1
|
Alonso-Alconada D, Chillida M, Catalan A, Gressens P, Robertson NJ. Sex dimorphism in brain cell death after hypoxia-ischemia in newborn piglets. Pediatr Res 2025:10.1038/s41390-025-04046-5. [PMID: 40240875 DOI: 10.1038/s41390-025-04046-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/06/2025] [Accepted: 03/05/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Clinical data suggest that females might be more resistant to hypoxia than males, with male sex recognized as a risk factor for suffering life-long neurological sequelae. However, the impact of hypoxia-ischemia in certain brain regions and its association with genetic sex remains unclear. METHODS Using the piglet model of neonatal brain injury, fifteen piglets (8 females and 7 males) were subjected to a global cerebral hypoxic-ischemic insult. After 48 h, total cell death and the number of necrotic, apoptotic and cleaved-caspase-3 positive cells was quantified in five brain regions. RESULTS Male piglets exposed to hypoxia-ischemia were more vulnerable than females (total cell death p < 0.01), also showing a region-specific response to brain injury depending on sex, with males being more affected in both deep gray (caudate p < 0.01; THAL p < 0.0001) and white (p < 0.01) matter. Despite necrosis was the primary form of cell death for both sexes, the pattern of cell death differed: while male piglets showed more necrosis (p < 0.0001), apoptosis (p < 0.0001) and caspase-3 activation (p < 0.0001) were higher in females. CONCLUSION Our results suggest that male piglets were globally and regionally more vulnerable than females after HI; further, both the pattern of cell death and the apoptotic molecular mechanisms were sexually dimorphic. IMPACT Clinical data suggest that females might be more resistant to perinatal asphyxia than male newborns. The impact of hypoxia-ischemia in certain brain regions and the association of cell death patterns with sex remain unclear. Hypoxic-ischemic male piglets were more vulnerable than females, showing also increased regional vulnerability in both deep gray and white matter areas. Although necrosis was the primary form of cell death for both sexes, male piglets showed more necrosis, whereas apoptosis and caspase-3 activation were higher in females. Neonatal brain injury and therapeutic responses may be sex-dependent due to differences in cell death patterns and molecular mechanisms.
Collapse
Affiliation(s)
- Daniel Alonso-Alconada
- Department of Cell Biology & Histology, School of Medicine & Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.
| | - Marc Chillida
- Department of Cell Biology & Histology, School of Medicine & Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Ana Catalan
- Psychiatry Department, OSI Bilbao-Basurto, Basurto University Hospital, Bilbao, Spain
- Neuroscience Department, University of the Basque Country (UPV/EHU), Leioa, Spain
- Biobizkaia Health Research Institute, Barakaldo, Spain
- CIBERSAM, Centro Investigación Biomédica en Red de Salud Mental, Madrid, Spain
| | | | - Nicola J Robertson
- Institute for Women's Health, University College London, London, UK
- Edinburgh Neuroscience & Centre for Clinical Brain Sciences (CCBS), University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Rana M, Vega Gonzales-Portillo JD, Hahn C, Dutt M, Sanchez-Fernandez I, Jonas R, Douglass L, Torres AR. Current Evidence: Seizures in Extremely Low Gestational Age Newborns (ELGANs). J Child Neurol 2024; 39:285-291. [PMID: 38836290 DOI: 10.1177/08830738241259052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Extremely low gestational age newborns (ELGANs) are born at or below 28 weeks of gestational age. Despite improved obstetric care, the incidence of preterm birth continues to rise in advanced countries. Preterm birth remains a major cause of infant mortality, and for infants who survive, neonatal seizures are a significant predictor of later neurologic morbidity. However, little is known about risk factors for neonatal seizures in ELGANs. Understanding the association between neonatal seizures and the development of other neurologic disorders is important given the increasing prevalence of ELGANs. Identifying risk factors that contribute to the development of neonatal seizures in ELGANs may offer insights into novel mechanisms of epileptogenesis in the developing brain and improvements in the prevention or treatment of seizures in preterm infants, including ELGANs. In this literature review, we outline the limitations of epidemiologic studies of neonatal seizures in ELGANs and discuss risk factors for neonatal seizures.
Collapse
Affiliation(s)
- Mandeep Rana
- Division of Pediatric Neurology and Sleep Medicine, Department of Pediatrics, Boston University School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Juan Diego Vega Gonzales-Portillo
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Cecil Hahn
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Monideep Dutt
- Division of Pediatric Neurology, Children's Healthcare of Atlanta: Pediatric Institute, Emory University, Atlanta, GA, USA
| | - Ivan Sanchez-Fernandez
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Rinat Jonas
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Laurie Douglass
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Alcy R Torres
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
3
|
Giovannini E, Bonasoni MP, Pascali JP, Giorgetti A, Pelletti G, Gargano G, Pelotti S, Fais P. Infection Induced Fetal Inflammatory Response Syndrome (FIRS): State-of- the-Art and Medico-Legal Implications-A Narrative Review. Microorganisms 2023; 11:microorganisms11041010. [PMID: 37110434 PMCID: PMC10142209 DOI: 10.3390/microorganisms11041010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/21/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Fetal inflammatory response syndrome (FIRS) represents the fetal inflammatory reaction to intrauterine infection or injury, potentially leading to multiorgan impairment, neonatal mortality, and morbidity. Infections induce FIRS after chorioamnionitis (CA), defined as acute maternal inflammatory response to amniotic fluid infection, acute funisitis and chorionic vasculitis. FIRS involves many molecules, i.e., cytokines and/or chemokines, able to directly or indirectly damage fetal organs. Therefore, due to FIRS being a condition with a complex etiopathogenesis and multiple organ dysfunction, especially brain injury, medical liability is frequently claimed. In medical malpractice, reconstruction of the pathological pathways is paramount. However, in cases of FIRS, ideal medical conduct is hard to delineate, due to uncertainty in diagnosis, treatment, and prognosis of this highly complex condition. This narrative review revises the current knowledge of FIRS caused by infections, maternal and neonatal diagnosis and treatments, the main consequences of the disease and their prognoses, and discusses the medico-legal implications.
Collapse
Affiliation(s)
- Elena Giovannini
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 49, 40126 Bologna, Italy
| | - Maria Paola Bonasoni
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Jennifer Paola Pascali
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35127 Padova, Italy
| | - Arianna Giorgetti
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 49, 40126 Bologna, Italy
| | - Guido Pelletti
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 49, 40126 Bologna, Italy
| | - Giancarlo Gargano
- Neonatal Intensive Care Unit, Azienda USL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Susi Pelotti
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 49, 40126 Bologna, Italy
| | - Paolo Fais
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 49, 40126 Bologna, Italy
| |
Collapse
|
4
|
Teo EJ, Chand KK, Miller SM, Wixey JA, Colditz PB, Bjorkman ST. Early evolution of glial morphology and inflammatory cytokines following hypoxic-ischemic injury in the newborn piglet brain. Sci Rep 2023; 13:282. [PMID: 36609414 PMCID: PMC9823001 DOI: 10.1038/s41598-022-27034-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/23/2022] [Indexed: 01/09/2023] Open
Abstract
Neuroinflammation is a hallmark of hypoxic-ischemic injury and can be characterized by the activation of glial cells and the expression of inflammatory cytokines and chemokines. Interleukin (IL)-1β and tumor necrosis factor (TNF)α are among the best-characterized early response cytokines and are often expressed concurrently. Several types of central nervous system cells secrete IL-1β and TNFα, including microglia, astrocytes, and neurons, and these cytokines convey potent pro-inflammatory actions. Chemokines also play a central role in neuroinflammation by controlling inflammatory cell trafficking. Our aim was to characterise the evolution of early neuroinflammation in the neonatal piglet model of hypoxic-ischemic encephalopathy (HIE). Piglets (< 24 h old) were exposed to HI insult, and recovered to 2, 4, 8, 12 or 24H post-insult. Brain tissue from the frontal cortex and basal ganglia was harvested for assessment of glial cell activation profiles and transcription levels of inflammatory markers in HI piglets with comparison to a control group of newborn piglets. Fluorescence microscopy was used to observe microglia, astrocytes, neurons, degenerating neurons and possibly apoptotic cells, and quantitative polymerase chain reaction was used to measure gene expression of several cytokines and chemokines. HI injury was associated with microglial activation and morphological changes to astrocytes at all time points examined. Gene expression analyses of inflammation-related markers revealed significantly higher expression of pro-inflammatory cytokines tumor necrosis factor-α (TNFα) and interleukin 1 beta (IL-1β), chemokines cxc-chemokine motif ligand (CXCL)8 and CXCL10, and anti-inflammatory cytokine transforming growth factor (TGF)β in every HI group, with some region-specific differences noted. No significant difference was observed in the level of C-X-C chemokine receptor (CCR)5 over time. This high degree of neuroinflammation was associated with a reduction in the number of neurons in piglets at 12H and 24H in the frontal cortex, and the putamen at 12H. This reduction of neurons was not associated with increased numbers of degenerating neurons or potentially apoptotic cells. HI injury triggered a robust early neuroinflammatory response associated with a reduction in neurons in cortical and subcortical regions in our piglet model of HIE. This neuroinflammatory response may be targeted using novel therapeutics to reduce neuropathology in our piglet model of neonatal HIE.
Collapse
Affiliation(s)
- Elliot J. Teo
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| | - Kirat. K. Chand
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| | - Stephanie M. Miller
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| | - Julie A. Wixey
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| | - Paul B. Colditz
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| | - S. Tracey. Bjorkman
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| |
Collapse
|
5
|
Lear BA, Lear CA, Dhillon SK, Davidson JO, Bennet L, Gunn AJ. Is late prevention of cerebral palsy in extremely preterm infants plausible? Dev Neurosci 2021; 44:177-185. [PMID: 34937030 DOI: 10.1159/000521618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/16/2021] [Indexed: 11/19/2022] Open
Abstract
Preterm birth continues to be associated with neurodevelopmental problems including cerebral palsy. Cystic white matter injury is still the major neuropathology underlying cerebral palsy, affecting 1-3% of preterm infants. Although rates have gradually fallen over time, the pathogenesis and evolution of cystic white matter injury are still poorly understood. Hypoxia-ischemia (HI) remains an important contributor yet there is no established treatment to prevent injury. Clinically, serial ultrasound and magnetic resonance imaging studies typically show delayed development of cystic lesions 2 to 4 weeks after birth. This raises the important and unresolved question as to whether this represents slow evolution of injury occurring around the time of birth, or repeated injury over many weeks after birth. There is increasing evidence that tertiary injury after HI can contribute to impairment of white and grey matter maturation. In the present review, we discuss preclinical evidence that severe, cystic white matter injury can evolve for many weeks after acute HI and is associated with microglia activity. This suggests the intriguing hypothesis that the tertiary phase of injury is not as subtle as often thought and that there may be a window of therapeutic opportunity for one to two weeks after hypoxic-ischemic injury to prevent delayed cystic lesions and so further reduce the risk of cerebral palsy after preterm birth.
Collapse
Affiliation(s)
- Benjamin A Lear
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Christopher A Lear
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | | | - Joanne O Davidson
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
6
|
Zhang P, Zhang Q, Zhu B, Xia S, Tai X, Tai X, Li B. Chinese Tuina Protects against Neonatal Hypoxia-Ischemia through Inhibiting the Neuroinflammatory Reaction. Neural Plast 2020; 2020:8828826. [PMID: 33488693 PMCID: PMC7790570 DOI: 10.1155/2020/8828826] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/30/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023] Open
Abstract
Aim. Neonatal hypoxic-ischemic encephalopathy (HIE) is a significant cause of perinatal morbidity and mortality. Chinese Tuina is an effective treatment for HIE, but the molecular mechanisms are yet unknown. This study investigated the effect and mechanisms of Chinese Tuina on the inflammatory response in neonatal HIE rats. Main Methods. 30 male neonatal rats were divided randomly into 3 groups: sham, HIE, and HIE with Chinese Tuina (CHT) groups. The HIE and CHT groups were subjected to left common carotid occlusion and hypoxia at 3 days postnatal (P3). The pups in the CHT group received Chinese Tuina treatment on the next day for 28 days. The weight was measured at P4, P9, P13, P21, and P31. The behavioral functions were determined at P21. The protein expression and the methylation level in promoter regions of TNF-α and IL-10 were determined by Western blotting, immunohistochemistry, and pyrosequencing, respectively, at P33. Key Findings. The weight gain in the HIE group was slow compared with that of the CHT group. The rats in the CHT group performed better both in the balance beam and hang plate experiment. Chinese Tuina inhibited the expression of TNF-α and upregulated the expression of IL-10. Neonatal hypoxic-ischemic injury downregulated the methylation level in promoter regions of TNF-α at all CpG points but not IL-10. However, Chinese Tuina did not change the methylation level in promoter regions of TNF-α and IL-10. Significance. Chinese Tuina protected against HIE through inhibiting the neuroinflammatory reaction. While HIE markedly downregulated the methylation level of TNF-α, the protective effects of Chinese Tuina were independent of the regulation of the methylation level of TNF-α and IL-10.
Collapse
Affiliation(s)
- Pengyue Zhang
- Key Laboratory of Acupuncture and Tuina for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| | - Qian Zhang
- Key Laboratory of Acupuncture and Tuina for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| | - Bowen Zhu
- Zhejiang Provincial Hospital of TCM, Zhejiang Hangzhou 310006, China
| | - Shijin Xia
- Department of Geriatrics, Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Xianyan Tai
- Department of Prevention and Health Care, The First Hospital Affiliated to Kunming Medical University, Kunming 650032, China
| | - Xiantao Tai
- Key Laboratory of Acupuncture and Tuina for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| | - Bing Li
- Jinshan Hospital of Fudan University, Shanghai 200054, China
| |
Collapse
|
7
|
Vankeshwaram V, Maheshwary A, Mohite D, Omole JA, Khan S. Is Stem Cell Therapy the New Savior for Cerebral Palsy Patients? A Review. Cureus 2020; 12:e10214. [PMID: 33042660 PMCID: PMC7535865 DOI: 10.7759/cureus.10214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cerebral Palsy (CP) is one of the foremost causes of childhood motor disability and disrupts the individual's development and ability to function. Several factors contribute to the development of CP such as preterm delivery, low birth weight, infection/inflammation, and additional pregnancy complications, both in preterm and term infants. As there is no specific treatment for CP, rehabilitation is the current option for the management of patients. The serious nature of this condition creates deficits that last a lifetime. We collected studies that were published in the past 10 years, using PubMed as our main database. We chose studies that were relevant to CP and stem cell therapy. We mainly focused on various types of stem cells that can be used in treatment, mechanism of action (MOA) of stem cells, routes, dosage, and adverse effects, their efficacy, and safety in CP patients. Of all the 38 studies we reviewed, we found that five articles discussed the utilization of human umbilical cord blood [hUCB], four articles discussed autologous bone marrow stem cells, and one discussed allogeneic umbilical cord blood usage. One article discussed neural stem-like cells (NSLCs) derived from bone marrow and the remaining 27 articles were about CP and its treatment. We reviewed detailed information about the possible stem cell therapies and their benefits in patients with CP. We found that immune modulation is the major mechanism of action of stem cells, and among all the types of stem cells. Autologous umbilical cord mesenchymal stem cells appear to be safe and most effective in treatment compared to other stem cell treatments. Among all symptoms, motor symptoms are best corrected by stem cell therapy. Still, it did not show any marked improvement in treating other symptoms like speech defects, sensory or cognitive defects, or visual impairment.
Collapse
Affiliation(s)
- Varun Vankeshwaram
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Medicine, Zaporozhye State Medical University, Zaporozhye, UKR
| | - Ankush Maheshwary
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Medicine, Government Medical College, Amritsar, IND
| | - Divya Mohite
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Janet A Omole
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
8
|
Goncalves LF, Cornejo P, Towbin R. Neuroimaging findings associated with the fetal inflammatory response syndrome. Semin Fetal Neonatal Med 2020; 25:101143. [PMID: 32800654 DOI: 10.1016/j.siny.2020.101143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The fetal inflammatory response syndrome (FIRS) is a condition whereby the fetus mounts an inflammatory response to intrauterine infection/inflammation. Clinical consequences include preterm premature rupture of membranes (PPROM), spontaneous preterm delivery, neonatal sepsis, bronchopulmonary dysplasia, and brain and other organ injury. Mechanisms leading to brain injury in FIRS have been investigated in animal and human studies. We review the neuroimaging findings of brain injury in FIRS, which overlap those of hypoxic-ischemic injury, and clinical correlation is necessary for a correct diagnosis. FIRS should be considered the primary diagnosis when neuroimaging findings such as periventricular leukomalacia are identified in preterm children born as a consequence of PPROM and spontaneous preterm labor. Additionally, FIRS should be considered in term infants who do not have the most common features of HIE (e.g. a sentinel event). Systematic histopathologic examination of the placenta and umbilical cord and/or detection of characteristic inflammatory markers in such cases are needed to establish the correct diagnosis.
Collapse
Affiliation(s)
- Luis F Goncalves
- Director of Fetal Imaging, Phoenix Children's Hospital, Professor of Radiology and Child Health, University of Arizona College of Medicine, Phoenix, AZ, Professor of Radiology, Mayo Clinic, Phoenix, AZ, Professor of Radiology, Creighton University, Director of MRI, Phoenix Children's Hospital, USA
| | - Patricia Cornejo
- Assistant Professor of Radiology and Child Health, University of Arizona College of Medicine, Phoenix, AZ, Assistant Professor of Pediatric Neuroradiology, Barrows Neurological Institute, Phoenix, AZ, Assistant Professor Radiology, Creighton University, USA
| | - Richard Towbin
- Emeritus-Radiologist-in-Chief, Phoenix Children's Hospital, Emeritus Professor of Radiology and Child Health, University of Arizona College of Medicine-Phoenix, Professor of Radiology Mayo Clinic, USA.
| |
Collapse
|
9
|
Jiang H, Liu H, He H, Yang J, Liu Z, Huang T, Lyu J, Li X. Specific White Matter Lesions Related to Motor Dysfunction in Spastic Cerebral Palsy: A Meta-analysis of Diffusion Tensor Imaging Studies. J Child Neurol 2020; 35:146-154. [PMID: 31646936 DOI: 10.1177/0883073819879844] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Assessing motor impairment in spastic cerebral palsy is a key factor in the treatment and rehabilitation of patients. We intend to investigate the correlation between diffusion tensor imaging properties of sensorimotor pathways and motor function in spastic cerebral palsy using meta-analysis, and to determine specific white matter lesions that are closely related to motor dysfunction in spastic cerebral palsy. METHODS We conducted a literature search of PubMed, Embase, Scopus, and Web of Science databases to identify trials published from January 1999 to January 2019 that had evaluated the correlation between fractional anisotropy and motor function scores in spastic cerebral palsy. Correlation coefficient (r) values were extracted for each study, and the extent of r was quantitatively explored. The r values between fractional anisotropy within different sensorimotor pathways and motor function scores were pooled respectively. RESULTS Nineteen studies involving 504 children with spastic cerebral palsy, were included. Fractional anisotropy in both sensory and motor pathways significantly correlated with motor function scores. However, compared with the corticospinal tract and thalamic radiation, fractional anisotropy in the posterior limb of the internal capsule correlated more strongly with gross motor function classification system and upper limb motor function (r = -0.71, 95% confidence interval [CI] -0.80 to -0.60; r = 0.73, 95% CI 0.60-0.82, respectively; P < .05). CONCLUSIONS Fractional anisotropy within the posterior limb of the internal capsule is more closely related to motor dysfunction and can potentially be a biomarker for evaluating the degree of motor impairment in spastic cerebral palsy.
Collapse
Affiliation(s)
- Haoxiang Jiang
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Department of Diagnostic Radiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Diagnostic Radiology, Xi'an Children Hospital, Xi'an, China
| | - Heng Liu
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Department of Diagnostic Radiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Hairong He
- Department of Clinical Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jian Yang
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Department of Diagnostic Radiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhe Liu
- Department of Diagnostic Radiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Tingting Huang
- Department of Diagnostic Radiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jun Lyu
- Department of Clinical Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xianjun Li
- Department of Diagnostic Radiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
10
|
Therapeutic Potential of Human Amniotic Epithelial Cells on Injuries and Disorders in the Central Nervous System. Stem Cells Int 2019; 2019:5432301. [PMID: 31827529 PMCID: PMC6886344 DOI: 10.1155/2019/5432301] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/02/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023] Open
Abstract
Despite recent advances in neurosurgery and pharmaceuticals, contemporary treatments are ineffective in restoring lost neurological functions in patients with injuries and disorders of the central nervous system (CNS). Therefore, novel and effective therapies are urgently needed. Recent studies have indicated that stem cells, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs), could repair/replace damaged or degenerative neurons and improve functional recovery in both preclinical and clinical trials. However, there are many unanswered questions and unsolved issues regarding stem cell therapy in terms of potency, stability, oncogenicity, immune response, cell sources, and ethics. Currently, human amniotic epithelial cells (hAECs) derived from the amnion exhibit considerable advantages over other stem cells and have drawn much attention from researchers. hAECs are readily available, pose no ethical concerns, and have little risk of tumorigenicity and immunogenicity. Mounting evidence has shown that hAECs can promote neural cell survival and regeneration, repair affected neurons, and reestablish damaged neural connections. It is suggested that hAECs may be the most promising candidate for cell-based therapy of neurological diseases. In this review, we mainly focus on recent advances and potential applications of hAECs for treating various CNS injuries and neurodegenerative disorders. We also discuss current hurdles and challenges regarding hAEC therapies.
Collapse
|
11
|
Parikh NA, Hershey A, Altaye M. Early Detection of Cerebral Palsy Using Sensorimotor Tract Biomarkers in Very Preterm Infants. Pediatr Neurol 2019; 98:53-60. [PMID: 31201071 PMCID: PMC6717543 DOI: 10.1016/j.pediatrneurol.2019.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/25/2019] [Accepted: 05/02/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Our objectives were to evaluate the brain's sensorimotor network microstructure using diffusion magnetic resonance imaging (MRI) at term-corrected age and test the ability of sensorimotor microstructural parameters to accurately predict cerebral palsy in extremely-low-birth-weight infants. METHODS We enrolled a prospective pilot cohort of extremely-low-birth-weight preterm infants (birth weight ≤ 1000 g) before neonatal intensive care unit discharge and studied them with structural and diffusion MRI at term-corrected age. Six sensorimotor tracts were segmented, and microstructural parameters from these tracts were evaluated for their ability to predict later development of cerebral palsy, diagnosed at 18 to 22 months corrected age. RESULTS We found significant differences in multiple diffusion MRI parameters from five of the six sensorimotor tracts in infants who developed cerebral palsy (n = 5) versus those who did not (n = 36). When compared with structural MRI or individual diffusion MRI biomarkers, the combination of two individual biomarkers-fractional anisotropy of superior thalamic radiations (sensory component) and radial diffusivity of the corticospinal tract-exhibited the highest sensitivity (80%), specificity (97%), and positive likelihood ratio (28.0) for prediction of cerebral palsy. This combination of diffusion MRI biomarkers accurately classified 95% of the study infants. CONCLUSIONS Development of cerebral palsy in very preterm infants is preceded by early brain injury or immaturity to one or more sensorimotor tracts. A larger study is warranted to evaluate if a combination of sensorimotor microstructural biomarkers could accurately facilitate early diagnosis of cerebral palsy.
Collapse
Affiliation(s)
- Nehal A Parikh
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio; The Research Institute at Nationwide Children's Hospital, Columbus, Ohio.
| | - Alexa Hershey
- The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Mekibib Altaye
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio; Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
12
|
Petrenko V, van de Looij Y, Mihhailova J, Salmon P, Hüppi PS, Sizonenko SV, Kiss JZ. Multimodal MRI Imaging of Apoptosis-Triggered Microstructural Alterations in the Postnatal Cerebral Cortex. Cereb Cortex 2019; 28:949-962. [PMID: 28158611 DOI: 10.1093/cercor/bhw420] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Indexed: 12/17/2022] Open
Abstract
Prematurely born children often develop neurodevelopmental delay that has been correlated with reduced growth and microstructural alterations in the cerebral cortex. Much research has focused on apoptotic neuronal cell death as a key neuropathological features following preterm brain injuries. How scattered apoptotic death of neurons may contribute to microstructural alterations remains unknown. The present study investigated in a rat model the effects of targeted neuronal apoptosis on cortical microstructure using in vivo MRI imaging combined with neuronal reconstruction and histological analysis. We describe that mild, targeted death of layer IV neurons in the developing rat cortex induces MRI-defined metabolic and microstructural alterations including increased cortical fractional anisotropy. Delayed architectural modifications in cortical gray matter and myelin abnormalities in the subcortical white matter such as hypomyelination and microglia activation follow the acute phase of neuronal death and axonal degeneration. These results establish the link between mild cortical apoptosis and MRI-defined microstructure changes that are reminiscent to those previously observed in preterm babies.
Collapse
Affiliation(s)
- Volodymyr Petrenko
- Department of Neurosciences, University of Geneva Medical School, Geneva, Switzerland
| | - Yohan van de Looij
- Division of Child Growth & Development, Department of Pediatrics, University of Geneva, Geneva, Switzerland.,Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jevgenia Mihhailova
- Department of Neurosciences, University of Geneva Medical School, Geneva, Switzerland
| | - Patrick Salmon
- Department of Neurosciences, University of Geneva Medical School, Geneva, Switzerland
| | - Petra S Hüppi
- Division of Child Growth & Development, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| | - Stéphane V Sizonenko
- Division of Child Growth & Development, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| | - Jozsef Z Kiss
- Department of Neurosciences, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
13
|
Pattern of intracranial findings detected on magnetic resonance imaging in surviving infants born before 29 weeks of gestation. PLoS One 2019; 14:e0214683. [PMID: 30946769 PMCID: PMC6448872 DOI: 10.1371/journal.pone.0214683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/18/2019] [Indexed: 11/27/2022] Open
Abstract
Despite the positive survival trend in infants born prematurely, the risk for development of intracranial lesions has remained unchanged. However, there are limitations to our understanding of the pattern of the magnetic resonance imaging (MRI) -detected brain pathology in the preterm infants surviving to discharge. The present study outlines the type of intracranial lesions and factors allied with the neonatal brain hemorrhage (NBH) and white matter injury (WMI) seen on MRI at term-equivalent age or close to discharge in infants born before 29 weeks of gestation. We obtained demographic and clinical data, and reports of serial cranial ultrasound (CUS) performed during first month of life and qualitative MRI at term-equivalent age or close to discharge. Statistical comparison was conducted with respect to the MRI results that were classified as normal, WMI, and NBH using univariate and logistic regression analysis. One hundred and ninety three infants with MRI at term-equivalent age or close to discharge were included in final analysis. They were less mature and had a higher prevalence of pathological findings on CUS as compared with 249 other survivors born with gestational ages less than 29 weeks during the assigned study period. MRI was normal in 72.5% [95% Confidence Interval (95% CI 65.9%-78.4%)], showed WMI in 9.8% (95%CI 6.4%-14.9%) and NBH in 17.6% (95%CI 12.9–23.6) of the studied infants. Intracranial hemorrhages had also been reported in 42.2% of the infants with WMI. Except for moderate agreement with prior CUS results, no other factors were associated with the MRI detected pathological findings. In general, the likelihood for detection of WMI and NBH on MRI at term-equivalent age or close to discharge was reduced by approximately 80% and 70%, respectively if the serial CUS had not shown any abnormalities during the first month of life.
Collapse
|
14
|
Ma Q, Dasgupta C, Li Y, Huang L, Zhang L. MicroRNA-210 Downregulates ISCU and Induces Mitochondrial Dysfunction and Neuronal Death in Neonatal Hypoxic-Ischemic Brain Injury. Mol Neurobiol 2019; 56:5608-5625. [PMID: 30656514 DOI: 10.1007/s12035-019-1491-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/10/2019] [Indexed: 02/01/2023]
Abstract
Neonatal hypoxic-ischemic (HI) brain injury causes significant mortality and long-term neurologic sequelae. We previously demonstrated that HI significantly increased microRNA-210 (miR-210) in the neonatal rat brain and inhibition of brain endogenous miR-210 was neuroprotective in HI brain injury. However, the molecular mechanisms underpinning this neuroprotection remain unclear. Using both in vivo and in vitro models, herein we uncover a novel mechanism mediating oxidative brain injury after neonatal HI, in which miR-210 induces mitochondrial dysfunction via downregulation of iron-sulfur cluster assembly protein (ISCU). Inhibition of miR-210 significantly ameliorates mitochondrial dysfunction, oxidative stress, and neuronal loss in the neonatal brain subjected to HI, as well as in primary cortical neurons exposed to oxygen-glucose deprivation (OGD). These effects are mediated through ISCU, in that miR-210 mimic decreases ISCU abundance in the brains of rat pups and primary cortical neurons, and inhibition of miR-210 protects ISCU against HI in vivo or OGD in vitro. Deletion of miR-210 binding sequences at the 3'UTR of ISCU transcript ablates miR-210-induced downregulation of ISCU protein abundance in PC12 cells. In primary cortical neurons, miR-210 mimic or silencing ISCU results in mitochondrial dysfunction, reactive oxygen species production, and activation of caspase-dependent death pathways. Of importance, knockdown of ISCU increases HI-induced injury in the neonatal rat brain and counteracts the neuroprotection of miR-210 inhibition. Therefore, miR-210 by downregulating ISCU and inducing mitochondrial dysfunction in neurons is a potent contributor of oxidative brain injury after neonatal HI.
Collapse
Affiliation(s)
- Qingyi Ma
- The Lawrence D. Longo Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| | - Chiranjib Dasgupta
- The Lawrence D. Longo Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Yong Li
- The Lawrence D. Longo Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Lei Huang
- The Lawrence D. Longo Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Lubo Zhang
- The Lawrence D. Longo Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
15
|
Aridas JDS, Yawno T, Sutherland AE, Nitsos I, Ditchfield M, Wong FY, Hunt RW, Fahey MC, Malhotra A, Wallace EM, Jenkin G, Miller SL. Systemic and transdermal melatonin administration prevents neuropathology in response to perinatal asphyxia in newborn lambs. J Pineal Res 2018; 64:e12479. [PMID: 29464766 PMCID: PMC5947141 DOI: 10.1111/jpi.12479] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/06/2018] [Indexed: 01/19/2023]
Abstract
Perinatal asphyxia remains a principal cause of infant mortality and long-term neurological morbidity, particularly in low-resource countries. No neuroprotective interventions are currently available. Melatonin (MLT), a potent antioxidant, anti-inflammatory and antiapoptotic agent, offers promise as an intravenous (IV) or transdermal therapy to protect the brain. We aimed to determine the effect of melatonin (IV or transdermal patch) on neuropathology in a lamb model of perinatal asphyxia. Asphyxia was induced in newborn lambs via umbilical cord occlusion at birth. Animals were randomly allocated to melatonin commencing 30 minutes after birth (60 mg in 24 hours; IV or transdermal patch). Brain magnetic resonance spectroscopy (MRS) was undertaken at 12 and 72 hours. Animals (control n = 9; control+MLT n = 6; asphyxia n = 16; asphyxia+MLT [IV n = 14; patch n = 4]) were euthanised at 72 hours, and cerebrospinal fluid (CSF) and brains were collected for analysis. Asphyxia resulted in severe acidosis (pH 6.9 ± 0.0; lactate 9 ± 2 mmol/L) and altered determinants of encephalopathy. MRS lactate:N-acetyl aspartate ratio was 2.5-fold higher in asphyxia lambs compared with controls at 12 hours and 3-fold higher at 72 hours (P < .05). Melatonin prevented this rise (3.5-fold reduced vs asphyxia; P = .02). Asphyxia significantly increased brain white and grey matter apoptotic cell death (activated caspase-3), lipid peroxidation (4HNE) and neuroinflammation (IBA-1). These changes were significantly mitigated by both IV and patch melatonin. Systemic or transdermal neonatal melatonin administration significantly reduces the neuropathology and encephalopathy signs associated with perinatal asphyxia. A simple melatonin patch, administered soon after birth, may improve outcome in infants affected by asphyxia, especially in low-resource settings.
Collapse
Affiliation(s)
- James D. S. Aridas
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
| | - Tamara Yawno
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVic.Australia
| | - Amy E. Sutherland
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
| | - Ilias Nitsos
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVic.Australia
| | | | - Flora Y. Wong
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Monash Children's HospitalMonash HealthClaytonVic.Australia
| | - Rod W. Hunt
- Murdoch Children's Research InstituteMelbourneVic.Australia
| | - Michael C. Fahey
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Monash Children's HospitalMonash HealthClaytonVic.Australia
| | - Atul Malhotra
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Monash Children's HospitalMonash HealthClaytonVic.Australia
| | - Euan M. Wallace
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVic.Australia
| | - Graham Jenkin
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVic.Australia
| | - Suzanne L. Miller
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVic.Australia
| |
Collapse
|
16
|
Li B, Concepcion K, Meng X, Zhang L. Brain-immune interactions in perinatal hypoxic-ischemic brain injury. Prog Neurobiol 2017; 159:50-68. [PMID: 29111451 PMCID: PMC5831511 DOI: 10.1016/j.pneurobio.2017.10.006] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/26/2017] [Indexed: 01/07/2023]
Abstract
Perinatal hypoxia-ischemia remains the primary cause of acute neonatal brain injury, leading to a high mortality rate and long-term neurological deficits, such as behavioral, social, attentional, cognitive and functional motor deficits. An ever-increasing body of evidence shows that the immune response to acute cerebral hypoxia-ischemia is a major contributor to the pathophysiology of neonatal brain injury. Hypoxia-ischemia provokes an intravascular inflammatory cascade that is further augmented by the activation of resident immune cells and the cerebral infiltration of peripheral immune cells response to cellular damages in the brain parenchyma. This prolonged and/or inappropriate neuroinflammation leads to secondary brain tissue injury. Yet, the long-term effects of immune activation, especially the adaptive immune response, on the hypoxic-ischemic brain still remain unclear. The focus of this review is to summarize recent advances in the understanding of post-hypoxic-ischemic neuroinflammation triggered by the innate and adaptive immune responses and to discuss how these mechanisms modulate the brain vulnerability to injury. A greater understanding of the reciprocal interactions between the hypoxic-ischemic brain and the immune system will open new avenues for potential immunomodulatory therapy in the treatment of neonatal brain injury.
Collapse
Affiliation(s)
- Bo Li
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Katherine Concepcion
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Xianmei Meng
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
17
|
Oxidative stress and endoplasmic reticulum (ER) stress in the development of neonatal hypoxic-ischaemic brain injury. Biochem Soc Trans 2017; 45:1067-1076. [PMID: 28939695 PMCID: PMC5652227 DOI: 10.1042/bst20170017] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/09/2017] [Accepted: 08/11/2017] [Indexed: 02/06/2023]
Abstract
Birth asphyxia in term neonates affects 1–2/1000 live births and results in the development of hypoxic–ischaemic encephalopathy with devastating life-long consequences. The majority of neuronal cell death occurs with a delay, providing the potential of a treatment window within which to act. Currently, treatment options are limited to therapeutic hypothermia which is not universally successful. To identify new interventions, we need to understand the molecular mechanisms underlying the injury. Here, we provide an overview of the contribution of both oxidative stress and endoplasmic reticulum stress in the development of neonatal brain injury and identify current preclinical therapeutic strategies.
Collapse
|
18
|
Lee YA. White Matter Injury of Prematurity: Its Mechanisms and Clinical Features. J Pathol Transl Med 2017; 51:449-455. [PMID: 28797157 PMCID: PMC5611534 DOI: 10.4132/jptm.2017.07.25] [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: 05/02/2017] [Revised: 07/19/2017] [Accepted: 07/25/2017] [Indexed: 11/17/2022] Open
Abstract
A developing central nervous system is vulnerable to various insults such as infection and ischemia. While increased understanding of the dynamic nature of brain development allows a deeper insight into the pathophysiology of perinatal brain injury, the precise nature of specific fetal and neonatal brain injuries and their short- and long-term clinical consequences need special attention and further elucidation. The current review will describe the pathophysiological aspects and clinical significance of white matter injury of prematurity, a main form of perinatal brain injury in premature newborns, with a particular emphasis on its potential antenatal components.
Collapse
Affiliation(s)
- Young Ah Lee
- Division of Pediatric Neurology, Department of Pediatrics, Beaumont Hospital, Oakland University School of Medicine, Royal Oak, MI, USA
| |
Collapse
|
19
|
Kubo KI, Deguchi K, Nagai T, Ito Y, Yoshida K, Endo T, Benner S, Shan W, Kitazawa A, Aramaki M, Ishii K, Shin M, Matsunaga Y, Hayashi K, Kakeyama M, Tohyama C, Tanaka KF, Tanaka K, Takashima S, Nakayama M, Itoh M, Hirata Y, Antalffy B, Armstrong DD, Yamada K, Inoue K, Nakajima K. Association of impaired neuronal migration with cognitive deficits in extremely preterm infants. JCI Insight 2017; 2:88609. [PMID: 28515367 DOI: 10.1172/jci.insight.88609] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 04/18/2017] [Indexed: 12/23/2022] Open
Abstract
Many extremely preterm infants (born before 28 gestational weeks [GWs]) develop cognitive impairment in later life, although the underlying pathogenesis is not yet completely understood. Our examinations of the developing human neocortex confirmed that neuronal migration continues beyond 23 GWs, the gestational week at which extremely preterm infants have live births. We observed larger numbers of ectopic neurons in the white matter of the neocortex in human extremely preterm infants with brain injury and hypothesized that altered neuronal migration may be associated with cognitive impairment in later life. To confirm whether preterm brain injury affects neuronal migration, we produced brain damage in mouse embryos by occluding the maternal uterine arteries. The mice showed delayed neuronal migration, ectopic neurons in the white matter, altered neuronal alignment, and abnormal corticocortical axonal wiring. Similar to human extremely preterm infants with brain injury, the surviving mice exhibited cognitive deficits. Activation of the affected medial prefrontal cortices of the surviving mice improved working memory deficits, indicating that decreased neuronal activity caused the cognitive deficits. These findings suggest that altered neuronal migration altered by brain injury might contribute to the subsequent development of cognitive impairment in extremely preterm infants.
Collapse
Affiliation(s)
- Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kimiko Deguchi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan.,Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan.,Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukiko Ito
- Department of Molecular Neuroscience, Medical Research Institute/School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keitaro Yoshida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Toshihiro Endo
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Seico Benner
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Wei Shan
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ayako Kitazawa
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan.,Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Michihiko Aramaki
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kazuhiro Ishii
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Minkyung Shin
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Matsunaga
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kanehiro Hayashi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Kakeyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,Laboratory for Systems Neuroscience & Preventive Medicine, Waseda University Faculty of Human Sciences, Tokorozawa, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kohichi Tanaka
- Department of Molecular Neuroscience, Medical Research Institute/School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sachio Takashima
- Division of Child Neurology, Yanagawa Institute of Developmental Disabilities, Yanagawa, Japan
| | - Masahiro Nakayama
- Department of Pathology, Osaka Medical Center and Research Institute for Maternal and Child Health, Izumi, Japan
| | - Masayuki Itoh
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Yukio Hirata
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Barbara Antalffy
- Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Dawna D Armstrong
- Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ken Inoue
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal Hypoxia Ischaemia: Mechanisms, Models, and Therapeutic Challenges. Front Cell Neurosci 2017; 11:78. [PMID: 28533743 PMCID: PMC5420571 DOI: 10.3389/fncel.2017.00078] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Neonatal hypoxia-ischaemia (HI) is the most common cause of death and disability in human neonates, and is often associated with persistent motor, sensory, and cognitive impairment. Improved intensive care technology has increased survival without preventing neurological disorder, increasing morbidity throughout the adult population. Early preventative or neuroprotective interventions have the potential to rescue brain development in neonates, yet only one therapeutic intervention is currently licensed for use in developed countries. Recent investigations of the transient cortical layer known as subplate, especially regarding subplate's secretory role, opens up a novel set of potential molecular modulators of neonatal HI injury. This review examines the biological mechanisms of human neonatal HI, discusses evidence for the relevance of subplate-secreted molecules to this condition, and evaluates available animal models. Neuroserpin, a neuronally released neuroprotective factor, is discussed as a case study for developing new potential pharmacological interventions for use post-ischaemic injury.
Collapse
Affiliation(s)
- Lancelot J. Millar
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Lei Shi
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan UniversityGuangzhou, China
| | | | - Zoltán Molnár
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| |
Collapse
|
21
|
Santos AS, Almeida W, Popik B, Sbardelotto BM, Torrejais MM, Souza MA, Centenaro LA. Characterization of a cerebral palsy‐like model in rats: Analysis of gait pattern and of brain and spinal cord motor areas. Int J Dev Neurosci 2017; 60:48-55. [DOI: 10.1016/j.ijdevneu.2017.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/29/2017] [Accepted: 04/29/2017] [Indexed: 12/23/2022] Open
Affiliation(s)
- Adriana Souza Santos
- Laboratório de Morfologia Experimental, Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Rua Universitária1619, CascavelParanáCEP: 85819‐110Brazil
| | - Wellington Almeida
- Laboratório de Morfologia Experimental, Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Rua Universitária1619, CascavelParanáCEP: 85819‐110Brazil
| | - Bruno Popik
- Laboratório de Morfologia Experimental, Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Rua Universitária1619, CascavelParanáCEP: 85819‐110Brazil
| | - Bruno Marques Sbardelotto
- Laboratório de Morfologia Experimental, Centro de Ciências Médicas e Farmacêuticas, Universidade Estadual do Oeste do Paraná, Rua Universitária1619, CascavelParanáCEP: 85819‐110Brazil
| | - Márcia Miranda Torrejais
- Laboratório de Morfologia Experimental, Programa de Pós‐Graduação em Biociências e Saúde, Universidade Estadual do Oeste do Paraná, Rua Universitária1619, CascavelParanáCEP: 85819‐110Brazil
| | - Marcelo Alves Souza
- Universidade Federal do Paraná, Rua General Rondon2195, ToledoParanáCEP: 85902‐090Brazil
| | - Lígia Aline Centenaro
- Laboratório de Morfologia Experimental, Centro de Ciências Médicas e Farmacêuticas, Universidade Estadual do Oeste do Paraná, Rua Universitária1619, CascavelParanáCEP: 85819‐110Brazil
| |
Collapse
|
22
|
LaRosa DA, Ellery SJ, Walker DW, Dickinson H. Understanding the Full Spectrum of Organ Injury Following Intrapartum Asphyxia. Front Pediatr 2017; 5:16. [PMID: 28261573 PMCID: PMC5313537 DOI: 10.3389/fped.2017.00016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/23/2017] [Indexed: 11/13/2022] Open
Abstract
Birth asphyxia is a significant global health problem, responsible for ~1.2 million neonatal deaths each year worldwide. Those who survive often suffer from a range of health issues including brain damage-manifesting as cerebral palsy (CP)-respiratory insufficiency, cardiovascular collapse, and renal dysfunction, to name a few. Although the majority of research is directed toward reducing the brain injury that results from intrapartum birth asphyxia, the multi-organ injury observed in surviving neonates is of equal importance. Despite the advent of hypothermia therapy for the treatment of hypoxic-ischemic encephalopathy (HIE), treatment options following asphyxia at birth remain limited, particularly in low-resource settings where the incidence of birth asphyxia is highest. Furthermore, although cooling of the neonate results in improved neurological outcomes for a small proportion of treated infants, it does not provide any benefit to the other organ systems affected by asphyxia at birth. The aim of this review is to summarize the current knowledge of the multi-organ effects of intrapartum asphyxia, with particular reference to the findings from our laboratory using the precocial spiny mouse to model birth asphyxia. Furthermore, we reviewed the current treatments available for neonates who have undergone intrapartum asphyxia, and highlight the emergence of maternal dietary creatine supplementation as a preventative therapy, which has been shown to provide multi-organ protection from birth asphyxia-induced injury in our preclinical studies. This cheap and effective nutritional supplement may be the key to reducing birth asphyxia-induced death and disability, particularly in low-resource settings where current treatments are unavailable.
Collapse
Affiliation(s)
- Domenic A LaRosa
- Ritchie Centre, Department of Obstetrics and Gynaecology, Hudson Institute of Medical Research, Monash University, Melbourne, VIC, Australia; Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Stacey J Ellery
- Ritchie Centre, Department of Obstetrics and Gynaecology, Hudson Institute of Medical Research, Monash University , Melbourne, VIC , Australia
| | - David W Walker
- Ritchie Centre, Department of Obstetrics and Gynaecology, Hudson Institute of Medical Research, Monash University , Melbourne, VIC , Australia
| | - Hayley Dickinson
- Ritchie Centre, Department of Obstetrics and Gynaecology, Hudson Institute of Medical Research, Monash University , Melbourne, VIC , Australia
| |
Collapse
|
23
|
Zhang Z, Bassam B, Thomas AG, Williams M, Liu J, Nance E, Rojas C, Slusher BS, Kannan S. Maternal inflammation leads to impaired glutamate homeostasis and up-regulation of glutamate carboxypeptidase II in activated microglia in the fetal/newborn rabbit brain. Neurobiol Dis 2016; 94:116-28. [PMID: 27326668 PMCID: PMC5394739 DOI: 10.1016/j.nbd.2016.06.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 06/05/2016] [Accepted: 06/16/2016] [Indexed: 12/12/2022] Open
Abstract
Astrocyte dysfunction and excessive activation of glutamatergic systems have been implicated in a number of neurologic disorders, including periventricular leukomalacia (PVL) and cerebral palsy (CP). However, the role of chorioamnionitis on glutamate homeostasis in the fetal and neonatal brains is not clearly understood. We have previously shown that intrauterine endotoxin administration results in intense microglial 'activation' and increased pro-inflammatory cytokines in the periventricular region (PVR) of the neonatal rabbit brain. In this study, we assessed the effect of maternal inflammation on key components of the glutamate pathway and its relationship to astrocyte and microglial activation in the fetal and neonatal New Zealand white rabbit brain. We found that intrauterine endotoxin exposure at gestational day 28 (G28) induced acute and prolonged glutamate elevation in the PVR of fetal (G29, 1day post-injury) and postnatal day 1 (PND1, 3days post-injury) brains along with prominent morphological changes in the astrocytes (soma hypertrophy and retracted processes) in the white matter tracts. There was a significant increase in glutaminase and N-Methyl-d-Aspartate receptor (NMDAR) NR2 subunit expression along with decreased glial L-glutamate transporter 1 (GLT-1) in the PVR at G29, that would promote acute dysregulation of glutamate homeostasis. This was accompanied with significantly decreased TGF-β1 at PND1 in CP kits indicating ongoing neuroinflammation. We also show for the first time that glutamate carboxypeptidase II (GCPII) was significantly increased in the activated microglia at the periventricular white matter area in both G29 and PND1 CP kits. This was confirmed by in vitro studies demonstrating that LPS activated primary microglia markedly upregulate GCPII enzymatic activity. These results suggest that maternal intrauterine endotoxin exposure results in early onset and long-lasting dysregulation of glutamate homeostasis, which may be mediated by impaired astrocyte function and GCPII upregulation in activated microglia.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Bassam Bassam
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Monica Williams
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Jinhuan Liu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Elizabeth Nance
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Barbara S Slusher
- Neurology, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA; Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA.
| |
Collapse
|
24
|
Aridas JDS, McDonald CA, Paton MCB, Yawno T, Sutherland AE, Nitsos I, Pham Y, Ditchfield M, Fahey MC, Wong F, Malhotra A, Castillo-Melendez M, Bhakoo K, Wallace EM, Jenkin G, Miller SL. Cord blood mononuclear cells prevent neuronal apoptosis in response to perinatal asphyxia in the newborn lamb. J Physiol 2015; 594:1421-35. [PMID: 26527561 DOI: 10.1113/jp271104] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/23/2015] [Indexed: 11/08/2022] Open
Abstract
Perinatal asphyxia is a significant cause of death or long-term neurodevelopmental impairment. Hypothermia, currently the only effective treatment, leads to modest improvements, but new therapeutic strategies are required. Umbilical cord blood (UCB) mononuclear cells have potent anti-inflammatory properties and may reduce neuropathology. This study examined whether autologous UCB mononuclear cells were neuroprotective when administered to newborn lambs at 12 h after birth asphyxia. At caesarean section, birth asphyxia was induced by clamping the umbilical cord until mean arterial blood pressure decreased to 18-20 mmHg. Asphyxia (n = 20) or control (n = 11) lambs were resuscitated and maintained, with magnetic resonance spectroscropy (MRS) performed at 12 and 72 h, and were then killed at 72 h. Cord blood was collected once the cord was clamped, and mononuclear cells were isolated and labelled fluorescently and administered to control (n = 3) or asphyxia (n = 8) lambs. Asphyxia induced a significant increase in cellular apoptosis (caspase-3 immunopositive) within all brain regions examined, including cortex, hippocampus, thalamus, striatum and subcortical white matter (P < 0.01 vs. control). Additionally, asphyxia induced significant and widespread astrogliosis and increased inflammatory cells (activated microglia and macrophages). The administration of UCB mononuclear cells (asphyxia+UCB) significantly decreased neuronal apoptosis, astrogliosis and inflammation (P < 0.05 vs. asphyxia alone). Asphyxia+UCB lambs also demonstrated decreased brain metabolites lactate:choline (P = 0.01) and lactate:N-acetylaspartate (P < 0.01) from 12 to 72 h, detected using MRS. Autologous UCB mononuclear cell treatment restores normal brain metabolism following perinatal asphyxia, and reduces brain inflammation, astrogliosis and neuronal apoptosis, supporting its use as a neuroprotective therapy following asphyxia.
Collapse
Affiliation(s)
- James D S Aridas
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Madison C B Paton
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Tamara Yawno
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Ilias Nitsos
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Michael Ditchfield
- Monash Children's, Monash Health, and Department of Paediatrics, Monash University, Clayton, Victoria, Australia.,Diagnostic Imaging, Monash Health, Clayton, Victoria, Australia
| | - Michael C Fahey
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Monash Children's, Monash Health, and Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Flora Wong
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Monash Children's, Monash Health, and Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Atul Malhotra
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Monash Children's, Monash Health, and Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Kishore Bhakoo
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Biopolis Way, Singapore
| | - Euan M Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
25
|
Panteliadis CP, Hagel C, Karch D, Heinemann K. Cerebral Palsy: A Lifelong Challenge Asks for Early Intervention. Open Neurol J 2015; 9:45-52. [PMID: 26191093 PMCID: PMC4503828 DOI: 10.2174/1874205x01509010045] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 03/09/2015] [Accepted: 04/05/2015] [Indexed: 01/03/2023] Open
Abstract
One of the oldest and probably well-known examples of cerebral palsy is the mummy of the Pharaoh Siptah about 1196-1190 B.C., and a letter from Hippocrates (460-390 B.C.). Cerebral palsy (CP) is one of the most common congenital or acquired neurological impairments in paediatric patients, and refers to a group of children with motor disability and related functional defects. The visible core of CP is characterized by abnormal coordination of movements and/or muscle tone which manifest very early in the development. Resulting from pre- or perinatal brain damage CP is not a progressive condition per se. However, without systematic medical and physiotherapeutic support the dystonia leads to muscle contractions and to deterioration of the handicap. Here we review the three general spastic manifestations of CP hemiplegia, diplegia and tetraplegia, describe the diagnostic procedures and delineate a time schedule for an early intervention.
Collapse
Affiliation(s)
- Christos P Panteliadis
- Paediatric, Division of Paediatric Neurology and Developmental Medicine, Aristotle University of Thessaloniki, Greece
| | - Christian Hagel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Dieter Karch
- Clinic of Paediatric Neurology and Social Paediatrics, Children Centre Maulbronn, Germany
| | - Karl Heinemann
- Clinic of Paediatric Neurology and Social Paediatrics, Children Centre Maulbronn, Germany
| |
Collapse
|
26
|
Reisinger S, Khan D, Kong E, Berger A, Pollak A, Pollak DD. The poly(I:C)-induced maternal immune activation model in preclinical neuropsychiatric drug discovery. Pharmacol Ther 2015; 149:213-26. [PMID: 25562580 DOI: 10.1016/j.pharmthera.2015.01.001] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 12/30/2014] [Indexed: 12/28/2022]
Abstract
Increasing epidemiological and experimental evidence implicates gestational infections as one important factor involved in the pathogenesis of several neuropsychiatric disorders. Corresponding preclinical model systems based upon maternal immune activation (MIA) by treatment of the pregnant female have been developed. These MIA animal model systems have been successfully used in basic and translational research approaches, contributing to the investigation of the underlying pathophysiological mechanisms at the molecular, cellular and behavioral levels. The present article focuses on the application of a specific MIA rodent paradigm, based upon treatment of the gestating dam with the viral mimic polyinosinic-polycytidilic acid (Poly(I:C)), a synthetic analog of double-stranded RNA (dsRNA) which activates the Toll-like receptor 3 (TLR3) pathway. Important advantages and constraints of this animal model will be discussed, specifically in light of gestational infection as one vulnerability factor contributing to the complex etiology of mood and psychotic disorders, which are likely the result of intricate multi-level gene×environment interactions. Improving our currently incomplete understanding of the molecular pathomechanistic principles underlying these disorders is a prerequisite for the development of alternative therapeutic approaches which are critically needed in light of the important drawbacks and limitations of currently available pharmacological treatment options regarding efficacy and side effects. The particular relevance of the Poly(I:C) MIA model for the discovery of novel drug targets for symptomatic and preventive therapeutic strategies in mood and psychotic disorders is highlighted in this review article.
Collapse
Affiliation(s)
- Sonali Reisinger
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Austria
| | - Deeba Khan
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Austria
| | - Eryan Kong
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Austria
| | - Angelika Berger
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Austria
| | - Arnold Pollak
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Austria
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Austria.
| |
Collapse
|
27
|
Barton SK, Melville JM, Tolcos M, Polglase GR, McDougall ARA, Azhan A, Crossley KJ, Jenkin G, Moss TJM. Human Amnion Epithelial Cells Modulate Ventilation-Induced White Matter Pathology in Preterm Lambs. Dev Neurosci 2015; 37:338-48. [PMID: 25720586 DOI: 10.1159/000371415] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/08/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Preterm infants can be inadvertently exposed to high tidal volumes (VT) during resuscitation in the delivery room due to limitations of available equipment. High VT ventilation of preterm lambs produces cerebral white matter (WM) pathology similar to that observed in preterm infants who develop cerebral palsy. We hypothesized that human amnion epithelial cells (hAECs), which have anti-inflammatory and regenerative properties, would reduce ventilation-induced WM pathology in neonatal late preterm lamb brains. METHODS Two groups of lambs (0.85 gestation) were used, as follows: (1) ventilated lambs (Vent; n = 8) were ventilated using a protocol that induces injury (VT targeting 15 ml/kg for 15 min, with no positive end-expiratory pressure) and were then maintained for another 105 min, and (2) ventilated + hAECs lambs (Vent+hAECs; n = 7) were similarly ventilated but received intravenous and intratracheal administration of 9 × 10(7) hAECs (18 × 10(7) hAECs total) at birth. Oxygenation and ventilation parameters were monitored in real time; cerebral oxygenation was measured using near-infrared spectroscopy. qPCR (quantitative real-time PCR) and immunohistochemistry were used to assess inflammation, vascular leakage and astrogliosis in both the periventricular and subcortical WM of the frontal and parietal lobes. An unventilated control group (UVC; n = 5) was also used for qPCR analysis of gene expression. Two-way repeated measures ANOVA was used to compare physiological data. Student's t test and one-way ANOVA were used for immunohistological and qPCR data comparisons, respectively. RESULTS Respiratory parameters were not different between groups. Interleukin (IL)-6 mRNA levels in subcortical WM were lower in the Vent+hAECs group than the Vent group (p = 0.028). IL-1β and IL-6 mRNA levels in periventricular WM were higher in the Vent+hAECs group than the Vent group (p = 0.007 and p = 0.001, respectively). The density of Iba-1-positive microglia was lower in the subcortical WM of the parietal lobes (p = 0.010) in the Vent+hAECs group but not in the periventricular WM. The number of vessels in the WM of the parietal lobe exhibiting protein extravasation was lower (p = 0.046) in the Vent+hAECs group. Claudin-1 mRNA levels were higher in the periventricular WM (p = 0.005). The density of GFAP-positive astrocytes was not different between groups. CONCLUSIONS Administration of hAECs at the time of birth alters the effects of injurious ventilation on the preterm neonatal brain. Further studies are required to understand the regional differences in the effects of hAECs on ventilation-induced WM pathology and their net effect on the developing brain.
Collapse
Affiliation(s)
- Samantha K Barton
- The Ritchie Centre, MIMR-PHI Institute of Medical Research, Clayton, Vic., Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
A clinical study of autologous bone marrow mononuclear cells for cerebral palsy patients: a new frontier. Stem Cells Int 2015; 2015:905874. [PMID: 25788947 PMCID: PMC4348592 DOI: 10.1155/2015/905874] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 01/15/2015] [Accepted: 01/20/2015] [Indexed: 01/01/2023] Open
Abstract
Cerebral palsy is a nonprogressive heterogeneous group of neurological disorders with a growing rate of prevalence. Recently, cellular therapy is emerging as a potential novel treatment strategy for cerebral palsy. The various mechanisms by which cellular therapy works include neuroprotection, immunomodulation, neurorestoration, and neurogenesis. We conducted an open label, nonrandomized study on 40 cases of cerebral palsy with an aim of evaluating the benefit of cellular therapy in combination with rehabilitation. These cases were administered autologous bone marrow mononuclear cells intrathecally. The follow-up was carried out at 1 week, 3 months, and 6 months after the intervention. Adverse events of the treatment were also monitored in this duration. Overall, at six months, 95% of patients showed improvements. The study population was further divided into diplegic, quadriplegic, and miscellaneous group of cerebral palsy. On statistical analysis, a significant association was established between the symptomatic improvements and cell therapy in diplegic and quadriplegic cerebral palsy. PET-CT scan done in 6 patients showed metabolic improvements in areas of the brain correlating to clinical improvements. The results of this study demonstrate that cellular therapy may accelerate the development, reduce disability, and improve the quality of life of patients with cerebral palsy.
Collapse
|
29
|
Zuculo GM, Knap CCF, Pinato L. Correlation between sleep and quality of life in cerebral palsy. Codas 2014; 26:447-56. [DOI: 10.1590/2317-1782/20140201435] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 07/01/2014] [Indexed: 11/21/2022] Open
Abstract
PURPOSE: To investigate and correlate the sleep pattern and quality of life in individuals with cerebral palsy (CP) compared to healthy controls. METHODS: Seventy-eight subjects (aged 4-18 years, both genders) comprised two groups: CP, composed of 43 individuals with CP, and control, composed of 35 individuals without neurodevelopmental disorders. General Sleep Habits Questionnaire, Sleep Disturbance Scale for Children, sleep diary, Child Health Questionnaire, and Children's Quality of Life Scale were used. RESULTS: This study identified that 60.5% individuals with CP had sleep disorders. The respiratory disorders (25.6%) and the sleep hyperhidrosis (34.9%) were the most common disturbances in the group with CP. Moreover, 23.2% individuals of the group with CP reported awaking in the middle of the night and 37.2% of them snore, both percentages were higher than those in the control group. The sleep diary showed that individuals in the group with CP spend more time to initiate sleep (around 21 minutes). The group with CP also showed deficits in all parameters analyzed by the Child Health Questionnaire, except in family activity and the sleep-wake pattern, and quality of life showed negative correlation in several respects. CONCLUSION: The altered pattern of sleep in individuals with CP directly affects their physical and emotional well-being.
Collapse
Affiliation(s)
| | | | - Luciana Pinato
- Universidade Estadual Paulista Júlio de Mesquita Filho, Brazil
| |
Collapse
|
30
|
Porambo M, Phillips AW, Marx J, Ternes K, Arauz E, Pletnikov M, Wilson MA, Rothstein JD, Johnston MV, Fatemi A. Transplanted glial restricted precursor cells improve neurobehavioral and neuropathological outcomes in a mouse model of neonatal white matter injury despite limited cell survival. Glia 2014; 63:452-65. [PMID: 25377280 DOI: 10.1002/glia.22764] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 10/15/2014] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Neonatal white matter injury (NWMI) is the leading cause of cerebral palsy and other neurocognitive deficits in prematurely-born children, and no restorative therapies exist. Our objective was to determine the fate and effect of glial restricted precursor cell (GRP) transplantation in an ischemic mouse model of NWMI. METHODS Neonatal CD-1 mice underwent unilateral carotid artery ligation on postnatal-Day 5 (P5). At P22, intracallosal injections of either enhanced green fluorescent protein (eGFP) + GRPs or saline were performed in control and ligated mice. Neurobehavioral and postmortem studies were performed at 4 and 8 weeks post-transplantation. RESULTS GRP survival was comparable at 1 month but significantly lower at 2 months post-transplantation in NWMI mice compared with unligated controls. Surviving cells showed better migration capability in controls; however, the differentiation capacity of transplanted cells was similar in control and NWMI. Saline-treated NWMI mice showed significantly altered response in startle amplitude and prepulse inhibition (PPI) paradigms compared with unligated controls, while these behavioral tests were completely normal in GRP-transplanted animals. Similarly, there was significant increase in hemispheric myelin basic protein density, along with significant decrease in pathologic axonal staining in cell-treated NWMI mice compared with saline-treated NWMI animals. INTERPRETATION The reduced long-term survival and migration of transplanted GRPs in an ischemia-induced NWMI model suggests that neonatal ischemia leads to long-lasting detrimental effects on oligodendroglia even months after the initial insult. Despite limited GRP-survival, behavioral, and neuropathological outcomes were improved after GRP-transplantation. Our results suggest that exogenous GRPs improve myelination through trophic effects in addition to differentiation into mature oligodendrocytes.
Collapse
Affiliation(s)
- Michael Porambo
- Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Aridas JDS, Yawno T, Sutherland AE, Nitsos I, Ditchfield M, Wong FY, Fahey MC, Malhotra A, Wallace EM, Jenkin G, Miller SL. Detecting brain injury in neonatal hypoxic ischemic encephalopathy: closing the gap between experimental and clinical research. Exp Neurol 2014; 261:281-90. [PMID: 25079368 DOI: 10.1016/j.expneurol.2014.07.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 07/03/2014] [Accepted: 07/20/2014] [Indexed: 12/11/2022]
Abstract
Moderate to severe neonatal hypoxic ischemic encephalopathy remains an important cause of infant death and childhood disability. Early and accurate diagnosis of encephalopathy is difficult but critical for timely intervention. Thus, we have utilized a clinically relevant large animal model of asphyxia in-utero, followed by immediate lamb delivery, resuscitation and clinical care over the next 72h for assessment of potential biomarkers of brain injury. In-utero asphyxia was induced in twelve near-term lambs and outcomes compared with seven controls. Asphyxia resulted in bradycardia (97±12beats/min), hypotension (12.1±1mm Hg) and metabolic acidosis (pH6.9±0.02; base-excess -13.8±0.8mmol/l). 72h following asphyxia, cerebrospinal concentrations of malondialdehyde and S100B were elevated 2-fold and 5-fold, respectively, in asphyxic lambs compared to control lambs. Magnetic resonance spectroscopy (MRS) at 72h showed a significant decrease in n-acetyl aspartate: choline ratio in asphyxia lambs compared to that observed at 12h (0.56±0.23 vs. 0.82±0.15, respectively); lactate:choline ratio was not changed over this time. Marked neuropathology was observed in asphyxia lambs with neuronal degeneration in the hippocampus, thalamus, striatum and cortex. Astrogliosis was observed in the hippocampus and thalamus. Early blood markers of metabolic state showed limited predictive value of histological damage at 72h. MRS outcomes at 72h showed a modest but significant correlation with histological evidence of neuronal brain injury (lactate:N-acetyl aspartate ratio in the thalamus r(2)=0.2, p<0.01). MRS at 72h was best able to detect established brain injury, but a combination of biomarkers over multiple phases of injury may be able to assess the evolution of neonatal brain injury.
Collapse
Affiliation(s)
- James D S Aridas
- The Ritchie Centre, MIMR-PHI Institute, Clayton, Victoria, Australia
| | - Tamara Yawno
- The Ritchie Centre, MIMR-PHI Institute, Clayton, Victoria, Australia
| | - Amy E Sutherland
- The Ritchie Centre, MIMR-PHI Institute, Clayton, Victoria, Australia
| | - Ilias Nitsos
- The Ritchie Centre, MIMR-PHI Institute, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | | | - Flora Y Wong
- The Ritchie Centre, MIMR-PHI Institute, Clayton, Victoria, Australia; Monash Children's, Monash Health, Clayton, Victoria, Australia
| | - Michael C Fahey
- The Ritchie Centre, MIMR-PHI Institute, Clayton, Victoria, Australia; Monash Children's, Monash Health, Clayton, Victoria, Australia
| | - Atul Malhotra
- The Ritchie Centre, MIMR-PHI Institute, Clayton, Victoria, Australia; Monash Children's, Monash Health, Clayton, Victoria, Australia
| | - Euan M Wallace
- The Ritchie Centre, MIMR-PHI Institute, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Graham Jenkin
- The Ritchie Centre, MIMR-PHI Institute, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Suzanne L Miller
- The Ritchie Centre, MIMR-PHI Institute, Clayton, Victoria, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
32
|
Gump WC, Mutchnick IS, Moriarty TM. Selective dorsal rhizotomy for spasticity not associated with cerebral palsy: reconsideration of surgical inclusion criteria. Neurosurg Focus 2014; 35:E6. [PMID: 24175866 DOI: 10.3171/2013.8.focus13294] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Children with spastic diplegia from cerebral palsy (CP) experience measurable improvement in their spasticity and motor function following selective dorsal rhizotomy (SDR). The role of this operation in the treatment of other spasticity causes is less well defined. A literature review was undertaken to survey outcomes from SDRs performed outside the CP population. Multiple sclerosis was the most common diagnosis found, accounting for 74 of 145 patients described. Selective dorsal rhizotomies have also been reported in patients with traumatic brain and spinal cord injuries, ischemic and hemorrhagic stroke, neurodegenerative disease, hypoxic encephalopathy, and other causes of spasticity. Outcomes from surgery are generally described as favorable, although postoperative assessments and follow-up times are not standardized across reports. Long-term outcomes are sparsely reported. Larger numbers of patients and more detailed outcomes data have the potential to form a basis for expanding the inclusion criteria for SDR.
Collapse
Affiliation(s)
- William C Gump
- Division of Pediatric Neurosurgery, Norton Neuroscience Institute and Kosair Children's Hospital; and
| | | | | |
Collapse
|
33
|
Tosun C, Koltz MT, Kurland DB, Ijaz H, Gurakar M, Schwartzbauer G, Coksaygan T, Ivanova S, Gerzanich V, Simard JM. The protective effect of glibenclamide in a model of hemorrhagic encephalopathy of prematurity. Brain Sci 2014; 3:215-38. [PMID: 23667741 PMCID: PMC3647482 DOI: 10.3390/brainsci3010215] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We studied a model of hemorrhagic encephalopathy of prematurity (EP) that closely recapitulates findings in humans with hemorrhagic EP. This model involves tandem insults of 20 min intrauterine ischemia (IUI) plus an episode of elevated venous pressure induced by intraperitoneal glycerol on post-natal day (P) 0. We examined Sur1 expression, which is upregulated after focal ischemia but has not been studied after brief global ischemia including IUI. We found that 20 min IUI resulted in robust upregulation of Sur1 in periventricular microvessels and tissues. We studied tandem insult pups from untreated or vehicle-treated dams (TI-CTR), and tandem insult pups from dams administered a low-dose, non-hypoglycemogenic infusion of the Sur1 blocker, glibenclamide, for 1 week after IUI (TI-GLIB). Compared to pups from the TI-CTR group, pups from the TI-GLIB group had significantly fewer and less severe hemorrhages on P1, performed significantly better on the beam walk and accelerating Rotarod on P35 and in tests of thigmotaxis and rapid learning on P35–49, and had significantly greater body and brain weights at P52. We conclude that low-dose glibenclamide administered to the mother at the end of pregnancy protects pups subjected to IUI from post-natal events of elevated venous pressure and its consequences.
Collapse
Affiliation(s)
- Cigdem Tosun
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Michael T. Koltz
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - David B. Kurland
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Hina Ijaz
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Melda Gurakar
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Gary Schwartzbauer
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Turhan Coksaygan
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mail:
| | - Svetlana Ivanova
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mail:
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-410-328-0850; Fax: +1-410-328-0124
| |
Collapse
|
34
|
Pogledic I, Kostovic I, Fallet-Bianco C, Adle-Biassette H, Gressens P, Verney C. Involvement of the subplate zone in preterm infants with periventricular white matter injury. Brain Pathol 2014; 24:128-41. [PMID: 25003178 DOI: 10.1111/bpa.12096] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Studies of periventricular white matter injury (PWMI) in preterm infants suggest the involvement of the transient cortical subplate zone. We studied the cortical wall of noncystic and cystic PWMI cases and controls. Non-cystic PWMI corresponded to diffuse white matter lesions, the predominant injury currently detected by imaging. Glial cell populations were analyzed in post-mortem human frontal lobes from very preterm [24–29 postconceptional weeks (pcw)] and preterm infants (30–34 pcw) using immunohistochemistry for glial fibrillary acidic protein (GFAP), monocarboxylate transporter 1(MCT1), ionized calcium-binding adapter molecule 1 (Iba1), CD68 and oligodendrocyte lineage (Olig2). Glial activation extended into the subplate in non-cystic PWMI but was restricted to the white matter in cystic PWMI. Two major age-related and laminar differences were observed in non-cystic PWMI: in very preterm cases, activated microglial cells were increased and extended into the subplate adjacent to the lesion, whereas in preterm cases, an astroglial reaction was seen not only in the subplate but throughout the cortical plate. There were no differences in Olig2-positive pre-oligodendrocytes in the subplate inPWMI cases compared with controls. The involvement of gliosis in the deep subplate supports the concept of the complex cellular vulnerability of the subplate zone during the preterm period and may explain widespread changes in magnetic resonance signal intensity in early PWMI.
Collapse
Affiliation(s)
- Ivana Pogledic
- Inserm U676, Paris; Croatian Institute for Brain Research, Medical School, University of Zagreb, Zagreb
| | | | | | | | | | | |
Collapse
|
35
|
Strickland AD. Prevention of cerebral palsy, autism spectrum disorder, and attention deficit-hyperactivity disorder. Med Hypotheses 2014; 82:522-8. [PMID: 24581674 DOI: 10.1016/j.mehy.2014.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 01/24/2014] [Accepted: 02/03/2014] [Indexed: 12/31/2022]
Abstract
This hypothesis states that cerebral palsy (CP), autism spectrum disorder (ASD), and attention-deficit/hyperactivity disorder (ADHD) are all caused by an exaggerated central nervous system inflammatory response to a prenatal insult. This prenatal insult may be one or more episodes of ischemia-reperfusion, an infectious disease of the mother or the fetus, or other causes of maternal inflammation such as allergy or autoimmune disease. The resultant fetal inflammatory hyper-response injures susceptible neurons in the developing white matter of the brain in specific areas at specific gestational ages. The exaggerated neuroinflammatory response is theorized to occur between about 19 and 34 post-conception weeks for CP, about 32 and 40 weeks for ADHD, and about 36 and 48 weeks (i.e. 2 months after delivery) for ASD. The exaggerated inflammatory response is hypothesized to occur because present diets limit intake of effective antioxidants and omega-3 polyunsaturated fatty acids while increasing intake of omega-6 polyunsaturated fatty acids. Oxidation products of the omega-3 fatty acids docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) limit neuroinflammation while oxidation products of the omega-6 fatty acid arachidonic acid exacerbate inflammation. Preventative treatment should begin in all pregnant women during the first trimester and should include both DHA and an effective antioxidant for prevention of neuroinflammation. The suggested antioxidant would be N-acetylcysteine, though melatonin could be chosen instead. Combined DHA and NAC therapy is theorized to decrease the incidence of the three disorders by more than 75%.
Collapse
|
36
|
Jansen LA, Hevner RF, Roden WH, Hahn SH, Jung S, Gospe SM. Glial localization of antiquitin: implications for pyridoxine-dependent epilepsy. Ann Neurol 2014; 75:22-32. [PMID: 24122892 DOI: 10.1002/ana.24027] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 08/26/2013] [Accepted: 09/10/2013] [Indexed: 11/10/2022]
Abstract
OBJECTIVE A high incidence of structural brain abnormalities has been reported in individuals with pyridoxine-dependent epilepsy (PDE). PDE is caused by mutations in ALDH7A1, also known as antiquitin. How antiquitin dysfunction leads to cerebral dysgenesis is unknown. In this study, we analyzed tissue from a child with PDE as well as control human and murine brain to determine the normal distribution of antiquitin, its distribution in PDE, and associated brain malformations. METHODS Formalin-fixed human brain sections were subjected to histopathology and fluorescence immunohistochemistry studies. Frozen brain tissue was utilized for measurement of PDE-associated metabolites and Western blot analysis. Comparative studies of antiquitin distribution were performed in developing mouse brain sections. RESULTS Histologic analysis of PDE cortex revealed areas of abnormal radial neuronal organization consistent with type Ia focal cortical dysplasia. Heterotopic neurons were identified in subcortical white matter, as was cortical astrogliosis, hippocampal sclerosis, and status marmoratus of the basal ganglia. Highly elevated levels of lysine metabolites were present in postmortem PDE cortex. In control human and developing mouse brain, antiquitin immunofluorescence was identified in radial glia, mature astrocytes, ependyma, and choroid plexus epithelium, but not in neurons. In PDE cortex, antiquitin immunofluorescence was greatly attenuated with evidence of perinuclear accumulation in astrocytes. INTERPRETATION Antiquitin is expressed within glial cells in the brain, and its dysfunction in PDE is associated with neuronal migration abnormalities and other structural brain defects. These malformations persist despite postnatal pyridoxine supplementation and likely contribute to neurodevelopmental impairments.
Collapse
Affiliation(s)
- Laura A Jansen
- Department of Neurology, University of Washington, Seattle, WA; Seattle Children's Research Institute, Seattle, WA
| | | | | | | | | | | |
Collapse
|
37
|
Sukal-Moulton T, Krosschell KJ, Gaebler-Spira DJ, Dewald JP. Motor impairment factors related to brain injury timing in early hemiparesis. Part I: expression of upper-extremity weakness. Neurorehabil Neural Repair 2014; 28:13-23. [PMID: 24009182 PMCID: PMC3974904 DOI: 10.1177/1545968313500564] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Extensive neuromotor development occurs early in human life, but the time that a brain injury occurs during development has not been rigorously studied when quantifying motor impairments. OBJECTIVE This study investigated the impact of timing of brain injury on the magnitude and distribution of weakness in the paretic arm of individuals with childhood-onset hemiparesis. METHODS A total of 24 individuals with hemiparesis were divided into time periods of injury before birth (PRE-natal, n = 8), around the time of birth (PERI-natal, n = 8), or after 6 months of age (POST-natal, n = 8). They, along with 8 typically developing peers, participated in maximal isometric shoulder, elbow, wrist, and finger torque generation tasks using a multiple-degree-of-freedom load cell to quantify torques in 10 directions. A mixed-model ANOVA was used to determine the effect of group and task on a calculated relative weakness ratio between arms. RESULTS There was a significant effect of both time of injury group (P < .001) and joint torque direction (P < .001) on the relative weakness of the paretic arm. Distal joints were more affected compared with proximal joints, especially in the POST-natal group. CONCLUSIONS The distribution of weakness provides evidence for the relative preservation of ipsilateral corticospinal motor pathways to the paretic limb in those individuals injured earlier, whereas those who sustained later injury may rely more on indirect ipsilateral corticobulbospinal projections during the generation of torques with the paretic arm.
Collapse
Affiliation(s)
- Theresa Sukal-Moulton
- Department of Biomedical Engineering, Northwestern University
- Department of Physical Therapy and Human Movement Sciences, Northwestern University
| | | | - Deborah J. Gaebler-Spira
- Department of Physical Medicine and Rehabilitation, Northwestern University
- The Rehabilitation Institute of Chicago
| | - Julius P.A. Dewald
- Department of Biomedical Engineering, Northwestern University
- Department of Physical Therapy and Human Movement Sciences, Northwestern University
- Department of Physical Medicine and Rehabilitation, Northwestern University
| |
Collapse
|
38
|
Collins KA, Byard RW. Pediatric Natural Deaths. FORENSIC PATHOLOGY OF INFANCY AND CHILDHOOD 2014. [PMCID: PMC7123209 DOI: 10.1007/978-1-61779-403-2_36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Autopsies are important in the investigation of childhood deaths. Most natural deaths are unlikely to come to the attention of the forensic pathologist, particularly in cases where death occurs in hospital. During the neonatal period (up to 28 days of age), deaths most commonly occur as a result of prematurity and related conditions, chromosomal abnormalities, or congenital malformations. Beyond the neonatal period, trauma-related deaths and sudden infant death syndrome are more common. In terms of natural acquired diseases of childhood, certain conditions are prevalent based on age and may be encountered at autopsy. Common acquired diseases that cause death in infants and children up to 5 years of age include pneumonia and other respiratory diseases, other infectious diseases, and malignancies. In older children, mortality due to natural disease declines substantially with trauma being the major cause of death, and malignancies the major cause of acquired disease. Sudden and/or unexpected deaths in which a natural disease state was previously unknown are most likely to come under the jurisdiction of the medical examiner or coroner and may be related to an underlying natural disease. Depending on the underlying disease process, the approach can differ, and therefore familiarity with common causes of death during childhood is important in order to focus the autopsy so that special techniques can be used along with obtaining proper ancillary testing to arrive at an accurate diagnosis and cause of death.
Collapse
Affiliation(s)
- Kim A. Collins
- Fulton County Medical Examiner's Office, Emory University School of Medicine, Atlanta, Georgia USA
| | - Roger W. Byard
- University of Adelaide, Adelaide, South Australia Australia
| |
Collapse
|
39
|
Englander ZA, Pizoli CE, Batrachenko A, Sun J, Worley G, Mikati MA, Kurtzberg J, Song AW. Diffuse reduction of white matter connectivity in cerebral palsy with specific vulnerability of long range fiber tracts. Neuroimage Clin 2013; 2:440-7. [PMID: 24179798 PMCID: PMC3777769 DOI: 10.1016/j.nicl.2013.03.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/07/2013] [Accepted: 03/11/2013] [Indexed: 11/25/2022]
Abstract
Cerebral palsy (CP) is a heterogeneous group of non-progressive motor disorders caused by injury to the developing fetal or infant brain. Although the defining feature of CP is motor impairment, numerous other neurodevelopmental disabilities are associated with CP and contribute greatly to its morbidity. The relationship between brain structure and neurodevelopmental outcomes in CP is complex, and current evidence suggests that motor and developmental outcomes are related to the spatial pattern and extent of brain injury. Given that multiple disabilities are frequently associated with CP, and that there is increasing burden of neurodevelopmental disability with increasing motor severity, global white matter (WM) connectivity was examined in a cohort of 17 children with bilateral CP to test the hypothesis that increased global WM damage will be seen in the group of severely affected (Gross Motor Function Classification Scale (GMFCS) level of IV) as compared to moderately affected (GMFCS of II or III) individuals. Diffusion tensor tractography was performed and the resulting fibers between anatomically defined brain regions were quantified and analyzed in relation to GMFCS levels. Overall, a reduction in total WM connectivity throughout the brain in severe versus moderate CP was observed, including but not limited to regions associated with the sensorimotor system. Our results also show a diffuse and significant reduction in global inter-regional connectivity between severity groups, represented by inter-regional fiber count, throughout the brain. Furthermore, it was also observed that there is a significant difference (p = 0.02) in long-range connectivity in patients with severe CP as compared to those with moderate CP, whereas short-range connectivity was similar between groups. This new finding, which has not been previously reported in the CP literature, demonstrates that CP may involve distributed, network-level structural disruptions.
Collapse
Affiliation(s)
- Zoë A. Englander
- Brain Imaging and Analysis Center, Duke University Medical Center, USA
| | - Carolyn E. Pizoli
- Brain Imaging and Analysis Center, Duke University Medical Center, USA
- Department of Pediatrics, Duke University Medical Center, USA
| | | | - Jessica Sun
- Department of Pediatrics, Duke University Medical Center, USA
- The Robertson Cell and Translational Therapy Center, Duke University Medical Center, USA
| | - Gordon Worley
- Department of Pediatrics, Duke University Medical Center, USA
| | | | - Joanne Kurtzberg
- Department of Pediatrics, Duke University Medical Center, USA
- The Robertson Cell and Translational Therapy Center, Duke University Medical Center, USA
| | - Allen W. Song
- Brain Imaging and Analysis Center, Duke University Medical Center, USA
| |
Collapse
|
40
|
Sukal-Moulton T, Murray TM, Dewald JPA. Loss of independent limb control in childhood hemiparesis is related to time of brain injury onset. Exp Brain Res 2013; 225:455-63. [PMID: 23411673 DOI: 10.1007/s00221-012-3385-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/15/2012] [Indexed: 10/27/2022]
Abstract
This study investigated the presence of inter-limb activity at the elbow joint in individuals with childhood-onset hemiparesis, including spontaneous mirror movements during unilateral tasks and the ability to suppress them during bilateral tasks. Eighteen individuals with hemiparesis were divided into three categories of injury timing: before birth (PRE-natal), around the time of birth (PERI-natal), and after 6 months of age (POST-natal). Individuals with hemiparesis, as well as 12 typically developing peers, participated in unilateral and bilateral elbow flexion and extension tasks completed at maximal and submaximal effort while muscle activity was monitored and motor output was quantified by two multiple degrees-of-freedom load cells. Significantly, higher levels of paretic elbow flexion were found only in the PRE- and PERI-natal groups during the flexion of the non-paretic limb, which was modulated by effort level in both unilateral and bilateral tasks. The bilateral activation of elbow flexors in the PRE-/PERI-natal groups indicates potential use of a common cortical command source to drive both upper extremities, while the POST-natal/typically developing groups' flexors appear to receive input from different supraspinal structures.
Collapse
Affiliation(s)
- Theresa Sukal-Moulton
- Department of Physical Therapy and Human Movement Sciences, Northwestern University, 645 N Michigan Avenue Suite 1100, Chicago, IL 60611, USA
| | | | | |
Collapse
|
41
|
Otellin VA, Khozhai LI, Vataeva LA. Effect of hypoxia in early perinatal ontogenesis on behavior and structural characteristics of the rat brain. J EVOL BIOCHEM PHYS+ 2013. [DOI: 10.1134/s0022093012050088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
The influence of spatial working memory on ipsilateral remembered proprioceptive matching in adults with cerebral palsy. Exp Brain Res 2012; 223:259-69. [DOI: 10.1007/s00221-012-3256-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 08/31/2012] [Indexed: 11/24/2022]
|
43
|
Harvey L, Boksa P. Prenatal and postnatal animal models of immune activation: Relevance to a range of neurodevelopmental disorders. Dev Neurobiol 2012; 72:1335-48. [DOI: 10.1002/dneu.22043] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 06/18/2012] [Indexed: 11/11/2022]
|
44
|
Toomela A. Short-Term Memory in Young Adults With Spastic Diplegic Cerebral Palsy. Dev Neuropsychol 2012; 37:317-32. [DOI: 10.1080/87565641.2011.632461] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
45
|
Microglial reaction in axonal crossroads is a hallmark of noncystic periventricular white matter injury in very preterm infants. J Neuropathol Exp Neurol 2012; 71:251-64. [PMID: 22318128 DOI: 10.1097/nen.0b013e3182496429] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Disabilities after brain injury in very preterm infants have mainly been attributed to noncystic periventricular white matter injury (PWMI). We analyzed spatiotemporal patterns of PWMI in the brains of 18 very preterm infants (25-29 postconceptional weeks [pcw]), 7 preterm infants (30-34 pcw), and 10 preterm controls without PWMI. In very preterm infants, we examined PWMI in detail in 2 axonal crossroad areas in the frontal lobe: C1 (lateral to the lateral angle of the anterior horn of the lateral ventricle, at the exit of the internal capsule radiations) and C2 (above the corpus callosum and dorsal angle of the anterior horn). These brains had greater microglia-macrophage densities and activation but lesser astroglial reaction (glial fibrillary acidic protein and monocarboxylate transporter 1 expression) than in preterm cases with PWMI. In preterm infants, scattered necrotic foci were rimmed by axonal spheroids and ionized calcium binding adaptor molecule 1-positive macrophages. Diffuse lesions near these foci consisted primarily of hypertrophic and reactive astrocytes associated with fewer microglia. No differences in Olig2-positive preoligodendrocytes between noncystic PWMI and control cases were found. These data show that the growing axonal crossroad areas are highly vulnerable to PWMI in very preterm infants and highlight differences in glial activation patterns between very preterm and preterm infants.
Collapse
|
46
|
Zhang T, Wang M, Pan L, Ding W, Wang JG, Yang L, Liu M, Li W, Yan Z. Study of Gene Expression Profiles and Biological Mechanism of Cerebral Palsy Using a Monozygotic Twin Pair. Twin Res Hum Genet 2012; 10:496-507. [PMID: 17564508 DOI: 10.1375/twin.10.3.496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AbstractThe gene expression profile of a normal-suffering monozygotic twin pair is investigated to explore biological mechanisms of spastic type cerebral palsy. Main works include following three aspects: First, a cDNA microarray test is carried out to get the differentially expressed genes of the patient with cerebral palsy compared to her monozygotic twin sister. Second, these differentially expressed genes are searched for their bioinformation within 4 biological databases: FatiGO, FatiGOPlus, KEGG, and SOURCE. Third, a set of special genes and gene families are screened out from the spastic type cerebral palsy patient. These biological analyses reveal that those genes for cell junction are mostly down-regulated, while those genes for metabolism are mostly up-regulated. The individual genes, gene family, and their associated biological functions can reflect the pathological and physiological characteristics of the cerebral palsy.
Collapse
Affiliation(s)
- Tiane Zhang
- Genetics Laboratory, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Koltz MT, Tosun C, Kurland DB, Coksaygan T, Castellani RJ, Ivanova S, Gerzanich V, Simard JM. Tandem insults of prenatal ischemia plus postnatal raised intrathoracic pressure in a novel rat model of encephalopathy of prematurity. J Neurosurg Pediatr 2011; 8:628-39. [PMID: 22132923 PMCID: PMC3465975 DOI: 10.3171/2011.9.peds11174] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Encephalopathy of prematurity (EP) is common in preterm, low birth weight infants who require postnatal mechanical ventilation. The worst types of EP are the hemorrhagic forms, including choroid plexus, germinal matrix, periventricular, and intraventricular hemorrhages. Survivors exhibit life-long cognitive, behavioral, and motor abnormalities. Available preclinical models do not fully recapitulate the salient features of hemorrhagic EP encountered in humans. In this study, the authors evaluated a novel model using rats that featured tandem insults of transient prenatal intrauterine ischemia (IUI) plus transient postnatal raised intrathoracic pressure (RIP). METHODS Timed-pregnant Wistar rats were anesthetized and underwent laparotomy on embryonic Day 19. Intrauterine ischemia was induced by clamping the uterine and ovarian vasculature for 20 minutes. Natural birth occurred on embryonic Day 22. Six hours after birth, the pups were subjected to an episode of RIP, induced by injecting glycerol (50%, 13 μl/g intraperitoneally). Control groups included naive, sham surgery, and IUI alone. Pathological, histological, and behavioral analyses were performed on pups up to postnatal Day 52. RESULTS Compared with controls, pups subjected to IUI+RIP exhibited significant increases in postnatal mortality and hemorrhages in the choroid plexus, germinal matrix, and periventricular tissues as well as intraventricularly. On postnatal Days 35-52, they exhibited significant abnormalities involving complex vestibulomotor function and rapid spatial learning. On postnatal Day 52, the brain and body mass were significantly reduced. CONCLUSIONS Tandem insults of IUI plus postnatal RIP recapitulate many features of the hemorrhagic forms of EP found in humans, suggesting that these insults in combination may play important roles in pathogenesis.
Collapse
Affiliation(s)
- Michael T. Koltz
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Cigdem Tosun
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - David B. Kurland
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Turhan Coksaygan
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rudolph J. Castellani
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Svetlana Ivanova
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland,Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
48
|
In vivo magnetization transfer MRI shows dysmyelination in an ischemic mouse model of periventricular leukomalacia. J Cereb Blood Flow Metab 2011; 31:2009-18. [PMID: 21540870 PMCID: PMC3208153 DOI: 10.1038/jcbfm.2011.68] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Periventricular leukomalacia, PVL, is the leading cause of cerebral palsy in prematurely born infants, and therefore more effective interventions are required. The objective of this study was to develop an ischemic injury model of PVL in mice and to determine the feasibility of in vivo magnetization transfer (MT) magnetic resonance imaging (MRI) as a potential monitoring tool for the evaluation of disease severity and experimental therapeutics. Neonatal CD-1 mice underwent unilateral carotid artery ligation on postnatal day 5 (P5); at P60, in vivo T2-weighted (T2w) and MT-MRI were performed and correlated with postmortem histopathology. In vivo T2w MRI showed thinning of the right corpus callosum, but no significant changes in hippocampal and hemispheric volumes. Magnetization transfer MRI revealed significant white matter abnormalities in the bilateral corpus callosum and internal capsule. These quantitative MT-MRI changes correlated highly with postmortem findings of reduced myelin basic protein in bilateral white matter tracts. Ventriculomegaly and persistent astrogliosis were observed on the ligated side, along with evidence of axonopathy and fewer oligodendrocytes in the corpus callosum. We present an ischemia-induced mouse model of PVL, which has pathologic abnormalities resembling autopsy reports in infants with PVL. We further validate in vivo MRI techniques as quantitative monitoring tools that highly correlate with postmortem histopathology.
Collapse
|
49
|
Tuzun F, Kumral A, Dilek M, Ozbal S, Ergur B, Yesilirmak DC, Duman N, Yılmaz O, Ozkan H. Maternal omega-3 fatty acid supplementation protects against lipopolysaccharide-induced white matter injury in the neonatal rat brain. J Matern Fetal Neonatal Med 2011; 25:849-54. [DOI: 10.3109/14767058.2011.587917] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
50
|
Northington FJ, Chavez-Valdez R, Martin LJ. Neuronal cell death in neonatal hypoxia-ischemia. Ann Neurol 2011; 69:743-58. [PMID: 21520238 DOI: 10.1002/ana.22419] [Citation(s) in RCA: 269] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Perinatal hypoxic-ischemic encephalopathy (HIE) is a significant cause of mortality and morbidity in infants and young children. Therapeutic opportunities are very limited for neonatal and pediatric HIE. Specific neural systems and populations of cells are selectively vulnerable in HIE; however, the mechanisms of degeneration are unresolved. These mechanisms involve oxidative stress, excitotoxicity, inflammation, and the activation of several different cell death pathways. Decades ago the structural and mechanistic basis of the cellular degeneration in HIE was thought to be necrosis. Subsequently, largely due to advances in cell biology and to experimental animal studies, emphasis has been switched to apoptosis or autophagy mediated by programmed cell death (PCD) mechanisms as important forms of degeneration in HIE. We have conceptualized based on morphological and biochemical data that this degeneration is better classified according to an apoptosis-necrosis cell death continuum and that programmed cell necrosis has prominent contribution in the neurodegeneration of HIE in animal models. It is likely that neonatal HIE evolves through many cell death chreodes influenced by the dynamic injury landscape. The relevant injury mechanisms remain to be determined in human neonatal HIE, though preliminary work suggests a complexity in the cell death mechanisms greater than that anticipated from experimental animal models. The accurate identification of the various cell death chreodes and their mechanisms unfolding within the immature brain matrix could provide fresh insight for developing meaningful therapies for neonatal and pediatric HIE.
Collapse
Affiliation(s)
- Frances J Northington
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | |
Collapse
|