a Test of Variables of Attention (TOVA). The TOVA test provides an objective measure of these two variables. It was found that mold-exposed subjects show statistically significant decreases in attention span and significant increases in reaction time to stimuli compared to controls. After ten sessions of hyperbaric oxygen treatment (HBOT), a statistically significant improvement was seen in both measures. This preliminary study suggests promising outcomes in treating mold-exposed patients with hyperbaric oxygen.
Collapse
Affiliation(s)
- N Ezra
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | | | | |
Collapse
|
34
|
Matchett GA, Martin RD, Zhang JH. Hyperbaric oxygen therapy and cerebral ischemia: neuroprotective mechanisms. Neurol Res 2009; 31:114-21. [PMID: 19298750 DOI: 10.1179/174313209x389857] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
INTRODUCTION Numerous studies have demonstrated a protective effect of hyperbaric oxygen therapy in experimental ischemic brain injury, and many physiological and molecular mechanisms of hyperbaric oxygen therapy-related neuroprotection have been identified. METHODS Review of articles pertaining to hyperbaric oxygen therapy and cerebral ischemia in the National Library of Medicine and National Institutes of Health database, emphasizing mechanisms of hyperbaric oxygen therapy-related neuroprotection. RESULTS Hyperbaric oxygen therapy has been shown to ameliorate brain injury in a variety of animal models including focal cerebral ischemia, global cerebral ischemia, neonatal hypoxia-ischemia and subarachnoid hemorrhage. Small human trials of hyperbaric oxygen therapy in focal ischemia have not shown benefit, although one trial of hyperbaric oxygen therapy before cardiopulmonary bypass demonstrated improved neuropsychological and inflammatory outcomes with hyperbaric oxygen therapy. Hyperbaric oxygen therapy is associated with improved cerebral oxygenation, reduced blood-brain barrier breakdown, decreased inflammation, reduced cerebral edema, decreased intracranial pressure, reduced oxidative burden, reduced metabolic derangement, decreased apoptotic cell death and increased neural regeneration. CONCLUSION On a molecular level, hyperbaric oxygen therapy leads to activation of ion channels, inhibition of hypoxia inducible factor-1alpha, up-regulation of Bcl-2, inhibition of MMP-9, decreased cyclooxygenase-2 activity, decreased myeloperoxidase activity, up-regulation of superoxide dismutase and inhibition of Nogo-A (an endogenous growth-inhibitory factor). Ongoing research will continue to describe the mechanisms of hyperbaric oxygen therapy-related neuroprotection, and possibly expand hyperbaric oxygen therapy use clinically.
Collapse
Affiliation(s)
- Gerald A Matchett
- Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | | | | |
Collapse
|
35
|
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: 2.6] [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
|
36
|
Abstract
Oxygen is frequently administered to patients with suspected stroke. However, the role of oxygen therapy in ischemic stroke remains controversial in light of the failure of three clinical trials of hyperbaric oxygen therapy to show efficacy, and the fear of exacerbating oxygen free radical injury. The previous trials had several shortcomings, perhaps because they were designed on basis of anecdotal case reports and little preclinical data. Most animal studies concerning oxygen therapy in stroke have been conducted over the last 6 years. Emerging data suggests that hyperbaric and even normobaric oxygen therapy can be effective if used appropriately, and raises the tantalizing possibility that hyperoxia can be used to extend the narrow therapeutic time window for stroke thrombolysis. This article reviews the history, rationale, mechanisms of action and adverse effects of hyperoxia, the key results of previous hyperoxia studies, and the potential role of oxygen therapy in contemporary stroke treatment.
Collapse
Affiliation(s)
- Aneesh B Singhal
- Massachusetts General Hospital, Stroke Research Center, 175 Cambridge Street, Suite 300, Boston, MA 02114, USA.
| |
Collapse
|
37
|
Blanco YC, Farias AS, Goelnitz U, Lopes SCP, Arrais-Silva WW, Carvalho BO, Amino R, Wunderlich G, Santos LMB, Giorgio S, Costa FTM. Hyperbaric oxygen prevents early death caused by experimental cerebral malaria. PLoS One 2008; 3:e3126. [PMID: 18769544 PMCID: PMC2518956 DOI: 10.1371/journal.pone.0003126] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Accepted: 08/14/2008] [Indexed: 01/12/2023] Open
Abstract
Background Cerebral malaria (CM) is a syndrome characterized by neurological signs, seizures and coma. Despite the fact that CM presents similarities with cerebral stroke, few studies have focused on new supportive therapies for the disease. Hyperbaric oxygen (HBO) therapy has been successfully used in patients with numerous brain disorders such as stroke, migraine and atherosclerosis. Methodology/Principal Findings C57BL/6 mice infected with Plasmodium berghei ANKA (PbA) were exposed to daily doses of HBO (100% O2, 3.0 ATA, 1–2 h per day) in conditions well-tolerated by humans and animals, before or after parasite establishment. Cumulative survival analyses demonstrated that HBO therapy protected 50% of PbA-infected mice and delayed CM-specific neurological signs when administrated after patent parasitemia. Pressurized oxygen therapy reduced peripheral parasitemia, expression of TNF-α, IFN-γ and IL-10 mRNA levels and percentage of γδ and αβ CD4+ and CD8+ T lymphocytes sequestered in mice brains, thus resulting in a reduction of blood-brain barrier (BBB) dysfunction and hypothermia. Conclusions/Significance The data presented here is the first indication that HBO treatment could be used as supportive therapy, perhaps in association with neuroprotective drugs, to prevent CM clinical outcomes, including death.
Collapse
Affiliation(s)
- Yara C. Blanco
- Department of Microbiology & Immunology, State University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
- Department of Parasitology, UNICAMP, State University of Campinas, Campinas, São Paulo, Brazil
| | - Alessandro S. Farias
- Department of Microbiology & Immunology, State University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
| | - Uta Goelnitz
- Department of Parasitology – ICB, University of São Paulo – USP, São Paulo, São Paulo, Brazil
| | - Stefanie C. P. Lopes
- Department of Microbiology & Immunology, State University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
- Department of Parasitology, UNICAMP, State University of Campinas, Campinas, São Paulo, Brazil
| | - Wagner W. Arrais-Silva
- Department of Parasitology, UNICAMP, State University of Campinas, Campinas, São Paulo, Brazil
| | - Bruna O. Carvalho
- Department of Microbiology & Immunology, State University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
- Department of Parasitology, UNICAMP, State University of Campinas, Campinas, São Paulo, Brazil
| | - Rogério Amino
- Department of Biochemistry, Federal University of São Paulo – UNIFESP, São Paulo, São Paulo, Brazil
| | - Gerhard Wunderlich
- Department of Parasitology – ICB, University of São Paulo – USP, São Paulo, São Paulo, Brazil
| | - Leonilda M. B. Santos
- Department of Microbiology & Immunology, State University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
| | - Selma Giorgio
- Department of Parasitology, UNICAMP, State University of Campinas, Campinas, São Paulo, Brazil
| | - Fabio T. M. Costa
- Department of Microbiology & Immunology, State University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
- Department of Parasitology, UNICAMP, State University of Campinas, Campinas, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
38
|
HIF-1alpha inhibition ameliorates neonatal brain injury in a rat pup hypoxic-ischemic model. Neurobiol Dis 2008; 31:433-41. [PMID: 18602008 DOI: 10.1016/j.nbd.2008.05.020] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 05/17/2008] [Accepted: 05/28/2008] [Indexed: 01/03/2023] Open
Abstract
Hypoxia-inducible factor-1alpha (HIF-1alpha) has been considered as a regulator of both prosurvival and prodeath pathways in the nervous system. The present study was designed to elucidate the role of HIF-1alpha in neonatal hypoxic-ischemic (HI) brain injury. Rice-Vannucci model of neonatal hypoxic-ischemic brain injury was used in seven-day-old rats, by subjecting unilateral carotid artery ligation followed by 2 h of hypoxia (8% O2 at 37 degrees C). HIF-1alpha activity was inhibited by 2-methoxyestradiol (2ME2) and enhanced by dimethyloxalylglycine (DMOG). Results showed that 2ME2 exhibited dose-dependent neuroprotection by decreasing infarct volume and reducing brain edema at 48 h post HI. The neuroprotection was lost when 2ME2 was administered 3 h post HI. HIF-1alpha upregulation by DMOG increased the permeability of the BBB and brain edema compared with HI group. 2ME2 attenuated the increase in HIF-1alpha and VEGF 24 h after HI. 2ME2 also had a long-term effect of protecting against the loss of brain tissue. The study showed that the early inhibition of HIF-1alpha acutely after injury provided neuroprotection after neonatal hypoxia-ischemia which was associated with preservation of BBB integrity, attenuation of brain edema, and neuronal death.
Collapse
|
39
|
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: 1.8] [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
|
40
|
Hyperbaric Oxygen Inhibits Growth But Not Differentiation of Normal and Irradiated Osteoblasts. J Craniofac Surg 2008; 19:757-65. [DOI: 10.1097/scs.0b013e31816aac19] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
41
|
Proliferation of neural stem cells correlates with Wnt-3 protein in hypoxic-ischemic neonate rats after hyperbaric oxygen therapy. Neuroreport 2008; 18:1753-6. [PMID: 17921881 DOI: 10.1097/wnr.0b013e3282f0ec09] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hyperbaric oxygen therapy promoted brain cell proliferation. Wnt-3 is closely associated with the proliferation of neural stem cells. We examined whether hyperbaric oxygen promoted neural stem cells to proliferate and its correlation with Wnt-3 protein in hypoxic-ischemic neonate rats. Hyperbaric oxygen therapy was administered 3 h after hypoxia ischemia daily for 7 days. The proliferating stem cells and Wnt-3 protein were examined dynamically in the subventricular zone. Results showed that stem cells proliferated and peaked 7 days after hyperbaric oxygen therapy. Wnt-3 protein increased to the higher levels 3 days after therapy. Linear regression analysis showed that nestin protein correlated with Wnt-3 protein. We propose that hyperbaric oxygen treatment promote stem cells to proliferate, which is correlated with Wnt-3 protein.
Collapse
|
42
|
Abstract
Hyperbaric oxygen (HBO) therapy is defined by the Undersea and Hyperbaric Medical Society (UHMS) as a treatment in which a patient intermittingly breathes 100% oxygen under a pressure that is greater than the pressure at sea level [a pressure greater than 1 atmosphere absolute (ATA)]. HBO has been shown to be a potent means to increase the oxygen content of blood and has been advocated for the treatment of various ailments, including air embolism, carbon monoxide poisoning, wound healing and ischemic stroke. However, definitive established mechanisms of action are still lacking. This has led to uncertainty among clinicians, who have understandingly become hesitant in regard to using HBO therapy, even in situations where it could prove beneficial. Therefore, this review will summarize the literature regarding the effects of HBO on brain oxygenation, cerebral blood flow and intracranial pressure in both the healthy and injured brains, as well as discuss how changes in these three factors can impart protection.
Collapse
Affiliation(s)
- John W Calvert
- Department of Physiology, Division of Neurosurgery, Loma Linda University Medical Center, Loma Linda, CA, USA.
| | | | | |
Collapse
|
43
|
Chen C, Hu Q, Yan J, Lei J, Qin L, Shi X, Luan L, Yang L, Wang K, Han J, Nanda A, Zhou C. Multiple effects of 2ME2 and D609 on the cortical expression of HIF-1alpha and apoptotic genes in a middle cerebral artery occlusion-induced focal ischemia rat model. J Neurochem 2007; 102:1831-1841. [PMID: 17532791 DOI: 10.1111/j.1471-4159.2007.04652.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Despite 2-methoxyestradiol (2ME2) and tricyclodecan-9-yl-xanthogenate (D609) having multiple effects on cancer cells, mechanistically, both of them down-regulate hypoxia-inducible factor-1alpha (HIF-1alpha) and vascular endothelial growth factor (VEGF). We hypothesize HIF-1alpha plays an essential role in cerebral ischemia as a pro-apoptosis regulator; 2ME2 and D609 decrease the levels of HIF-1alpha and VEGF, that might contribute to protecting brain from ischemia injury. A total of 102 male Sprague-Dawley rats were split into five groups: sham, middle cerebral artery occlusion (MCAO), MCAO + dimethyl sulfoxide, MCAO + 2ME2, and MCAO + D609. 2ME2 and D609 were injected intraperitoneally 1 h after reperfusion. Rats were killed at 24 h and 7 days. At 24 h, 2ME2 and D609 reduce the levels of HIF-1alpha and VEGF (enzyme-linked immunosorbent assay), depress the expression of HIF-1alpha, VEGF, BCL2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3) and cleaved caspase 3 (western blot and immunohistochemistry) in the brain infarct area. Double fluorescence labeling shows HIF-1alpha positive immunoreactive materials are co-localized with BNIP3 and terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling inside the nuclei of neurons. At 7 days, 2ME2 and D609 reduce the infarct volume (2,3,7-triphenyltetrazolium chloride) and blood-brain barrier extravasation, decrease the mortality and improve the neurological deficits. In conclusion, 2ME2 and D609 are powerful agents to protect brain from cerebral ischemic injury by inhibiting HIF-1alpha expression, attenuating the superfluous expression of VEGF to avoid blood-brain barrier disruption and suppressing neuronal apoptosis via BNIP3 pathway.
Collapse
Affiliation(s)
- Chunhua Chen
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Qin Hu
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Junhao Yan
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Jiliang Lei
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Lihua Qin
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Xianzhong Shi
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Liju Luan
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Lei Yang
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Ke Wang
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Jingyan Han
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Anil Nanda
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| | - Changman Zhou
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, ChinaCenter of Tasly Microcirculation, Peking University Health Science Center, Beijing, ChinaDepartment of Neurosurgery, Louisiana State University Health Science Center in Shreveport, Louisiana, USA
| |
Collapse
|
44
|
Abstract
OBJECTIVE The purpose of this study was to determine whether oxygen treatment could attenuate the alterations in cerebral energy metabolism found in the brain following hypoxia-ischemia. DESIGN Seven-day-old rat pups were subjected to unilateral carotid artery ligation followed by 2 hrs of hypoxia (8% oxygen at 37 degrees C). The concentrations of high-energy phosphate compounds and glycolytic intermediates and the activity of Na+/K+-adenosine triphosphatase were measured at 4-72 hrs of recovery. Brain weight was used to determine the severity of the brain injury at 2 wks after insult. SETTING Experimental setting. SUBJECTS Rat pups. INTERVENTIONS Pups were treated with 100% oxygen 1 hr after the insult at 2.5 atmospheres absolute (hyperbaric oxygen) or at normobaric pressure for a duration of 2 hrs. MEASUREMENTS AND MAIN RESULTS During the initial period of recovery from hypoxia-ischemia, values of adenosine triphosphate and phosphocreatine remained at levels below normal, whereas the levels of glucose and other glycolytic intermediates were elevated. Hyperbaric oxygen and normobaric oxygen both attenuated brain injury, restored the levels of adenosine triphosphate and phosphocreatine, decreased the levels of the glycolytic intermediates, and increased the utilization of energy. CONCLUSIONS These results suggest that oxygen treatment during the initial period of recovery from a hypoxia-ischemic insult is able to attenuate energy deficits in the brain, which ultimately leads to a reduction in brain injury.
Collapse
Affiliation(s)
- John W Calvert
- Department of Physiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | | |
Collapse
|
45
|
Yata K, Matchett GA, Tsubokawa T, Tang J, Kanamaru K, Zhang JH. Granulocyte-colony stimulating factor inhibits apoptotic neuron loss after neonatal hypoxia-ischemia in rats. Brain Res 2007; 1145:227-38. [PMID: 17359943 PMCID: PMC1888563 DOI: 10.1016/j.brainres.2007.01.144] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Revised: 01/27/2007] [Accepted: 01/30/2007] [Indexed: 11/30/2022]
Abstract
Neonatal hypoxia-ischemia (HI) is an important clinical problem with few effective treatments. Granulocyte-colony stimulating factor (G-CSF) is an endogenous peptide hormone of the hematopoietic system that has been shown to be neuroprotective in focal ischemia in vivo and is currently in phase I/II clinical trials for ischemic stroke in humans. We tested G-CSF in a rat model of neonatal hypoxia-ischemia in postnatal day 7 unsexed rat pups. Three groups of animals were used: hypoxia-ischemia (HI, n=67), hypoxia-ischemia with G-CSF treatment (HI+G, n=65), and healthy control (C, n=53). G-CSF (50 microg/kg, subcutaneous) was administered 1 h after HI and given on four subsequent days (five total injections). Animals were euthanized 24 h, 1, 2, and 3 weeks after HI. Assessment included brain weight, histology, immunohistochemistry, and Western blotting. G-CSF treatment was associated with improved quantitative brain weight and qualitative Nissl histology after hypoxia-ischemia. TUNEL demonstrated reduced apoptosis in group HI+G. Western blot demonstrated decreased expression of Bax and cleaved caspase-3 in group HI+G. G-CSF treatment was also associated with increased expression of STAT3, Bcl-2, and Pim-1, all of which may have participated in the anti-apoptotic effect of the drug. We conclude that G-CSF ameliorates hypoxic-ischemic brain injury and that this may occur in part by an inhibition of apoptotic cell death.
Collapse
Affiliation(s)
- Kenichiro Yata
- Department of Physiology, Loma Linda University, Loma Linda, CA 92354, USA
| | | | | | | | | | | |
Collapse
|
46
|
Yogaratnam JZ, Laden G, Guvendik L, Cowen M, Cale A, Griffin S. Can hyperbaric oxygen be used as adjunctive heart failure therapy through the induction of endogenous heat shock proteins? Adv Ther 2007; 24:106-18. [PMID: 17526467 DOI: 10.1007/bf02849998] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Heart failure (HF) is a chronic condition that is expected to increase in incidence along with increased life expectancy and an aging population. As the incidence of HF increases, the cost to national healthcare budgets is expected to run into the billions. The costs of lost productivity and increased social reliance on state support must also be considered. Recently, acute myocardial infarction (AMI) has come to be seen as the major contributing factor to HF. Although thrombolysis may restore coronary perfusion after an AMI, it may also introduce ischemic reperfusion injury (IRI). In an attempt to ameliorate sustained protein damage caused by IRI, endogenous chaperone proteins known as heat shock proteins (HSPs) are induced as a consequence of the stress of IRI. Recently, hyperbaric oxygen has been shown to induce the production of HSPs in noncardiac tissue, with a resultant protective effect. This current opinion review article suggests a possible role for hyperbaric oxygen, as a technologically modern drug, in augmenting the induction of endogenous HSPs to repair and improve the function of failing hearts that have been damaged by AMI and IRI. In addition, this simple, safe, noninvasive drug may prove useful in easing the economic burden of HF on already overextended health resources.
Collapse
|
47
|
Evaluation of hyperbaric oxygen treatment of neuropsychiatric disorders following traumatic brain injury. Chin Med J (Engl) 2006. [DOI: 10.1097/00029330-200612010-00007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
48
|
Calvert JW, Cahill J, Yamaguchi-Okada M, Zhang JH. Oxygen treatment after experimental hypoxia-ischemia in neonatal rats alters the expression of HIF-1alpha and its downstream target genes. J Appl Physiol (1985) 2006; 101:853-65. [PMID: 16728520 DOI: 10.1152/japplphysiol.00268.2006] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Recently, mounting evidence has emerged to suggest that hyperbaric oxygenation (HBOT)-induced neuroprotection after experimental global ischemia and subarachnoid hemorrhage entails a decrease in the expression of hypoxia-inducible factor-1alpha (HIF-1alpha). Therefore, the purpose of this study was to test the hypothesis that oxygen-induced neuroprotection after neonatal hypoxia-ischemia involves alterations in the expression of HIF-1alpha. Seven-day-old rat pups were subjected to unilateral carotid artery ligation followed by 2 h of hypoxia (8% O(2) at 37 degrees C). Pups were then treated with HBOT (2.5 ATA) or normobaric oxygenation treatment (NBOT) for 2 h. The expression and phosphorylation status of HIF-1alpha was evaluated at intervals up to 24 h after the insult, as was the expression of glucose transporter (GLUT)-1, GLUT-3, lactate dehydrogenase (LDH), aldolase (Ald), and p53. The protein-protein interaction of HIF-1alpha and p53 was also examined. An elevated expression of HIF-1alpha, GLUT-1, GLUT-3, Ald, and LDH was observed after the insult. An increase in the dephosphorylated form of HIF-1alpha was followed by an increase in the association of HIF-1alpha with p53 and an increase in p53 levels. Both HBOT and NBOT reduced the elevated expression of HIF-1alpha and decreased its dephosphorylated form. Furthermore, both treatments promoted a transient increase in the expression of GLUT-1, GLUT-3, LDH, and Ald, while decreasing the HIF-1alpha-p53 interaction and decreasing the expression of p53. Therefore, the alteration of the HIF-1alpha phenotype by a single oxygen treatment may be one of the underlying mechanisms for the observed oxygen-induced neuroprotection seen when oxygen is administered after a neonatal hypoxic-ischemic insult.
Collapse
Affiliation(s)
- John W Calvert
- Division of Neurosurgery, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | | | | | | |
Collapse
|
49
|
Presti AL, Kishkurno SV, Slinko SK, Randis TM, Ratner VI, Polin RA, Ten VS. Reoxygenation with 100% oxygen versus room air: late neuroanatomical and neurofunctional outcome in neonatal mice with hypoxic-ischemic brain injury. Pediatr Res 2006; 60:55-9. [PMID: 16690944 DOI: 10.1203/01.pdr.0000223766.98760.88] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Study investigated neuroutcome in mice subjected at 7-8 d of life to hypoxic-ischemic brain injury (HI) followed by 30 min of reoxygenation with 100% O(2) (Re-O(2)) or room air (Re-Air). At 24 h of recovery, mouse reflexes were tested. At 7 wks after HI spatial orientation and memory were assessed in the same mice. Mortality rate was recorded at 24 h and at 7 wks of recovery. In separate cohort of mice, changes in cerebral blood flow (CBF) during HI-insult and reoxygenation were recorded. Re-O(2)versus Re-Air mice exhibited significantly delayed geotaxis reflex. Adult Re-O(2)versus Re-Air mice exhibited significantly better spatial learning and orientation with strong tendency toward better preserved memory. Histopathology revealed significantly less hippocampal atrophy in Re-O(2)versus Re-Air mice. Following a hypoxia-induced hypoperfusion, Re-O(2) re-established CBF in the ipsilateral side to the prehypoxic level significantly faster than Re-Air. The mortality was higher among Re-O2 versus Re-Air mice, although, it did not reach statistical significance. Re-O(2)versus Re-Air restores CBF significantly faster and results in better late neuroutcome. However, greater early motor deficit and higher mortality rate among Re-O(2)versus Re-Air mice suggest that Re-O(2) may be deleterious at the early stage of recovery.
Collapse
Affiliation(s)
- Amy L Presti
- Department of Pediatrics, Weill- Cornell University, NY, NY 10021, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Rossignol DA, Rossignol LW. Hyperbaric oxygen therapy may improve symptoms in autistic children. Med Hypotheses 2006; 67:216-28. [PMID: 16554123 DOI: 10.1016/j.mehy.2006.02.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2006] [Revised: 02/03/2006] [Accepted: 02/07/2006] [Indexed: 01/25/2023]
Abstract
Autism is a neurodevelopmental disorder that currently affects as many as 1 out of 166 children in the United States. Recent research has discovered that some autistic individuals have decreased cerebral perfusion, evidence of neuroinflammation, and increased markers of oxidative stress. Multiple independent single photon emission computed tomography (SPECT) and positron emission tomography (PET) research studies have revealed hypoperfusion to several areas of the autistic brain, most notably the temporal regions and areas specifically related to language comprehension and auditory processing. Several studies show that diminished blood flow to these areas correlates with many of the clinical features associated with autism including repetitive, self-stimulatory and stereotypical behaviors, and impairments in communication, sensory perception, and social interaction. Hyperbaric oxygen therapy (HBOT) has been used with clinical success in several cerebral hypoperfusion syndromes including cerebral palsy, fetal alcohol syndrome, closed head injury, and stroke. HBOT can compensate for decreased blood flow by increasing the oxygen content of plasma and body tissues and can even normalize oxygen levels in ischemic tissue. In addition, animal studies have shown that HBOT has potent anti-inflammatory effects and reduces oxidative stress. Furthermore, recent evidence demonstrates that HBOT mobilizes stem cells from human bone marrow, which may aid recovery in neurodegenerative diseases. Based upon these findings, it is hypothesized that HBOT will improve symptoms in autistic individuals. A retrospective case series is presented that supports this hypothesis.
Collapse
Affiliation(s)
- Daniel A Rossignol
- Blue Ridge Medical Center, 4038 Thomas Nelson Highway, Arrington, VA 22922, USA.
| | | |
Collapse
|