1
|
Nikpour F, Salimi A, Saghazadeh A, Rezaei N. Blood and CSF levels of brain-derived neurotrophic factor in patients with encephalopathy/encephalitis: a systematic review and meta-analysis. Acta Neurol Belg 2024; 124:533-542. [PMID: 38267724 DOI: 10.1007/s13760-023-02442-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 11/20/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is critical for enhancing the survival and growth of neurons and modulating the synaptic plasticity. BDNF levels have been demonstrated to be changed in plasma and cerebrospinal fluid (CSF) following brain insults such as inflammation or ischemia or infection in several studies. Currently, there is no systematic review regarding BDNF levels in encephalitis or encephalopathy patients. Considering inconsistency between studies, we aimed to pool the data from existing studies to determine whether blood or CSF levels of BDNF are different in patients with encephalopathy/encephalitis. METHODS We comprehensively searched Web of Science, PubMed, Scopus, and Embase databases to identify eligible studies. The last search occurred in December 2022. RESULTS 12 studies met our inclusion criteria and ten studies including 283 patients and 323 healthy controls were enrolled in this meta-analysis. In comparison to controls, patients with encephalitis/encephalopathy had higher levels of BDNF in their CSF [standardized mean difference (SMD) = 1.48, 95% CI 0.18-2.77; P = 0.03)], while their blood levels of BDNF did not differ significantly [standardized mean difference (SMD) = 0.27, 95% CI = - 0.71 to 1.25; P = 0.58)]. Moreover, regarding the heterogeneity among studies reporting BDNF blood levels, we performed two subgroup analyses based on the disease etiology and the specimen (plasma and serum); none of them indicated statistically significant difference in BDNF levels between the subgroups (P = 0.41 and 0.20, respectively). CONCLUSION Meta-analysis provides evidence that patients with encephalopathy/encephalitis have higher CSF levels of BDNF compared to controls.
Collapse
Affiliation(s)
- Fatemeh Nikpour
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amir Salimi
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amene Saghazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
2
|
Gelen V, Özkanlar S, Kara A, Yeşildağ A. Citrate-coated silver nanoparticles loaded with agomelatine provide neuronal therapy in acute cerebral ischemia/reperfusion of rats by inhibiting the oxidative stress, endoplasmic reticulum stress, and P2X7 receptor-mediated inflammasome. ENVIRONMENTAL TOXICOLOGY 2024; 39:1531-1543. [PMID: 38009636 DOI: 10.1002/tox.24021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
Cerebral ischemia and reperfusion are related to various situations like injuries after various traumas, oxidative stress, increased calcium ion, capillary hypoperfusion, microvascular hyperpermeability, leukocyte infiltration, and blood-brain barrier disruption. An antidepressant Agomelatine which is a melatonin receptor (MT1/MT2) agonist and serotonin receptor (5-HT2C) antagonist has been reported by studies to have antioxidant and anti-inflammatory effects. In our study, we aimed to detect the effects of citrate-coated silver nanoparticle-loaded agomelatine application on neurodegeneration, endoplasmic reticulum stress, autophagic and apoptotic cell death, inflammation, and P2X7R expression in the cerebral ischemia-reperfusion model to facilitate the passage of blood-brain barrier. Forty two Sprague-Dawley rats in total were divided into six equal groups (n:7) and applications were performed. Acute cerebral injury in the ischemia-reperfusion model was created 2 h after internal carotid artery ligation in rats and then at the 2nd hour of reperfusion citrate-coated silver nanoparticles loaded with Agomelatine were applied. Twenty four hours later, neurologic analysis on animals in experimental groups was performed, animals were decapitated and GSH, GPx, SOD, CAT, MDA, IL-1β, and TNF-α parameters were examined after taking blood and the cerebral tissue samples. As a result, it was determined that ischemia-reperfusion caused endoplasmic reticulum stress in the cerebral tissues and thus caused cellular injury.
Collapse
Affiliation(s)
- Volkan Gelen
- Department of Physiology, Faculty of Veterinary Medicine, Kafkas University, Kars, Turkey
| | - Seçkin Özkanlar
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Adem Kara
- Department of Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Turkey
| | - Ali Yeşildağ
- Department of Bioengineering, Faculty of Engineering and Architecture, Kafkas University, Kars, Turkey
| |
Collapse
|
3
|
Bilchak JN, Caron G, Côté MP. Exercise-Induced Plasticity in Signaling Pathways Involved in Motor Recovery after Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms22094858. [PMID: 34064332 PMCID: PMC8124911 DOI: 10.3390/ijms22094858] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) leads to numerous chronic and debilitating functional deficits that greatly affect quality of life. While many pharmacological interventions have been explored, the current unsurpassed therapy for most SCI sequalae is exercise. Exercise has an expansive influence on peripheral health and function, and by activating the relevant neural pathways, exercise also ameliorates numerous disorders of the central nervous system (CNS). While the exact mechanisms by which this occurs are still being delineated, major strides have been made in the past decade to understand the molecular underpinnings of this essential treatment. Exercise rapidly and prominently affects dendritic sprouting, synaptic connections, neurotransmitter production and regulation, and ionic homeostasis, with recent literature implicating an exercise-induced increase in neurotrophins as the cornerstone that binds many of these effects together. The field encompasses vast complexity, and as the data accumulate, disentangling these molecular pathways and how they interact will facilitate the optimization of intervention strategies and improve quality of life for individuals affected by SCI. This review describes the known molecular effects of exercise and how they alter the CNS to pacify the injury environment, increase neuronal survival and regeneration, restore normal neural excitability, create new functional circuits, and ultimately improve motor function following SCI.
Collapse
|
4
|
Visco DB, Toscano AE, Juárez PAR, Gouveia HJCB, Guzman-Quevedo O, Torner L, Manhães-de-Castro R. A systematic review of neurogenesis in animal models of early brain damage: Implications for cerebral palsy. Exp Neurol 2021; 340:113643. [PMID: 33631199 DOI: 10.1016/j.expneurol.2021.113643] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
Brain damage during early life is the main factor in the development of cerebral palsy (CP), which is one of the leading neurodevelopmental disorders in childhood. Few studies, however, have focused on the mechanisms of cell proliferation, migration, and differentiation in the brain of individuals with CP. We thus conducted a systematic review of preclinical evidence of structural neurogenesis in early brain damage and the underlying mechanisms involved in the pathogenesis of CP. Studies were obtained from Embase, Pubmed, Scopus, and Web of Science. After screening 2329 studies, 29 studies, covering a total of 751 animals, were included. Prenatal models based on oxygen deprivation, inflammatory response and infection, postnatal models based on oxygen deprivation or hypoxic-ischemia, and intraventricular hemorrhage models showed varying neurogenesis responses according to the nature of the brain damage, the time period during which the brain injury occurred, proliferative capacity, pattern of migration, and differentiation profile in neurogenic niches. Results mainly from rodent studies suggest that prenatal brain damage impacts neurogenesis and curbs generation of neural stem cells, while postnatal models show increased proliferation of neural precursor cells, improper migration, and reduced survival of new neurons.
Collapse
Affiliation(s)
- Diego Bulcão Visco
- Post Graduate Program in Nutrition, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Ana Elisa Toscano
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Department of Nursing, CAV, Federal University of Pernambuco, Vitória de Santo Antão, Pernambuco, Brazil; Post Graduate Program in Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Pedro Alberto Romero Juárez
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Henrique José Cavalcanti Bezerra Gouveia
- Post Graduate Program in Nutrition, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Omar Guzman-Quevedo
- Instituto Tecnológico Superior de Tacámbaro, Tacámbaro, Michoacán, Mexico; Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico; Post Graduate Program in Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Raul Manhães-de-Castro
- Post Graduate Program in Nutrition, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil.
| |
Collapse
|
5
|
Sun C, Zhang AD, Chen HH, Bian J, Liu ZJ. Magnet-targeted delivery of bone marrow-derived mesenchymal stem cells improves therapeutic efficacy following hypoxic-ischemic brain injury. Neural Regen Res 2021; 16:2324-2329. [PMID: 33818519 PMCID: PMC8354132 DOI: 10.4103/1673-5374.310942] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Stem cell transplantation may represent a feasible therapeutic option for the recovery of neurological function in children with hypoxic-ischemic brain injury; however, the therapeutic efficacy of bone marrow-derived mesenchymal stem cells largely depends on the number of cells that are successfully transferred to the target. Magnet-targeted drug delivery systems can use a specific magnetic field to attract the drug to the target site, increasing the drug concentration. In this study, we found that the double-labeling using superparamagnetic iron oxide nanoparticle and poly-L-lysine (SPIO-PLL) of bone marrow-derived mesenchymal stem cells had no effect on cell survival but decreased cell proliferation 48 hours after labeling. Rat models of hypoxic-ischemic brain injury were established by ligating the left common carotid artery. One day after modeling, intraventricular and caudal vein injections of 1 × 105 SPIO-PLL-labeled bone marrow-derived mesenchymal stem cells were performed. Twenty-four hours after the intraventricular injection, magnets were fixed to the left side of the rats’ heads for 2 hours. Intravoxel incoherent motion magnetic resonance imaging revealed that the perfusion fraction and the diffusion coefficient of rat brain tissue were significantly increased in rats treated with SPIO-PLL-labeled cells through intraventricular injection combined with magnetic guidance, compared with those treated with SPIO-PLL-labeled cells through intraventricular or tail vein injections without magnetic guidance. Hematoxylin-eosin and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining revealed that in rats treated with SPIO-PLL-labeled cells through intraventricular injection under magnetic guidance, cerebral edema was alleviated, and apoptosis was decreased. These findings suggest that targeted magnetic guidance can be used to improve the therapeutic efficacy of bone marrow-derived mesenchymal stem cell transplantation for hypoxic-ischemic brain injury. This study was approved by the Animal Care and Use Committee of The Second Hospital of Dalian Medical University, China (approval No. 2016-060) on March 2, 2016.
Collapse
Affiliation(s)
- Chuang Sun
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Ao-Dan Zhang
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Hong-Hai Chen
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Jie Bian
- Department of Radiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Zheng-Juan Liu
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
6
|
Zaghloul N, Kurepa D, Bader MY, Nagy N, Ahmed MN. Prophylactic inhibition of NF-κB expression in microglia leads to attenuation of hypoxic ischemic injury of the immature brain. J Neuroinflammation 2020; 17:365. [PMID: 33261624 PMCID: PMC7709340 DOI: 10.1186/s12974-020-02031-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/10/2020] [Indexed: 11/29/2022] Open
Abstract
Background Periventricular leukomalacia (PVL), a devastating brain injury affecting premature infants, is the most common cause of cerebral palsy. PVL is caused by hypoxia ischemia (HI) and is characterized by white matter necrotic lesions, microglial activation, upregulation of NF-κB, and neuronal death. The microglia is the main cell involved in PVL pathogenesis. The goal of this study was to investigate the role of microglial NF-κB activity and its prophylactic inhibition in a neonate mouse model of HI. Methods Transgenic mice with specific knockout NF-κB in microglia and colony stimulating factor 1 receptor Cre with floxed IKKβ (CSF-1R Cre + IKKβflox/wt ) were used. Postnatal day 5 (P5) mice underwent sham or bilateral temporary carotid artery ligation followed by hypoxia. After HI insult, inflammatory cytokines, volumetric MRI, histopathology, and immunohistochemistry for oligodendroglia and microglial activation markers were analyzed. Long-term neurobehavioral assessment, including grip strength, rotarod, and open field testing, was performed at P60. Results We demonstrate that selective inhibition of NF-κB in microglia decreases HI-induced brain injury by decreasing microglial activation, proinflammatory cytokines, and nitrative stress. Rescue of oligodendroglia is evidenced by immunohistochemistry, decreased ventriculomegaly on MRI, and histopathology. This selective inhibition leads to attenuation of paresis, incoordination, and improved grip strength, gait, and locomotion. Conclusion We conclude that NF-κb activation in microglia plays a major role in the pathogenesis of hypoxic ischemic injury of the immature brain, and its prophylactic inhibition offers significant neuroprotection. Using a specific inhibitor of microglial NF-κB may offer a new prophylactic or therapeutic alternative in preterm infants affected by HI and possibly other neurological diseases in which microglial activation plays a role.
Collapse
Affiliation(s)
- Nahla Zaghloul
- Department of Pediatrics, Division of Neonatology, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ, USA.
| | - Dalibor Kurepa
- Department of Pediatrics, Division of Neonatology, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Mohammad Y Bader
- Department of Pediatrics, Division of Neonatology, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ, USA
| | - Nadia Nagy
- Department of Pediatrics, Division of Neonatology, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ, USA
| | - Mohamed N Ahmed
- Department of Pediatrics, Division of Neonatology, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ, USA
| |
Collapse
|
7
|
Zárraga-Granados G, Muciño-Hernández G, Sánchez-Carbente MR, Villamizar-Gálvez W, Peñas-Rincón A, Arredondo C, Andrés ME, Wood C, Covarrubias L, Castro-Obregón S. The nuclear receptor NR4A1 is regulated by SUMO modification to induce autophagic cell death. PLoS One 2020; 15:e0222072. [PMID: 32210435 PMCID: PMC7094859 DOI: 10.1371/journal.pone.0222072] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/08/2020] [Indexed: 12/31/2022] Open
Abstract
NR4A is a nuclear receptor protein family whose members act as sensors of cellular environment and regulate multiple processes such as metabolism, proliferation, migration, apoptosis, and autophagy. Since the ligand binding domains of these receptors have no cavity for ligand interaction, their function is most likely regulated by protein abundance and post-translational modifications. In particular, NR4A1 is regulated by protein abundance, phosphorylation, and subcellular distribution (nuclear-cytoplasmic translocation), and acts both as a transcription factor and as a regulator of other interacting proteins. SUMOylation is a post-translational modification that can affect protein stability, transcriptional activity, alter protein-protein interactions and modify intracellular localization of target proteins. In the present study we evaluated the role of SUMOylation as a posttranslational modification that can regulate the activity of NR4A1 to induce autophagy-dependent cell death. We focused on a model potentially relevant for neuronal cell death and demonstrated that NR4A1 needs to be SUMOylated to induce autophagic cell death. We observed that a triple mutant in SUMOylation sites has reduced SUMOylation, increased transcriptional activity, altered intracellular distribution, and more importantly, its ability to induce autophagic cell death is impaired.
Collapse
Affiliation(s)
- Gabriela Zárraga-Granados
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Gabriel Muciño-Hernández
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - María R. Sánchez-Carbente
- Biotechnology Research Center, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | - Wendy Villamizar-Gálvez
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Ana Peñas-Rincón
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Cristian Arredondo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María E. Andrés
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christopher Wood
- Laboratorio Nacional de Microscopía Avanzada, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico
| | - Luis Covarrubias
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico
| | - Susana Castro-Obregón
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
- * E-mail:
| |
Collapse
|
8
|
Intracellular Neuroprotective Mechanisms in Neuron-Glial Networks Mediated by Glial Cell Line-Derived Neurotrophic Factor. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1036907. [PMID: 31827666 PMCID: PMC6885812 DOI: 10.1155/2019/1036907] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 10/19/2019] [Indexed: 12/28/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) has a pronounced neuroprotective effect in various nervous system pathologies, including ischaemic brain damage and neurodegenerative diseases. In this work, we studied the effect of GDNF on the ultrastructure and functional activity of neuron-glial networks during acute hypoxic exposure, a key damaging factor in numerous brain pathologies. We analysed the molecular mechanisms most likely involved in the positive effects of GDNF. Hypoxia modelling was performed on day 14 of culturing primary hippocampal cells obtained from mouse embryos (E18). GDNF (1 ng/ml) was added to the culture medium 20 min before oxygen deprivation. Acute hypoxia-induced irreversible changes in the ultrastructure of neurons and astrocytes led to the loss of functional Сa2+ activity and neural network disruption. Destructive changes in the mitochondrial apparatus and its functional activity characterized by an increase in the basal oxygen consumption rate and respiratory chain complex II activity during decreased stimulated respiration intensity were observed 24 hours after hypoxic injury. At a concentration of 1 ng/ml, GDNF maintained the functional metabolic network activity in primary hippocampal cultures and preserved the structure of the synaptic apparatus and number of mature chemical synapses, confirming its neuroprotective effect. GDNF maintained the normal structure of mitochondria in neuronal outgrowth but not in the soma. Analysis of the possible GDNF mechanism revealed that RET kinase, a component of the receptor complex, and the PI3K/Akt pathway are crucial for the neuroprotective effect of GDNF. The current study also revealed the role of GDNF in the regulation of HIF-1α transcription factor expression under hypoxic conditions.
Collapse
|
9
|
Krüger A, Fabrizius A, Mikkelsen B, Siebert U, Folkow LP, Burmester T. Transcriptome analysis reveals a high aerobic capacity in the whale brain. Comp Biochem Physiol A Mol Integr Physiol 2019; 240:110593. [PMID: 31676411 DOI: 10.1016/j.cbpa.2019.110593] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/30/2019] [Accepted: 10/22/2019] [Indexed: 01/04/2023]
Abstract
The brain of diving mammals is repeatedly exposed to low oxygen conditions (hypoxia) that would have caused severe damage to most terrestrial mammals. Some whales may dive for >2 h with their brain remaining active. Many of the physiological adaptations of whales to diving have been investigated, but little is known about the molecular mechanisms that enable their brain to survive sometimes prolonged periods of hypoxia. Here, we have used an RNA-Seq approach to compare the mRNA levels in the brains of whales with those of cattle, which serves as a terrestrial relative. We sequenced the transcriptomes of the brains from cattle (Bos taurus), killer whale (Orcinus orca), and long-finned pilot whale (Globicephala melas). Further, the brain transcriptomes of cattle, minke whale (Balaenoptera acutorostrata) and bowhead whale (Balaena mysticetus), which were available in the databases, were included. We found a high expression of genes related to oxidative phosphorylation and the respiratory electron chain in the whale brains. In the visual cortex of whales, transcripts related to the detoxification of reactive oxygen species were more highly expressed than in the visual cortex of cattle. These findings indicate a high oxidative capacity in the whale brain that might help to maintain aerobic metabolism in periods of reduced oxygen availability during dives.
Collapse
Affiliation(s)
- Alena Krüger
- Institute of Zoology, University of Hamburg, Germany.
| | | | | | - Ursula Siebert
- Institute for Terrestrial and Aquatic Wildlife Research, University of Veterinary Medicine Hannover, D-25761 Büsum, Germany.
| | - Lars P Folkow
- University of Tromsø - The Arctic University of Norway, NO-9037 Tromsø, Norway.
| | | |
Collapse
|
10
|
Kletkiewicz H, Maliszewska J, Jaworski K, Jermacz Ł, Smoliński DJ, Rogalska J. Thermal conditions during neonatal anoxia affect the endogenous level of brain-derived neurotrophic factor. J Neurosci Res 2019; 97:1266-1277. [PMID: 31257630 DOI: 10.1002/jnr.24486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022]
Abstract
Anoxia during delivery is a complication that can disturb infant brain development leading to various types of neurological disorders. Our studies have shown that increased body temperature of newborn rats of both sexes intensifies the postanoxic oxidative stress and prevents triggering the endogenous adaptive response such as HIF-1α activation. Currently, brain-derived neurotrophic factor-BDNF is considered to be a modulator of neuronal plasticity. In the developing brain, mature BDNF and its precursor exhibit prosurvival action through the TrkB receptor and proapoptotic functions binding to p75NTR , respectively. The aim of our experiments was to check the effects of body temperature on the postanoxic level of BDNF and on the expression of its receptors as well as on the marker of apoptosis-caspase-3 in the rat brain. Two-day-old Wistar Han rats (male/female ratio, 1:1) were exposed to anoxia in 100% nitrogen atmosphere for 10 min in different thermal conditions, which allowed them to regulate their rectal temperature at the following levels: normothermic-33°C; hyperthermic-37°C; and extremely hyperthermic-39°C. Thermal conditions during neonatal anoxia affected the level of proBDNF, BDNF as well as their receptors and caspase-3 in the forebrain. The increased BDNF protein level followed by decreased caspase-3 protein level was probably dependent on body temperature under anoxic conditions and was observed only in rats maintaining decreased body temperature. The positive effect of BDNF was not observed under hyperthermic conditions. Moreover, BDNF level changes correlated with body temperature probably affected the learning and spatial memory in juvenile rats.
Collapse
Affiliation(s)
- Hanna Kletkiewicz
- Department of Animal Physiology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| | - Justyna Maliszewska
- Department of Animal Physiology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| | - Krzysztof Jaworski
- Chair of Plant Physiology and Biotechnology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| | - Łukasz Jermacz
- Department of Hydrobiology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| | - Dariusz J Smoliński
- Department of Cellular and Molecular Biology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland.,Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Toruń, Poland
| | - Justyna Rogalska
- Department of Animal Physiology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, Toruń, Poland
| |
Collapse
|
11
|
Barks AK, Beeson MM, Matveeva T, Gale JJ, Rao R, Tran PV. Perinatal Ischemia Alters Global Expression of Synaptosomal Proteins Critical for Neural Plasticity in the Developing Mouse Brain. Dev Neurosci 2019; 40:1-13. [PMID: 31207599 DOI: 10.1159/000499126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/25/2019] [Indexed: 11/19/2022] Open
Abstract
Ischemic perinatal stroke (IPS) affects 1 in 2,300-5,000 live births. Despite a survival rate >95%, approximately 60% of IPS infants develop motor and cognitive impairments. Given the importance of axonal growth and synaptic plasticity in neurocognitive development, our objective was to identify the molecular pathways underlying IPS-associated synaptic dysfunction using a mouse model. IPS was induced by unilateral ligation of the common carotid artery of postnatal day 10 (P10) mice. Five days after ischemia, sensorimotor and motor functions were assessed by vibrissae-evoked forepaw placement and the tail suspension test respectively, showing evidence of greater impairments in male pups than in female pups. Twenty-four hours after ischemia, both hemispheres were collected and synaptosomal proteins then prepared for quantification, using isobaric tags for relative and absolute quantitation. Seventy-two of 1,498 qualified proteins were altered in the ischemic hemisphere. Ingenuity Pathway Analysis was used to map these proteins onto molecular networks indicative of reduced neuronal proliferation, survival, and synaptic plasticity, accompanied by reduced PKCα signaling in male, but not female, pups. These effects also occurred in the non-ischemic hemisphere when compared with sham controls. The altered signaling effects may contribute to the sex-specific neurodevelopmental dysfunction following IPS, highlighting potential pathways for targeting during treatment.
Collapse
Affiliation(s)
- Amanda K Barks
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Montana M Beeson
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Tatyana Matveeva
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jonathan J Gale
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Raghavendra Rao
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Phu V Tran
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA,
| |
Collapse
|
12
|
Intrauterine growth restriction and development of the hippocampus: implications for learning and memory in children and adolescents. THE LANCET CHILD & ADOLESCENT HEALTH 2018; 2:755-764. [PMID: 30236384 DOI: 10.1016/s2352-4642(18)30245-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/13/2018] [Accepted: 07/20/2018] [Indexed: 12/15/2022]
Abstract
Intrauterine growth restriction (IUGR) is often the result of compromised placental function and suboptimal uteroplacental blood flow. Children born with IUGR have impaired cognitive functioning and specific memory deficits, indicating long-lasting impairments in hippocampal functioning; indeed, hippocampal volume is reduced in infants with IUGR. Animal studies have provided valuable insight into the nature of deficits in hippocampal-dependent functions observed in children born with IUGR; outcomes of experimental IUGR reveal reduced neuron numbers and morphological alterations in the cornu ammonis fields 1 and 3 and dentate gyrus subregions of the hippocampus. However, whether such early and ongoing structural changes in the hippocampus could account for deficits in spatial memory reported in adolescent rats with IUGR is yet to be established. Understanding the association between hippocampal structural and functional alterations in IUGR will aid in the development of interventions to minimise the effect of IUGR on the hippocampus and long-term cognitive outcomes.
Collapse
|
13
|
Synaptic transmission and excitability during hypoxia with inflammation and reoxygenation in hippocampal CA1 neurons. Neuropharmacology 2018; 138:20-31. [PMID: 29775678 DOI: 10.1016/j.neuropharm.2018.05.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/19/2018] [Accepted: 05/08/2018] [Indexed: 12/30/2022]
Abstract
Although a number of experimental and clinical studies have shown that hypoxia typically accompanies acute inflammatory responses, the combinatorial effect of the two insults on basic neural function has not been thoroughly investigated. Previous studies have predominantly suggested that hypoxia reduces network activity; however, several studies suggest the opposite effect. Of note, inflammation is known to increase neural activity. In the current study, we examined the effects of limited oxygen in combination with an inflammatory stimulus, as well as the effects of reoxygenation, on synaptic transmission and excitability. We observed a significant reduction of both synaptic transmission and excitability when hypoxia and inflammation occurred in combination, whereas reoxygenation caused hyperexcitability of neurons. Further, we found that the observed reduction in synaptic transmission was due to compromised presynaptic release efficiency based on an adenosine-receptor-dependent increase in synaptic facilitation. Excitability changes in both directions were attributable to dynamic regulation of the hyperpolarization-activated cation current (Ih) and to changes in the input resistance and the voltage difference between resting membrane potential and action potential threshold. We found that zatebradine, an Ih current inhibitor, reduced the fluctuation in excitability, suggesting that it may have potential as a drug to ameliorate reperfusion brain injury.
Collapse
|
14
|
Massaro AN, Wu YW, Bammler TK, Comstock B, Mathur A, McKinstry RC, Chang T, Mayock DE, Mulkey SB, Van Meurs K, Juul S. Plasma Biomarkers of Brain Injury in Neonatal Hypoxic-Ischemic Encephalopathy. J Pediatr 2018; 194:67-75.e1. [PMID: 29478510 DOI: 10.1016/j.jpeds.2017.10.060] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/21/2017] [Accepted: 10/25/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVES To evaluate plasma brain specific proteins and cytokines as biomarkers of brain injury in newborns with hypoxic-ischemic encephalopathy (HIE) and, secondarily, to assess the effect of erythropoietin (Epo) treatment on the relationship between biomarkers and outcomes. STUDY DESIGN A study of candidate brain injury biomarkers was conducted in the context of a phase II multicenter randomized trial evaluating Epo for neuroprotection in HIE. Plasma was collected at baseline (<24 hours) and on day 5. Brain injury was assessed by magnetic resonance imaging (MRI) and neurodevelopmental assessments at 1 year. The relationships between Epo, brain-specific proteins (S100B, ubiquitin carboxy-terminal hydrolase-L1 [UCH-L1], total Tau, neuron specific enolase), cytokines (interleukin [IL]-1β, IL-6, IL-8, IL-10, IL-12P70, IL-13, interferon-gamma [IFN-γ], tumor necrosis factor alpha [TNF-α], brain-derived neurotrophic factor [BDNF], monocyte chemoattractant protein-1), and brain injury were assessed. RESULTS In 50 newborns with encephalopathy, elevated baseline S100B, Tau, UCH-L1, IL-1β, IL-6, IL-8, IL-10, IL-13, TNF-α, and IFN-γ levels were associated with increasing brain injury severity by MRI. Higher baseline Tau and lower day 5 BDNF were associated with worse 1 year outcomes. No statistically significant evidence of Epo treatment modification on biomarkers was detected in this small cohort. CONCLUSIONS Elevated plasma brain-specific proteins and cytokine levels in the first 24 hours of life are associated with worse brain injury by MRI in newborns with HIE. Only Tau and BDNF levels were found to be related to neurodevelopmental outcomes. The effect of Epo treatment on the relationships between biomarkers and brain injury in HIE requires further study. TRIAL REGISTRATION ClinicalTrials.gov: 01913340.
Collapse
Affiliation(s)
- An N Massaro
- Department of Pediatrics, The George Washington University School of Medicine and Children's National Health Systems, Washington, DC.
| | - Yvonne W Wu
- Department of Neurology, University of California, San Francisco, San Francisco, CA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| | - Theo K Bammler
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA
| | - Bryan Comstock
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Amit Mathur
- Department of Pediatrics, Washington University, St Louis, MO
| | - Robert C McKinstry
- Department of Pediatrics, Washington University, St Louis, MO; Department of Radiology, Washington University, St Louis, MO
| | - Taeun Chang
- Department of Pediatrics, The George Washington University School of Medicine and Children's National Health Systems, Washington, DC; Department of Neurology, The George Washington University School of Medicine and Children's National Health Systems, Washington, DC
| | - Dennis E Mayock
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Sarah B Mulkey
- Department of Pediatrics, The George Washington University School of Medicine and Children's National Health Systems, Washington, DC; Department of Neurology, The George Washington University School of Medicine and Children's National Health Systems, Washington, DC
| | - Krisa Van Meurs
- Department of Pediatrics, Stanford University, Palo Alto, CA
| | - Sandra Juul
- Department of Pediatrics, University of Washington, Seattle, WA
| |
Collapse
|
15
|
Cardoso FDS, França EF, Serra FT, Victorino AB, de Almeida AA, Fernandes J, Cabral FR, Venancio DP, Arida RM, Gomes da Silva S. Aerobic exercise reduces hippocampal ERK and p38 activation and improves memory of middle-aged rats. Hippocampus 2017; 27:899-905. [DOI: 10.1002/hipo.22740] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 04/25/2017] [Accepted: 05/02/2017] [Indexed: 12/28/2022]
Affiliation(s)
| | | | | | | | | | - Jansen Fernandes
- Universidade Federal de São Paulo (UNIFESP); São Paulo - SP Brazil
| | | | - Daniel Paulino Venancio
- Departamento de Morfologia e Fisiologia; Faculdade de Medicina do ABC; Santo André - SP Brazil
| | | | - Sérgio Gomes da Silva
- Universidade de Mogi das Cruzes (UMC). Mogi das Cruzes - SP; Brazil
- Hospital Israelita Albert Einstein; São Paulo - SP Brazil
| |
Collapse
|
16
|
Astroglial Activation by an Enriched Environment after Transplantation of Mesenchymal Stem Cells Enhances Angiogenesis after Hypoxic-Ischemic Brain Injury. Int J Mol Sci 2016; 17:ijms17091550. [PMID: 27649153 PMCID: PMC5037823 DOI: 10.3390/ijms17091550] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/25/2016] [Accepted: 09/05/2016] [Indexed: 12/12/2022] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) has paracrine effects; however, the effects are known to be largely limited. Here we investigated the combination effects of cell transplantation and enriched environment (EE) in a model of hypoxic-ischemic brain injury. Brain damage was induced in seven-day-old mice by unilateral carotid artery ligation and exposure to hypoxia (8% O2 for 90 min). At six weeks of age, the mice were randomly assigned to four groups: phosphate-buffered saline (PBS)-control (CON), PBS-EE, MSC-CON, and MSC-EE. Rotarod and grip strength tests were performed to evaluate neurobehavioral functions. Histologic evaluations were also performed to confirm the extent of astrocyte activation and endogenous angiogenesis. An array-based multiplex ELISA and Western blot were used to identify growth factors in vivo and in vitro. Two weeks after treatment, levels of astrocyte density and angiogenic factors were increased in MSC-EE mice, but glial scarring was not increased. Eight weeks after treatment, angiogenesis was increased, and behavioral outcomes were synergistically improved in the MSC-EE group. Astrocytes co-cultured with MSCs expressed higher levels of angiogenic factors than astrocytes cultured alone. The mechanisms of this synergistic effect included enhanced repair processes, such as increased endogenous angiogenesis and upregulation of angiogenic factors released from activated astrocytes.
Collapse
|
17
|
Logica T, Riviere S, Holubiec MI, Castilla R, Barreto GE, Capani F. Metabolic Changes Following Perinatal Asphyxia: Role of Astrocytes and Their Interaction with Neurons. Front Aging Neurosci 2016; 8:116. [PMID: 27445788 PMCID: PMC4921470 DOI: 10.3389/fnagi.2016.00116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 05/03/2016] [Indexed: 11/13/2022] Open
Abstract
Perinatal Asphyxia (PA) represents an important cause of severe neurological deficits including delayed mental and motor development, epilepsy, major cognitive deficits and blindness. The interaction between neurons, astrocytes and endothelial cells plays a central role coupling energy supply with changes in neuronal activity. Traditionally, experimental research focused on neurons, whereas astrocytes have been more related to the damage mechanisms of PA. Astrocytes carry out a number of functions that are critical to normal nervous system function, including uptake of neurotransmitters, regulation of pH and ion concentrations, and metabolic support for neurons. In this work, we aim to review metabolic neuron-astrocyte interactions with the purpose of encourage further research in this area in the context of PA, which is highly complex and its mechanisms and pathways have not been fully elucidated to this day.
Collapse
Affiliation(s)
- Tamara Logica
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - Stephanie Riviere
- Laboratorio de Biología Molecular, Facultad de Medicina, Instituto de Investigaciones cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - Mariana I Holubiec
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - Rocío Castilla
- Laboratorio de Biología Molecular, Facultad de Medicina, Instituto de Investigaciones cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABA Buenos Aires, Argentina
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá Bogotá, Colombia
| | - Francisco Capani
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, CABABuenos Aires, Argentina; Departamento de Biología, Universidad Argentina JF KennedyBuenos Aires, Argentina; Investigador Asociado, Universidad Autónoma de ChileSantiago, Chile
| |
Collapse
|
18
|
Gopagondanahalli KR, Li J, Fahey MC, Hunt RW, Jenkin G, Miller SL, Malhotra A. Preterm Hypoxic-Ischemic Encephalopathy. Front Pediatr 2016; 4:114. [PMID: 27812521 PMCID: PMC5071348 DOI: 10.3389/fped.2016.00114] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/05/2016] [Indexed: 11/18/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a recognizable and defined clinical syndrome in term infants that results from a severe or prolonged hypoxic-ischemic episode before or during birth. However, in the preterm infant, defining hypoxic-ischemic injury (HII), its clinical course, monitoring, and outcomes remains complex. Few studies examine preterm HIE, and these are heterogeneous, with variable inclusion criteria and outcomes reported. We examine the available evidence that implies that the incidence of hypoxic-ischemic insult in preterm infants is probably higher than recognized and follows a more complex clinical course, with higher rates of adverse neurological outcomes, compared to term infants. This review aims to elucidate the causes and consequences of preterm hypoxia-ischemia, the subsequent clinical encephalopathy syndrome, diagnostic tools, and outcomes. Finally, we suggest a uniform definition for preterm HIE that may help in identifying infants most at risk of adverse outcomes and amenable to neuroprotective therapies.
Collapse
Affiliation(s)
| | - Jingang Li
- The Ritchie Centre, Hudson Institute of Medical Research , Melbourne, VIC , Australia
| | - Michael C Fahey
- Monash Children's Hospital, Melbourne, VIC, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia; Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Rod W Hunt
- The Royal Children's Hospital, Melbourne, VIC, Australia; Murdoch Childrens Research Institute, Melbourne, VIC, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia; Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia; Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Atul Malhotra
- Monash Children's Hospital, Melbourne, VIC, Australia; The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia; Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
19
|
Mesenchymal stem cells stabilize the blood-brain barrier through regulation of astrocytes. Stem Cell Res Ther 2015; 6:187. [PMID: 26420371 PMCID: PMC4588687 DOI: 10.1186/s13287-015-0180-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 08/31/2015] [Accepted: 09/07/2015] [Indexed: 12/18/2022] Open
Abstract
Introduction The blood–brain barrier (BBB) protects the brain against potentially neurotoxic molecules in the circulation, and loss of its integrity may contribute to disease progression in neurodegenerative conditions. Recently, the active role of reactive astrocytes in BBB disruption has become evident in the inflamed brain. In the present study, we investigated whether mesenchymal stem cell (MSC) treatment might modulate reactive astrocytes and thus stabilize BBB integrity through vascular endothelial growth factor A (VEGF-A) signaling in inflammatory conditions. Methods For the inflamed brain, we injected LPS using a stereotaxic apparatus and MSCs were injected into the tail vein. At 6 hours and 7 days after LPS injection, we analyzed modulatory effects of MSCs on the change of BBB permeability through VEGF-A signaling using immunochemistry and western blot. To determine the effects of MSCs on VEGF-A-related signaling in cellular system, we had used endothelial cells treated with VEGF-A and co-cultured astrocyte and BV 2 cells treated with lipopolysaccharide (LPS) and then these cells were co-cultured with MSCs. Results In LPS-treated rats, MSCs restored Evans blue infiltration and the number of endothelial-barrier antigen (EBA) and P-glycoprotein (p-gp)-expressing cells, which were significantly altered in LPS-treated animals. Additionally, MSC administration following LPS treatment markedly increased the density of astrocytic filaments around vessels and reversed LPS-induced elevations in VEGF-A levels as well as endothelial nitric oxide synthase (eNOS)-dependent downregulation of tight junction proteins in the endothelium. Consequently, MSC treatment reduced neutrophil infiltration and enhanced survival of midbrain dopaminergic neurons in LPS-treated animals. In cellular system, MSC treatment led to a significant reversion of VEGF-A-induced eNOS and tight junction protein expression in endothelial cells, which led to increased EBA expressing cells. Additionally, MSC treatment significantly attenuated LPS-induced increased expressions of IL-1β in microglia and VEGF-A in astrocytes with an increase in IL-10 levels. Conclusion The present study indicated that MSCs may stabilize BBB permeability by modulating astrocytic endfeet and VEGF-A signaling, which may be relevant to the treatment of Parkinsonian diseases as a candidate for disease modifying therapeutics. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0180-4) contains supplementary material, which is available to authorized users.
Collapse
|
20
|
Huang W, Liu X, Cao J, Meng F, Li M, Chen B, Zhang J. miR-134 Regulates Ischemia/Reperfusion Injury-Induced Neuronal Cell Death by Regulating CREB Signaling. J Mol Neurosci 2014; 55:821-9. [DOI: 10.1007/s12031-014-0434-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 09/29/2014] [Indexed: 12/21/2022]
|
21
|
Dale EA, Ben Mabrouk F, Mitchell GS. Unexpected benefits of intermittent hypoxia: enhanced respiratory and nonrespiratory motor function. Physiology (Bethesda) 2014; 29:39-48. [PMID: 24382870 DOI: 10.1152/physiol.00012.2013] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Intermittent hypoxia (IH) is most often thought of for its role in morbidity associated with sleep-disordered breathing, including central nervous system pathology. However, recent evidence suggests that the nervous system fights back in an attempt to minimize pathology by increasing the expression of growth/trophic factors that confer neuroprotection and neuroplasticity. For example, even modest ("low dose") IH elicits respiratory motor plasticity, increasing the strength of respiratory contractions and breathing. These low IH doses upregulate hypoxia-sensitive growth/trophic factors within respiratory motoneurons but do not elicit detectable pathologies such as hippocampal cell death, neuroinflammation, or systemic hypertension. Recent advances have been made toward understanding cellular mechanisms giving rise to IH-induced respiratory plasticity, and attempts have been made to harness the benefits of low-dose IH to treat respiratory insufficiency after cervical spinal injury. Our recent realization that IH also upregulates growth/trophic factors in nonrespiratory motoneurons and improves limb (or leg) function after incomplete chronic spinal injuries suggests that IH-induced plasticity is a general feature of motor systems. Collectively, available evidence suggests that low-dose IH may represent a safe and effective treatment to restore lost motor function in diverse clinical disorders that impair motor function.
Collapse
Affiliation(s)
- E A Dale
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| | | | | |
Collapse
|
22
|
Villapol S, Byrnes KR, Symes AJ. Temporal dynamics of cerebral blood flow, cortical damage, apoptosis, astrocyte-vasculature interaction and astrogliosis in the pericontusional region after traumatic brain injury. Front Neurol 2014; 5:82. [PMID: 24926283 PMCID: PMC4044679 DOI: 10.3389/fneur.2014.00082] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 05/14/2014] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) results in a loss of brain tissue at the moment of impact in the cerebral cortex. Subsequent secondary injury involves the release of molecular signals with dramatic consequences for the integrity of damaged tissue, leading to the evolution of a pericontusional-damaged area minutes to days after in the initial injury. The mechanisms behind the progression of tissue loss remain under investigation. In this study, we analyzed the spatial–temporal profile of blood flow, apoptotic, and astrocytic–vascular events in the cortical regions around the impact site at time points ranging from 5 h to 2 months after TBI. We performed a mild–moderate controlled cortical impact injury in young adult mice and analyzed the glial and vascular response to injury. We observed a dramatic decrease in perilesional cerebral blood flow (CBF) immediately following the cortical impact that lasted until days later. CBF finally returned to baseline levels by 30 days post-injury (dpi). The initial impact also resulted in an immediate loss of tissue and cavity formation that gradually increased in size until 3 dpi. An increase in dying cells localized in the pericontusional region and a robust astrogliosis were also observed at 3 dpi. A strong vasculature interaction with astrocytes was established at 7 dpi. Glial scar formation began at 7 dpi and seemed to be compact by 60 dpi. Altogether, these results suggest that TBI results in a progression from acute neurodegeneration that precedes astrocytic activation, reformation of the neurovascular unit to glial scar formation. Understanding the multiple processes occurring after TBI is critical to the ability to develop neuroprotective therapeutics to ameliorate the short and long-term consequences of brain injury.
Collapse
Affiliation(s)
- Sonia Villapol
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, MD , USA ; Department of Pharmacology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - Kimberly R Byrnes
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, MD , USA ; Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - Aviva J Symes
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, MD , USA ; Department of Pharmacology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| |
Collapse
|
23
|
Sato Y, Bernier F, Suzuki I, Kotani S, Nakagawa M, Oda Y. Comparative lipidomics of mouse brain exposed to enriched environment. J Lipid Res 2013; 54:2687-96. [PMID: 23833247 DOI: 10.1194/jlr.m038075] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Several studies have shown that housing conditions and environmental exposure to a series of stimuli lead to behavior improvement in several species. While more works have been focused on illustrating changes of the proteome and transcriptome following enriched environment exposure in mice, little has been done to understand changes in the brain metabolome in this paradigm due to the complexity of this type of analysis. In this paper, lipidomics focused on phospholipids and gangliosides were conducted for brain tissues of mice exposed to enriched or impoverished conditions. We optimized previously reported method and established a reliable relative comparison method for phospholipids and gangliosides in brain tissue using prefractionation with weak anion exchange cartridge. We used liquid chromatography mass spectrometry to explore metabolic signatures of the cerebral cortex and hippocampus after confirming the animals had significant memory differences using the fear conditioning paradigm and brain immunohistochemistry. Although both cerebral cortex and hippocampus regions did not show major alterations in ganglioside composition, we found significant differences in a series of phospholipids containing 22:6 fatty acid in the prefrontal cortex, indicating that environmental enrichment and impoverished housing conditions might be a relevant paradigm to study aberrant lipid metabolism of docosahexaenoic acid consumption. Our study highlights the hypothesis-generating potential of lipidomics and identifies novel region-specific lipid changes possibly linked not only to change of memory function in these models, but also to help us better understand how lipid changes may contribute to memory disorders.
Collapse
Affiliation(s)
- Yoshiaki Sato
- Eisai Company, Limited, Ibaraki 300-2635, Japan; and
| | | | | | | | | | | |
Collapse
|
24
|
Ke XJ, Zhang JJ. Changes in HIF-1α, VEGF, NGF and BDNF levels in cerebrospinal fluid and their relationship with cognitive impairment in patients with cerebral infarction. ACTA ACUST UNITED AC 2013; 33:433-437. [DOI: 10.1007/s11596-013-1137-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Indexed: 11/28/2022]
|
25
|
Lara-Celador I, Goñi-de-Cerio F, Alvarez A, Hilario E. Using the endocannabinoid system as a neuroprotective strategy in perinatal hypoxic-ischemic brain injury. Neural Regen Res 2013; 8:731-44. [PMID: 25206720 PMCID: PMC4146074 DOI: 10.3969/j.issn.1673-5374.2013.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 02/07/2013] [Indexed: 12/30/2022] Open
Abstract
One of the most important causes of brain injury in the neonatal period is a perinatal hypoxic-ischemic event. This devastating condition can lead to long-term neurological deficits or even death. After hypoxic-ischemic brain injury, a variety of specific cellular mechanisms are set in motion, triggering cell damage and finally producing cell death. Effective therapeutic treatments against this phenomenon are still unavailable because of complex molecular mechanisms underlying hypoxic-ischemic brain injury. After a thorough understanding of the mechanism underlying neural plasticity following hypoxic-ischemic brain injury, various neuroprotective therapies have been developed for alleviating brain injury and improving long-term outcomes. Among them, the endocannabinoid system emerges as a natural system of neuroprotection. The endocannabinoid system modulates a wide range of physiological processes in mammals and has demonstrated neuroprotective effects in different paradigms of acute brain injury, acting as a natural neuroprotectant. The aim of this review is to study the use of different therapies to induce long-term therapeutic effects after hypoxic-ischemic brain injury, and analyze the important role of the endocannabinoid system as a new neuroprotective strategy against perinatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- I. Lara-Celador
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, Leioa 48949, Bizkaia, Spain
| | - F. Goñi-de-Cerio
- GAIKER Technology Centre, Bizkaia Science and Technology Park, Building 202, Zamudio 48170, Bizkaia, Spain
| | - Antonia Alvarez
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, Leioa 48949, Bizkaia, Spain
| | - Enrique Hilario
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, Leioa 48949, Bizkaia, Spain
| |
Collapse
|
26
|
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: 2.4] [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
|
27
|
Dynamic Changes of Cerebral-Specific Proteins in Full-Term Newborns with Hypoxic–Ischemic Encephalopathy. Cell Biochem Biophys 2012. [DOI: 10.1007/s12013-012-9478-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
28
|
Bouzas-Rodríguez J, Zárraga-Granados G, Sánchez-Carbente MDR, Rodríguez-Valentín R, Gracida X, Anell-Rendón D, Covarrubias L, Castro-Obregón S. The nuclear receptor NR4A1 induces a form of cell death dependent on autophagy in mammalian cells. PLoS One 2012; 7:e46422. [PMID: 23071566 PMCID: PMC3465341 DOI: 10.1371/journal.pone.0046422] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/29/2012] [Indexed: 11/18/2022] Open
Abstract
The control of cell death is a biological process essential for proper development, and for preventing devastating pathologies like cancer and neurodegeneration. On the other hand, autophagy regulation is essential for protein and organelle degradation, and its dysfunction is associated with overlapping pathologies like cancer and neurodegeneration, but also for microbial infection and aging. In the present report we show that two evolutionarily unrelated receptors--Neurokinin 1 Receptor (NK(1)R,) a G-protein coupled receptor, and Insulin-like Growth Factor 1 Receptor (IGF1R), a tyrosine kinase receptor--both induce non-apoptotic cell death with autophagic features and requiring the activity of the autophagic core machinery proteins PI3K-III, Beclin-1 and Atg7. Remarkably, this form of cell death occurs in apoptosis-competent cells. The signal transduction pathways engaged by these receptors both converged on the activation of the nuclear receptor NR4A1, which has previously been shown to play a critical role in some paradigms of apoptosis and in NK(1)R-induced cell death. The activity of NR4A1 was necessary for IGF1R-induced cell death, as well as for a canonical model of cell death by autophagy induced by the presence of a pan-caspase inhibitor, suggesting that NR4A1 is a general modulator of this kind of cell death. During cell death by autophagy, NR4A1 was transcriptionally competent, even though a fraction of it was present in the cytoplasm. Interestingly, NR4A1 interacts with the tumor suppressor p53 but not with Beclin-1 complex. Therefore the mechanism to promote cell death by autophagy might involve regulation of gene expression, as well as protein interactions. Understanding the molecular basis of autophagy and cell death mediation by NR4A1, should provide novel insights and targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jimena Bouzas-Rodríguez
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Gabriela Zárraga-Granados
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Maria del Rayo Sánchez-Carbente
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Rocío Rodríguez-Valentín
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Xicotencatl Gracida
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Dámaris Anell-Rendón
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Luis Covarrubias
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Susana Castro-Obregón
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
- * E-mail:
| |
Collapse
|
29
|
Lara-Celador I, Castro-Ortega L, Alvarez A, Goñi-de-Cerio F, Lacalle J, Hilario E. Endocannabinoids reduce cerebral damage after hypoxic-ischemic injury in perinatal rats. Brain Res 2012; 1474:91-9. [PMID: 22841538 DOI: 10.1016/j.brainres.2012.07.045] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 06/25/2012] [Accepted: 07/23/2012] [Indexed: 12/19/2022]
Abstract
Hypoxic-ischemic (HI) insult during the perinatal period remains as one of the most common causes of brain injury and produces long-term neurological deficits, and there is a growing need for effective therapies. The aim of the present work was to perform a prospective study designed to assess the possible protector effect of two endocannabinoids: 2-arachidonoylglycerol (2AG) and anandamide (AEA) in the brain after HI injury in perinatal rat model. We evaluate their effects on cell death and check several cellular parameters. 7-days-old Wistar rats were assigned to four different experimental groups (n=7-10): Sham, HI, and HI treated with 2AG or AEA. The injury was induced by the left carotid artery ligature and subsequent exposure to 8% O(2) for 120 min. Immediately after the injury, treated groups received a single dose of 2AG (1mg/kg) or AEA (5mg/kg) and then animals were sacrificed 24, 72 h or 7 days after the HI event. Brains fixed by perfusion were stained with Nissl for morphological studies, and non-fixed brains were dissociated and analyzed by flow cytometry to quantify apoptosis, mitochondrial state, intracellular calcium and reactive oxygen species. Our results show that both 2AG and AEA have beneficial effects after HI injury in this rat model, producing a remarkable amelioration of brain injury, reducing apoptotic cell death, contributing to the maintenance of mitochondrial functionality, and improving cellular parameters such as the influx of calcium and ROS production.
Collapse
Affiliation(s)
- Idoia Lara-Celador
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, E-48940 Leioa, Vizcaya, Spain.
| | | | | | | | | | | |
Collapse
|
30
|
Fusco FR, Anzilotti S, Giampà C, Dato C, Laurenti D, Leuti A, Colucci D'Amato L, Perrone L, Bernardi G, Melone MA. Changes in the expression of extracellular regulated kinase (ERK 1/2) in the R6/2 mouse model of Huntington's disease after phosphodiesterase IV inhibition. Neurobiol Dis 2012; 46:225-33. [DOI: 10.1016/j.nbd.2012.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 01/09/2012] [Accepted: 01/21/2012] [Indexed: 11/29/2022] Open
|
31
|
Alonso-Alconada D, Alvarez A, Hilario E. Cannabinoid as a neuroprotective strategy in perinatal hypoxic-ischemic injury. Neurosci Bull 2011; 27:275-85. [PMID: 21788999 DOI: 10.1007/s12264-011-1008-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Perinatal hypoxia-ischemia remains the single most important cause of brain injury in the newborn, leading to death or lifelong sequelae. Because of the fact that there is still no specific treatment for perinatal brain lesions due to the complexity of neonatal hypoxic-ischemic pathophysiology, the search of new neuroprotective therapies is of great interest. In this regard, therapeutic possibilities of the endocannabinoid system have grown lately. The endocannabinoid system modulates a wide range of physiological processes in mammals and has demonstrated neuroprotective effects in different paradigms of acute brain injury, acting as a natural neuroprotectant. Concerning perinatal asphyxia, the neuroprotective role of this endogenous system is emerging these years. The present review mainly focused on the current knowledge of the cannabinoids as a new neuroprotective strategy against perinatal hypoxic-ischemic brain injury.
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
|
32
|
Enciu AM, Constantinescu SN, Popescu LM, Mureşanu DF, Popescu BO. Neurobiology of vascular dementia. J Aging Res 2011; 2011:401604. [PMID: 21876809 PMCID: PMC3160011 DOI: 10.4061/2011/401604] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 06/27/2011] [Accepted: 06/28/2011] [Indexed: 01/22/2023] Open
Abstract
Vascular dementia is, in its current conceptual form, a distinct type of dementia with a spectrum of specific clinical and pathophysiological features. However, in a very large majority of cases, these alterations occur in an already aged brain, characterized by a milieu of cellular and molecular events common for different neurodegenerative diseases. The cell signaling defects and molecular dyshomeostasis might lead to neuronal malfunction prior to the death of neurons and the alteration of neuronal networks. In the present paper, we explore some of the molecular mechanisms underlying brain malfunction triggered by cerebrovascular disease and risk factors. We suggest that, in the age of genetic investigation and molecular diagnosis, the concept of vascular dementia needs a new approach.
Collapse
Affiliation(s)
- Ana-Maria Enciu
- Department of Cellular and Molecular Medicine, School of Medicine, "Carol Davila" University of Medicine and Pharmacy, 8 Eroilor Sanitari, Sector 5, 050474 Bucharest, Romania
| | | | | | | | | |
Collapse
|
33
|
Suganuma H, Arai Y, Kitamura Y, Hayashi M, Okumura A, Shimizu T. Maternal docosahexaenoic acid-enriched diet prevents neonatal brain injury. Neuropathology 2011; 30:597-605. [PMID: 20408962 DOI: 10.1111/j.1440-1789.2010.01114.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hypoxic-ischemic encephalopathy due to neonatal asphyxia is one of the most important causes of delayed neurological development. Prolonged neuronal apoptosis plays an important role in the processes contributing to neuronal degeneration. Docosahexaenoic acid (DHA), a major component of brain membrane phospholipids, prevents neuronal cell apoptosis and plays an important role as an anti-oxidant agent. We investigated the neuroprotective and anti-oxidant effects of maternal DHA supplementation during pregnancy in a model of neonatal hypoxic-ischemic encephalopathy. Pregnant rats were randomly assigned to two experimental groups: a control group or a DHA-enriched diet group. Hypoxic-ischemic encephalopathy was produced by left common carotid artery occlusion and exposure to 8% oxygen for 1.5 h. TUNEL assay, immunohistochemistry for caspase-3 and 8-hydroxy-deoxyguanosine (8-OHdG), and Western blot for caspase-3 were performed at postnatal days 8, 10 and 14. Fatty acid composition of brain was estimated on postnatal day 7. Maternal diet clearly influenced brain fatty acid composition in pups. Numbers of apoptotic neuronal cells and 8-OHdG immunoreactivity were significantly decreased in the DHA-enriched group. Our findings indicate that maternal DHA-enriched diet during pregnancy provides neuroprotection by inhibiting oxidative stress and apoptotic neuronal death. Dietary supplementation of DHA during pregnancy may thus be beneficial in preventing neonatal brain injury.
Collapse
Affiliation(s)
- Hiroki Suganuma
- Department of Pediatrics, Juntendo University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
34
|
Orellana JA, Hernández DE, Ezan P, Velarde V, Bennett MVL, Giaume C, Sáez JC. Hypoxia in high glucose followed by reoxygenation in normal glucose reduces the viability of cortical astrocytes through increased permeability of connexin 43 hemichannels. Glia 2010; 58:329-43. [PMID: 19705457 DOI: 10.1002/glia.20926] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Brain ischemia causes more extensive injury in hyperglycemic than normoglycemic subjects, and the increased damage is to astroglia as well as neurons. In the present work, we found that in cortical astrocytes from rat or mouse, reoxygenation after hypoxia in a medium mimicking interstitial fluid during ischemia increases hemichannel activity and decreases cell-cell communication via gap junctions as indicated by dye uptake and dye coupling, respectively. These effects were potentiated by high glucose during the hypoxia in a concentration-dependent manner (and by zero glucose) and were not observed in connexin 43(-/-) astrocytes. The responses were transient and persistent after short and long periods of hypoxia, respectively. The persistent responses were associated with a progressive reduction in cell viability that was prevented by La(3+) or peptides that block connexin 43 (Cx43) hemichannels or by inhibition of p38 MAP kinase prior to hypoxia-reoxygenation but not by treatments that block pannexin hemichannels. Block of Cx43 hemichannels did not affect the reduction in gap junction mediated dye coupling observed during reoxygenation. Cx43 hemichannels may be a novel therapeutic target to reduce cell death following stroke, particularly in hyperglycemic conditions.
Collapse
Affiliation(s)
- Juan A Orellana
- Departamento de Ciencias Fisiológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile.
| | | | | | | | | | | | | |
Collapse
|
35
|
Rogalska J. Mineralocorticoid and glucocorticoid receptors in hippocampus: their impact on neurons survival and behavioral impairment after neonatal brain injury. VITAMINS AND HORMONES 2010; 82:391-419. [PMID: 20472149 DOI: 10.1016/s0083-6729(10)82020-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Glucocorticoids (GC) exert multiple effects within the central nervous system via mineralocorticoid receptors (MR) and glucocorticoid receptors (GR) activation. MR expression is associated with a neuroprotective phenotype, whereas GR activation is implicated in the induction of an endangered neural phenotype and the opposite actions are most evident in hippocampus, where these receptors are predominantly present. Hippocampus has an overall inhibitory influence on the activity of the hypothalamic-pituitary-adrenal (HPA) axis and it has been suggested that efficient learning and adequate stress response depend on the appropriate functioning of the axis brought by coordinated activation of MR and GR in this region. There is a growing body of evidence that perinatal asphyxia causes irreversible damage to the brain leading to neurons loss in regions vulnerable to oxygen shortage especially in hippocampus. In the present review, some aspects of recently acquired insight in the role of GC receptors in promoting neuronal death and survival after hippocampal injury are discussed. Since the unbalance of MR and GR in hippocampus creates a condition of disturbed neuroendocrine regulation their potential impact on behavioral impairment will also be reviewed.
Collapse
Affiliation(s)
- Justyna Rogalska
- Department of Animal Physiology, Institute of General and Molecular Biology, N. Copernicus University, Torun, Poland
| |
Collapse
|
36
|
A history of our understanding of cerebral vascular development and pathogenesis of perinatal brain damage over the past 30 years. Semin Pediatr Neurol 2009; 16:226-36. [PMID: 19945657 DOI: 10.1016/j.spen.2009.09.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This article reviews our studies focusing on cerebral vascular development, the pathogenesis of subependymal/intraventricular hemorrhage (SEH/IVH), periventricular leukomalacia (PVL), and pontosubicular neuron necrosis (PSN). Their pathogenesis consists of predisposing developmental and causal factors. SEH/IVH may be caused by reperfusion or overperfusion following ischemia in the subependymal germinal matrix with characteristic vasculature. The cause of PVL is multifactorial (ie, ischemia and inflammation), predisposed by the maturational status of the vasculature and oligodendroglia in the white matter. Focal PVL is ischemic necrosis, and diffuse PVL or white matter injury may include cytotoxic damage. PSN has an apoptotic character, and may be induced by ischemic and oxidative stress on specific immature neurons. Further studies on preventive and therapeutic measures are necessary in clinical, pathologic, and experimental fields. The monitoring and control methods of brain hemodynamics and cellular stability should be more developed to prevent brain damages.
Collapse
|
37
|
Duman RS. Neuronal damage and protection in the pathophysiology and treatment of psychiatric illness: stress and depression. DIALOGUES IN CLINICAL NEUROSCIENCE 2009. [PMID: 19877493 PMCID: PMC3181922 DOI: 10.31887/dcns.2009.11.3/rsduman] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The discovery that stress and depression, as well as other psychiatric illnesses, are characterized by structural alterations, and that these changes result from atrophy and loss of neurons and glia in specific limbic regions and circuits, has contributed to a fundamental change in our understanding of these illnesses. These structural changes are accompanied by dysregulation of neuroprotective and neurotrophic signaling mechanisms that are required for the maturation, growth, and survival of neurons and glia. Conversely, behavioral and therapeutic interventions can reverse these structural alterations by stimulating neuroprotective and neurotrophic pathways and by blocking the damaging, excitotoxic, and inflammatory effects of stress. Lifetime exposure to cellular and environmental stressors and interactions with genetic factors contribute to individual susceptibility or resilience. This exciting area of research holds promise and potential for further elucidating the pathophysiology of psychiatric illness and for development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06508, USA.
| |
Collapse
|
38
|
Massaro CM, Pielage J, Davis GW. Molecular mechanisms that enhance synapse stability despite persistent disruption of the spectrin/ankyrin/microtubule cytoskeleton. ACTA ACUST UNITED AC 2009; 187:101-17. [PMID: 19805631 PMCID: PMC2762090 DOI: 10.1083/jcb.200903166] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Neuromuscular junctions crippled by a disrupted microtubule cytoskeleton are rescued by stress-induced activation of MAPK-JNK-Fos signaling. Loss of spectrin or ankyrin in the presynaptic motoneuron disrupts the synaptic microtubule cytoskeleton and leads to disassembly of the neuromuscular junction (NMJ). Here, we demonstrate that NMJ disassembly after loss of α-spectrin can be suppressed by expression of a WldS transgene, providing evidence for a Wallerian-type degenerative mechanism. We then identify a second signaling system. Enhanced MAPK-JNK-Fos signaling suppresses NMJ disassembly despite loss of presynaptic α-spectrin or ankyrin2-L. This signaling system is activated after an acute cytoskeletal disruption, suggesting an endogenous role during neurological stress. This signaling system also includes delayed, negative feedback via the JNK phosphatase puckered, which inhibits JNK-Fos to allow NMJ disassembly in the presence of persistent cytoskeletal stress. Finally, the MAPK-JNK pathway is not required for baseline NMJ stabilization during normal NMJ growth. We present a model in which signaling via JNK-Fos functions as a stress response system that is transiently activated after cytoskeletal disruption to enhance NMJ stability, and is then shut off allowing NMJ disassembly during persistent cytoskeletal disruption.
Collapse
Affiliation(s)
- Catherine M Massaro
- Department of Biochemistry and Biophysics, Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | |
Collapse
|
39
|
Zhu C, Huang Z, Gao J, Zhang Y, Wang X, Karlsson N, Li Q, Lannering B, Björk-Eriksson T, Georg Kuhn H, Blomgren K. Irradiation to the immature brain attenuates neurogenesis and exacerbates subsequent hypoxic-ischemic brain injury in the adult. J Neurochem 2009; 111:1447-56. [PMID: 19799713 DOI: 10.1111/j.1471-4159.2009.06413.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cranial radiotherapy is common in pediatric oncology. Our purpose was to investigate if irradiation (IR) to the immature brain would increase the susceptibility to hypoxic-ischemic injury in adulthood. The left hemisphere of postnatal day 10 (P10) mice was irradiated with 8 Gy and subjected to hypoxia-ischemia (HI) on P60. Brain injury, neurogenesis and inflammation were evaluated 30 days after HI. IR alone caused significant hemispheric tissue loss, or lack of growth (2.8 +/- 0.42 mm(3), p < 0.001). Tissue loss after HI (18.2 +/- 5.8 mm(3), p < 0.05) was synergistically increased if preceded by IR (32.0 +/- 3.5 mm(3), p < 0.05). Infarct volume (5.1 +/- 1.6 mm(3)) nearly doubled if HI was preceded by IR (9.8 +/- 1.2 mm(3), p < 0.05). Pathological scoring revealed that IR aggravated hippocampal, cortical and striatal, but not thalamic, injury. Hippocampal neurogenesis decreased > 50% after IR but was unchanged by HI alone. The number of newly formed microglia was three times higher after IR + HI than after HI alone. In summary, IR to the immature brain produced long-lasting changes, including decreased hippocampal neurogenesis, subsequently rendering the adult brain more susceptible to HI, resulting in larger infarcts, increased hemispheric tissue loss and more inflammation than in non-irradiated brains.
Collapse
Affiliation(s)
- Changlian Zhu
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Blanquet PR, Mariani J, Fournier B. Temporal assessment of histone H3 phospho-acetylation and casein kinase 2 activation in dentate gyrus from ischemic rats. Brain Res 2009; 1302:10-20. [PMID: 19765564 DOI: 10.1016/j.brainres.2009.09.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 08/31/2009] [Accepted: 09/08/2009] [Indexed: 11/30/2022]
Abstract
Hippocampal dentate gyrus possesses an exceptional capacity of adaptation to ischemic insults. Recently, using a transient global ischemic model in the adult rat, we identified a neuroprotective signalling cascade in the dentate gyrus involving calcium/calmodulin-dependent protein kinase IV (CaMKIV), cyclic AMP response element (CRE)-binding protein (CREB) and brain-derived neurotrophic factor (BDNF), a major regulator of survival. We have shown that intracerebroventricular injections of anti-BDNF and anti-CREB are sufficient to cause substantial tissular damages and apoptotic deaths in late periods (48-72 h) after ischemia. Herein, we provide immunohistochemical and biochemical evidence that antibody-induced impairment of the protective CaMKIV/CREB/BDNF pathway induces an apparent duality of response in the dentate gyrus. The experimental protocol is performed as follows: (a) rats are anesthetized and vertebral arteries are occluded by electrocauterization; (b) on the following day, transient global ischemia is produced by occlusion of carotid arteries for 25 min; (c) finally, rats are infused with the pharmacologic agents into the left cerebral ventricle and then perfusion-fixed at different time points after ischemia for immunohistochemical and immunoblotting analyses. After infusion with anti-CaMKIV, phosphorylation of mitogen-activated protein kinases (MAPK) MKK3, MKK6 and p38 and phospho-acetylation of histone H3 occur at 6 h after ischemia without presence of any caspase-9 activation and cellular injuries. In contrast, infusion of anti-BDNF or anti-CREB surprisingly results in a remarkable stimulation of casein kinase 2 (CK2) and caspase-9 activities at 48-72 h post-insult. This is accompanied by the disappearance of phosphorylation of MKK(3/6) and p38 and phospho-acetylation of histone H3. These results suggest that: (1) activation of a MKK(3/6)/p38/H3 cascade at early periods post-ischemia may be capable of causing a short transient protective effect in the dentate gyrus; (2) CK2 might be implicated in inhibition of activity of molecules such as MKK(3/6), p38 and deacetylases at late periods post-insult, thereby promoting injuries and cell deaths in the dentate cell layer.
Collapse
Affiliation(s)
- P R Blanquet
- Laboratoire Développement et Vieillissement du Système Nerveux, UMR 7102 CNRS-UPMC (Neurobiologie des Processus Adaptatifs), Université P & M Curie, 9 Quai Saint-Bernard, Bâtiment B, 4(e)Etage, Boîte 14, 75005, Paris, France.
| | | | | |
Collapse
|
41
|
Beltran WA, Allore HG, Johnson E, Towle V, Tao W, Acland GM, Aguirre GD, Zeiss CJ. CREB1/ATF1 activation in photoreceptor degeneration and protection. Invest Ophthalmol Vis Sci 2009; 50:5355-63. [PMID: 19643965 DOI: 10.1167/iovs.09-3741] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The cAMP response element binding protein 1 (CREB1) and activating transcription factor 1 (ATF1) are closely related members of the bZIP superfamily of transcription factors. Both are activated in response to a wide array of stimuli, including cellular stress. This study was conducted to assess the CREB1/ATF1 pathway in photoreceptor disease and protection. METHODS The expression levels of p-CREB1, CREB1, and ATF1 were examined by immunoblot and immunohistochemistry in normal canine retina and retinas of several canine models of retinal degeneration (rcd1, rcd2, erd, prcd, XLPRA1, XLPRA2, T4R RHO). Humans retinas affected with age-related macular degeneration (AMD) were also examined. p-CREB1/ATF1 immunolabeling was assessed in normal and rcd1 dogs treated with ciliary neurotrophic factor (CNTF), to examine the effect of a neuroprotective stimulus on activation of CREB1/ATF1. RESULTS Native CREB1 and ATF1 as well as phosphorylated CREB1/ATF1 was examined in normal canine retina by immunoblot. The p-CREB1 antibody identified phosphorylated CREB1 and ATF1 and labeled the inner retina only in normal dogs. In degenerate canine and human retinas, strong immunolabeling appeared in rod and cone photoreceptors, indicating increased expression of native CREB1 and ATF1, as well as increased phosphorylation of these proteins. Retinal protection by CNTF in rcd1 dogs was accompanied by a significant increase in the number of p-CREB1/ATF1-labeled photoreceptor nuclei. CONCLUSIONS Positive association of CREB1/ATF1 phosphorylation with photoreceptor protection suggests that it may contribute to an innate protective response. These data identify a signaling mechanism in rods and cones of potential importance for therapies of RP and AMD.
Collapse
Affiliation(s)
- William A Beltran
- Section of Ophthalmology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhong J, Zhao L, Du Y, Wei G, Yao WG, Lee WH. Delayed IGF-1 treatment reduced long-term hypoxia-ischemia-induced brain damage and improved behavior recovery of immature rats. Neurol Res 2009; 31:483-9. [PMID: 19500451 DOI: 10.1179/174313208x338133] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Cerebral hypoxia-ischemia during the perinatal period is the single most important cause of acute newborn mortality and chronic disability. Despite our increasing understanding of the mechanisms of neuronal injury, an effective clinical therapy has yet to be established to mitigate brain damage and improve the prognosis and well-being of these newborn patients. Insulin-like growth factor 1 (IGF-1) is a well-known neurotrophic factor, essential for the survival and functional maturation of immature neurons. This study demonstrated that subcutaneous administration of IGF-1 at 24 and 48 hours of recovery significantly reduced hypoxia-ischemia-induced injury to immature rat brains and improved long-term memory and cognitive behavior. IGF-1's therapeutic effects likely involve its ability to prevent delayed apoptosis, as we demonstrated in primary cortical neuronal cultures under oxygen and glucose deprivation. IGF-1's neuroprotective effects parallel the activities of phosphatidylinositol-3/Akt and its down-stream signaling pathway, suggesting a potential mechanistic link. Overall, evidence from this investigation strongly supports IGF-1's potential therapeutic use in the treatment of hypoxic-ischemic encephalopathy in newborn patients.
Collapse
Affiliation(s)
- Jin Zhong
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | |
Collapse
|
43
|
Lai M, Bae SE, Bell JE, Seckl JR, Macleod MR. Mineralocorticoid receptor mRNA expression is increased in human hippocampus following brief cerebral ischaemia. Neuropathol Appl Neurobiol 2009; 35:156-64. [PMID: 19284477 DOI: 10.1111/j.1365-2990.2008.00980.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND We have previously reported that neuronal endangerment in vitro and hypothermic transient global ischaemia in vivo each result in increased mineralocorticoid receptor (MR) expression. In both models MR induction is associated with increased neuronal survival, and blocking MR signalling reduces neuronal survival. Furthermore, transgenic overexpression of human MR promotes neuronal survival both in vitro and in vivo. AIMS Here we have assessed whether brief periods of cerebral ischaemia in human subjects, such as occurs in cardiac arrest from which successful resuscitation is achieved, are associated with a sustained increase in hippocampal MR mRNA expression. METHODS Human post-mortem brain sections from patients who had died in the weeks following cardiac arrest were analysed for MR mRNA expression by in situ hybridization. RESULTS Sustained upregulation of MR mRNA expression was observed in the dentate gyrus region of human hippocampus following a brief episode of cerebral ischaemia. CONCLUSIONS This confirms that MR mRNA expression is regulated following neuronal injury in human brain, and suggests that the benefits of increased MR expression seen in animal models of ischaemia may also be observed in humans.
Collapse
Affiliation(s)
- M Lai
- Endocrinology Unit, Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK.
| | | | | | | | | |
Collapse
|
44
|
Gill MB, Bockhorst K, Narayana P, Perez-Polo JR. Bax shuttling after neonatal hypoxia-ischemia: hyperoxia effects. J Neurosci Res 2009; 86:3584-604. [PMID: 18655197 DOI: 10.1002/jnr.21795] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Perinatal hypoxia-ischemia (HI) occurs in 0.2%-0.4% of all live births, with 100% O(2) resuscitation (HHI) remaining a standard clinical treatment. HI produces a broad spectrum of neuronal death phenotypes ranging from a more noninflammatory apoptotic death to a more inflammatory necrotic cell death that may be responsible for the broad spectrum of reported dysfunctional outcomes. However, the mechanisms that would account for this phenotypic spectrum of cell death are not fully understood. Here, we provide evidence that Bcl-2-associated X protein (Bax) can shuttle to different subcellular compartments in response to HI, thus triggering the different organelle-associated cell death signaling cascades resulting in cell death phenotype diversity. There was an early increase in intranuclear and total nuclear Bax protein levels followed by a later Bax redistribution to the mitochondria and endoplasmic reticulum (ER). Associated with the organelle-specific Bax shuttling time course, there was an increase in nuclear phosphorylated p53, cytosolic cleaved caspase-3, and caspase-12. When HI-treated P7 rats were resuscitated with 100% O(2) (HHI), there were increased lesion volumes as determined by T2-weighted magnetic resonance imaging with no change in cortical apoptotic signaling compared with HI treatment alone. There was, however, increased inflammatory (cytosolic-cleaved interleukin-1beta) and necrotic (increased nuclear 55-kDa-cleaved PARP-1 [poly-ADP-ribose 1] and decreased nuclear HMGB1 [nuclear high-mobility group box 1]) after HHI. Furthermore, HHI increased ER calpain activation and ER Bax protein levels compared with HI alone. These data suggest that 100% O(2) resuscitation increases Bax-mediated activation of ER cell death signaling, inflammation, and lesion volume by increasing necrotic-like cell death. In light of these findings, the use of 100% O(2) treatment for neonatal HI should be reevaluated.
Collapse
Affiliation(s)
- Martin B Gill
- Department of Neuroscience and Cell Biology, University of Texas-Medical Branch, Galveston, TX 77555-1072, USA
| | | | | | | |
Collapse
|
45
|
Nishigori H, Mazzuca DM, Nygard KL, Han VK, Richardson BS. BDNF and TrkB in the preterm and near-term ovine fetal brain and the effect of intermittent umbilical cord occlusion. Reprod Sci 2009; 15:895-905. [PMID: 19050322 DOI: 10.1177/1933719108324135] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We have determined the developmental change in immunoreactivity for brain-derived neurotrophic factor and its high-affinity tyrosine kinase receptor, TrkB, in the ovine fetal brain with advancing gestation and in response to intermittent umbilical cord occlusion, which might then contribute to adverse neurodevelopment. Fetal sheep (control and experimental groups at 0.75 and 0.90 of gestation) were studied over 4 days with umbilical cord occlusions performed in the experimental group animals by complete inflation of an occluder cuff for 90 seconds every 30 minutes for 3 to 5 hours each day. Animals were then euthanized and the fetal brains perfusion fixed and prepared for subsequent histology with the distribution of brain-derived neurotrophic factor and tyrosine kinase receptor immunoreactivity determined by immunohistochemistry. In the control group animals brain-derived neurotrophic factor immunoreactivity decreased in the gray matter, thalamus, and hippocampus but increased in the white matter, while tyrosine kinase receptor immunoreactivity decreased in all regions (most P < .01), with advancing gestation consistent with the developmental change from neurogenesis/gliagenesis to myelination over this time period. Intermittent umbilical cord occlusion as studied with severe but limited hypoxemia resulted in a variable decrease in brain-derived neurotrophic factor and tyrosine kinase receptor immunoreactivity for all brain regions in the preterm animals (most P < .01) when protein turnover is higher, but a selective increase in brain-derived neurotrophic factor immunoreactivity in the hippocampus of the near-term animals consistent with a heightened vulnerability for necrotic/apoptotic injury at this time. As such, brain-derived neurotrophic factor-tyrosine kinase receptor in the ovine fetal brain may be altered with intermittent hypoxic insults over the latter part of pregnancy with potential for longer term neurologic consequences.
Collapse
Affiliation(s)
- Hidekazu Nishigori
- Department of Obstetrics and Gynaecology, The Children's Health Research Institute, The University of Western Ontario, London, Canada
| | | | | | | | | |
Collapse
|
46
|
Schmid-Brunclik N, Bürgi-Taboada C, Antoniou X, Gassmann M, Ogunshola OO. Astrocyte responses to injury: VEGF simultaneously modulates cell death and proliferation. Am J Physiol Regul Integr Comp Physiol 2008; 295:R864-73. [PMID: 18614764 DOI: 10.1152/ajpregu.00536.2007] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypoxia is linked to changes in blood-brain barrier (BBB) permeability, and loss of BBB integrity is characteristic of many pathological brain diseases including stroke. In particular, astrocytes play a central role in brain homeostasis and BBB function. We investigated how hypoxia affects astrocyte survival and assessed whether VEGF release through hypoxia-inducible factor-1alpha (HIF-1alpha) induction plays a role in tolerance of these cells to insult. Thus primary astrocytes were subjected to normoxic (21% O(2)), hypoxic (1% O(2)), or near-anoxic (<0.1% O(2)) conditions in the presence or absence of glucose. Cell death was significantly initiated after combined oxygen glucose deprivation, and, surprisingly, astrocyte proliferation increased concomitantly. Near anoxic, but not hypoxic, conditions stabilized HIF-1alpha protein and provoked DNA binding activity, whereas oxygen and glucose deprivation accelerated HIF-1alpha accumulation. Unexpectedly, Hif-1alpha knockdown studies showed that elevated VEGF levels following increased insult was only partially due to HIF-1alpha induction, suggesting alternative mechanisms of VEGF regulation. Notably, endogenous VEGF signaling during insult was essential for cell fate since VEGF inhibition appreciably augmented cell death and reduced proliferation. These data suggest Hif-1 only partially contributes to VEGF-mediated astrocyte responses during chronic injury (as occurs in clinical hypoxic/ischemic insults) that may ultimately be responsible for disrupting BBB integrity.
Collapse
Affiliation(s)
- Nicole Schmid-Brunclik
- Institute of Veterinary Physiology, Vetsuisse Faculty, Univ. of Zurich, Winterthurerstrasse 260, Zurich CH 8057, Switzerland
| | | | | | | | | |
Collapse
|
47
|
Ying Z, Roy RR, Zhong H, Zdunowski S, Edgerton VR, Gomez-Pinilla F. BDNF-exercise interactions in the recovery of symmetrical stepping after a cervical hemisection in rats. Neuroscience 2008; 155:1070-8. [PMID: 18672032 DOI: 10.1016/j.neuroscience.2008.06.057] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Revised: 06/07/2008] [Accepted: 06/28/2008] [Indexed: 10/21/2022]
Abstract
Clinical evidence indicates that motor training facilitates functional recovery after a spinal cord injury (SCI). Brain-derived neurotrophic factor (BDNF) is a powerful synaptic facilitator and likely plays a key role in motor and sensory functions. Spinal cord hemisection decreases the levels of BDNF below the injury site, and exercise can counteract this decrease [Ying Z, Roy RR, Edgerton VR, Gomez-Pinilla F (2005) Exercise restores levels of neurotrophins and synaptic plasticity following spinal cord injury. Exp Neurol 193:411-419]. It is not clear, however, whether the exercise-induced increases in BDNF play a role in mediating the recovery of locomotion after a SCI. We performed a lateral cervical ( approximately C4) hemisection in adult rats. Seven days after hemisection, the BDNF inhibitor trkB IgG was injected into the cervical spinal cord below the lesion ( approximately C5-C6). Half of the rats were exposed to voluntary running wheels for 14 days. Locomotor ability was assessed by determining the symmetry between the contralateral (unaffected) vs. the ipsilateral (affected) forelimb at the most optimum treadmill speed for each rat. Sedentary and exercised rats with BDNF inhibition showed a higher level of asymmetry during the treadmill locomotion test than rats not treated with the BDNF inhibitor. In hemisected rats, exercise normalized the levels of molecules important for synaptic function, such as cyclic AMP response element binding protein (CREB) and synapsin I, in the ipsilateral cervical enlargement, whereas the BDNF blocker lessened these exercise-associated effects. The results indicate that BDNF levels play an important role in shaping the synaptic plasticity and in defining the level of recovery of locomotor performance after a SCI.
Collapse
Affiliation(s)
- Z Ying
- Department of Physiological Science, University of California, Los Angeles, CA 90095-1527, USA
| | | | | | | | | | | |
Collapse
|
48
|
Neuroprotection of brain‐derived neurotrophic factor against hypoxic injury
in vitro
requires activation of extracellular signal‐regulated kinase and phosphatidylinositol 3‐kinase. Int J Dev Neurosci 2007; 26:363-70. [PMID: 18243629 DOI: 10.1016/j.ijdevneu.2007.11.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 11/16/2007] [Accepted: 11/29/2007] [Indexed: 11/22/2022] Open
|
49
|
Abstract
A atividade física é conhecida por promover saúde e bem-estar. O exercício também é responsável por aumentar a produção de Espécies Reativas de Oxigênio (ERO) pelo acréscimo do consumo de oxigênio mitocondrial nos tecidos. O desequilíbrio entre a produção de EROs e as defesas oxidantes dos tecidos pode provocar danos oxidativos a proteínas, lipídios e DNA. O dano oxidativo cerebral é um mecanismo etiopatológico comum da apoptose e da neurodegeneração. O fator de crescimento cérebro-derivado desempenha um importante papel neste contexto. Nesta revisão, apresentamos os resultados de diferentes modelos de exercício físico no metabolismo oxidativo e neurotrófico do Sistema Nervoso Central (SNC). Também revisamos estudos que utilizaram suplementação antioxidante para prevenir danos oxidativos exercício-induzido ao SNC. Os modelos de exercício físico mais comuns foram as rodas de correr, a natação e a esteira com configurações de treinamento muito diferentes como a duração e a intensidade. Os resultados do treinamento físico no tecido cerebral são muito controversos, mas geralmente demonstram ganhos na plasticidade sináptica e na função cognitiva com exercícios de intensidade moderada e baixa.
Collapse
|
50
|
Wang C, Wang H. Protective roles of heat stress on the neurons in hippocampal CA1 region of mice. ACTA ACUST UNITED AC 2007; 1:418-22. [DOI: 10.1007/s11684-007-0082-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|