51
|
The Association between NOS3 Gene Polymorphisms and Hypoxic-Ischemic Encephalopathy Susceptibility and Symptoms in Chinese Han Population. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1957374. [PMID: 28070505 PMCID: PMC5192303 DOI: 10.1155/2016/1957374] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/09/2016] [Accepted: 11/27/2016] [Indexed: 11/22/2022]
Abstract
Endothelial NOS (NOS3) has a potential role in the prevention of neuronal injury in hypoxic-ischemic encephalopathy (HIE). Thus, we aimed to explore the association between NOS3 gene polymorphisms and HIE susceptibility and symptoms in a Chinese Han population. Three single nucleotide polymorphisms (SNPs) in the NOS3 gene, rs1800783, rs1800779, and rs2070744, were detected in 226 children with HIE and 212 healthy children in a Chinese Han population. Apgar scores and magnetic resonance image scans were used to estimate the symptoms and brain damage. The association analyses were conducted by using SNPStats and SPSS 18.0 software. The genotype and allele distributions of rs1800779 and rs1799983 displayed no significant differences between the patients and the controls, while the rs2070744 allele distribution was significantly different (corrected P = 0.009). For clinical characteristics, the rs2070744 genotype distribution was significantly different in patients with different Apgar scores (≤5, TT/TC/CC = 6/7/5; 6~7, TT/TC/CC = 17/0/0; 8~9, TT/TC/CC = 6/2/0; 10, TT/TC/CC = 7/1/0; corrected P = 0.006) in the 1001 to 1449 g birth weight subgroup. The haplotype test did not show any associations with the risk and clinical characteristics of HIE. The results suggest that NOS3 gene SNP rs2070744 was significantly associated with HIE susceptibility and symptom expression in Chinese Han population.
Collapse
|
52
|
Wu H, Li Z, Yang X, Liu J, Wang W, Liu G. SBDPs and Tau proteins for diagnosis and hypothermia therapy in neonatal hypoxic ischemic encephalopathy. Exp Ther Med 2016; 13:225-229. [PMID: 28123494 PMCID: PMC5245122 DOI: 10.3892/etm.2016.3911] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/03/2016] [Indexed: 12/14/2022] Open
Abstract
The use of spectrin breakdown products (SBDPs) and Tau protein levels for diagnosis and a mild hypothermia therapy for treatment of neonatal hypoxic-ischemic encephalopathy (HIE) was evaluated. One hundred and fifty infants, with HIE within 12 h after birth, participated in the study. There were 30 newborns with mild symptoms, 60 with moderate symptoms, 60 with severe symptoms, and 30 in a control group. Regular therapy was used for the control and the mild HIE groups, and also for 30 cases in the group with moderate symptoms and for 30 in the group with severe symptoms. For the remaining infants, with moderate and severe symptoms, mild hypothermia therapy was used instead. A sandwich ELISA measured plasma concentrations of SBDPs and Tau proteins, at different time-points. For clinical follow-up, the neonatal behavioral neurological assessment (NBNA) assay and the Gesell development scale were performed at different time-points. The levels of SBDP and Tau proteins increased with the exacerbation of HIE, and decreased with the prolongation of therapy with statistically significant differences amongst groups. After treatment, the levels of SBDP and Tau proteins in groups with moderate and severe symptoms treated with mild hypothermia therapy were significantly lower than those of the groups treated with regular therapy. NBNA scores and the developmental quotient (DQ) were both worse with the increase in severity of HIE, however, the scores of groups with moderate and severe symptoms treated with mild hypothermia therapy were significantly better than those of groups treated with regular therapy (P<0.05). A gradual improvement of DQ was seen in the process of therapy in each group (P<0.05). According to a receiver operating characteristic (ROC) curve analysis, at a critical plasma concentration of SBDPs of 1.58 ng/ml, the sensitivity and specificity for HIE diagnosis was 84.6 and 87.5%, respectively. The ROC analysis for Tau protein yielded a sensitivity and specificity of 79.5 and 96.9%, respectively, at a critical plasma concentration of 4.76 pg/ml. Given our results, SBDPs and Tau proteins are very useful for the early diagnosis of HIE. Early application of mild hypothermia therapy for the treatment of HIE can greatly improve the function of neural development. These findings should greatly improve the evaluation and treatment approaches for HIE.
Collapse
Affiliation(s)
- Hongwei Wu
- Department of Neonatology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Zhenguang Li
- Department of Neonatology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Xia Yang
- Department of Neonatology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Jinfeng Liu
- Department of Neonatology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Wei Wang
- Department of Neonatology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Gang Liu
- Department of Neonatology, Xuzhou Children's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
53
|
Netto CA, Sanches E, Odorcyk FK, Duran-Carabali LE, Weis SN. Sex-dependent consequences of neonatal brain hypoxia-ischemia in the rat. J Neurosci Res 2016; 95:409-421. [DOI: 10.1002/jnr.23828] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Carlos Alexandre Netto
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Eduardo Sanches
- Division of Child Development and Growth, Department of Pediatrics; University of Geneva; Geneva Switzerland
| | - Felipe Kawa Odorcyk
- Postgraduate Program of Neurosciences, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Luz Elena Duran-Carabali
- Postgraduate Program of Physiology, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Simone Nardin Weis
- Department of Cellular Biology; Universidade de Brasília; Brasilia Distrito Federal Brazil
| |
Collapse
|
54
|
Chafer-Pericas C, Cernada M, Rahkonen L, Stefanovic V, Andersson S, Vento M. Preliminary case control study to establish the correlation between novel peroxidation biomarkers in cord serum and the severity of hypoxic ischemic encephalopathy. Free Radic Biol Med 2016; 97:244-249. [PMID: 27296840 DOI: 10.1016/j.freeradbiomed.2016.06.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/01/2016] [Accepted: 06/09/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) has deleterious neurological consequences. To identify patients at risk of neuronal damage deserving implementation of neuroprotective strategies clinicians have relied on prenatal sentinel events, postnatal clinical assessment (Apgar score), and blood gas analysis. This feasibility study aimed to assess if lipid peroxidation byproducts associated with neuronal damage correlated with cord blood metabolic acidemia in patients with HIE. POPULATION AND METHODS This is a case/control study in which cases were newborn infants with severe acidemia (pH<7.00; base excess ≥12mmol/L) while control babies exhibited normal gases (pH=7.20-7.40; base excess=-4 to +4mmol/L) in the first cord blood analysis performed immediately after birth. Concomitantly, lipid peroxidation byproducts were determined using ultra performance liquid chromatography coupled to mass spectrometry in the same cord blood sample. RESULTS A total of 19 controls and 20 cases were recruited. No differences in gestational characteristics were present. However, cases exhibited profound metabolic alterations as compared to controls (Cases vs. CONTROL pH=6.90±0.1 vs. 7.33±0.03; base excess=-15±3 vs. -1±2mmol/L), 85% were admitted to the NICU, and 50% developed symptoms of HIE. 8-iso-15(R)-PGF2α (P=0.01) and total isoprostanes (P=0.045) presented statistically significant differences between cases and control groups and correlated with level of HIE. CONCLUSIONS The 8-iso-15(R)-PGF2α and isoprostanes reflecting oxidative damage are significantly increased in severe postnatal acidemia. Follow up studies with adequate power are necessary to confirm if these biomarkers measured in cord blood serum could be predictive of neonatal encephalopathy.
Collapse
Affiliation(s)
| | - María Cernada
- Neonatal Research Group, Health Research Institute La Fe, Valencia, Spain
| | - Leena Rahkonen
- Department of Obstetrics and Gynecology, Fetomaternal Medical Center, Helsinki University Hospital, Finland
| | - Vedran Stefanovic
- Department of Obstetrics and Gynecology, Fetomaternal Medical Center, Helsinki University Hospital, Finland
| | - Sture Andersson
- Children׳s Hospital, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Máximo Vento
- Neonatal Research Group, Health Research Institute La Fe, Valencia, Spain; Division of Neonatology, University & Polytechnic Hospital La Fe, Valencia, Spain.
| |
Collapse
|
55
|
Wong DM, Jeffery N, Hepworth-Warren KL, Wiechert SA, Miles K. Magnetic resonance imaging of presumptive neonatal encephalopathy in a foal. EQUINE VET EDUC 2016. [DOI: 10.1111/eve.12590] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- D. M. Wong
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; Iowa State University; Ames USA
| | - N. Jeffery
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; Iowa State University; Ames USA
| | - K. L. Hepworth-Warren
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; Iowa State University; Ames USA
| | - S. A. Wiechert
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; Iowa State University; Ames USA
| | - K. Miles
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; Iowa State University; Ames USA
| |
Collapse
|
56
|
Dixon BJ, Reis C, Ho WM, Tang J, Zhang JH. Neuroprotective Strategies after Neonatal Hypoxic Ischemic Encephalopathy. Int J Mol Sci 2015; 16:22368-401. [PMID: 26389893 PMCID: PMC4613313 DOI: 10.3390/ijms160922368] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/31/2015] [Accepted: 09/06/2015] [Indexed: 12/21/2022] Open
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) is a devastating disease that primarily causes neuronal and white matter injury and is among the leading cause of death among infants. Currently there are no well-established treatments; thus, it is important to understand the pathophysiology of the disease and elucidate complications that are creating a gap between basic science and clinical translation. In the development of neuroprotective strategies and translation of experimental results in HIE, there are many limitations and challenges to master based on an appropriate study design, drug delivery properties, dosage, and use in neonates. We will identify understudied targets after HIE, as well as neuroprotective molecules that bring hope to future treatments such as melatonin, topiramate, xenon, interferon-beta, stem cell transplantation. This review will also discuss some of the most recent trials being conducted in the clinical setting and evaluate what directions are needed in the future.
Collapse
Affiliation(s)
- Brandon J Dixon
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | - Cesar Reis
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Wing Mann Ho
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Medical University Innsbruck, Tyrol 6020, Austria.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|
57
|
Lv H, Wang Q, Wu S, Yang L, Ren P, Yang Y, Gao J, Li L. Neonatal hypoxic ischemic encephalopathy-related biomarkers in serum and cerebrospinal fluid. Clin Chim Acta 2015; 450:282-97. [PMID: 26320853 DOI: 10.1016/j.cca.2015.08.021] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/22/2015] [Accepted: 08/25/2015] [Indexed: 12/15/2022]
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) is a common disease caused by perinatal asphyxia, a major cause of neonatal death, neurological behavior, and long-term disability. Currently, the diagnosis and prognosis of neonatal HIE are based on nervous system clinical manifestations, imaging and electrophysiological examination. These take time and late diagnosis allows brain injury to occur in newborns, so that infants of many brain injury missed the best treatment time, left with varying degrees of neurological sequelae. The use of biomarkers to monitor brain injury and evaluate neuroprotective effects might allow the early intervention and treatment of neonatal HIE to reduce mortality rates. This study reviewed the mechanism of neonatal hypoxic ischemic encephalopathy in relation to numerous brain-related biomarkers including NSE, S-100β, GFAP, UCH-L1, Tau protein, miRNA, LDH, and CK-BB. In early diagnosis of neonatal HIE, S-100β and activin A seems to be better biomarkers. Biomarkers with the greatest potential to predict long-term neurologic handicap of neonates with HIE are GFAP and UCH-L1 and when combined with other markers or brain imaging can increase the detection rate of HIE. Tau protein is a unique biological component of nervous tissues, and might have value for neonatal HIE diagnosis. Combination of more than two biological markers should be a future research direction.
Collapse
Affiliation(s)
- Hongyan Lv
- Department of Neonatology, Handan Maternal and Child Care Centers, Handan 056002, Hebei Province, PR China; Department of Neonatal pathology, Handan Maternal and Child Care Centers, Handan 056002, Hebei Province, PR China
| | - Qiuli Wang
- Department of Neonatology, Handan Maternal and Child Care Centers, Handan 056002, Hebei Province, PR China
| | - Sujing Wu
- Department of Neonatology, Handan Maternal and Child Care Centers, Handan 056002, Hebei Province, PR China; Department of Neonatal pathology, Handan Maternal and Child Care Centers, Handan 056002, Hebei Province, PR China
| | - Lihong Yang
- Department of Neonatology, Handan Maternal and Child Care Centers, Handan 056002, Hebei Province, PR China
| | - Pengshun Ren
- Department of Neonatology, Handan Maternal and Child Care Centers, Handan 056002, Hebei Province, PR China
| | - Yihui Yang
- Department of Neural development and neural pathology, Hebei University of Engineering School of Medicine, Handan 056029, Hebei Province, PR China
| | - Jinsheng Gao
- Department of Pathology, Hebei University of Engineering School of Medicine, Handan 056029, Hebei Province, PR China
| | - Lianxiang Li
- Department of Neonatal pathology, Handan Maternal and Child Care Centers, Handan 056002, Hebei Province, PR China; Department of Neural development and neural pathology, Hebei University of Engineering School of Medicine, Handan 056029, Hebei Province, PR China.
| |
Collapse
|
58
|
Ek CJ, D'Angelo B, Baburamani AA, Lehner C, Leverin AL, Smith PLP, Nilsson H, Svedin P, Hagberg H, Mallard C. Brain barrier properties and cerebral blood flow in neonatal mice exposed to cerebral hypoxia-ischemia. J Cereb Blood Flow Metab 2015; 35:818-27. [PMID: 25627141 PMCID: PMC4420855 DOI: 10.1038/jcbfm.2014.255] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 12/05/2014] [Accepted: 12/08/2014] [Indexed: 11/09/2022]
Abstract
Insults to the developing brain often result in irreparable damage resulting in long-term deficits in motor and cognitive functions. The only treatment today for hypoxic-ischemic encephalopathy (HIE) in newborns is hypothermia, which has limited clinical benefit. We have studied changes to the blood-brain barriers (BBB) as well as regional cerebral blood flow (rCBF) in a neonatal model of HIE to further understand the underlying pathologic mechanisms. Nine-day old mice pups, brain roughly equivalent to the near-term human fetus, were subjected to hypoxia-ischemia. Hypoxia-ischemia increased BBB permeability to small and large molecules within hours after the insult, which normalized in the following days. The opening of the BBB was associated with changes to BBB protein expression whereas gene transcript levels were increased showing direct molecular damage to the BBB but also suggesting compensatory mechanisms. Brain pathology was closely related to reductions in rCBF during the hypoxia as well as the areas with compromised BBB showing that these are intimately linked. The transient opening of the BBB after the insult is likely to contribute to the pathology but at the same time provides an opportunity for therapeutics to better reach the infarcted areas in the brain.
Collapse
Affiliation(s)
- C Joakim Ek
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Barbara D'Angelo
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ana A Baburamani
- 1] Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden [2] Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Christine Lehner
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Department of Traumatology and Sport Injuries, Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria; Austrian Cluster for Tissue Regeneration
| | - Anna-Lena Leverin
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter L P Smith
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Holger Nilsson
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Svedin
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- 1] Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK [2] Departments of Obstetrics and Gynecology, Institute for Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
59
|
Moretti R, Pansiot J, Bettati D, Strazielle N, Ghersi-Egea JF, Damante G, Fleiss B, Titomanlio L, Gressens P. Blood-brain barrier dysfunction in disorders of the developing brain. Front Neurosci 2015; 9:40. [PMID: 25741233 PMCID: PMC4330788 DOI: 10.3389/fnins.2015.00040] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/27/2015] [Indexed: 12/22/2022] Open
Abstract
Disorders of the developing brain represent a major health problem. The neurological manifestations of brain lesions can range from severe clinical deficits to more subtle neurological signs or behavioral problems and learning disabilities, which often become evident many years after the initial damage. These long-term sequelae are due at least in part to central nervous system immaturity at the time of the insult. The blood-brain barrier (BBB) protects the brain and maintains homeostasis. BBB alterations are observed during both acute and chronic brain insults. After an insult, excitatory amino acid neurotransmitters are released, causing reactive oxygen species (ROS)-dependent changes in BBB permeability that allow immune cells to enter and stimulate an inflammatory response. The cytokines, chemokines and other molecules released as well as peripheral and local immune cells can activate an inflammatory cascade in the brain, leading to secondary neurodegeneration that can continue for months or even years and finally contribute to post-insult neuronal deficits. The role of the BBB in perinatal disorders is poorly understood. The inflammatory response, which can be either acute (e.g., perinatal stroke, traumatic brain injury) or chronic (e.g., perinatal infectious diseases) actively modulates the pathophysiological processes underlying brain injury. We present an overview of current knowledge about BBB dysfunction in the developing brain during acute and chronic insults, along with clinical and experimental data.
Collapse
Affiliation(s)
- Raffaella Moretti
- INSERM U1141, Robert Debre's Hospital Paris, France ; Université Paris Diderot, Sorbonne Paris Cité, UMRS 1141-PROTECT Paris, France ; PremUP Paris, France ; S. Maria della Misericordia Hospital, Università degli Studi di Udine Udine, Italy
| | - Julien Pansiot
- INSERM U1141, Robert Debre's Hospital Paris, France ; Université Paris Diderot, Sorbonne Paris Cité, UMRS 1141-PROTECT Paris, France ; PremUP Paris, France
| | - Donatella Bettati
- INSERM U1141, Robert Debre's Hospital Paris, France ; Université Paris Diderot, Sorbonne Paris Cité, UMRS 1141-PROTECT Paris, France ; PremUP Paris, France
| | - Nathalie Strazielle
- Lyon Neurosciences Research Center, INSERM U1028, CNRS UMR5292 - Lyon University Lyon, France ; Brain-i Lyon, France
| | | | - Giuseppe Damante
- S. Maria della Misericordia Hospital, Università degli Studi di Udine Udine, Italy
| | - Bobbi Fleiss
- INSERM U1141, Robert Debre's Hospital Paris, France ; Université Paris Diderot, Sorbonne Paris Cité, UMRS 1141-PROTECT Paris, France ; PremUP Paris, France ; Department of Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, St. Thomas' Hospital London, UK
| | - Luigi Titomanlio
- INSERM U1141, Robert Debre's Hospital Paris, France ; Université Paris Diderot, Sorbonne Paris Cité, UMRS 1141-PROTECT Paris, France ; PremUP Paris, France ; Pediatric Emergency Department, APHP, Robert Debré Hospital Paris, France
| | - Pierre Gressens
- INSERM U1141, Robert Debre's Hospital Paris, France ; Université Paris Diderot, Sorbonne Paris Cité, UMRS 1141-PROTECT Paris, France ; PremUP Paris, France ; Department of Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, St. Thomas' Hospital London, UK
| |
Collapse
|
60
|
Zhang J, Sadowska GB, Chen X, Park SY, Kim JE, Bodge CA, Cummings E, Lim YP, Makeyev O, Besio WG, Gaitanis J, Banks WA, Stonestreet BS. Anti-IL-6 neutralizing antibody modulates blood-brain barrier function in the ovine fetus. FASEB J 2015; 29:1739-53. [PMID: 25609424 DOI: 10.1096/fj.14-258822] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 12/22/2014] [Indexed: 12/15/2022]
Abstract
Impaired blood-brain barrier function represents an important component of hypoxic-ischemic brain injury in the perinatal period. Proinflammatory cytokines could contribute to ischemia-related blood-brain barrier dysfunction. IL-6 increases vascular endothelial cell monolayer permeability in vitro. However, contributions of IL-6 to blood-brain barrier abnormalities have not been examined in the immature brain in vivo. We generated pharmacologic quantities of ovine-specific neutralizing anti-IL-6 mAbs and systemically infused mAbs into fetal sheep at 126 days of gestation after exposure to brain ischemia. Anti-IL-6 mAbs were measured by ELISA in fetal plasma, cerebral cortex, and cerebrospinal fluid, blood-brain barrier permeability was quantified using the blood-to-brain transfer constant in brain regions, and IL-6, tight junction proteins, and plasmalemma vesicle protein (PLVAP) were detected by Western immunoblot. Anti-IL-6 mAb infusions resulted in increases in mAb (P < 0.05) in plasma, brain parenchyma, and cerebrospinal fluid and decreases in brain IL-6 protein. Twenty-four hours after ischemia, anti-IL-6 mAb infusions attenuated ischemia-related increases in blood-brain barrier permeability and modulated tight junction and PLVAP protein expression in fetal brain. We conclude that inhibiting the effects of IL-6 protein with systemic infusions of neutralizing antibodies attenuates ischemia-related increases in blood-brain barrier permeability by inhibiting IL-6 and modulates tight junction proteins after ischemia.
Collapse
Affiliation(s)
- Jiyong Zhang
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Grazyna B Sadowska
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Xiaodi Chen
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Seon Yeong Park
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jeong-Eun Kim
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Courtney A Bodge
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Erin Cummings
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Yow-Pin Lim
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Oleksandr Makeyev
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Walter G Besio
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - John Gaitanis
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - William A Banks
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Barbara S Stonestreet
- *Department of Pediatrics, Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA; ProThera Biologics, Incorporated, Providence, Rhode Island, USA; Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island, USA; Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA; and Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
61
|
Douglas-Escobar MV, Heaton SC, Bennett J, Young LJ, Glushakova O, Xu X, Barbeau DY, Rossignol C, Miller C, Old Crow AM, Hayes RL, Weiss MD. UCH-L1 and GFAP Serum Levels in Neonates with Hypoxic-Ischemic Encephalopathy: A Single Center Pilot Study. Front Neurol 2014; 5:273. [PMID: 25566179 PMCID: PMC4271579 DOI: 10.3389/fneur.2014.00273] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 12/03/2014] [Indexed: 01/10/2023] Open
Abstract
Objective: We examined two potential biomarkers of brain damage in hypoxic–ischemic encephalopathy (HIE) neonates: glial fibrillary acidic protein (GFAP; a marker of gliosis) and ubiquitin C-terminal hydrolase L1 (UCH-L1; a marker of neuronal injury). We hypothesized that the biomarkers would be measurable in cord blood of healthy neonates and could serve as a normative reference for brain injury in HIE infants. We further hypothesized that higher levels would be detected in serum samples of HIE neonates and would correlate with brain damage on magnetic resonance imaging (MRI) and later developmental outcomes.? Study Design: Serum UCH-L1 and GFAP concentrations from HIE neonates (n = 16) were compared to controls (n = 11). The relationship between biomarker concentrations of HIE neonates and brain damage (MRI) and developmental outcomes (Bayley-III) was examined using Pearson correlation coefficients and a mixed model design. Result: Both biomarkers were detectable in cord blood from control subjects. UCH-L1 concentrations were higher in HIE neonates (p < 0.001), and associated with cortical injury (p < 0.055) and later motor and cognitive developmental outcomes (p < 0.05). The temporal change in GFAP concentrations during (from birth to 96 h of age) predicted motor developmental outcomes (p < 0.05) and injury to the basal ganglia and white matter. Conclusion: Ubiquitin C-terminal hydrolase L1 and GFAP should be explored further as promising serum biomarkers of brain damage and later neurodevelopmental outcomes in neonates with HIE.
Collapse
Affiliation(s)
- Martha V Douglas-Escobar
- Department of Pediatrics, University of Florida , Gainesville, FL , USA ; Department of Pediatrics, University of California San Francisco , San Francisco, CA , USA
| | - Shelley C Heaton
- Department of Clinical and Health Psychology, University of Florida , Gainesville, FL , USA
| | - Jeffrey Bennett
- Department of Radiology, University of Florida , Gainesville, FL , USA
| | - Linda J Young
- Department of Statistics, University of Florida , Gainesville, FL , USA
| | | | - Xiaohui Xu
- Department of Biostatistics, University of Florida , Gainesville, FL , USA
| | | | - Candice Rossignol
- Department of Pediatrics, University of Florida , Gainesville, FL , USA
| | - Cindy Miller
- Department of Pediatrics, University of Florida , Gainesville, FL , USA
| | - Alissa M Old Crow
- Department of Radiology, University of Florida , Gainesville, FL , USA
| | | | - Michael D Weiss
- Department of Pediatrics, University of Florida , Gainesville, FL , USA
| |
Collapse
|
62
|
Yu L, Yang B, Wang J, Zhao L, Luo W, Jiang Q, Yang J. Time course change of COX2-PGI2/TXA2 following global cerebral ischemia reperfusion injury in rat hippocampus. Behav Brain Funct 2014; 10:42. [PMID: 25388440 PMCID: PMC4240876 DOI: 10.1186/1744-9081-10-42] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 10/22/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroinflammation plays pivotal roles in the progression of cerebral ischemia injury. Prostaglandins (PGs) as the major inflammatory mediators in the brain participate in the pathophysiological processes of cerebral ischemia injury. Cyclooxygenase-2 (COX2) is the rate-limiting enzyme of PGs, and thus it is necessary to characterize of the expression patterns of COX2 and its downstream products at the same time in a cerebral ischemia/reperfusion (I/R) model. METHODS The levels of prostacyclin (PGI2) and thromboxane (TXA2) and the expression of COX2 were detected in the rat hippocampus at different time points after reperfusion (30 min, 2 h, 6 h, 24 h, 48 h, 7 d, and 15 d). RESULTS The COX2 mRNA and protein expressions in hippocampus both remarkably increased at 30 min, and peaked at 7 d after global cerebral I/R compared with the sham-operated group. The level of PGI2 significantly increased at 2 h after reperfusion, with a peak at 48 h, but was still significantly higher than the sham-operated animals at 15 d. TXA2 level decreased at 30 min and 2 h after reperfusion, but significantly increased at 6 h and peaked at 48 h. PGI2/TXA2 ratio increased at 30 min after reperfusion, and peaked at 48 h compared with the sham-operated animals. CONCLUSIONS I/R injury significantly increased the COX2 expression, PGI2 and TXA2 levels, and the PGI2/TXA2 ratio in rat hippocampus in a time-dependent manner. As a consequence, the increased PGI2 level and PGI2/TXA2 ratio may represent a physiological mechanism to protect the brain against the neuronal damage produced by I/R injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Junqing Yang
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Medical College Rd, No 1, Chongqing 400016, China.
| |
Collapse
|
63
|
Hagberg H, Ek C. Targeting the vasculature for cerebroprotection in the immature brain. Exp Neurol 2014; 261:551-2. [DOI: 10.1016/j.expneurol.2014.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 08/05/2014] [Indexed: 11/29/2022]
|
64
|
Abstract
Therapeutic hypothermia is the only treatment that has been shown to be of benefit to infant's ≥ 36 weeks of gestation with hypoxic-ischemic encephalopathy. The evidence for the benefit is based on multiple, well-designed randomized clinical trials. Based on this data, the use of therapeutic hypothermia has been widely disseminated throughout the neonatal community. An important concept in hypoxic-ischemic brain injury is the functioning of the neurovascular unit which links neurons, non-neuronal cellular elements and the capillary endothelial cells to promote optimal barrier maintenance between the brain and systemic circulation, regulation of blood flow and neuro-immunologic functioning. Hypoxic-ischemic injury can trigger increased permeability of the blood-brain-barrier via molecular events within the neurovascular unit and initiate pathways to brain injury. In addition, exposure of the brain to cellular elements from the systemic circulation can further propagate the neuro-inflammatory response. The influence of temperature on injury to the neurovascular unit has received relatively little attention. This review will focus on one component of the neurovascular unit, the blood-brain barrier and its constituents. Specifically, this review will address the effects of hypoxia-ischemia and temperature on the neurovascular unit and potential knowledge gaps which may serve as areas for further investigation.
Collapse
Affiliation(s)
- Abbot Laptook
- Warren Alpert Medical School of Brown University, United States; Neonatal Intensive Care Unit, Women and Infants Hospital of Rhode Island, 101 Dudley Street, Providence, RI 02905, United States.
| |
Collapse
|
65
|
Golomb BA, Allison M, Koperski S, Koslik HJ, Devaraj S, Ritchie JB. Coenzyme Q10 benefits symptoms in Gulf War veterans: results of a randomized double-blind study. Neural Comput 2014; 26:2594-651. [PMID: 25149705 DOI: 10.1162/neco_a_00659] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
We sought to assess whether coenzyme Q10 (CoQ10) benefits the chronic multisymptom problems that affect one-quarter to one-third of 1990-1 Gulf War veterans, using a randomized, double-blind, placebo-controlled study. Participants were 46 veterans meeting Kansas and Centers for Disease Control criteria for Gulf War illness. Intervention was PharmaNord (Denmark) CoQ10 100 mg per day (Q100), 300 mg per day (Q300), or an identical-appearing placebo for 3.5 ± 0.5 months. General self-rated health (GSRH), the primary outcome, differed across randomization arms at baseline, and sex significantly predicted GSRH change, compelling adjustment for baseline GSRH and prompting sex-stratified analysis. GSRH showed no significant benefit in the combined-sex sample. Among males (85% of participants), Q100 significantly benefited GSRH versus placebo and versus Q300, providing emphasis on Q100. Physical function (summary performance score, SPS) improved on Q100 versus placebo. A rise in CoQ10 approached significance as a predictor of improvement in GSRH and significantly predicted SPS improvement. Among 20 symptoms each present in half or more of the enrolled veterans, direction-of-difference on Q100 versus placebo was favorable for all except sleep problems; sign test 19:1, p=0.00004) with several symptoms individually significant. Significance for these symptoms despite the small sample underscores large effect sizes, and an apparent relation of key outcomes to CoQ10 change increases prospects for causality. In conclusion, Q100 conferred benefit to physical function and symptoms in veterans with Gulf War illness. Examination in a larger sample is warranted, and findings from this study can inform the conduct of a larger trial.
Collapse
Affiliation(s)
- Beatrice A Golomb
- Departments of Medicine and of Family and Preventive Medicine, University of California, San Diego, La Jolla, CA 92093, U.S.A.
| | | | | | | | | | | |
Collapse
|
66
|
Chen X, Deng A, Zhou T, Ding F. Pretreatment with 2-(4-methoxyphenyl)ethyl-2-acetamido-2-deoxy-β-D-pyranoside attenuates cerebral ischemia/reperfusion-induced injury in vitro and in vivo. PLoS One 2014; 9:e100126. [PMID: 24991917 PMCID: PMC4084628 DOI: 10.1371/journal.pone.0100126] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 05/22/2014] [Indexed: 01/06/2023] Open
Abstract
Salidroside, extracted from the root of Rhodiola rosea L, is known for its pharmacological properties, in particular its neuroprotective effects. 2-(4-Methoxyphenyl) ethyl-2-acetamido-2-deoxy-β-D-pyranoside (GlcNAc-Sal), an analog of salidroside, was recently synthesized and shown to possess neuroprotective properties. The purpose of the current study was to investigate the neuroprotective effects of GlcNAc-Sal against oxygen-glucose deprivation-reperfusion (OGD-R)-induced neurotoxicity in vitro and global cerebral ischemia-reperfusion (GCI-R) injury in vivo. Cell viability tests and Hoechst 33342 staining confirmed that GlcNAc-Sal pretreatment markedly attenuated OGD-R induced apoptotic cell death in immortalized mouse hippocampal HT22 cells. Western blot, immunofluorescence and PCR analyses revealed that GlcNAc-Sal pretreatment restored the balance of pro- and anti-apoptotic proteins and inhibited the activation of caspase-3 and PARP induced by OGD-R treatment. Further analyses showed that GlcNAc-Sal pretreatment antagonized reactive oxygen species (ROS) generation, iNOS-derived NO production and NO-related apoptotic cell death during OGD-R stimulation. GCI-R was induced by bilateral common carotid artery occlusion (BCCAO) and reperfusion in mice in vivo. Western blot analysis showed that GlcNAc-Sal pretreatment decreased the expression of caspase-3 and increased the expression of Bcl-2 (B-cell lymphoma 2)/Bax (Bcl-2-associated X protein) induced by GCI-R treatment. Our findings suggest that GlcNAc-Sal pretreatment prevents brain ischemia reperfusion injury by the direct or indirect suppression of cell apoptosis and GlcNAc-Sal could be developed as a broad-spectrum agent for the prevention and/or treatment of cerebral ischemic injury.
Collapse
Affiliation(s)
- Xia Chen
- Basic Medical Research Centre, Medical School, Nantong University, Nantong, China
| | - Aiqing Deng
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, China
| | - Tianqiu Zhou
- Department of ophtalmology, Affiliated Hospital of Nantong University, Nantong, China
| | - Fei Ding
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
- * E-mail:
| |
Collapse
|
67
|
Reinke SN, Walsh BH, Boylan GB, Sykes BD, Kenny LC, Murray DM, Broadhurst DI. 1H NMR derived metabolomic profile of neonatal asphyxia in umbilical cord serum: implications for hypoxic ischemic encephalopathy. J Proteome Res 2013; 12:4230-9. [PMID: 23931672 DOI: 10.1021/pr400617m] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) is a severe consequence of perinatal asphyxia (PA) that can result in life-long neurological disability. Disease mechanisms, including the role and interaction of individual metabolic pathways, remain unclear. As hypoxia is an acute condition, aerobic energy metabolism is central to global metabolic pathways, and these metabolites are detectable using 1H NMR spectroscopy, we hypothesized that characterizing the NMR-derived umbilical cord serum metabolome would offer insight into the consequences of PA that lead to HIE. Fifty-nine at-risk infants were enrolled, together with 1:1 matched healthy controls, and stratified by disease severity (n=25, HIE; n=34, non-HIE PA). Eighteen of 37 reproducibly detectable metabolites were significantly altered between study groups. Acetone, 3-hydroxybutyrate, succinate, and glycerol were significantly differentially altered in severe HIE. Multivariate data analysis revealed a metabolite profile associated with both asphyxia and HIE. Multiple-linear regression modeling using 4 metabolites (3-hydroxybutyrate, glycerol, O-phosphocholine, and succinate) predicted HIE severity with an adjusted R2 of 0.4. Altered ketones suggest that systemic metabolism may play a critical role in preventing neurological injury, while altered succinate provides a possible explanation for hypoxia-inducible factor 1-α (HIF-1α) stabilization in HI injury.
Collapse
Affiliation(s)
- Stacey N Reinke
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
68
|
Abstract
The treatment of neonatal seizures has not changed significantly over the last 50 years despite advances in antiepileptic drug (AED) development for older children and adults. Recently new drugs have emerged some of which address age-specific challenges or mechanisms and will be discussed in this review. The loop diuretic bumetanide blocks the neuronal NKCC1 co-transporter and is thought specifically to supress seizures in the immature brain. Levetiracetam has been used in children and infants with good efficacy, an excellent safety profile, and near-ideal pharmacokinetic characteristics. Randomised controlled trials are now underway to test the efficacy of some newer AEDs for neonatal seizures. Topiramate has been shown to have neuroprotective properties in addition to its antiepileptic action and trials in babies with hypoxic-ischaemic encephalopathy are now planned. There is an urgent need to develop age-specific AEDs for preterm and term babies. These drugs must be evaluated with multicentre, collaborative trials using innovative methods and high ethical standards to overcome age-specific challenges with the ultimate aim of improving the outcome for neonates with seizures.
Collapse
|
69
|
Abstract
OBJECTIVE To explore the association between multiple umbilical cord blood proteins and severity of hypoxic-ischemic encephalopathy as defined by continuous multichannel electroencephalography. DESIGN A prospective case-control cohort study, which was divided into separate exploratory and validation cohorts. SETTING A single tertiary neonatal intensive care facility. PATIENTS The study recruited full-term infants with perinatal asphyxia and controls. Identical procedures were used to recruit a representative exploratory sample (n = 30) and a subsequent validation cohort (n = 100). INTERVENTION All had umbilical cord blood drawn and biobanked at delivery, continuous multichannel electroencephalography commenced in the first 24 hours, and a modified Sarnat score assigned. Analysis of 37 potential cord blood protein markers of hypoxic-ischemic encephalopathy was performed using Luminex multiplex assays. MEASUREMENTS AND RESULTS Cord blood from 130 infants was analyzed. Interleukin-16 and interleukin-6 significantly differentiated between a moderate-severely abnormal and normal-mildly abnormal electroencephalography background in both exploratory (p = 0.005 and p = 0.016, respectively) and validation cohorts (p = 0.039 and p = 0.024, respectively). To develop a predictive model for a moderate-severely abnormal electroencephalography, stepwise regression analysis was used to combine these analytes with current standard clinical markers of asphyxia (pH, base deficit, and 10-min Apgar). Only Apgar score and interleukin-16 remained in the model, which was highly predictive of an abnormal electroencephalography (area under the curve [AUC] = 0.956, p < 0.001, positive predictive value = 89%, and negative predictive value = 94%). CONCLUSIONS Cord blood interleukin-6 and interleukin-16 were associated with electrographic grade of hypoxic-ischemic encephalopathy. To predict an abnormal electroencephalography, interleukin-16 and 10-minute Apgar used in combination performed better than current markers.
Collapse
|
70
|
Takechi R, Pallebage-Gamarallage MM, Lam V, Giles C, Mamo JC. Nutraceutical agents with anti-inflammatory properties prevent dietary saturated-fat induced disturbances in blood-brain barrier function in wild-type mice. J Neuroinflammation 2013; 10:73. [PMID: 23782872 PMCID: PMC3693897 DOI: 10.1186/1742-2094-10-73] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 06/08/2013] [Indexed: 12/11/2022] Open
Abstract
Background Emerging evidence suggests that disturbances in the blood–brain barrier (BBB) may be pivotal to the pathogenesis and pathology of vascular-based neurodegenerative disorders. Studies suggest that heightened systemic and central inflammations are associated with BBB dysfunction. This study investigated the effect of the anti-inflammatory nutraceuticals garlic extract-aged (GEA), alpha lipoic acid (ALA), niacin, and nicotinamide (NA) in a murine dietary-induced model of BBB dysfunction. Methods C57BL/6 mice were fed a diet enriched in saturated fatty acids (SFA, 40% fat of total energy) for nine months to induce systemic inflammation and BBB disturbances. Nutraceutical treatment groups included the provision of either GEA, ALA, niacin or NA in the positive control SFA-group and in low-fat fed controls. Brain parenchymal extravasation of plasma derived immunoglobulin G (IgG) and large macromolecules (apolipoprotein (apo) B lipoproteins) measured by quantitative immunofluorescent microscopy, were used as markers of disturbed BBB integrity. Parenchymal glial fibrillar acidic protein (GFAP) and cyclooxygenase-2 (COX-2) were considered in the context of surrogate markers of neurovascular inflammation and oxidative stress. Total anti-oxidant status and glutathione reductase activity were determined in plasma. Results Brain parenchymal abundance of IgG and apoB lipoproteins was markedly exaggerated in mice maintained on the SFA diet concomitant with significantly increased GFAP and COX-2, and reduced systemic anti-oxidative status. The nutraceutical GEA, ALA, niacin, and NA completely prevented the SFA-induced disturbances of BBB and normalized the measures of neurovascular inflammation and oxidative stress. Conclusions The anti-inflammatory nutraceutical agents GEA, ALA, niacin, or NA are potent inhibitors of dietary fat-induced disturbances of BBB induced by systemic inflammations.
Collapse
Affiliation(s)
- Ryusuke Takechi
- School of Public Health, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Faculty of Health Sciences, Curtin University, Kent st, Bentley, WA, 6102, Australia
| | | | | | | | | |
Collapse
|
71
|
Hahn GH, Hyttel-Sorensen S, Petersen SM, Pryds O, Greisen G. Cerebral effects of commonly used vasopressor-inotropes: a study in newborn piglets. PLoS One 2013; 8:e63069. [PMID: 23700412 PMCID: PMC3659109 DOI: 10.1371/journal.pone.0063069] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/27/2013] [Indexed: 11/21/2022] Open
Abstract
Background Despite widespread use in sick infants, it is still debated whether vasopressor-inotropes have direct cerebral effects that might affect neurological outcome. We aimed to test direct cerebrovascular effects of three commonly used vasopressor-inotropes (adrenaline, dopamine and noradrenaline) by comparing the responses to those of nonpharmacologically induced increases in blood pressure. We also searched for reasons for a mismatch between the response in perfusion and oxygenation. Methods Twenty-four piglets had long and short infusions of the three vasopressor-inotropes titrated to raise mean arterial blood pressure (MAP) 10 mmHg in random order. Nonpharmacological increases in MAP were induced by inflation of a balloon in the descending aorta. We measured cerebral oxygenation (near-infrared spectroscopy), perfusion (laser-Doppler), oxygen consumption (co-oximetry of arterial and superior sagittal sinus blood), and microvascular heterogeneity (side stream dark field video microscopy). Results Vasopressor-inotropes increased cerebral oxygenation significantly less (p≤0.01) compared to non-pharmacological MAP increases, whereas perfusion was similar. Furthermore, cerebral total hemoglobin concentration increased significantly less during vasopressor-inotrope infusions (p = 0.001). These physiologic responses were identical between the three vasopressor-inotropes (p>0.05). Furthermore, they induced a mild, although insignificant increase in cerebral metabolism and microvascular heterogeneity (p>0.05). Removal of the scalp tissue did not influence the mismatch (p>0.05). Conclusion We demonstrated a moderate vasopressor-inotrope induced mismatch between cerebral perfusion and oxygenation. Scalp removal did not affect this mismatch, why vasopressor-inotropes appear to have direct cerebral actions. The statistically nonsignificant increases in cerebral metabolism and/or microvascular heterogeneity may explain the mismatch. Alternatively, it may simply reflect a vasopressor-inotrope-induced decrease in the arterial-to-venous volume ratio as detected by near-infrared spectroscopy.
Collapse
Affiliation(s)
- Gitte H Hahn
- Department of Neonatology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.
| | | | | | | | | |
Collapse
|
72
|
Neuroprotective effect of melatonin: a novel therapy against perinatal hypoxia-ischemia. Int J Mol Sci 2013; 14:9379-95. [PMID: 23629670 PMCID: PMC3676788 DOI: 10.3390/ijms14059379] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 01/01/2023] Open
Abstract
One of the most common causes of mortality and morbidity in children is perinatal hypoxia-ischemia (HI). In spite of the advances in neonatology, its incidence is not diminishing, generating a pediatric population that will require an extended amount of chronic care throughout their lifetime. For this reason, new and more effective neuroprotective strategies are urgently required, in order to minimize as much as possible the neurological consequences of this encephalopathy. In this sense, interest has grown in the neuroprotective possibilities of melatonin, as this hormone may help to maintain cell survival through the modulation of a wide range of physiological functions. Although some of the mechanisms by which melatonin is neuroprotective after neonatal asphyxia remain a subject of investigation, this review tries to summarize some of the most recent advances related with its use as a therapeutic drug against perinatal hypoxic-ischemic brain injury, supporting the high interest in this indoleamine as a future feasible strategy for cerebral asphyctic events.
Collapse
|
73
|
Cerio FGD, Lara-Celador I, Alvarez A, Hilario E. Neuroprotective therapies after perinatal hypoxic-ischemic brain injury. Brain Sci 2013; 3:191-214. [PMID: 24961314 PMCID: PMC4061821 DOI: 10.3390/brainsci3010191] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/13/2013] [Accepted: 02/22/2013] [Indexed: 12/29/2022] Open
Abstract
Hypoxic-ischemic (HI) brain injury is one of the main causes of disabilities in term-born infants. It is the result of a deprivation of oxygen and glucose in the neural tissue. As one of the most important causes of brain damage in the newborn period, the neonatal HI event is a devastating condition that can lead to long-term neurological deficits or even death. The pattern of this injury occurs in two phases, the first one is a primary energy failure related to the HI event and the second phase is an energy failure that takes place some hours later. Injuries that occur in response to these events are often manifested as severe cognitive and motor disturbances over time. Due to difficulties regarding the early diagnosis and treatment of HI injury, there is an increasing need to find effective therapies as new opportunities for the reduction of brain damage and its long term effects. Some of these therapies are focused on prevention of the production of reactive oxygen species, anti-inflammatory effects, anti-apoptotic interventions and in a later stage, the stimulation of neurotrophic properties in the neonatal brain which could be targeted to promote neuronal and oligodendrocyte regeneration.
Collapse
Affiliation(s)
- Felipe Goñi de Cerio
- Biotechnology Area, GAIKER Technology Centre, Parque Tecnológico de Zamudio Ed 202, 48170 Zamudio, Vizcaya, Spain.
| | - Idoia Lara-Celador
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48949 Leioa, Bizkaia, Spain.
| | - Antonia Alvarez
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48949 Leioa, Bizkaia, Spain.
| | - Enrique Hilario
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48949 Leioa, Bizkaia, Spain.
| |
Collapse
|
74
|
Hypoxia-ischemia alters nucleotide and nucleoside catabolism and Na+,K+-ATPase activity in the cerebral cortex of newborn rats. Neurochem Res 2013; 38:886-94. [PMID: 23397287 DOI: 10.1007/s11064-013-0994-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 01/22/2013] [Accepted: 01/29/2013] [Indexed: 12/21/2022]
Abstract
It is well known that the levels of adenosine in the brain increase dramatically during cerebral hypoxic-ischemic (HI) insults. Its levels are tightly regulated by physiological and pathophysiological changes that occur during the injury acute phase. The aim of the present study was to examine the effects of the neonatal HI event on cytosolic and ecto-enzymes of purinergic system--NTPDase, 5'-nucleotidase (5'-NT) and adenosine deaminase (ADA)--in cerebral cortex of rats immediately post insult. Furthermore, the Na(+)/K(+)-ATPase activity, adenosine kinase (ADK) expression and thiobarbituric acid reactive species (TBARS) levels were assessed. Immediately after the HI event the cytosolic NTPDase and 5'-NT activities were increased in the cerebral cortex. In synaptosomes there was an increase in the ecto-ADA activity while the Na(+)/K(+) ATPase activity presented a decrease. The difference between ATP, ADP, AMP and adenosine degradation in synaptosomal and cytosolic fractions could indicate that NTPDase, 5'-NT and ADA were differently affected after insult. Interestingly, no alterations in the ADK expression were observed. Furthermore, the Na(+)/K(+)-ATPase activity was correlated negatively with the cytosolic NTPDase activity and TBARS content. The increased hydrolysis of nucleotides ATP, ADP and AMP in the cytosol could contribute to increased adenosine levels, which could be related to a possible innate neuroprotective mechanism aiming at potentiating the ambient levels of adenosine. Together, these results may help the understanding of the mechanism by which adenosine is produced following neonatal HI injury, therefore highlighting putative therapeutical targets to minimize ischemic injury and enhance recovery.
Collapse
|
75
|
Jellema RK, Lima Passos V, Zwanenburg A, Ophelders DRMG, De Munter S, Vanderlocht J, Germeraad WTV, Kuypers E, Collins JJP, Cleutjens JPM, Jennekens W, Gavilanes AWD, Seehase M, Vles HJ, Steinbusch H, Andriessen P, Wolfs TGAM, Kramer BW. Cerebral inflammation and mobilization of the peripheral immune system following global hypoxia-ischemia in preterm sheep. J Neuroinflammation 2013; 10:13. [PMID: 23347579 PMCID: PMC3614445 DOI: 10.1186/1742-2094-10-13] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 01/07/2013] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) is one of the most important causes of brain injury in preterm infants. Preterm HIE is predominantly caused by global hypoxia-ischemia (HI). In contrast, focal ischemia is most common in the adult brain and known to result in cerebral inflammation and activation of the peripheral immune system. These inflammatory responses are considered to play an important role in the adverse outcomes following brain ischemia. In this study, we hypothesize that cerebral and peripheral immune activation is also involved in preterm brain injury after global HI. METHODS Preterm instrumented fetal sheep were exposed to 25 minutes of umbilical cord occlusion (UCO) (n = 8) at 0.7 gestation. Sham-treated animals (n = 8) were used as a control group. Brain sections were stained for ionized calcium binding adaptor molecule 1 (IBA-1) to investigate microglial proliferation and activation. The peripheral immune system was studied by assessment of circulating white blood cell counts, cellular changes of the spleen and influx of peripheral immune cells (MPO-positive neutrophils) into the brain. Pre-oligodendrocytes (preOLs) and myelin basic protein (MBP) were detected to determine white matter injury. Electro-encephalography (EEG) was recorded to assess functional impairment by interburst interval (IBI) length analysis. RESULTS Global HI resulted in profound activation and proliferation of microglia in the hippocampus, periventricular and subcortical white matter. In addition, non-preferential mobilization of white blood cells into the circulation was observed within 1 day after global HI and a significant influx of neutrophils into the brain was detected 7 days after the global HI insult. Furthermore, global HI resulted in marked involution of the spleen, which could not be explained by increased splenic apoptosis. In concordance with cerebral inflammation, global HI induced severe brain atrophy, region-specific preOL vulnerability, hypomyelination and persistent suppressed brain function. CONCLUSIONS Our data provided evidence that global HI in preterm ovine fetuses resulted in profound cerebral inflammation and mobilization of the peripheral innate immune system. These inflammatory responses were paralleled by marked injury and functional loss of the preterm brain. Further understanding of the interplay between preterm brain inflammation and activation of the peripheral immune system following global HI will contribute to the development of future therapeutic interventions in preterm HIE.
Collapse
Affiliation(s)
- Reint K Jellema
- School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
| | - Valéria Lima Passos
- Department of Methodology and Statistics, Maastricht University, P. Debyeplein 1, Maastricht, 6229 HA, The Netherlands
| | - Alex Zwanenburg
- Department of Biomedical Technology, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
- Department of Clinical Physics, Maxima Medical Centre, De Run 4600, Veldhoven, 5504 DB, The Netherlands
| | - Daan RMG Ophelders
- School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
| | - Stephanie De Munter
- School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
| | - Joris Vanderlocht
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
| | - Wilfred TV Germeraad
- Department of Internal Medicine, Division of Haematology, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
| | - Elke Kuypers
- School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
| | - Jennifer JP Collins
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
| | - Jack PM Cleutjens
- Department of Pathology, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
| | - Ward Jennekens
- Neonatal Intensive Care Unit, Maxima Medical Centre, De Run 4600, Veldhoven, 5504 DB, The Netherlands
- Department of Clinical Physics, Maxima Medical Centre, De Run 4600, Veldhoven, 5504 DB, The Netherlands
| | - Antonio WD Gavilanes
- School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
| | - Matthias Seehase
- School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
| | - Hans J Vles
- Department of Child Neurology, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
| | - Harry Steinbusch
- School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
| | - Peter Andriessen
- Neonatal Intensive Care Unit, Maxima Medical Centre, De Run 4600, Veldhoven, 5504 DB, The Netherlands
- Department of Clinical Physics, Maxima Medical Centre, De Run 4600, Veldhoven, 5504 DB, The Netherlands
| | - Tim GAM Wolfs
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
| | - Boris W Kramer
- School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
| |
Collapse
|
76
|
Walsh BH, Broadhurst DI, Mandal R, Wishart DS, Boylan GB, Kenny LC, Murray DM. The metabolomic profile of umbilical cord blood in neonatal hypoxic ischaemic encephalopathy. PLoS One 2012; 7:e50520. [PMID: 23227182 PMCID: PMC3515614 DOI: 10.1371/journal.pone.0050520] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Accepted: 10/23/2012] [Indexed: 02/08/2023] Open
Abstract
Background Hypoxic ischaemic encephalopathy (HIE) in newborns can cause significant long-term neurological disability. The insult is a complex injury characterised by energy failure and disruption of cellular homeostasis, leading to mitochondrial damage. The importance of individual metabolic pathways, and their interaction in the disease process is not fully understood. The aim of this study was to describe and quantify the metabolomic profile of umbilical cord blood samples in a carefully defined population of full-term infants with HIE. Methods and Findings The injury severity was defined using both the modified Sarnat score and continuous multichannel electroencephalogram. Using these classification systems, our population was divided into those with confirmed HIE (n = 31), asphyxiated infants without encephalopathy (n = 40) and matched controls (n = 71). All had umbilical cord blood drawn and biobanked at −80°C within 3 hours of delivery. A combined direct injection and LC-MS/MS assay (AbsolutIDQ p180 kit, Biocrates Life Sciences AG, Innsbruck, Austria) was used for the metabolomic analyses of the samples. Targeted metabolomic analysis showed a significant alteration between study groups in 29 metabolites from 3 distinct classes (Amino Acids, Acylcarnitines, and Glycerophospholipids). 9 of these metabolites were only significantly altered between neonates with Hypoxic ischaemic encephalopathy and matched controls, while 14 were significantly altered in both study groups. Multivariate Discriminant Analysis models developed showed clear multifactorial metabolite associations with both asphyxia and HIE. A logistic regression model using 5 metabolites clearly delineates severity of asphyxia and classifies HIE infants with AUC = 0.92. These data describe wide-spread disruption to not only energy pathways, but also nitrogen and lipid metabolism in both asphyxia and HIE. Conclusion This study shows that a multi-platform targeted approach to metabolomic analyses using accurately phenotyped and meticulously biobanked samples provides insight into the pathogenesis of perinatal asphyxia. It highlights the potential for metabolomic technology to develop a diagnostic test for HIE.
Collapse
Affiliation(s)
- Brian H Walsh
- Neonatal Brain Research Group, Department of Paediatrics and Child Health, Cork University Maternity Hospital, Wilton, Cork, Ireland.
| | | | | | | | | | | | | |
Collapse
|
77
|
Baburamani AA, Ek CJ, Walker DW, Castillo-Melendez M. Vulnerability of the developing brain to hypoxic-ischemic damage: contribution of the cerebral vasculature to injury and repair? Front Physiol 2012; 3:424. [PMID: 23162470 PMCID: PMC3493883 DOI: 10.3389/fphys.2012.00424] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 10/17/2012] [Indexed: 11/13/2022] Open
Abstract
As clinicians attempt to understand the underlying reasons for the vulnerability of different regions of the developing brain to injury, it is apparent that little is known as to how hypoxia-ischemia may affect the cerebrovasculature in the developing infant. Most of the research investigating the pathogenesis of perinatal brain injury following hypoxia-ischemia has focused on excitotoxicity, oxidative stress and an inflammatory response, with the response of the developing cerebrovasculature receiving less attention. This is surprising as the presentation of devastating and permanent injury such as germinal matrix-intraventricular haemorrhage (GM-IVH) and perinatal stroke are of vascular origin, and the origin of periventricular leukomalacia (PVL) may also arise from poor perfusion of the white matter. This highlights that cerebrovasculature injury following hypoxia could primarily be responsible for the injury seen in the brain of many infants diagnosed with hypoxic-ischemic encephalopathy (HIE). Interestingly the highly dynamic nature of the cerebral blood vessels in the fetus, and the fluctuations of cerebral blood flow and metabolic demand that occur following hypoxia suggest that the response of blood vessels could explain both regional protection and vulnerability in the developing brain. However, research into how blood vessels respond following hypoxia-ischemia have mostly been conducted in adult models of ischemia or stroke, further highlighting the need to investigate how the developing cerebrovasculature responds and the possible contribution to perinatal brain injury following hypoxia. This review discusses the current concepts on the pathogenesis of perinatal brain injury, the development of the fetal cerebrovasculature and the blood brain barrier (BBB), and key mediators involved with the response of cerebral blood vessels to hypoxia.
Collapse
Affiliation(s)
- Ana A Baburamani
- The Ritchie Centre, Monash Medical Centre, Monash Institute of Medical Research, Clayton Melbourne, VIC, Australia ; Sahlgrenska Academy, Gothenburg University Göteborg, Sweden
| | | | | | | |
Collapse
|
78
|
Alonso-Alconada D, Hilario E, Álvarez FJ, Álvarez A. Apoptotic cell death correlates with ROS overproduction and early cytokine expression after hypoxia-ischemia in fetal lambs. Reprod Sci 2012; 19:754-63. [PMID: 22378862 DOI: 10.1177/1933719111432868] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite advances in neonatology, the hypoxic-ischemic injury in the perinatal period remains the single most important cause of brain injury in the newborn, leading to death or lifelong sequelae. Using a sheep model of intrauterine asphyxia, we evaluated the correlation between reactive oxygen species (ROS) overproduction, cytokine expression, and apoptotic cell death. Fetal lambs were assigned to sham group, nonasphyctic animals; and hypoxia-ischemia (HI) group, lambs subjected to 60 minutes of HI) by partial cord occlusion and sacrificed 3 hours later. Different brain regions were separated to quantify the number of apoptotic cells and the same territories were dissociated for flow cytometry studies. Our results suggest that the overproduction of ROS and the early increase in cytokine production after HI in fetal lambs correlate in a significant manner with the apoptotic index, as well as with each brain region evaluated.
Collapse
Affiliation(s)
- Daniel Alonso-Alconada
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, Leioa, Vizcaya, Spain.
| | | | | | | |
Collapse
|
79
|
Lehner C, Gehwolf R, Tempfer H, Krizbai I, Hennig B, Bauer HC, Bauer H. Oxidative stress and blood-brain barrier dysfunction under particular consideration of matrix metalloproteinases. Antioxid Redox Signal 2011; 15:1305-23. [PMID: 21294658 PMCID: PMC6464004 DOI: 10.1089/ars.2011.3923] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A cell's "redox" (oxidation and reduction) state is determined by the sum of all redox processes yielding reactive oxygen species (ROS), reactive nitrogen species (RNS), and other reactive intermediates. Low amounts of ROS/RNS are generated by different mechanisms in every cell and are important regulatory mediators in many signaling processes (redox signaling). When the physiological balance between the generation and elimination of ROS/RNS is disrupted, oxidative/nitrosative stress with persistent oxidative damage of the organism occurs. Oxidative stress has been suggested to act as initiator and/or mediator of many human diseases. The cerebral vasculature is particularly susceptible to oxidative stress, which is critical since cerebral endothelial cells play a major role in the creation and maintenance of the blood-brain barrier (BBB). This article will only contain a focused introduction on the biochemical background of redox signaling, since this has been reported already in a series of excellent recent reviews. The goal of this work is to increase the understanding of basic mechanisms underlying ROS/RNS-induced BBB disruption, with a focus on the role of matrix metalloproteinases, which, after all, appear to be a key mediator in the initiation and progression of BBB damage elicited by oxidative stress.
Collapse
Affiliation(s)
- Christine Lehner
- Department of Organismic Biology, Development Biology Group, University Hospital of Salzburg, Salzburg, Austria
| | | | | | | | | | | | | |
Collapse
|
80
|
Elabiad MT, Hook RE. Mercury content of blood transfusions for infants with extremely low birth weight. Pediatrics 2011; 128:331-4. [PMID: 21746718 DOI: 10.1542/peds.2010-3712] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES To assess whether blood transfusions for infants with birth weights of 1000 g or less are a source of mercury and whether any mercury delivered through the transfusion is above the currently set oral reference dose. PATIENTS AND METHODS We studied an observational cohort. Inclusion criteria included birth weight 1000 g or less and receipt of 1 or more packed red blood cell transfusions. Packed red blood cell units were tested prospectively for mercury levels. The quantity of transfused mercury was calculated on the basis of transfused volume and packed red blood cell mercury level. The resulting mercury level was compared with the reference dose as set by the Agency for Toxic Substances and Disease Registry, the World Health Organization, and the US Environmental Protection Agency. RESULTS Thirty-seven infants (birth weight: 736 ± 157 g; gestational age: 25.5 ± 1.5 weeks) met the inclusion criteria. A total of 325 transfusions from 49 packed red blood cell units were administered. Mercury was detected in 40 units. The average mercury level in a packed red blood cell unit was 1.9 ± 2.6 μg/L (median: 0.9 μg/L [interquartile range: 0.3-2.5]). None of the infants received any mercury above the reference dose set by the Agency for Toxic Substances and Disease Registry and the World Health Organization. Twelve infants received 1 transfusion, and 5 infants received 2 transfusions above the Environmental Protection Agency reference dose during their entire hospitalization. CONCLUSIONS Packed red blood cells are a source of mercury for infants. However, the amount delivered is low compared with currently set safety levels. The episodes in which mercury intake exceeded the reference dose were rare. However, without long-term follow-up, no conclusions can be made about the cognitive implications of these episodes.
Collapse
Affiliation(s)
- Mohamad T Elabiad
- Division of Neonatology, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| | | |
Collapse
|
81
|
Morales P, Bustamante D, Espina-Marchant P, Neira-Peña T, Gutiérrez-Hernández MA, Allende-Castro C, Rojas-Mancilla E. Pathophysiology of perinatal asphyxia: can we predict and improve individual outcomes? EPMA J 2011. [PMID: 23199150 PMCID: PMC3405380 DOI: 10.1007/s13167-011-0100-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Perinatal asphyxia occurs still with great incidence whenever delivery is prolonged, despite improvements in perinatal care. After asphyxia, infants can suffer from short- to long-term neurological sequelae, their severity depend upon the extent of the insult, the metabolic imbalance during the re-oxygenation period and the developmental state of the affected regions. Significant progresses in understanding of perinatal asphyxia pathophysiology have achieved. However, predictive diagnostics and personalised therapeutic interventions are still under initial development. Now the emphasis is on early non-invasive diagnosis approach, as well as, in identifying new therapeutic targets to improve individual outcomes. In this review we discuss (i) specific biomarkers for early prediction of perinatal asphyxia outcome; (ii) short and long term sequelae; (iii) neurocircuitries involved; (iv) molecular pathways; (v) neuroinflammation systems; (vi) endogenous brain rescue systems, including activation of sentinel proteins and neurogenesis; and (vii) therapeutic targets for preventing or mitigating the effects produced by asphyxia.
Collapse
Affiliation(s)
- Paola Morales
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Diego Bustamante
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Pablo Espina-Marchant
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Tanya Neira-Peña
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Manuel A. Gutiérrez-Hernández
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Camilo Allende-Castro
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Edgardo Rojas-Mancilla
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| |
Collapse
|
82
|
Anderson V, Spencer-Smith M, Wood A. Do children really recover better? Neurobehavioural plasticity after early brain insult. Brain 2011; 134:2197-221. [PMID: 21784775 DOI: 10.1093/brain/awr103] [Citation(s) in RCA: 362] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Vicki Anderson
- Department of Psychology, Royal Children's Hospital, Parkville, Victoria 3052, Australia.
| | | | | |
Collapse
|
83
|
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: 3.7] [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
|
84
|
Greggio S, de Paula S, de Oliveira IM, Trindade C, Rosa RM, Henriques JAP, DaCosta JC. NAP prevents acute cerebral oxidative stress and protects against long-term brain injury and cognitive impairment in a model of neonatal hypoxia-ischemia. Neurobiol Dis 2011; 44:152-9. [PMID: 21757007 DOI: 10.1016/j.nbd.2011.06.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 06/17/2011] [Accepted: 06/26/2011] [Indexed: 10/18/2022] Open
Abstract
Hypoxia-ischemia (HI) is a common cause of neonatal brain damage with lifelong morbidities in which current therapies are limited. In this study, we investigated the effect of neuropeptide NAP (NAPVSIPQ) on early cerebral oxidative stress, long-term neurological function and brain injury after neonatal HI. Seven-day-old rat pups were subjected to an HI model by applying a unilateral carotid artery occlusion and systemic hypoxia. The animals were randomly assigned to groups receiving an intraperitoneal injection of NAP (3 μg/g) or vehicle immediately (0 h) and 24 h after HI. Brain DNA damage, lipid peroxidation and reduced glutathione (GSH) content were determined 24 h after the last NAP injection. Cognitive impairment was assessed on postnatal day 60 using the spatial version of the Morris water maze learning task. Next, the animals were euthanized to assess the cerebral hemispheric volume using the Cavalieri principle associated with the counting point method. We observed that NAP prevented the acute HI-induced DNA and lipid membrane damage and also recovered the GSH levels in the injured hemisphere of the HI rat pups. Further, NAP was able to prevent impairments in learning and long-term spatial memory and to significantly reduce brain damage up to 7 weeks following the neonatal HI injury. Our findings demonstrate that NAP confers potent neuroprotection from acute brain oxidative stress, long-term cognitive impairment and brain lesions induced by neonatal HI through, at least in part, the modulation of the glutathione-mediated antioxidant system.
Collapse
Affiliation(s)
- Samuel Greggio
- Laboratório de Neurociências, Instituto do Cérebro e Instituto de Pesquisas Biomédicas, Programa de Pós-Graduação em Pediatria e Saúde da Criança, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
85
|
Birth asphyxia as the major complication in newborns: moving towards improved individual outcomes by prediction, targeted prevention and tailored medical care. EPMA J 2011. [PMID: 23199149 PMCID: PMC3405378 DOI: 10.1007/s13167-011-0087-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Perinatal Asphyxia—oxygen deficit at delivery—can lead to severe hypoxic ischaemic organ damage in newborns followed by a fatal outcome or severe life-long pathologies. The severe insults often cause neurodegenerative diseases, mental retardation and epilepsies. The mild insults lead to so-called “minimal brain-damage disorders” such as attention deficits and hyperactivity, but can also be associated with the development of schizophrenia and life-long functional psychotic syndromes. Asphyxia followed by re-oxygenation can potentially lead to development of several neurodegenerative pathologies, diabetes type 2 and cancer. The task of individual prediction, targeted prevention and personalised treatments before a manifestation of the life-long chronic pathologies usually developed by newborns with asphyxic deficits, should be given the extraordinary priority in neonatology and paediatrics. Socio-economical impacts of educational measures and advanced strategies in development of robust diagnostic approaches targeted at effected molecular pathways, biomarker-candidates and potential drug-targets for tailored treatments are reviewed in the paper.
Collapse
|
86
|
Hydrogen-rich saline is cerebroprotective in a rat model of deep hypothermic circulatory arrest. Neurochem Res 2011; 36:1501-11. [PMID: 21512745 DOI: 10.1007/s11064-011-0476-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2011] [Indexed: 01/03/2023]
Abstract
Deep hypothermic circulatory arrest (DHCA) has been widely used in the operations involving the aortic arch and brain aneurysm since 1950s; but prolonged DHCA contributes significantly to neurological deficit which remains a major cause of postoperative morbidity and mortality. It has been reported that hydrogen exerts a therapeutic antioxidant activity by selectively reducing hydroxyl radical. In this study, DHCA treated rats developed a significant oxidative stress, inflammatory reaction and apoptosis. The administration of HRS resulted in a significant decrease in the brain injury, together with lower production of IL-1β, TNF-α, 8-OHdG and MDA as well as decreased activity of NOS while increased activity of SOD. The apoptotic index as well as the expressions of caspase-3 in brain tissue was significantly decreased after treatment. HRS administration significantly attenuated the severity of DHCA induced brain injury by mechanisms involving amelioration of oxidative stress, down-regulation of inflammatory factors and reduction of apoptosis.
Collapse
|
87
|
Solaroglu A, Suat Dede F, Gelisen O, Secilmis O, Dede H. Neuroprotective effect of magnesium sulfate treatment on fetal brain in experimental intrauterine ischemia reperfusion injury. J Matern Fetal Neonatal Med 2011; 24:1259-61. [DOI: 10.3109/14767058.2011.572202] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
88
|
Abstract
The developing brain is particularly vulnerable to reactive oxygen and reactive nitrogen species-mediated damage because of its high concentrations of unsaturated fatty acids, high rate of oxygen consumption, low concentrations of antioxidants, high content of metals catalyzing free radical formation, and large proportion of sensitive immature cells. In this review, we outline the dynamic changes of energy resources, metabolic requirements, and endogenous free radical scavenging systems during physiologic brain development. We further discuss the involvement of oxidative stress in the pathogenesis of neuronal death after exposure of the infant brain to hyperoxia, hypoxia/ischemia, sedative drugs, ethanol, and mechanical trauma. Several approaches have been developed to combat oxidative stress, but neuroprotective treatment strategies are limited in the clinical setting.
Collapse
Affiliation(s)
- Chrysanthy Ikonomidou
- Department of Neurology, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792, USA.
| | | |
Collapse
|
89
|
Affiliation(s)
- Susan Givens Bell
- College of Nursing, University of South Florida, Tampa, Florida, USA.
| |
Collapse
|
90
|
Weis SN, Schunck RVA, Pettenuzzo LF, Krolow R, Matté C, Manfredini V, do Carmo R Peralba M, Vargas CR, Dalmaz C, Wyse ATS, Netto CA. Early biochemical effects after unilateral hypoxia-ischemia in the immature rat brain. Int J Dev Neurosci 2011; 29:115-20. [PMID: 21255637 DOI: 10.1016/j.ijdevneu.2010.12.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 11/25/2010] [Accepted: 12/26/2010] [Indexed: 11/30/2022] Open
Abstract
Perinatal hypoxia-ischemia (HI) gives rise to inadequate substrate supply to the brain tissue, resulting in damage to neural cells. Previous studies at different time points of development, and with different animal species, suggest that the HI insult causes oxidative damage and changes Na+, K+-ATPase activity, which is known to be very susceptible to free radical-related lipid peroxidation. The aim of the present study was to establish the onset of the oxidative damage response in neonatal Wistar rats subjected to brain HI, evaluating parameters of oxidative stress, namely nitric oxide production, lipoperoxidation by thiobarbituric acid reactive substances (TBA-RS) production and malondialdehyde (MDA) levels, reactive species production by DCFH oxidation, antioxidant enzymatic activities of catalase, glutathione peroxidase, superoxide dismutase as well as Na+, K+-ATPase activity in hippocampus and cerebral cortex. Rat pups were subjected to right common carotid ligation followed by exposure to a hypoxic atmosphere (8% oxygen and 92% nitrogen) for 90 min. Animals were sacrificed by decapitation 0, 1 and 2 h after HI and both hippocampus and cerebral cortex from the right hemisphere (ipsilateral to the carotid occlusion) were dissected out for further experimentation. Results show an early decrease of Na+, K+-ATPase activity (at 0 and 1 h), as well as a late increase in MDA levels (2 h) and superoxide dismutase activity (1 and 2 h after HI) in the hippocampus. There was a late increase in both MDA levels and DCFH oxidation (1 and 2 h) and an increase in superoxide dismutase activity (2 h after HI) in cortex; however Na+, K+-ATPase activity remained unchanged. We suggest that neonatal HI induces oxidative damage to both hippocampus and cortex, in addition to a decrease in Na+, K+-ATPase activity in hippocampus early after the insult. These events might contribute to the later morphological damage in the brain and indicate that it would be essential to pursue neuroprotective strategies, aimed to counteract oxidative stress, as early as possible after the HI insult.
Collapse
Affiliation(s)
- Simone N Weis
- Programa de Pós-Graduação em Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Jiao L, Zhang J, Li Z, Liu H, Chen Y, Xu S. Edaravone alleviates delayed neuronal death and long-dated cognitive dysfunction of hippocampus after transient focal ischemia in Wistar rat brains. Neuroscience 2011; 182:177-83. [PMID: 21241778 DOI: 10.1016/j.neuroscience.2011.01.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 01/06/2011] [Accepted: 01/07/2011] [Indexed: 01/02/2023]
Abstract
Edaravone is currently being used in acute ischemic stroke both in clinical and experimental research as a potent antioxidant. Here we explore the effects of edaravone on delayed neuronal death (DND) and long-dated cognitive dysfunction of hippocampus after cerebral ischemia-reperfusion (IR) injury and explain the underlying mechanisms and pathways. Our findings suggested that edaravone not only significantly alleviated delayed neuronal death and cognitive dysfunction of hippocampus after cerebral focal ischemia, but also markedly decreased malondialdehyde (MDA) levels. In addition, edaravone increased superoxide dismutase (SOD) levels and reduced the levels of inflammatory cytokines such as IL-1β and TNF-α expression; edaravone, also suppressed glial fibrillary acidic protein (GFAP) proliferation at days 3, 7 and 30 after reperfusion. Overall, the consensus emerging from this body of data indicated that edaravone exerts a later neuroprotective effect to hippocampus through its ability to inhibit inflammation, suppression of astrocyte activation and scavenging free radicals in stroke events.
Collapse
Affiliation(s)
- L Jiao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | | | | | | | | | | |
Collapse
|
92
|
Cooper DJ. Induced hypothermia for neonatal hypoxic-ischemic encephalopathy: pathophysiology, current treatment, and nursing considerations. Neonatal Netw 2011; 30:29-35. [PMID: 21317095 DOI: 10.1891/0730-0832.30.1.29] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) can lead to devastating neurodevelopmental consequences such as cerebral palsy, seizure disorders, and significant developmental delays. HIE in the newborn is often the result of a hypoxic event, such as uterine rupture, placental abruption, or cord prolapse. Biphasic brain injury occurs in HIE. The first phase involves activation of the sympathetic nervous system as a compensatory mechanism. The second phase, known as reperfusion brain injury, occurs hours later. Induced hypothermia, a neuroprotective strategy for treating HIE, targets the second phase to prevent reperfusion injury. NICU nurses are in a unique position to detect patient instability and to maintain the therapeutic interventions that contribute to the healing process. This article highlights the significant role nurses play in the management of infants diagnosed with HIE who are treated with induced hypothermia.
Collapse
|
93
|
Distefano G, Praticò AD. Actualities on molecular pathogenesis and repairing processes of cerebral damage in perinatal hypoxic-ischemic encephalopathy. Ital J Pediatr 2010; 36:63. [PMID: 20846380 PMCID: PMC2954868 DOI: 10.1186/1824-7288-36-63] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 09/16/2010] [Indexed: 11/12/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is the most important cause of cerebral damage and long-term neurological sequelae in the perinatal period both in term and preterm infant. Hypoxic-ischemic (H-I) injuries develop in two phases: the ischemic phase, dominated by necrotic processes, and the reperfusion phase, dominated by apoptotic processes extending beyond ischemic areas. Due to selective ischemic vulnerability, cerebral damage affects gray matter in term newborns and white matter in preterm newborns with the typical neuropathological aspects of laminar cortical necrosis in the former and periventricular leukomalacia in the latter. This article summarises the principal physiopathological and biochemical processes leading to necrosis and/or apoptosis of neuronal and glial cells and reports recent insights into some endogenous and exogenous cellular and molecular mechanisms aimed at repairing H-I cerebral damage.
Collapse
Affiliation(s)
- Giuseppe Distefano
- Department of Pediatrics, Division of Neonatology, University of Catania, Catania, Italy.
| | | |
Collapse
|
94
|
Assessment of uric acid and lipid peroxidation in serum and urine after hypoxia–ischemia neonatal in rats. Neurol Sci 2010; 32:59-65. [DOI: 10.1007/s10072-010-0393-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 07/23/2010] [Indexed: 01/29/2023]
|
95
|
Fan X, Heijnen CJ, van der Kooij MA, Groenendaal F, van Bel F. The role and regulation of hypoxia-inducible factor-1alpha expression in brain development and neonatal hypoxic-ischemic brain injury. ACTA ACUST UNITED AC 2009; 62:99-108. [PMID: 19786048 DOI: 10.1016/j.brainresrev.2009.09.006] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Revised: 09/17/2009] [Accepted: 09/19/2009] [Indexed: 01/10/2023]
Abstract
During neonatal hypoxic-ischemic brain injury, activation of transcription of a series of genes is induced to stimulate erythropoiesis, anti-apoptosis, apoptosis, necrosis and angiogenesis. A key factor mediating these gene transcriptions is hypoxia-inducible factor-1alpha (HIF-1alpha). During hypoxia, HIF-1alpha protein is stabilized and heterodimerizes with HIF-1beta to form HIF-1, subsequently regulating the expression of target genes. HIF-1alpha participates in early brain development and proliferation of neuronal precursor cells. Under pathological conditions, HIF-1alpha is known to play an important role in neonatal hypoxic-ischemic brain injury: on the one hand, HIF-1alpha has neuroprotective effects whereas it can also have neurotoxic effects. HIF-1alpha regulates the transcription of erythropoietin (EPO), which induces several pathways associated with neuroprotection. HIF-1alpha also promotes the expression of vascular endothelial cell growth factor (VEGF), which is related to neovascularization in hypoxic-ischemic brain areas. In addition, HIF-1alpha has an anti-apoptotic effect by increasing the expression of anti-apoptotic factors such as EPO during mild hypoxia. The neurotoxic effects of HIF-1alpha are represented by its participation in the apoptotic process by increasing the stability of the tumor suppressor protein p53 during severe hypoxia. Moreover, HIF-1alpha plays a role in cell necrosis, by interacting with calcium and calpain. HIF-1alpha can also exacerbate brain edema via increasing the permeability of the blood-brain barrier (BBB). Given these properties, HIF-1alpha has both neuroprotective and neurotoxic effects after hypoxia-ischemia. These events are cell type specific and related to the severity of hypoxia. Unravelling of the complex functions of HIF-1alpha may be important when designing neuroprotective therapies for hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Xiyong Fan
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan, The Netherlands
| | | | | | | | | |
Collapse
|
96
|
The Effect of Inhaled Nitric Oxide on the Course of Extracorporeal Membrane Oxygenation and the Occurrence of Hemorrhagic Complications. ASAIO J 2009; 55:213-6. [DOI: 10.1097/mat.0b013e31819901a5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
97
|
Yan EB, Baburamani AA, Walker AM, Walker DW. Changes in cerebral blood flow, cerebral metabolites, and breathing movements in the sheep fetus following asphyxia produced by occlusion of the umbilical cord. Am J Physiol Regul Integr Comp Physiol 2009; 297:R60-9. [PMID: 19403864 DOI: 10.1152/ajpregu.00047.2009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Severe global fetal asphyxia, if caused by a brief occlusion of the umbilical cord, results in prolonged cerebral hypoperfusion in fetal sheep. In this study, we sought evidence to support the hypothesis that cerebral hypoperfusion is a consequence of suppressed cerebral metabolism. In the 24 h following complete occlusion of the umbilical cord for 10 min, sagittal sinus blood flow velocity was significantly decreased for up to 12 h. Capillary blood flow, measured using microspheres, decreased at 1 and 5 h after cord occlusion in many brain regions, including cortical gray and white matter. Microdialysis probes implanted in the cerebral cortex revealed an increase in extracellular glucose concentrations in gray matter for 7-8 h postasphyxia, while lactate increased only briefly, suggesting decreased cerebral glucose utilization over this time. Although these data, as well as the concurrent suppression of breathing movements and electrocortical activity, support the concept of hypometabolic hypoperfusion, the significant increase of pyruvate and glycerol concentrations in dialysate fluid obtained from the cerebral cortex at 3-8 h after cord occlusion suggests an eventual loss of membrane integrity. The prolonged increase of breathing movements for many hours suggests loss of the pontine/thalamic control that produces the distinct pattern of fetal breathing movements.
Collapse
Affiliation(s)
- Edwin B Yan
- National Trauma Research Institute, The Alfred Hospital, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
98
|
Hydrogen gas is ineffective in moderate and severe neonatal hypoxia–ischemia rat models. Brain Res 2009; 1259:90-7. [DOI: 10.1016/j.brainres.2008.12.066] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 12/10/2008] [Accepted: 12/12/2008] [Indexed: 01/26/2023]
|
99
|
Giza CC, Kolb B, Harris NG, Asarnow RF, Prins ML. Hitting a moving target: Basic mechanisms of recovery from acquired developmental brain injury. Dev Neurorehabil 2009; 12:255-68. [PMID: 19956795 PMCID: PMC2772114 DOI: 10.3109/17518420903087558] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Acquired brain injuries represent a major cause of disability in the pediatric population. Understanding responses to developmental acquired brain injuries requires knowledge of the neurobiology of normal development, age-at-injury effects and experience-dependent neuroplasticity. In the developing brain, full recovery cannot be considered as a return to the premorbid baseline, since ongoing maturation means that cerebral functioning in normal individuals will continue to advance. Thus, the recovering immature brain has to 'hit a moving target' to achieve full functional recovery, defined as parity with age-matched uninjured peers. This review will discuss the consequences of developmental injuries such as focal lesions, diffuse hypoxia and traumatic brain injury (TBI). Underlying cellular and physiological mechanisms relevant to age-at-injury effects will be described in considerable detail, including but not limited to alterations in neurotransmission, connectivity/network functioning, the extracellular matrix, response to oxidative stress and changes in cerebral metabolism. Finally, mechanisms of experience-dependent plasticity will be reviewed in conjunction with their effects on neural repair and recovery.
Collapse
Affiliation(s)
- Christopher C Giza
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA.
| | | | | | | | | |
Collapse
|