1
|
Harris JP, Mietus CJ, Browne KD, Wofford KL, Keating CE, Brown DP, Johnson BN, Wolf JA, Smith DH, Cohen AS, Duda JE, Cullen DK. Neuronal somatic plasmalemmal permeability and dendritic beading caused by head rotational traumatic brain injury in pigs-An exploratory study. Front Cell Neurosci 2023; 17:1055455. [PMID: 37519631 PMCID: PMC10381956 DOI: 10.3389/fncel.2023.1055455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 06/23/2023] [Indexed: 08/01/2023] Open
Abstract
Closed-head traumatic brain injury (TBI) is induced by rapid motion of the head, resulting in diffuse strain fields throughout the brain. The injury mechanism(s), loading thresholds, and neuroanatomical distribution of affected cells remain poorly understood, especially in the gyrencephalic brain. We utilized a porcine model to explore the relationships between rapid head rotational acceleration-deceleration loading and immediate alterations in plasmalemmal permeability within cerebral cortex, sub-cortical white matter, and hippocampus. To assess plasmalemmal compromise, Lucifer yellow (LY), a small cell-impermeant dye, was delivered intraventricularly and diffused throughout the parenchyma prior to injury in animals euthanized at 15-min post-injury; other animals (not receiving LY) were survived to 8-h or 7-days. Plasmalemmal permeability preferentially occurred in neuronal somata and dendrites, but rarely in white matter axons. The burden of LY+ neurons increased based on head rotational kinematics, specifically maximum angular velocity, and was exacerbated by repeated TBI. In the cortex, LY+ cells were prominent in both the medial and lateral gyri. Neuronal membrane permeability was observed within the hippocampus and entorhinal cortex, including morphological changes such as beading in dendrites. These changes correlated with reduced fiber volleys and synaptic current alterations at later timepoints in the hippocampus. Further histological observations found decreased NeuN immunoreactivity, increased mitochondrial fission, and caspase pathway activation in both LY+ and LY- cells, suggesting the presence of multiple injury phenotypes. This exploratory study suggests relationships between plasmalemmal disruptions in neuronal somata and dendrites within cortical and hippocampal gray matter as a primary response in closed-head rotational TBI and sets the stage for future, traditional hypothesis-testing experiments.
Collapse
Affiliation(s)
- James P. Harris
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Constance J. Mietus
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kevin D. Browne
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Kathryn L. Wofford
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Carolyn E. Keating
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Daniel P. Brown
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Brian N. Johnson
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Research Institute, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - John A. Wolf
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Douglas H. Smith
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Akiva S. Cohen
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Research Institute, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - John E. Duda
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - D. Kacy Cullen
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
2
|
Traumatic Brain Injury Preserves Firing Rates But Disrupts Laminar Oscillatory Coupling and Neuronal Entrainment in Hippocampal CA1. eNeuro 2020; 7:ENEURO.0495-19.2020. [PMID: 32737188 PMCID: PMC7477953 DOI: 10.1523/eneuro.0495-19.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 07/15/2020] [Accepted: 07/19/2020] [Indexed: 11/21/2022] Open
Abstract
While hippocampal-dependent learning and memory are particularly vulnerable to traumatic brain injury (TBI), the functional status of individual hippocampal neurons and their interactions with oscillations are unknown following injury. Using the most common rodent TBI model and laminar recordings in CA1, we found a significant reduction in oscillatory input into the radiatum layer of CA1 after TBI. Surprisingly, CA1 neurons maintained normal firing rates despite attenuated input, but did not maintain appropriate synchronization with this oscillatory input or with local high-frequency oscillations. Normal synchronization between these coordinating oscillations was also impaired. Simultaneous recordings of medial septal neurons known to participate in theta oscillations revealed increased GABAergic/glutamatergic firing rates postinjury under anesthesia, potentially because of a loss of modulating feedback from the hippocampus. These results suggest that TBI leads to a profound disruption of connectivity and oscillatory interactions, potentially disrupting the timing of CA1 neuronal ensembles that underlie aspects of learning and memory.
Collapse
|
3
|
Ali ZS, Johnson VE, Stewart W, Zager EL, Xiao R, Heuer GG, Weber MT, Mallela AN, Smith DH. Neuropathological Characteristics of Brachial Plexus Avulsion Injury With and Without Concomitant Spinal Cord Injury. J Neuropathol Exp Neurol 2016; 75:69-85. [PMID: 26671984 PMCID: PMC6322589 DOI: 10.1093/jnen/nlv002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neonatal brachial plexus avulsion injury (BPAI) commonly occurs as a consequence of birth trauma and can result in lifetime morbidity; however, little is known regarding the evolving neuropathological processes it induces. In particular, mechanical forces during BPAI can concomittantly damage the spinal cord and may contribute to outcome. Here, we describe the functional and neuropathological outcome following BPAI, with or without spinal cord injury, in a novel pediatric animal model. Twenty-eight-day-old piglets underwent unilateral C5–C7 BPAI with and without limited myelotomy. Following avulsion, all animals demonstrated right forelimb monoparesis. Injury extending into the spinal cord conferred greater motor deficit, including long tract signs. Consistent with clinical observations, avulsion with myelotomy resulted in more severe neuropathological changes with greater motor neuron death, progressive axonopathy, and persistent glial activation. These data demonstrate neuropathological features of BPAI associated with poor functional outcome. Interestingly, in contrast to adult small animal models of BPAI, a degree of motor neuron survival was observed, even following severe injury in this neonatal model. If this is also the case in human neonatal BPAI, repair may permit functional restoration. This model also provides a clinically relevant platform for exploring the complex postavulsion neuropathological responses that may inform therapeutic strategies.
Collapse
Affiliation(s)
- Zarina S. Ali
- From the Department of Neurosurgery (ZAS, VEJ, WS, ELZ, MTW, ANM, DHS), Penn Center for Brain Injury and Repair (ZAS, VEJ, WS, MTW, ANM, DHS), Department of Biostatistics and Epidemiology (RX), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, UK (WS); Division of Neurosurgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania (GGH)
| | - Victoria E. Johnson
- From the Department of Neurosurgery (ZAS, VEJ, WS, ELZ, MTW, ANM, DHS), Penn Center for Brain Injury and Repair (ZAS, VEJ, WS, MTW, ANM, DHS), Department of Biostatistics and Epidemiology (RX), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, UK (WS); Division of Neurosurgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania (GGH)
| | - William Stewart
- From the Department of Neurosurgery (ZAS, VEJ, WS, ELZ, MTW, ANM, DHS), Penn Center for Brain Injury and Repair (ZAS, VEJ, WS, MTW, ANM, DHS), Department of Biostatistics and Epidemiology (RX), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, UK (WS); Division of Neurosurgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania (GGH)
| | - Eric L. Zager
- From the Department of Neurosurgery (ZAS, VEJ, WS, ELZ, MTW, ANM, DHS), Penn Center for Brain Injury and Repair (ZAS, VEJ, WS, MTW, ANM, DHS), Department of Biostatistics and Epidemiology (RX), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, UK (WS); Division of Neurosurgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania (GGH)
| | - Rui Xiao
- From the Department of Neurosurgery (ZAS, VEJ, WS, ELZ, MTW, ANM, DHS), Penn Center for Brain Injury and Repair (ZAS, VEJ, WS, MTW, ANM, DHS), Department of Biostatistics and Epidemiology (RX), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, UK (WS); Division of Neurosurgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania (GGH)
| | - Gregory G. Heuer
- From the Department of Neurosurgery (ZAS, VEJ, WS, ELZ, MTW, ANM, DHS), Penn Center for Brain Injury and Repair (ZAS, VEJ, WS, MTW, ANM, DHS), Department of Biostatistics and Epidemiology (RX), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, UK (WS); Division of Neurosurgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania (GGH)
| | - Maura T. Weber
- From the Department of Neurosurgery (ZAS, VEJ, WS, ELZ, MTW, ANM, DHS), Penn Center for Brain Injury and Repair (ZAS, VEJ, WS, MTW, ANM, DHS), Department of Biostatistics and Epidemiology (RX), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, UK (WS); Division of Neurosurgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania (GGH)
| | - Arka N. Mallela
- From the Department of Neurosurgery (ZAS, VEJ, WS, ELZ, MTW, ANM, DHS), Penn Center for Brain Injury and Repair (ZAS, VEJ, WS, MTW, ANM, DHS), Department of Biostatistics and Epidemiology (RX), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, UK (WS); Division of Neurosurgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania (GGH)
| | - Douglas H. Smith
- From the Department of Neurosurgery (ZAS, VEJ, WS, ELZ, MTW, ANM, DHS), Penn Center for Brain Injury and Repair (ZAS, VEJ, WS, MTW, ANM, DHS), Department of Biostatistics and Epidemiology (RX), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, UK (WS); Division of Neurosurgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania (GGH)
| |
Collapse
|
4
|
Alekseeva OS, Gusel’nikova VV, Beznin GV, Korzhevskii DE. Prospects for the application of neun nuclear protein as a marker of the functional state of nerve cells in vertebrates. J EVOL BIOCHEM PHYS+ 2015. [DOI: 10.1134/s0022093015050014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
5
|
Kobeissy FH, Liu MC, Yang Z, Zhang Z, Zheng W, Glushakova O, Mondello S, Anagli J, Hayes RL, Wang KK. Degradation of βII-Spectrin Protein by Calpain-2 and Caspase-3 Under Neurotoxic and Traumatic Brain Injury Conditions. Mol Neurobiol 2015; 52:696-709. [PMID: 25270371 PMCID: PMC4383741 DOI: 10.1007/s12035-014-8898-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/10/2014] [Indexed: 12/22/2022]
Abstract
A major consequence of traumatic brain injury (TBI) is the rapid proteolytic degradation of structural cytoskeletal proteins. This process is largely reflected by the interruption of axonal transport as a result of extensive axonal injury leading to neuronal cell injury. Previous work from our group has described the extensive degradation of the axonally enriched cytoskeletal αII-spectrin protein which results in molecular signature breakdown products (BDPs) indicative of injury mechanisms and to specific protease activation both in vitro and in vivo. In the current study, we investigated the integrity of βII-spectrin protein and its proteolytic profile both in primary rat cerebrocortical cell culture under apoptotic, necrotic, and excitotoxic challenge and extended to in vivo rat model of experimental TBI (controlled cortical impact model). Interestingly, our results revealed that the intact 260-kDa βII-spectrin is degraded into major fragments (βII-spectrin breakdown products (βsBDPs)) of 110, 108, 85, and 80 kDa in rat brain (hippocampus and cortex) 48 h post-injury. These βsBDP profiles were further characterized and compared to an in vitro βII-spectrin fragmentation pattern of naive rat cortex lysate digested by calpain-2 and caspase-3. Results revealed that βII-spectrin was degraded into major fragments of 110/85 kDa by calpain-2 activation and 108/80 kDa by caspase-3 activation. These data strongly support the hypothesis that in vivo activation of multiple protease system induces structural protein proteolysis involving βII-spectrin proteolysis via a specific calpain and/or caspase-mediated pathway resulting in a signature, protease-specific βsBDPs that are dependent upon the type of neural injury mechanism. This work extends on previous published work that discusses the interplay spectrin family (αII-spectrin and βII-spectrin) and their susceptibility to protease proteolysis and their implication to neuronal cell death mechanisms.
Collapse
Affiliation(s)
- Firas H Kobeissy
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Ming Cheng Liu
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Zhihui Yang
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Zhiqun Zhang
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Wenrong Zheng
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| | - Olena Glushakova
- Banyan Laboratory, Banyan Biomarkers, Inc., Alachua, FL 32615,
USA
| | - Stefania Mondello
- Department of Neurosciences, University of Messina, 98125
Messina, Italy
| | - John Anagli
- Banyan Laboratory, Banyan Biomarkers, Inc., Alachua, FL 32615,
USA
| | - Ronald L. Hayes
- Banyan Laboratory, Banyan Biomarkers, Inc., Alachua, FL 32615,
USA
| | - Kevin K.W. Wang
- Center for Neuroproteomics & Biomarkers Research, Department
of Psychiatry, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
6
|
Siman R, Lin YG, Malthankar-Phatak G, Dong Y. A rapid gene delivery-based mouse model for early-stage Alzheimer disease-type tauopathy. J Neuropathol Exp Neurol 2013; 72:1062-71. [PMID: 24128676 PMCID: PMC3815088 DOI: 10.1097/nen.0000000000000006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The perforant pathway projection from the entorhinal cortex (EC) to the hippocampal dentate gyrus is critically important for long-term memory and develops tau and amyloid pathologies and progressive degeneration starting in the early stages of Alzheimer disease (AD). However, perforant pathway function has not been assessed in experimental models of AD, and a therapeutic agent that protects its structure and function has not yet been identified. Therefore, we developed a new adeno-associated virus-based mouse model for perforant pathway tauopathy. Microinjection into the lateral EC of vectors designed to express either human tau bearing a pathogenic P301L mutation or enhanced green fluorescent protein as a control selectively drove transgene expression in lateral EC layer II perikarya and along the entire rostrocaudal extent of the lateral perforant pathway afferents and dentate terminal field. After human tau expression, hyperphosphorylated tau accumulated only within EC layer II perikarya, thereby modeling Braak stage I of transentorhinal AD tauopathy. Expression of pathologic human tau but not enhanced green fluorescent protein led to specific dose-dependent apoptotic death of perforant pathway neurons and loss of synapses in as little as 2 weeks. This novel adeno-associated virus-based method elicits rapid tauopathy and tau-mediated neurodegeneration localized to the mouse perforant pathway and represents a new experimental approach for studying tau-driven pathogenic processes and tau-based treatment strategies in a highly vulnerable neural circuit.
Collapse
Affiliation(s)
- Robert Siman
- From the Department of Neurosurgery and Center for Brain Injury and Repair, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | | | | |
Collapse
|
7
|
Peng G, Yuan Y, He Q, Wu W, Luo BY. MicroRNA let-7e regulates the expression of caspase-3 during apoptosis of PC12 cells following anoxia/reoxygenation injury. Brain Res Bull 2011; 86:272-6. [PMID: 21827835 DOI: 10.1016/j.brainresbull.2011.07.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 06/28/2011] [Accepted: 07/24/2011] [Indexed: 12/30/2022]
Abstract
This study aimed to investigate the role and mechanism of action of microRNA (miR) let-7e in PC12 cells undergoing apoptosis following anoxia/reoxygenation (A/R) injury. The putative binding site of let-7e in the 3' UTR of caspase-3 (Casp3) mRNA was analyzed using the miRanda algorithm. Precursor let-7e (pre-miRNA), let-7e miR and anti-let-7e oligonucleotides were transfected into PC12 cells, which were then subjected to A/R injury. The levels of Casp3 mRNA and let-7e miRNA, the total protein levels of Casp3, Casp8 and Casp9 and levels of cellular apoptosis were measured. It was found that let-7e expression in PC12 cells was decreased, whereas the expression of Casp3 was significantly increased after A/R injury. The transfection of pre-miRNA or let-7e miR into PC12 cells decreased Casp3 expression levels and cellular apoptosis following A/R injury, while co-transfection of anti-let-7e strikingly alleviated the effects of let-7e miR. These results indicate that let-7e may protect PC12 cells against apoptosis following A/R injury by negatively regulating the expression of Casp3.
Collapse
Affiliation(s)
- Guoping Peng
- Department of Neurology, Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| | | | | | | | | |
Collapse
|
8
|
Morales P, Bustamante D, Espina-Marchant P, Neira-Peña T, Gutiérrez-Hernández MA, Allende-Castro C, Rojas-Mancilla E. Pathophysiology of perinatal asphyxia: can we predict and improve individual outcomes? EPMA J 2011. [PMID: 23199150 PMCID: PMC3405380 DOI: 10.1007/s13167-011-0100-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Perinatal asphyxia occurs still with great incidence whenever delivery is prolonged, despite improvements in perinatal care. After asphyxia, infants can suffer from short- to long-term neurological sequelae, their severity depend upon the extent of the insult, the metabolic imbalance during the re-oxygenation period and the developmental state of the affected regions. Significant progresses in understanding of perinatal asphyxia pathophysiology have achieved. However, predictive diagnostics and personalised therapeutic interventions are still under initial development. Now the emphasis is on early non-invasive diagnosis approach, as well as, in identifying new therapeutic targets to improve individual outcomes. In this review we discuss (i) specific biomarkers for early prediction of perinatal asphyxia outcome; (ii) short and long term sequelae; (iii) neurocircuitries involved; (iv) molecular pathways; (v) neuroinflammation systems; (vi) endogenous brain rescue systems, including activation of sentinel proteins and neurogenesis; and (vii) therapeutic targets for preventing or mitigating the effects produced by asphyxia.
Collapse
Affiliation(s)
- Paola Morales
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Diego Bustamante
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Pablo Espina-Marchant
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Tanya Neira-Peña
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Manuel A. Gutiérrez-Hernández
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Camilo Allende-Castro
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| | - Edgardo Rojas-Mancilla
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70.000, Santiago 7, Chile
| |
Collapse
|
9
|
Abstract
Brain plasticity describes the potential of the organ for adaptive changes involved in various phenomena in health and disease. A substantial amount of experimental evidence, received in animal and cell models, shows that a cascade of plastic changes at the molecular, cellular, and tissue levels, is initiated in different regions of the postischemic brain. Underlying mechanisms include neurochemical alterations, functional changes in excitatory and inhibitory synapses, axonal and dendritic sprouting, and reorganization of sensory and motor central maps. Multiple lines of evidence indicate numerous points in which the process of postischemic recovery may be influenced with the aim to restore the full capacity of the brain tissue injured by an ischemic episode.
Collapse
Affiliation(s)
- Galyna G Skibo
- Department of Cytology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | | |
Collapse
|
10
|
Morales P, Simola N, Bustamante D, Lisboa F, Fiedler J, Gebicke-Haerter PJ, Morelli M, Tasker RA, Herrera-Marschitz M. Nicotinamide prevents the long-term effects of perinatal asphyxia on apoptosis, non-spatial working memory and anxiety in rats. Exp Brain Res 2009; 202:1-14. [PMID: 20012537 DOI: 10.1007/s00221-009-2103-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 11/13/2009] [Indexed: 01/04/2023]
Abstract
There is no established treatment for the long-term effects produced by perinatal asphyxia. Thus, we investigated the neuroprotection provided by nicotinamide against the effects elicited by perinatal asphyxia on hippocampus and behaviour observed at 30-90 days of age. Asphyxia was induced by immersing foetuses-containing uterine horns, removed from ready-to-deliver rats into a water bath at 37 degrees C for 20 min. Caesarean-delivered siblings were used as controls. Saline or nicotinamide (0.8 mmol/kg, i.p.) was administered to control and asphyxia-exposed animals 24, 48, and 72 h after birth. The animals were examined for morphological changes in hippocampus, focusing on delayed cell death and mossy fibre sprouting, and behaviour, focusing on cognitive behaviour and anxiety. At the age of 30-45 days, asphyxia-exposed rats displayed (1) increased apoptosis, assessed in whole hippocampus by nuclear Hoechst staining, and (2) increased mossy fibre sprouting, restricted to the stratum oriens of dorsal hippocampus, assessed by Timm's staining. Rats from the same cohorts displayed (3) deficits in non-spatial working memory, assessed by a novel object recognition task, and (4) increased anxiety, assessed by an elevated plus-maze test when examined at the age of 90 days. Nicotinamide prevented the effects elicited by perinatal asphyxia on apoptosis, working memory, and anxiety.
Collapse
Affiliation(s)
- Paola Morales
- Programme of Molecular and Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, PO Box 70,000, Santiago 7, Chile.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Bai Y, Meng Z, Cui M, Zhang X, Chen F, Xiao J, Shen L, Zhang Y. An Ang1-Tie2-PI3K axis in neural progenitor cells initiates survival responses against oxygen and glucose deprivation. Neuroscience 2009; 160:371-81. [PMID: 19409199 DOI: 10.1016/j.neuroscience.2009.01.076] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 01/23/2009] [Accepted: 01/23/2009] [Indexed: 12/25/2022]
Abstract
Neural progenitor cells (NPCs) have the potential to survive brain ischemia and participate in neurogenesis after stroke. However, it is not clear how survival responses are initiated in NPCs. Using embryonic mouse NPCs and the in vitro oxygen and glucose deprivation (OGD) model, we found that angiopoietin-1 (Ang1) could prevent NPCs from OGD-induced apoptosis, as evidenced by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and annexin V labeling. Ang1 significantly elevated tunica intima endothelial kinase 2 (Tie2) autophosphorylation level, suggesting the existence of functional Tie2 receptors on NPCs. NPCs under OGD conditions exhibited reduction of Akt phosphorylation, decrease of the Bcl-2/Bax ratio, activation of caspase-3, cleavage of PARP, and downregulation of beta-catenin and nestin. Ang1 reversed the above changes concomitantly with significant rising of survival rates of NPCs under OGD, but all these effects of Ang1 could be blocked by either soluble extracellular domain of Tie2 Fc fusion protein (sTie2Fc) or the phosphoinositide 3-kinase (PI3K) inhibitor 2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one (LY294002). Our findings suggest the existence of an Ang1-Tie2-PI3K signaling axis that is essential in initiation of survival responses in NPCs against cerebral ischemia and hypoxia.
Collapse
Affiliation(s)
- Y Bai
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 XueYuan Road, Beijing, 100191, China
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Ryu JS, Ko K, Lee JW, Park SB, Byun SJ, Jeong EJ, Ko K, Choo YK. Gangliosides are involved in neural differentiation of human dental pulp-derived stem cells. Biochem Biophys Res Commun 2009; 387:266-71. [PMID: 19580786 DOI: 10.1016/j.bbrc.2009.07.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Accepted: 07/01/2009] [Indexed: 01/09/2023]
Abstract
Human dental pulp-derived stem cells (hDPSCs) have been considered alternative sources of adult stem cells because of their potential to differentiate into multiple cell lineages. This study investigated the possible role of gangliosides in the neural differentiation of hDPSCs. When hDPSCs were cultured under neural differentiation conditions, expression of neural cell marker genes such as Nestin, MAP-2, and NeuN was detected. Immunostaining and high-performance thin-layer chromatography analysis showed that an increase in ganglioside biosynthesis was associated with neural differentiation of hDPSCs. Specifically, a significant increase in GD3 and GD1a expression was observed during neural differentiation. To confirm the role of gangliosides in neural differentiation, ganglioside biosynthesis was inhibited in hDPSCs by knockdown of UDP-glucose ceramide glucosyltransferase (Ugcg), which prevented differentiation into neural cells. These results suggest that gangliosides may play a role in the neural differentiation process of hDPSCs.
Collapse
Affiliation(s)
- Jae-Sung Ryu
- Department of Biological Science, College of Natural Sciences, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Morales P, Fiedler JL, Andrés S, Berrios C, Huaiquín P, Bustamante D, Cardenas S, Parra E, Herrera-Marschitz M. Plasticity of hippocampus following perinatal asphyxia: effects on postnatal apoptosis and neurogenesis. J Neurosci Res 2009; 86:2650-62. [PMID: 18512760 DOI: 10.1002/jnr.21715] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Asphyxia during delivery produces long-term deficits in brain development, including hippocampus. We investigated hippocampal plasticity after perinatal asphyxia, measuring postnatal apoptosis and neurogenesis. Asphyxia was performed by immersing rat fetuses with uterine horns removed from ready-to-deliver rats into a water bath for 20 min. Caesarean-delivered pups were used as controls. The animals were euthanized 1 week or 1 month after birth. Apoptotic nuclear morphology and DNA breaks were assessed by Hoechst and TUNEL assays. Neurogenesis was estimated by bromodeoxyuridine/MAP-2 immunocytochemistry, and the levels and expression of proteins related to apoptosis and cell proliferation were measured by Western blots and in situ hybridization, respectively. There was an increase of apoptosis in CA1, CA3, and dentate gyrus (DG) and cell proliferation and neurogenesis in CA1, DG, and hilus regions of hippocampus 1 week after asphyxia. The increase of apoptosis in CA3 and cell proliferation in the suprapyramidal band of DG was still observed 1 month following asphyxia. There was an increase of BAD, BCL-2, ERK2, and bFGF levels in whole hippocampus and bFGF expression in CA1 and CA2 and hilus at P7 and P30. There was a concomitant decrease of phosphorylated-BAD (Ser112) levels. The increase of BAD levels supports the idea of delayed cell death after perinatal asphyxia, whereas the increases of BCL-2, ERK2, and bFGF levels suggest the activation of neuroprotective and repair pathways. In conclusion, perinatal asphyxia induces short- and long-term regionally specific plastic changes, including delayed cell death and neurogenesis, involving pro- and antiapoptotic as well as mitogenic proteins, favoring hippocampal functional recovery.
Collapse
Affiliation(s)
- P Morales
- Programme of Molecular and Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Kuwagata M, Ogawa T, Nagata T, Shioda S. The evaluation of early embryonic neurogenesis after exposure to the genotoxic agent 5-bromo-2'-deoxyuridine in mice. Neurotoxicology 2006; 28:780-9. [PMID: 16956662 DOI: 10.1016/j.neuro.2006.07.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 07/25/2006] [Accepted: 07/26/2006] [Indexed: 11/26/2022]
Abstract
Developmental neurotoxicity (DNT) is an important issue in children's health. Neurogenesis occurs throughout the early fetal to the postnatal period. The proliferation of embryonic stem cells can be a target for toxicants, especially genotoxic compounds. 5-Bromo-2'-deoxyuridine (BrdU), a thymidine analogue, has been used as a marker for proliferating cells. However, we reported that prenatal BrdU exposure induced behavioral abnormalities such as hyperactivity in rat and mouse offspring. In this study, to further clarify the toxic effect of BrdU on the early neurogenesis and to examine the usefulness of the evaluation of this process in DNT, C57BL/6 mice were exposed to 100 mg/kg of BrdU once on gestational day (GD) 9 or 11, and serial sections from a wide variety of areas of the embryonic brains 24 h after the exposure were examined. BrdU exposure on GD11 induced cell death in some specific areas, such as the neocortex and striatum, but not in the substantia nigra, raphe and pons, even though BrdU was incorporated into those cells. BrdU decreased the number of cells positive for phosphorylated histone 3 (phospho-histone 3), a marker for proliferating cells at metaphase of mitosis, in the cortex, mammillary body and cerebellum, suggesting that BrdU affected the proliferation of neural stem cells. Exposure on GD9 did not induce cell death in the fetal brain. These results indicate that BrdU actually impaired the early neurogenesis, supporting the postnatal results, and demonstrated that embryonic neurogenesis has heterogeneous sensitivity to the genotoxic agents BrdU that differs according to the area and developmental stage. The evaluation of events in early neurogenesis such as the proliferation of neural stem cells shortly after chemical exposure will be one of the valuable endpoints for studying postnatal neurodevelopmental disorders.
Collapse
|