1
|
Impieri C, Ancona C, Bortolatto B, Laghetto I, Galzignato S, Nosadini M, Toldo I, D'Errico I, Sartori S, Calignano G, Cavicchiolo ME, Cavaliere E. Neonatal subpial hemorrhage: Padua experience and systematic review. Eur J Pediatr 2025; 184:265. [PMID: 40126707 PMCID: PMC11933206 DOI: 10.1007/s00431-025-06021-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 03/26/2025]
Abstract
Subpial hemorrhage (SPH) is a rare subtype of intracranial hemorrhage, predominantly affecting term neonates and often associated with cortical-subcortical infarction. We described the epidemiology of SPH by analyzing cases referred to our hospital and concurrently conducting a systematic review of the cases reported in the literature. We also illustrated factors associated with adverse outcomes. A retrospective study was conducted on neonates with SPH referred to our hospital from 2013 to 2023 (cohort 1). Additionally, a systematic literature review on neonatal SPH was performed using PubMed, Scopus, Cochrane, and Web of Science up to April 2024 (cohort 2). Cohorts 1 and 2 were pooled for combined analysis. A total of 173 cases were analyzed, 10 original cases (cohort 1) and 163 literature cases (cohort 2). Ninety-two percent was term/late preterm neonates (59% male). Clinical presentations included seizures (36%), apnea (36%), and encephalopathy (18%). Ninety-four percent was diagnosed with brain magnetic resonance imaging and/or cranial ultrasound. Lesions were located in the temporal lobe in 60%, with infarctions adjacent to SPH in 90%. Sixteen percent died, 53% was diagnosed with neurological impairment, and 8% with epilepsy. In a subcohort of 67 patients (cohort 3) with available individual data (10/10 from cohort 1, 57/163 from cohort 2), low birth weight (LBW), seizures, neonatal infections, and parenchymal hemorrhage were significantly associated with adverse outcomes. CONCLUSION Neonatal SPH is rare, predominantly located in the temporal lobe, and frequently presents with seizures and apneas. Neurologic sequelae are common, and parenchymal hemorrhage was strongly associated with neurological impairment in our study. WHAT IS KNOWN • Subpial hemorrhage is a rare subtype of intracranial extra-axial bleeding, often associated with cortical-subcortical infarction in the adjacent parenchyma, predominantly affecting male term neonates. The temporal lobe is the most commonly involved area, frequently exhibiting the "yin-yang sign" on brain MRI. WHAT IS NEW • This is the first systematic review of neonatal subpial hemorrhage, emphasizing a distinctive clinical presentation marked by seizures and apneas (potentially of ictal origin), consistent with a high prevalence of temporal lobe involvement. Prognostically, a significant incidence of neurological impairment was observed, and the occurrence of parenchymal hemorrhage adjacent to subpial hemorrhage was strongly associated with adverse outcomes.
Collapse
Affiliation(s)
- Cristina Impieri
- Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Claudio Ancona
- Department of Women's and Children's Health, University of Padua, Padua, Italy.
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy.
| | | | - Irene Laghetto
- Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Sofia Galzignato
- Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Margherita Nosadini
- Department of Women's and Children's Health, University of Padua, Padua, Italy
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Irene Toldo
- Department of Women's and Children's Health, University of Padua, Padua, Italy
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | | | - Stefano Sartori
- Department of Women's and Children's Health, University of Padua, Padua, Italy
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Giulia Calignano
- Department of Developmental Psychology and Socialization (DPSS), University of Padua, Padua, Italy
| | - Maria Elena Cavicchiolo
- Neonatal Intensive Care Unit, Department of Woman's and Child's Health, University Hospital of Padua, Padua, Italy
| | - Elena Cavaliere
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| |
Collapse
|
2
|
Pan S, Hale AT, Lemieux ME, Raval DK, Garton TP, Sadler B, Mahaney KB, Strahle JM. Iron homeostasis and post-hemorrhagic hydrocephalus: a review. Front Neurol 2024; 14:1287559. [PMID: 38283681 PMCID: PMC10811254 DOI: 10.3389/fneur.2023.1287559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Iron physiology is regulated by a complex interplay of extracellular transport systems, coordinated transcriptional responses, and iron efflux mechanisms. Dysregulation of iron metabolism can result in defects in myelination, neurotransmitter synthesis, and neuronal maturation. In neonates, germinal matrix-intraventricular hemorrhage (GMH-IVH) causes iron overload as a result of blood breakdown in the ventricles and brain parenchyma which can lead to post-hemorrhagic hydrocephalus (PHH). However, the precise mechanisms by which GMH-IVH results in PHH remain elusive. Understanding the molecular determinants of iron homeostasis in the developing brain may lead to improved therapies. This manuscript reviews the various roles iron has in brain development, characterizes our understanding of iron transport in the developing brain, and describes potential mechanisms by which iron overload may cause PHH and brain injury. We also review novel preclinical treatments for IVH that specifically target iron. Understanding iron handling within the brain and central nervous system may provide a basis for preventative, targeted treatments for iron-mediated pathogenesis of GMH-IVH and PHH.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Andrew T. Hale
- Department of Neurosurgery, University of Alabama at Birmingham School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mackenzie E. Lemieux
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Dhvanii K. Raval
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Thomas P. Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Brooke Sadler
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Hematology and Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Orthopedic Surgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
3
|
Warming H, Deinhardt K, Garland P, More J, Bulters D, Galea I, Vargas-Caballero M. Functional effects of haemoglobin can be rescued by haptoglobin in an in vitro model of subarachnoid haemorrhage. J Neurochem 2023; 167:90-103. [PMID: 37702203 DOI: 10.1111/jnc.15936] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023]
Abstract
During subarachnoid haemorrhage, a blood clot forms in the subarachnoid space releasing extracellular haemoglobin (Hb), which causes oxidative damage and cell death in surrounding tissues. High rates of disability and cognitive decline in SAH survivors are attributed to loss of neurons and functional connections during secondary brain injury. Haptoglobin sequesters Hb for clearance, but this scavenging system is overwhelmed after a haemorrhage. Whilst exogenous haptoglobin application can attenuate cytotoxicity of Hb in vitro and in vivo, the functional effects of sub-lethal Hb concentrations on surviving neurons and whether cellular function can be protected with haptoglobin treatment remain unclear. Here we use cultured neurons to investigate neuronal health and function across a range of Hb concentrations to establish the thresholds for cellular damage and investigate synaptic function. Hb impairs ATP concentrations and cytoskeletal structure. At clinically relevant but sub-lethal Hb concentrations, we find that synaptic AMPAR-driven currents are reduced, accompanied by a reduction in GluA1 subunit expression. Haptoglobin co-application can prevent these deficits by scavenging free Hb to reduce it to sub-threshold concentrations and does not need to be present at stoichiometric amounts to achieve efficacy. Haptoglobin itself does not impair measures of neuronal health and function at any concentration tested. Our data highlight a role for Hb in modifying synaptic function in surviving neurons, which may link to impaired cognition or plasticity after SAH and support the development of haptoglobin as a therapy for subarachnoid haemorrhage.
Collapse
Affiliation(s)
- Hannah Warming
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Katrin Deinhardt
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | | | - John More
- Bio Products Laboratory Limited, Elstree, UK
| | - Diederik Bulters
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Mariana Vargas-Caballero
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
4
|
Ramagiri S, Pan S, DeFreitas D, Yang PH, Raval DK, Wozniak DF, Esakky P, Strahle JM. Deferoxamine Prevents Neonatal Posthemorrhagic Hydrocephalus Through Choroid Plexus-Mediated Iron Clearance. Transl Stroke Res 2023; 14:704-722. [PMID: 36308676 PMCID: PMC10147846 DOI: 10.1007/s12975-022-01092-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 12/14/2022]
Abstract
Posthemorrhagic hydrocephalus occurs in up to 30% of infants with high-grade intraventricular hemorrhage and is associated with the worst neurocognitive outcomes in preterm infants. The mechanisms of posthemorrhagic hydrocephalus after intraventricular hemorrhage are unknown; however, CSF levels of iron metabolic pathway proteins including hemoglobin have been implicated in its pathogenesis. Here, we develop an animal model of intraventricular hemorrhage using intraventricular injection of hemoglobin at post-natal day 4 that results in acute and chronic hydrocephalus, pathologic choroid plexus iron accumulation, and subsequent choroid plexus injury at post-natal days 5, 7, and 15. This model also results in increased expression of aquaporin-1, Na+/K+/Cl- cotransporter 1, and Na+/K+/ATPase on the apical surface of the choroid plexus 24 h post-intraventricular hemorrhage. We use this model to evaluate a clinically relevant treatment strategy for the prevention of neurological sequelae after intraventricular hemorrhage using intraventricular administration of the iron chelator deferoxamine at the time of hemorrhage. Deferoxamine treatment prevented posthemorrhagic hydrocephalus for up to 11 days after intraventricular hemorrhage and prevented the development of sensorimotor gating deficits. In addition, deferoxamine treatment facilitated acute iron clearance through the choroid plexus and subsequently reduced choroid plexus iron levels at 24 h with reversal of hemoglobin-induced aquaporin-1 upregulation on the apical surface of the choroid plexus. Intraventricular administration of deferoxamine at the time of intraventricular hemorrhage may be a clinically relevant treatment strategy for preventing posthemorrhagic hydrocephalus and likely acts through promoting iron clearance through the choroid plexus to prevent hemoglobin-induced injury.
Collapse
Affiliation(s)
- Sruthi Ramagiri
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - Dakota DeFreitas
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - Peter H Yang
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - Dhvanii K Raval
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - David F Wozniak
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110-1093, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110-1093, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, 63110-1093, USA
| | - Prabagaran Esakky
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - Jennifer M Strahle
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA.
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
5
|
Wahjoepramono POP, Sasongko AB, Halim D, Aviani JK, Lukito PP, Adam A, Tsai YT, Wahjoepramono EJ, July J, Achmad TH. Hydrocephalus is an independent factor affecting morbidity and mortality of ICH patients: Systematic review and meta-analysis. World Neurosurg X 2023; 19:100194. [PMID: 37359762 PMCID: PMC10288487 DOI: 10.1016/j.wnsx.2023.100194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/04/2023] [Indexed: 06/28/2023] Open
Abstract
Background Despite advances in our knowledge of the causes, preventions, and treatments of stroke, it continues to be a leading cause of death and disability. The most common type of stroke-related morbidity and mortality is intracerebral haemorrhage (ICH). Many prognostication scores include an intraventricular extension (IVH) after ICH because it affects mortality independently. Although it is a direct result of IVH and results in significant damage, hydrocephalus (HC) has never been taken into account when calculating prognostication scores. This study aimed to evaluate the significance of hydrocephalus on the outcomes of ICH patients by meta-analysis. Methods Studies that compared the rates of mortality and/or morbidity in patients with ICH, ICH with IVH (ICH + IVH), and ICH with IVH and HC (ICH + IVH + HC) were identified. A meta-analysis was performed by using Mantel-Haezel Risk Ratio at 95% significance. Results This meta-analysis included thirteen studies. The findings indicate that ICH + IVH + HC has higher long-term (90-day) and short-term (30-day) mortality risks than ICH (4.26 and 2.30 higher risks, respectively) and ICH + IVH (1.96 and 1.54 higher risks). Patients with ICH + IVH + HC have lower rates of short-term (3 months) and long-term (6 months) good functional outcomes than those with ICH (0.66 and 0.38 times) or ICH + IVH (0.76 and 0.54 times). Confounding variables included vascular comorbidities, haemorrhage volume, midline shift, and an initial GCS score below 8. Conclusion Hydrocephalus causes a poorer prognosis in ICH patients. Thus, it is reasonable to suggest the inclusion of hydrocephalus in ICH prognostication scoring systems.
Collapse
Affiliation(s)
- Petra Octavian Perdana Wahjoepramono
- Department of Neurosurgery, Faculty of Medicine, Pelita Harapan University/Siloam Hospitals, Tangerang, Banten, Indonesia
- Post Graduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Aloysius Bagus Sasongko
- Department of Neurosurgery, Faculty of Medicine, Pelita Harapan University/Siloam Hospitals, Tangerang, Banten, Indonesia
- Post Graduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Danny Halim
- Department of Neurosurgery, Faculty of Medicine, Universitas Padjadjaran/Dr. Hasan Sadikin General Hospital, Bandung, West Java, Indonesia
- Research Center for Medical Genetics, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Jenifer Kiem Aviani
- Research Center for Medical Genetics, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| | - Patrick Putra Lukito
- Department of Neurosurgery, Faculty of Medicine, Pelita Harapan University/Siloam Hospitals, Tangerang, Banten, Indonesia
| | - Achmad Adam
- Department of Neurosurgery, Faculty of Medicine, Universitas Padjadjaran/Dr. Hasan Sadikin General Hospital, Bandung, West Java, Indonesia
| | - Yeo Tseng Tsai
- Division of Neurosurgery, Department of Surgery, National University Hospital, Singapore
| | - Eka Julianta Wahjoepramono
- Department of Neurosurgery, Faculty of Medicine, Pelita Harapan University/Siloam Hospitals, Tangerang, Banten, Indonesia
| | - Julius July
- Department of Neurosurgery, Faculty of Medicine, Pelita Harapan University/Siloam Hospitals, Tangerang, Banten, Indonesia
| | - Tri Hanggono Achmad
- Research Center for Medical Genetics, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
- Department of Basic Medical Science, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
| |
Collapse
|
6
|
Garcia-Bonilla M, Nair A, Moore J, Castaneyra-Ruiz L, Zwick SH, Dilger RN, Fleming SA, Golden RK, Talcott MR, Isaacs AM, Limbrick DD, McAllister JP. Impaired neurogenesis with reactive astrocytosis in the hippocampus in a porcine model of acquired hydrocephalus. Exp Neurol 2023; 363:114354. [PMID: 36822393 PMCID: PMC10411821 DOI: 10.1016/j.expneurol.2023.114354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/03/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND Hydrocephalus is a neurological disease with an incidence of 0.3-0.7 per 1000 live births in the United States. Ventriculomegaly, periventricular white matter alterations, inflammation, and gliosis are among the neuropathologies associated with this disease. We hypothesized that hippocampus structure and subgranular zone neurogenesis are altered in untreated hydrocephalus and correlate with recognition memory deficits. METHODS Hydrocephalus was induced by intracisternal kaolin injections in domestic juvenile pigs (43.6 ± 9.8 days). Age-matched sham controls received similar saline injections. MRI was performed to measure ventricular volume, and/or hippocampal and perirhinal sizes at 14 ± 4 days and 36 ± 8 days post-induction. Recognition memory was assessed one week before and after kaolin induction. Histology and immunohistochemistry in the hippocampus were performed at sacrifice. RESULTS The hippocampal width and the perirhinal cortex thickness were decreased (p < 0.05) in hydrocephalic pigs 14 ± 4 days post-induction. At sacrifice (36 ± 8 days post-induction), significant expansion of the cerebral ventricles was detected (p = 0.005) in hydrocephalic pigs compared with sham controls. The area of the dorsal hippocampus exhibited a reduction (p = 0.035) of 23.4% in the hydrocephalic pigs at sacrifice. Likewise, in hydrocephalic pigs, the percentages of neuronal precursor cells (doublecortin+ cells) and neurons decreased (p < 0.01) by 32.35%, and 19.74%, respectively, in the subgranular zone of the dorsal hippocampus. The percentage of reactive astrocytes (vimentin+) was increased (p = 0.041) by 48.7%. In contrast, microglial cells were found to decrease (p = 0.014) by 55.74% in the dorsal hippocampus in hydrocephalic pigs. There was no difference in the recognition index, a summative measure of learning and memory, one week before and after the induction of hydrocephalus. CONCLUSION In untreated juvenile pigs, acquired hydrocephalus caused morphological alterations, reduced neurogenesis, and increased reactive astrocytosis in the hippocampus and perirhinal cortex.
Collapse
Affiliation(s)
- Maria Garcia-Bonilla
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA.
| | - Arjun Nair
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Jason Moore
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | | | - Sarah H Zwick
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Ryan N Dilger
- Neuroscience Program, Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Stephen A Fleming
- Neuroscience Program, Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA; Traverse Science, Champaign, IL 61801, USA
| | - Rebecca K Golden
- Neuroscience Program, Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Michael R Talcott
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; AbbVie, Inc., North Chicago, IL 60064, USA
| | - Albert M Isaacs
- Department of Neurological Surgery, Vanderbilt, University Medical Center, Nashville, TN 37232, USA
| | - David D Limbrick
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - James P McAllister
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Fernández de Gamarra-Oca L, Kvanta H, Broström L, Nosko D, Eklöf E, Ojeda N, Zubiaurre-Elorza L, Padilla N, Ådén U. Hippocampal volumes and cognitive performance in children born extremely preterm with and without low-grade intraventricular haemorrhage. Brain Struct Funct 2023:10.1007/s00429-023-02643-w. [PMID: 37081204 DOI: 10.1007/s00429-023-02643-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/10/2023] [Indexed: 04/22/2023]
Abstract
Children born extremely preterm, especially those with intraventricular haemorrhage (IVH), are at increased risk of adverse cognitive outcomes during childhood. The present study aimed to explore the effects of IVH (grades I-II) on hippocampal volumes, and their correlates with cognitive performance. The sample consisted of 94 participants, including 54 children born extremely preterm (19 with IVH, grades I-II), and 40 children born at term. All participants underwent a magnetic resonance imaging study at the age of 10 (Mage = 10.20 years; SDage = 0.78), and 74 of them (45 extremely preterm and 29 full-term) carried out a cognitive assessment at 12 years old. Children born extremely preterm had lower scores in cognitive performance compared to their full-term peers. Significant positive partial correlations were observed between global bilateral hippocampus, left CA-field, and left subiculum volumes with processing speed in the full-term group, while no significant correlations were found in the extremely preterm group. Moderation analyses in the extremely preterm sample revealed that low-grade IVH moderated the relationship between right hippocampal volume and full-IQ (F(4,40) = 5.42, p = 0.001, R2 = 0.35). Having greater right hippocampal volume had a protective effect on full-IQ in those children born extremely preterm with low-grade IVH.
Collapse
Affiliation(s)
- L Fernández de Gamarra-Oca
- Department of Psychology, Faculty of Health Sciences, University of Deusto, Avda de Las Universidades 24, 48007, Bilbao, Bizkaia, Spain.
| | - H Kvanta
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - L Broström
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - D Nosko
- Department of Paediatrics, Örebro University Hospital, Örebro, Sweden
| | - E Eklöf
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - N Ojeda
- Department of Psychology, Faculty of Health Sciences, University of Deusto, Avda de Las Universidades 24, 48007, Bilbao, Bizkaia, Spain
| | - L Zubiaurre-Elorza
- Department of Psychology, Faculty of Health Sciences, University of Deusto, Avda de Las Universidades 24, 48007, Bilbao, Bizkaia, Spain
| | - N Padilla
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - U Ådén
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Department of Neonatal Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
8
|
Wu Y, Sun Y, Wang X, Zhu C. The Regulated Cell Death and Potential Interventions in Preterm Infants after Intracerebral Hemorrhage. Curr Neuropharmacol 2023; 21:1488-1503. [PMID: 36397619 PMCID: PMC10472811 DOI: 10.2174/1570159x21666221117155209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/21/2022] Open
Abstract
Intracerebral hemorrhage (ICH) in preterm infants is one of the major co-morbidities of preterm birth and is associated with long-term neurodevelopmental deficits. There are currently no widely accepted treatments to prevent ICH or therapies for the neurological sequelae. With studies broadening the scope of cell death, the newly defined concept of regulated cell death has enriched our understanding of the underlying mechanisms of secondary brain injury after ICH and has suggested potential interventions in preterm infants. In this review, we will summarize the current evidence for regulated cell death pathways in preterm infants after ICH, including apoptosis, necroptosis, pyroptosis, ferroptosis, autophagy, and PANoptosis as well as several potential intervention strategies that may protect the immature brain from secondary injury after ICH through regulating regulated cell death.
Collapse
Affiliation(s)
- Yanan Wu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
| | - Yanyan Sun
- Department of Human Anatomy, School of Basic Medical Science, Zhengzhou University, Zhengzhou, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
9
|
Lee SY, Schmit BD, Kurpad SN, Budde MD. Acute Magnetic Resonance Imaging Predictors of Chronic Motor Function and Tissue Sparing in Rat Cervical Spinal Cord Injury. J Neurotrauma 2022; 39:1727-1740. [PMID: 35708112 PMCID: PMC9734017 DOI: 10.1089/neu.2022.0034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Predicting functional outcomes from spinal cord injury (SCI) at the acute setting is important for patient management. This work investigated the relationship of early magnetic resonance imaging (MRI) biomarkers in a rat model of cervical contusion SCI with long-term functional outcome and tissue sparing. Forty rats with contusion injury at C5 at either the spinal cord midline (bilateral) or over the lateral cord (unilateral) were examined using in vivo multi-modal quantitative MRI at 1 day post-injury. The extent of T2-weighted hyperintensity reflecting edema was greater in the bilateral model compared with the unilateral injury. Diffusion tensor imaging (DTI) exhibited microscopic damage in similar regions of the cord as reductions in fractional anisotropy (FA) and mean diffusivity (MD), but DTI parameter maps were also confounded by the presence of vasogenic edema that locally increased FA and MD. In comparison, filtered diffusion-weighted imaging (fDWI) more clearly delineated the location of acute axonal damage without effects of vasogenic edema. Pairwise correlation analysis revealed that 28-day motor functional outcomes were most strongly associated with the extent of edema (R = -0.69). Principal component analysis identified close associations of motor functional score with tissue sparing, the extent of edema, lesion area, and injury type (unilateral or bilateral). Among the diffusion MRI parameters, lesion areas measured with fDWI had the strongest association with functional outcome (R = -0.41). Voxelwise correlation analysis identified a locus of white matter damage associated with function in the dorsal white matter, although this was likely driven by variance across the two injury patterns (unilateral and bilateral injury). Nonetheless, correlation with motor function within the damaged region found in the voxelwise analysis outperformed morphological lesion area measurement as a predictor of chronic function. Collectively, this study characterized anatomical and diffusion MRI signatures of acute SCI at cervical spine and their association with chronic functional outcomes and histological results.
Collapse
Affiliation(s)
- Seung-Yi Lee
- Neuroscience Doctoral Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Biophysics Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Brian D. Schmit
- Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Shekar N. Kurpad
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Matthew D. Budde
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
10
|
Lubinsky M, Encha-Razavi F. Delineating septo-optic dysplasia. Birth Defects Res 2022; 114:1343-1353. [PMID: 36200678 DOI: 10.1002/bdr2.2095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/05/2022] [Accepted: 09/08/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Septo-optic dysplasia (SOD), once a variable triad of septum pellucidum defects (SPDs), optic nerve hypoplasia (ONH), and hypopituitarism, has had multiple findings added, with uncertain causes, definitions, and limits. METHOD Literature review. RESULTS SOD is a complex vascular sequence with confounders. CONCLUSIONS Proximal anterior cerebral artery trunk disruptions cause overlapping primary effects, giving ONH alone most often, and isolated SPD less. ONH disruptions can spread to pituitary, SPD disruptions to the cerebral cortex, causing schizencephaly and related anomalies. Pituitary defects are rare without ONH, and cortical findings are rare without SPD. Extensions are unidirectional, so isolated pituitary or cortical defects are separate from SOD. Micro- an- ophthalmia, a suggested ONH variant, is not part of SOD. Disruption by-products can affect development, causing cognitive and endocrine issues, and structural anomalies such as corpus callosum thinning, ventriculomegaly, and hippocampal and olfactory findings. Limbic extensions may also contribute to the same structural defects as by-products. Midline CNS developmental anomalies can act as disruptive foci, most likely through vascular variants, but have separate pathogenesis. Relative frequencies of specific pituitary hormone defects change as SOD rates increase. Increasing relative rates of midline CNS developmental defects and cortical anomalies are consistent with rising levels of exogenous exposures sensitizing to midline predispositions.
Collapse
|
11
|
Miller BA, Pan S, Yang PH, Wang C, Trout AL, DeFreitas D, Ramagiri S, Olson SD, Strahle JM. Modeling Neonatal Intraventricular Hemorrhage through Intraventricular Injection of Hemoglobin. J Vis Exp 2022:10.3791/63345. [PMID: 36094266 PMCID: PMC10028568 DOI: 10.3791/63345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Neonatal intraventricular hemorrhage (IVH) is a common consequence of premature birth and leads to brain injury, posthemorrhagic hydrocephalus (PHH), and lifelong neurological deficits. While PHH can be treated by temporary and permanent cerebrospinal fluid (CSF) diversion procedures (ventricular reservoir and ventriculoperitoneal shunt, respectively), there are no pharmacological strategies to prevent or treat IVH-induced brain injury and hydrocephalus. Animal models are needed to better understand the pathophysiology of IVH and test pharmacological treatments. While there are existing models of neonatal IVH, those that reliably result in hydrocephalus are often limited by the necessity for large-volume injections, which may complicate modeling of the pathology or introduce variability in the clinical phenotype observed. Recent clinical studies have implicated hemoglobin and ferritin in causing ventricular enlargement after IVH. Here, we develop a straightforward animal model that mimics the clinical phenotype of PHH utilizing small-volume intraventricular injections of the blood breakdown product hemoglobin. In addition to reliably inducing ventricular enlargement and hydrocephalus, this model results in white matter injury, inflammation, and immune cell infiltration in periventricular and white matter regions. This paper describes this clinically relevant, simple method for modeling IVH-PHH in neonatal rats using intraventricular injection and presents methods for quantifying ventricle size post injection.
Collapse
Affiliation(s)
- Brandon A Miller
- Department of Neurosurgery, University of Kentucky; Department of Pediatric Surgery, University of Texas
| | - Shelei Pan
- Department of Neurological Surgery, Washington University in St. Louis School of Medicine
| | - Peter H Yang
- Department of Neurological Surgery, Washington University in St. Louis School of Medicine
| | | | | | - Dakota DeFreitas
- Department of Neurological Surgery, Washington University in St. Louis School of Medicine
| | - Sruthi Ramagiri
- Department of Neurological Surgery, Washington University in St. Louis School of Medicine
| | - Scott D Olson
- Department of Pediatric Surgery, University of Texas
| | - Jennifer M Strahle
- Department of Neurological Surgery, Washington University in St. Louis School of Medicine; Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine; Department of Pediatrics, Washington University in St. Louis School of Medicine;
| |
Collapse
|
12
|
Vinukonda G, La Gamma EF. Emerging therapies for brain recovery after IVH in neonates: Cord blood derived Mesenchymal Stem Cells (MSC) and Unrestricted Somatic Stem Cells (USSC). Semin Perinatol 2022; 46:151598. [PMID: 35589461 DOI: 10.1016/j.semperi.2022.151598] [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] [Indexed: 11/16/2022]
Abstract
In this report, we summarize evidence on mechanisms of injury after intraventricular hemorrhage resulting in post-hemorrhagic white matter injury and hydrocephalus and correlate that with the possibility of cellular therapy. We describe how two stem cell lines (MSC & USSC) acting in a paracrine fashion offer promise for attenuating the magnitude of injury in animal models and for improved functional recovery by: lowering the magnitude of apoptosis and neuronal cell death, reducing inflammation, and thus, mitigating white matter injury that culminates in improved motor and neurocognitive outcomes. Animal models of IVH are analyzed for their similarity to the human condition and we discuss merits of each approach. Studies on stem cell therapy for IVH in human neonates is described. Lastly, we offer suggestions on what future studies are needed to better understand mechanisms of injury and recovery and argue that human trials need to be expanded in parallel to animal research.
Collapse
Affiliation(s)
- Govindaiah Vinukonda
- Department of Pediatrics, Cell Biology & Anatomy New York Medical College, Valhalla, NY
| | - Edmund F La Gamma
- Department of Pediatrics, Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY.
| |
Collapse
|
13
|
Prediction of adult post-hemorrhagic hydrocephalus: a risk score based on clinical data. Sci Rep 2022; 12:12213. [PMID: 35842469 PMCID: PMC9288433 DOI: 10.1038/s41598-022-16577-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/12/2022] [Indexed: 11/08/2022] Open
Abstract
There is lacking research on risk factors and prediction models associated with Post-hemorrhagic hydrocephalus (PHH). Thus, this present study aimed to analyze the risk factors of PHH and establish a risk-scoring system through a large-scale study. A retrospective study of 382 patients with intracranial hemorrhage assessed age, history and diagnosis, Glasgow coma score (GCS), and fever time. After univariate and logistic regression analysis, a risk scoring system was established according to independent risk factors and evaluated using the area under the curve (AUC). Of the 382 patients, 133 (34.8%) had PHH, 43 (11.3%) received surgical treatment. Factor classification showed that age > 60 years old [odds ratio (OR): 0.347, II = 5 points], GCS < 5 (OR: 0.09, IV = 10 points), GCS 6‒8 (OR = 0.232, III = 6 points), fever time > 9 (OR: 0.202, III = 7 points), fever time 5-9 (OR: 0.341, II = 5 points), CSF-TP x time > 14,4000 group (OR: 0.267, IV = 6 points), and CSF-TP x time 9,601‒14,400 group (OR: 0.502, III = 3 points) were independent risk factors. The result of the receiver operating characteristic (ROC) prediction showed that AUC = 0.790 (0.744‒0.836). Low-risk (IV-VII), moderate (VIII-X), and high-risk group (XI-XIII) incidence of PHH were 11.76%, 50.55%, and 70.00% (p < 0.001), respectively. The coincidence rates in the validation cohort were 26.00%, 74.07%, and 100.0% (p < 0.001), respectively. AUC value was 0.860 (0.780‒0.941). The predictive model was conducive to determining the occurrence of PHH and facilitating early intervention.
Collapse
|
14
|
Lubinsky M. Hypothesis: By-products of vascular disruption carried in the CSF affect prenatal brain development. Birth Defects Res 2022; 114:847-854. [PMID: 35775635 DOI: 10.1002/bdr2.2064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/31/2022] [Accepted: 06/12/2022] [Indexed: 01/24/2023]
Abstract
Prenatal CNS disruptions can be associated with physically separate findings. Examples include cognitive issues in septo-optic dysplasia and sporadic and WNT1-related unilateral cerebellar hypoplasia, and physical findings such as thinning of the corpus callosum, ventriculomegaly, hippocampal abnormalities, olfactory tract and bulb hypoplasia, and distant cortical dysplasias with schizencephaly. Similar effects to toxicities with intraventricular hemorrhage in prematurity could occur earlier in development. CSF transportation of disruption by-products would provide access to vulnerable areas through inflammatory effects on blood-brain barrier permeability. Outcomes are influenced by location and volume of byproducts in the CSF, timing, transport, and inflammatory responses. A particular association of vermis disruption with cognitive issues may be related to CSF flow distortions that avoid toxin dilutions in the third ventricle. Symmetrical contralateral cortical dysplasia with schizencephaly may reflect immunovascular field-related vulnerabilities seen in situations such as vitiligo.
Collapse
|
15
|
Nadeem T, Bommareddy A, Bolarinwa L, Cuervo H. Pericyte dynamics in the mouse germinal matrix angiogenesis. FASEB J 2022; 36:e22339. [PMID: 35506590 DOI: 10.1096/fj.202200120r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022]
Abstract
Germinal matrix-intraventricular hemorrhage (GM-IVH) is the most devastating neurological complication in premature infants. GM-IVH usually begins in the GM, a highly vascularized region of the developing brain where glial and neuronal precursors reside underneath the lateral ventricular ependyma. Previous studies using human fetal tissue have suggested increased angiogenesis and paucity of pericytes as key factors contributing to GM-IVH pathogenesis. Yet, despite its relevance, the mechanisms underlying the GM vasculature's susceptibility to hemorrhage remain poorly understood. To gain better understanding on the vascular dynamics of the GM, we performed a comprehensive analysis of the mouse GM vascular endothelium and pericytes during development. We hypothesize that vascular development of the mouse GM will provide a good model for studies of human GM vascularization and provide insights into the role of pericytes in GM-IVH pathogenesis. Our findings show that the mouse GM presents significantly greater vascular area and vascular branching compared to the developing cortex (CTX). Analysis of pericyte coverage showed abundance in PDGFRβ-positive and NG2-positive pericyte coverage in the GM similar to the developing CTX. However, we found a paucity in Desmin-positive pericyte coverage of the GM vasculature. Our results underscore the highly angiogenic nature of the GM and reveal that pericytes in the developing mouse GM exhibit distinct phenotypical and likely functional characteristics compared to other brain regions which might contribute to the high susceptibility of the GM vasculature to hemorrhage.
Collapse
Affiliation(s)
- Taliha Nadeem
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Apoorva Bommareddy
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Lolade Bolarinwa
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Henar Cuervo
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
16
|
Tamakoshi K, Maeda M, Murohashi N, Saito A. Effect of exercise from a very early stage after intracerebral hemorrhage on microglial and macrophage reactivity states in rats. Neuroreport 2022; 33:304-311. [PMID: 35594443 DOI: 10.1097/wnr.0000000000001782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE This study investigated the effects of exercise, starting very early after intracerebral hemorrhage (ICH), on microglia and macrophages in a rat model. Collagenase solution was injected into the left striatum to induce ICH. METHODS Rats were randomly assigned to receive placebo surgery without exercise (sham surgery), ICH without exercise (ICH), or ICH with very early exercise (ICH + VET). The ICH + VET group was subjected to treadmill running 6 h, 24 h, and days 2-6 after ICH. Motor function assessment was performed using the ladder test and rotarod test 3 h, 25 h, and 7 days after ICH. Postexercise brain tissue was collected on day 8 after surgery to investigate the lesion volume. Very early exercise temporarily worsened motor dysfunction. The protein expression levels of the macrophage and microglial markers CD80, CD163, and TMEM119 were analyzed 6 h, 24 h, and 8 days after ICH. Protein analysis of NeuN, GFAP, and PSD95 was also performed on day 8 after ICH. RESULTS There was no significant difference in lesion volume between the ICH and ICH + VET groups on day 8 after ICH. Exercise from very early stage prevented elevated CD163 protein expression. CONCLUSION Very early exercise may inhibit the activation of anti-inflammatory-associated macrophages/microglia.
Collapse
Affiliation(s)
- Keigo Tamakoshi
- Department of Physical Therapy, Niigata University of Health and Welfare
- Institute for Human Movement and Medical Sciences, Niigata University of Health and Welfare
| | | | - Nae Murohashi
- Niigata Seiro Hospital, Rehabilitation, Seiro, Japan
| | - Ami Saito
- Department of Physical Therapy, Niigata University of Health and Welfare
| |
Collapse
|
17
|
Holste KG, Xia F, Ye F, Keep RF, Xi G. Mechanisms of neuroinflammation in hydrocephalus after intraventricular hemorrhage: a review. Fluids Barriers CNS 2022; 19:28. [PMID: 35365172 PMCID: PMC8973639 DOI: 10.1186/s12987-022-00324-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023] Open
Abstract
Intraventricular hemorrhage (IVH) is a significant cause of morbidity and mortality in both neonatal and adult populations. IVH not only causes immediate damage to surrounding structures by way of mass effect and elevated intracranial pressure; the subsequent inflammation causes additional brain injury and edema. Of those neonates who experience severe IVH, 25-30% will go on to develop post-hemorrhagic hydrocephalus (PHH). PHH places neonates and adults at risk for white matter injury, seizures, and death. Unfortunately, the molecular determinants of PHH are not well understood. Within the past decade an emphasis has been placed on neuroinflammation in IVH and PHH. More information has come to light regarding inflammation-induced fibrosis and cerebrospinal fluid hypersecretion in response to IVH. The aim of this review is to discuss the role of neuroinflammation involving clot-derived neuroinflammatory factors including hemoglobin/iron, peroxiredoxin-2 and thrombin, as well as macrophages/microglia, cytokines and complement in the development of PHH. Understanding the mechanisms of neuroinflammation after IVH may highlight potential novel therapeutic targets for PHH.
Collapse
Affiliation(s)
- Katherine G Holste
- Department of Neurosurgery, University of Michigan, 3470 Taubman Center, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109-5338, USA.
| | - Fan Xia
- Department of Neurosurgery, University of Michigan, 3470 Taubman Center, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109-5338, USA
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fenghui Ye
- Department of Neurosurgery, University of Michigan, 3470 Taubman Center, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109-5338, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, 3470 Taubman Center, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109-5338, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, 3470 Taubman Center, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109-5338, USA.
- , 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
18
|
Paturu M, Triplett RL, Thukral S, Alexopoulos D, Smyser CD, Limbrick DD, Strahle JM. Does ventricle size contribute to cognitive outcomes in posthemorrhagic hydrocephalus? Role of early definitive intervention. J Neurosurg Pediatr 2022; 29:10-20. [PMID: 34653990 PMCID: PMC8743027 DOI: 10.3171/2021.4.peds212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/28/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Posthemorrhagic hydrocephalus (PHH) is associated with significant morbidity, smaller hippocampal volumes, and impaired neurodevelopment in preterm infants. The timing of temporary CSF (tCSF) diversion has been studied; however, the optimal time for permanent CSF (pCSF) diversion is unknown. The objective of this study was to determine whether cumulative ventricle size or timing of pCSF diversion is associated with neurodevelopmental outcome and hippocampal size in preterm infants with PHH. METHODS Twenty-five very preterm neonates (born at ≤ 32 weeks' gestational age) with high-grade intraventricular hemorrhage (IVH), subsequent PHH, and pCSF diversion with a ventriculoperitoneal shunt (n = 20) or endoscopic third ventriculostomy (n = 5) were followed until 2 years of age. Infants underwent serial cranial ultrasounds from birth until 1 year after pCSF diversion, brain MRI at term-equivalent age, and assessment based on the Bayley Scales of Infant and Toddler Development, Third Edition, at 2 years of age. Frontooccipital horn ratio (FOHR) measurements were derived from cranial ultrasounds and term-equivalent brain MRI. Hippocampal volumes were segmented and calculated from term-equivalent brain MRI. Cumulative ventricle size until the time of pCSF diversion was estimated using FOHR measurements from each cranial ultrasound performed prior to permanent intervention. RESULTS The average gestational ages at tCSF and pCSF diversion were 28.9 and 39.0 weeks, respectively. An earlier chronological age at the time of pCSF diversion was associated with larger right hippocampal volumes on term-equivalent MRI (Pearson's r = -0.403, p = 0.046) and improved cognitive (r = -0.554, p = 0.047), motor (r = -0.487, p = 0.048), and language (r = -0.414, p = 0.021) outcomes at 2 years of age. Additionally, a smaller cumulative ventricle size from birth to pCSF diversion was associated with larger right hippocampal volumes (r = -0.483, p = 0.014) and improved cognitive (r = -0.711, p = 0.001), motor (r = -0.675, p = 0.003), and language (r = -0.618, p = 0.011) outcomes. There was no relationship between time to tCSF diversion or cumulative ventricle size prior to tCSF diversion and neurodevelopmental outcome or hippocampal size. Finally, a smaller cumulative ventricular size prior to either tCSF diversion or pCSF diversion was associated with a smaller ventricular size 1 year after pCSF diversion (r = 0.422, p = 0.040, R2 = 0.178 and r = 0.519, p = 0.009, R2 = 0.269, respectively). CONCLUSIONS In infants with PHH, a smaller cumulative ventricular size and shorter time to pCSF diversion were associated with larger right hippocampal volumes, improved neurocognitive outcomes, and reduced long-term ventriculomegaly. Future prospective randomized studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Mounica Paturu
- Department of Neurological Surgery, Washington University in St. Louis, Missouri
| | | | - Siddhant Thukral
- Department of Neurological Surgery, Washington University in St. Louis, Missouri
| | | | - Christopher D. Smyser
- Department of Neurology, Washington University in St. Louis, Missouri
- Department of Pediatrics, Washington University in St. Louis, Missouri
- Department of Radiology, Washington University in St. Louis, Missouri
| | - David D. Limbrick
- Department of Neurological Surgery, Washington University in St. Louis, Missouri
| | - Jennifer M. Strahle
- Department of Neurological Surgery, Washington University in St. Louis, Missouri
- Department of Pediatrics, Washington University in St. Louis, Missouri
| |
Collapse
|
19
|
Akeret K, Buzzi RM, Schaer CA, Thomson BR, Vallelian F, Wang S, Willms J, Sebök M, Held U, Deuel JW, Humar R, Regli L, Keller E, Hugelshofer M, Schaer DJ. Cerebrospinal fluid hemoglobin drives subarachnoid hemorrhage-related secondary brain injury. J Cereb Blood Flow Metab 2021; 41:3000-3015. [PMID: 34102922 PMCID: PMC8545037 DOI: 10.1177/0271678x211020629] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Secondary brain injury after aneurysmal subarachnoid hemorrhage (SAH-SBI) contributes to poor outcomes in patients after rupture of an intracranial aneurysm. The lack of diagnostic biomarkers and novel drug targets represent an unmet need. The aim of this study was to investigate the clinical and pathophysiological association between cerebrospinal fluid hemoglobin (CSF-Hb) and SAH-SBI. In a cohort of 47 patients, we collected daily CSF-samples within 14 days after aneurysm rupture. There was very strong evidence for a positive association between spectrophotometrically determined CSF-Hb and SAH-SBI. The accuracy of CSF-Hb to monitor for SAH-SBI markedly exceeded that of established methods (AUC: 0.89 [0.85-0.92]). Temporal proteome analysis revealed erythrolysis accompanied by an adaptive macrophage response as the two dominant biological processes in the CSF-space after aneurysm rupture. Ex-vivo experiments on the vasoconstrictive and oxidative potential of Hb revealed critical inflection points overlapping CSF-Hb thresholds in patients with SAH-SBI. Selective depletion and in-solution neutralization by haptoglobin or hemopexin efficiently attenuated the vasoconstrictive and lipid peroxidation activities of CSF-Hb. Collectively, the clinical association between high CSF-Hb levels and SAH-SBI, the underlying pathophysiological rationale, and the favorable effects of haptoglobin and hemopexin in ex-vivo experiments position CSF-Hb as a highly attractive biomarker and potential drug target.
Collapse
Affiliation(s)
- Kevin Akeret
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland
| | - Raphael M Buzzi
- Division of Internal Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Christian A Schaer
- Department of Anesthesiology, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Bart R Thomson
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland
| | - Florence Vallelian
- Division of Internal Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Sophie Wang
- Neurointensive Care Unit, Department of Neurosurgery and Institute of Intensive Care Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Jan Willms
- Neurointensive Care Unit, Department of Neurosurgery and Institute of Intensive Care Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Martina Sebök
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland
| | - Ulrike Held
- Epidemiology, Biostatistics and Prevention Institute, Department of Biostatistics, University of Zurich; Zurich, Switzerland
| | - Jeremy W Deuel
- Division of Internal Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Rok Humar
- Division of Internal Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland
| | - Emanuela Keller
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland.,Neurointensive Care Unit, Department of Neurosurgery and Institute of Intensive Care Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| | - Michael Hugelshofer
- Department of Neurosurgery, Clinical Neuroscience Center, Universitätsspital und University of Zurich; Zurich, Switzerland
| | - Dominik J Schaer
- Division of Internal Medicine, Universitätsspital and University of Zurich; Zurich, Switzerland
| |
Collapse
|
20
|
Wang M, Xia F, Wan S, Hua Y, Keep RF, Xi G. Role of Complement Component 3 in Early Erythrolysis in the Hematoma After Experimental Intracerebral Hemorrhage. Stroke 2021; 52:2649-2660. [PMID: 34176310 DOI: 10.1161/strokeaha.121.034372] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ming Wang
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.).,Brain Center, Zhejiang Hospital, Zhejiang University Medical School, Hangzhou, China (M.W., S.W.)
| | - Fan Xia
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.)
| | - Shu Wan
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.).,Brain Center, Zhejiang Hospital, Zhejiang University Medical School, Hangzhou, China (M.W., S.W.)
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.)
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.)
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor (M.W., F.X., S.W., Y.H., R.F.K., G.X.)
| |
Collapse
|
21
|
Strahle JM, Mahaney KB, Morales DM, Buddhala C, Shannon CN, Wellons JC, Kulkarni AV, Jensen H, Reeder RW, Holubkov R, Riva-Cambrin JK, Whitehead WE, Rozzelle CJ, Tamber M, Pollack IF, Naftel RP, Kestle JRW, Limbrick DD. Longitudinal CSF Iron Pathway Proteins in Posthemorrhagic Hydrocephalus: Associations with Ventricle Size and Neurodevelopmental Outcomes. Ann Neurol 2021; 90:217-226. [PMID: 34080727 DOI: 10.1002/ana.26133] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/27/2021] [Accepted: 05/15/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Iron has been implicated in the pathogenesis of brain injury and hydrocephalus after preterm germinal matrix hemorrhage-intraventricular hemorrhage, however, it is unknown how external or endogenous intraventricular clearance of iron pathway proteins affect the outcome in this group. METHODS This prospective multicenter cohort included patients with posthemorrhagic hydrocephalus (PHH) who underwent (1) temporary and permanent cerebrospinal fluid (CSF) diversion and (2) Bayley Scales of Infant Development-III testing around 2 years of age. CSF proteins in the iron handling pathway were analyzed longitudinally and compared to ventricle size and neurodevelopmental outcomes. RESULTS Thirty-seven patients met inclusion criteria with a median estimated gestational age at birth of 25 weeks; 65% were boys. Ventricular CSF levels of hemoglobin, iron, total bilirubin, and ferritin decreased between temporary and permanent CSF diversion with no change in CSF levels of ceruloplasmin, transferrin, haptoglobin, and hepcidin. There was an increase in CSF hemopexin during this interval. Larger ventricle size at permanent CSF diversion was associated with elevated CSF ferritin (p = 0.015) and decreased CSF hemopexin (p = 0.007). CSF levels of proteins at temporary CSF diversion were not associated with outcome, however, higher CSF transferrin at permanent CSF diversion was associated with improved cognitive outcome (p = 0.015). Importantly, longitudinal change in CSF iron pathway proteins, ferritin (decrease), and transferrin (increase) were associated with improved cognitive (p = 0.04) and motor (p = 0.03) scores and improved cognitive (p = 0.04), language (p = 0.035), and motor (p = 0.008) scores, respectively. INTERPRETATION Longitudinal changes in CSF transferrin (increase) and ferritin (decrease) are associated with improved neurodevelopmental outcomes in neonatal PHH, with implications for understanding the pathogenesis of poor outcomes in PHH. ANN NEUROL 2021;90:217-226.
Collapse
Affiliation(s)
- Jennifer M Strahle
- Department of Neurosurgery, Washington University St. Louis, St. Louis, MO, USA
| | - Kelly B Mahaney
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Diego M Morales
- Department of Neurosurgery, Washington University St. Louis, St. Louis, MO, USA
| | - Chandana Buddhala
- Department of Neurosurgery, Washington University St. Louis, St. Louis, MO, USA
| | - Chevis N Shannon
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John C Wellons
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Abhaya V Kulkarni
- Department of Neurosurgery, University of Toronto, Toronto, Ontario, Canada
| | - Hailey Jensen
- Data Coordinating Center, University of Utah, Salt Lake City, UT, USA
| | - Ron W Reeder
- Data Coordinating Center, University of Utah, Salt Lake City, UT, USA
| | - Richard Holubkov
- Data Coordinating Center, University of Utah, Salt Lake City, UT, USA
| | - Jay K Riva-Cambrin
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | | | - Curtis J Rozzelle
- Department of Neurosurgery, University of Alabama - Birmingham, Birmingham, AL, USA
| | - Mandeep Tamber
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ian F Pollack
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Robert P Naftel
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John R W Kestle
- Department of Neurosurgery, University of Utah, Salt Lake City, UT, USA
| | - David D Limbrick
- Department of Neurosurgery, Washington University St. Louis, St. Louis, MO, USA
| | | |
Collapse
|
22
|
Stokum JA, Cannarsa GJ, Wessell AP, Shea P, Wenger N, Simard JM. When the Blood Hits Your Brain: The Neurotoxicity of Extravasated Blood. Int J Mol Sci 2021; 22:5132. [PMID: 34066240 PMCID: PMC8151992 DOI: 10.3390/ijms22105132] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 12/15/2022] Open
Abstract
Hemorrhage in the central nervous system (CNS), including intracerebral hemorrhage (ICH), intraventricular hemorrhage (IVH), and aneurysmal subarachnoid hemorrhage (aSAH), remains highly morbid. Trials of medical management for these conditions over recent decades have been largely unsuccessful in improving outcome and reducing mortality. Beyond its role in creating mass effect, the presence of extravasated blood in patients with CNS hemorrhage is generally overlooked. Since trials of surgical intervention to remove CNS hemorrhage have been generally unsuccessful, the potent neurotoxicity of blood is generally viewed as a basic scientific curiosity rather than a clinically meaningful factor. In this review, we evaluate the direct role of blood as a neurotoxin and its subsequent clinical relevance. We first describe the molecular mechanisms of blood neurotoxicity. We then evaluate the clinical literature that directly relates to the evacuation of CNS hemorrhage. We posit that the efficacy of clot removal is a critical factor in outcome following surgical intervention. Future interventions for CNS hemorrhage should be guided by the principle that blood is exquisitely toxic to the brain.
Collapse
Affiliation(s)
- Jesse A. Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
| | - Gregory J. Cannarsa
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
| | - Aaron P. Wessell
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
| | - Phelan Shea
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
| | - Nicole Wenger
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (G.J.C.); (A.P.W.); (P.S.); (N.W.); (J.M.S.)
- Departments of Pathology and Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
23
|
Goulding DS, Vogel RC, Gensel JC, Morganti JM, Stromberg AJ, Miller BA. Acute brain inflammation, white matter oxidative stress, and myelin deficiency in a model of neonatal intraventricular hemorrhage. J Neurosurg Pediatr 2020; 26:613-623. [PMID: 32858507 PMCID: PMC10193502 DOI: 10.3171/2020.5.peds20124] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/18/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Neonatal intraventricular hemorrhage (IVH) leads to posthemorrhagic hydrocephalus (PHH), brain injury, and long-term disability. Current therapy for IVH is based on treating PHH but does not address the underlying brain injury. In order to develop pharmacological treatment for IVH, there must be a better understanding of the underlying pathology of this disease. This study was designed to determine the time course of the acute inflammation and oxidative stress that may underlie the progressive pathology of IVH. The authors sought to understand the temporal relationships among inflammation, oxidative stress, and white matter pathology in a rat model of IVH. METHODS A rat model of IVH consisting of hemoglobin injection into the lateral ventricle was used. Tissue was analyzed via biochemical and histological methods to map the spatiotemporal distribution of innate immune activation and oxidative stress. White matter was quantified using both immunohistochemistry and Western blot for myelin basic protein (MBP) in the corpus callosum. RESULTS IVH led to acute induction of inflammatory cytokines, followed by oxidative stress. Oxidative stress was concentrated in white matter, adjacent to the lateral ventricles. Animals with IVH initially gained weight at a lower rate than control animals and had larger ventricles and less MBP than control animals. CONCLUSIONS Experimental IVH induces global inflammation throughout the brain and oxidative stress concentrated in the white matter. Both of these phenomena occur early after IVH. This has implications for human neonates with immature white matter that is exquisitely sensitive to inflammation and oxidative stress. Antiinflammatory or antioxidant therapy for IVH may need to be initiated early in order to protect developing white matter.
Collapse
Affiliation(s)
- Danielle S. Goulding
- Departments of Neurosurgery
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| | - R. Caleb Vogel
- Departments of Neurosurgery
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| | - John C. Gensel
- Physiology
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| | - Josh M. Morganti
- Neuroscience, and
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| | | | - Brandon A. Miller
- Departments of Neurosurgery
- Neuroscience, and
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| |
Collapse
|
24
|
Volpe J. Commentary – Severe IVH: Time for newer, earlier interventions to prevent brain injury? J Neonatal Perinatal Med 2020; 13:435-439. [PMID: 32925116 PMCID: PMC7836057 DOI: 10.3233/npm-200539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- J.J. Volpe
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Newborn Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Germinal Matrix-Intraventricular Hemorrhage of the Preterm Newborn and Preclinical Models: Inflammatory Considerations. Int J Mol Sci 2020; 21:ijms21218343. [PMID: 33172205 PMCID: PMC7664434 DOI: 10.3390/ijms21218343] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022] Open
Abstract
The germinal matrix-intraventricular hemorrhage (GM-IVH) is one of the most important complications of the preterm newborn. Since these children are born at a critical time in brain development, they can develop short and long term neurological, sensory, cognitive and motor disabilities depending on the severity of the GM-IVH. In addition, hemorrhage triggers a microglia-mediated inflammatory response that damages the tissue adjacent to the injury. Nevertheless, a neuroprotective and neuroreparative role of the microglia has also been described, suggesting that neonatal microglia may have unique functions. While the implication of the inflammatory process in GM-IVH is well established, the difficulty to access a very delicate population has lead to the development of animal models that resemble the pathological features of GM-IVH. Genetically modified models and lesions induced by local administration of glycerol, collagenase or blood have been used to study associated inflammatory mechanisms as well as therapeutic targets. In the present study we review the GM-IVH complications, with special interest in inflammatory response and the role of microglia, both in patients and animal models, and we analyze specific proteins and cytokines that are currently under study as feasible predictors of GM-IVH evolution and prognosis.
Collapse
|
26
|
El-Dib M, Limbrick DD, Inder T, Whitelaw A, Kulkarni AV, Warf B, Volpe JJ, de Vries LS. Management of Post-hemorrhagic Ventricular Dilatation in the Infant Born Preterm. J Pediatr 2020; 226:16-27.e3. [PMID: 32739263 PMCID: PMC8297821 DOI: 10.1016/j.jpeds.2020.07.079] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Mohamed El-Dib
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| | - David D Limbrick
- Department of Neurological Surgery, St Louis Children's Hospital, Washington University School of Medicine, St Louis, MO
| | - Terrie Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Andrew Whitelaw
- Neonatal Neuroscience, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Abhaya V Kulkarni
- Department of Neurosurgery, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Warf
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Joseph J Volpe
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Linda S de Vries
- Department of Neonatology, University Medical Center Utrecht, the Netherlands; University Medical Center Utrecht, Utrecht Brain Center, the Netherlands
| |
Collapse
|
27
|
Mechanical injury and blood are drivers of spatial memory deficits after rapid intraventricular hemorrhage. Neurobiol Dis 2020; 145:105084. [PMID: 32941979 DOI: 10.1016/j.nbd.2020.105084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 08/23/2020] [Accepted: 09/09/2020] [Indexed: 12/27/2022] Open
Abstract
Aneurysmal intraventricular hemorrhage (IVH) survivors may recover with significant deficits in learning and memory. The goal of this study was to investigate the mechanism of memory decline after intraventricular aneurysm rupture. We developed an aneurysmal IVH rat model by injecting autologous, arterial blood over the period of two minutes into the right lateral ventricle. We also evaluated the effects of a volume-matched artificial cerebrospinal fluid (CSF) control, thrombin and the mode of delivery (pulsed hand injection versus continuous pump infusion). We performed magnetic resonance brain imaging after 1 and 5 weeks to evaluate for hydrocephalus and histological analysis of the dentate gyrus after 6 weeks. Only animals which underwent a whole blood pulsed hand injection had a spatial memory acquisition and retention deficit 5 weeks later. These animals had larger ventricles at 1 and 5 weeks than animals which underwent a continuous pump infusion of whole blood. We did not find a decline in dentate gyrus granule cell neurons or an impairment in dentate gyrus neurogenesis or differentiation 6 weeks after IVH. Rapid injections of blood or volume resulted in microglial activation in the dentate gyrus. In conclusion, our results point to mechanical injury as the predominant mechanism of memory decline after intraventricular aneurysmal rupture. However, volume-matched pulsed injections of artificial CSF did not create a spatial memory deficit at 5 weeks. Therefore, whole blood itself must play a role in the mechanism. Further research is required to evaluate whether the viscosity of blood causes additional mechanical disruption and hydrocephalus through a primary injury mechanism or whether the toxicity of blood causes a secondary injury mechanism that leads to the observed spatial memory deficit after 5 weeks.
Collapse
|
28
|
Romantsik O, Bruschettini M, Ley D. Intraventricular Hemorrhage and White Matter Injury in Preclinical and Clinical Studies. Neoreviews 2020; 20:e636-e652. [PMID: 31676738 DOI: 10.1542/neo.20-11-e636] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Germinal matrix-intraventricular hemorrhage (IVH) occurs in nearly half of infants born at less than 26 weeks' gestation. Up to 50% of survivors with IVH develop cerebral palsy, cognitive deficits, behavioral disorders, posthemorrhagic ventricular dilatation, or a combination of these sequelae. After the initial bleeding and the primary brain injury, inflammation and secondary brain injury might lead to periventricular leukomalacia or diffuse white matter injury. Potential factors that are involved include microglia and astrocyte activation, degradation of blood components with release of "toxic" products, infiltration of the brain by systemic immune cells, death of neuronal and glial cells, and arrest of preoligodendrocyte maturation. In addition, impairment of the blood-brain barrier may play a major role in the pathophysiology. A wide range of animal models has been used to explore causes and mechanisms leading to IVH-induced brain injury. Preclinical studies have identified potential targets for enhancing brain repair. However, little has been elucidated about the effectiveness of potential interventions in clinical studies. A systematic review of available preclinical and clinical studies might help identify research gaps and which types of interventions may be prioritized. Future trials should report clinically robust and long-term outcomes after IVH.
Collapse
Affiliation(s)
- Olga Romantsik
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| | - David Ley
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| |
Collapse
|
29
|
Gilard V, Tebani A, Bekri S, Marret S. Intraventricular Hemorrhage in Very Preterm Infants: A Comprehensive Review. J Clin Med 2020; 9:E2447. [PMID: 32751801 PMCID: PMC7465819 DOI: 10.3390/jcm9082447] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/20/2020] [Accepted: 07/25/2020] [Indexed: 11/30/2022] Open
Abstract
Germinal matrix-intraventricular-intraparenchymal hemorrhage (GMH-IVH-IPH) is a major complication of very preterm births before 32 weeks of gestation (WG). Despite progress in clinical management, its incidence remains high before 27 WG. In addition, severe complications may occur such as post-hemorrhagic hydrocephalus and/or periventricular intraparenchymal hemorrhage. IVH is strongly associated with subsequent neurodevelopmental disabilities. For this review, an automated literature search and a clustering approach were applied to allow efficient filtering as well as topic clusters identification. We used a programmatic literature search for research articles related to intraventricular hemorrhage in preterms that were published between January 1990 and February 2020. Two queries ((Intraventricular hemorrhage) AND (preterm)) were used in PubMed. This search resulted in 1093 articles. The data manual curation left 368 documents that formed 12 clusters. The presentation and discussion of the clusters provide a comprehensive overview of existing data on the pathogenesis, complications, neuroprotection and biomarkers of GMH-IVH-IPH in very preterm infants. Clinicians should consider that the GMH-IVH-IPH pathogenesis is mainly due to developmental immaturity of the germinal matrix and cerebral autoregulation impairment. New multiomics investigations of intraventricular hemorrhage could foster the development of predictive biomarkers for the benefit of very preterm newborns.
Collapse
Affiliation(s)
- Vianney Gilard
- Department of Pediatric Neurosurgery, Rouen University Hospital, 76000 Rouen, France;
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000 Rouen, France;
| | - Abdellah Tebani
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000 Rouen, France;
| | - Soumeya Bekri
- Department of Metabolic Biochemistry, Rouen University Hospital, 76000 Rouen, France;
- Normandie University, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France;
| | - Stéphane Marret
- Normandie University, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, France;
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, 76000 Rouen, France
| |
Collapse
|
30
|
Pan P, Xu L, Zhang H, Liu Y, Lu X, Chen G, Tang H, Wu J. A Review of Hematoma Components Clearance Mechanism After Subarachnoid Hemorrhage. Front Neurosci 2020; 14:685. [PMID: 32733194 PMCID: PMC7358443 DOI: 10.3389/fnins.2020.00685] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/04/2020] [Indexed: 12/18/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a complicated clinical syndrome, which is caused by several kinds of cerebrovascular disorders, with high morbidity, disability and mortality rate. In recent years, several studies have shown that early brain injury (EBI) is an important factor leading to the poor prognosis of SAH. A major cause of EBI has been attributed that hematoma components invade into the brain parenchyma, resulting in neuronal cell death. Therefore, the clearance of hematoma components is essential in the clinical outcome of patients after SAH. Here, in the review, we provide a summary of the current known hematoma components clearance mechanisms and simultaneously propose a new hypothesis for hematoma components clearance.
Collapse
Affiliation(s)
- Pengjie Pan
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Xu
- Intensive Care Unit of Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongrong Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuan Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaocheng Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hailiang Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
31
|
Mahaney KB, Buddhala C, Paturu M, Morales D, Limbrick DD, Strahle JM. Intraventricular Hemorrhage Clearance in Human Neonatal Cerebrospinal Fluid: Associations With Hydrocephalus. Stroke 2020; 51:1712-1719. [PMID: 32397930 DOI: 10.1161/strokeaha.119.028744] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Preterm neonates with intraventricular hemorrhage (IVH) are at risk for posthemorrhagic hydrocephalus and poor neurological outcomes. Iron has been implicated in ventriculomegaly, hippocampal injury, and poor outcomes following IVH. We hypothesized that levels of cerebrospinal fluid blood breakdown products and endogenous iron clearance proteins in neonates with IVH differ from those of neonates with IVH who subsequently develop posthemorrhagic hydrocephalus. Methods- Premature neonates with an estimated gestational age at birth <30 weeks who underwent lumbar puncture for clinical evaluation an average of 2 weeks after birth were evaluated. Groups consisted of controls (n=16), low-grade IVH (grades I-II; n=4), high-grade IVH (grades III-IV; n=6), and posthemorrhagic hydrocephalus (n=9). Control subjects were preterm neonates born at <30 weeks' gestation without brain abnormality or hemorrhage on cranial ultrasound, who underwent lumbar puncture for clinical purposes. Cerebrospinal fluid hemoglobin, total bilirubin, total iron, ferritin, ceruloplasmin, transferrin, haptoglobin, and hemopexin were quantified. Results- Cerebrospinal fluid hemoglobin levels were increased in posthemorrhagic hydrocephalus compared with high-grade IVH (9.45 versus 6.06 µg/mL, P<0.05) and cerebrospinal fluid ferritin levels were increased in posthemorrhagic hydrocephalus compared with controls (511.33 versus 67.08, P<0.01). No significant group differences existed for the other cerebrospinal fluid blood breakdown and iron-handling proteins tested. We observed positive correlations between ventricular enlargement (frontal occipital horn ratio) and ferritin (Pearson r=0.67), hemoglobin (Pearson r=0.68), and total bilirubin (Pearson r=0.69). Conclusions- Neonates with posthemorrhagic hydrocephalus had significantly higher levels of hemoglobin than those with high-grade IVH. Levels of blood breakdown products, hemoglobin, ferritin, and bilirubin correlated with ventricular size. There was no elevation of several iron-scavenging proteins in cerebrospinal fluid in neonates with posthemorrhagic hydrocpehalus, indicative of posthemorrhagic hydrocephalus as a disease state occurring when endogenous iron clearance mechanisms are overwhelmed.
Collapse
Affiliation(s)
- Kelly B Mahaney
- Department of Neurosurgery, Stanford University, Stanford, CA (K.B.M.)
| | - Chandana Buddhala
- From the Department of Neurological Surgery, Washington University in St Louis, MO (C.B., M.P., D.M., D.D.L., J.M.S.)
| | - Mounica Paturu
- From the Department of Neurological Surgery, Washington University in St Louis, MO (C.B., M.P., D.M., D.D.L., J.M.S.)
| | - Diego Morales
- From the Department of Neurological Surgery, Washington University in St Louis, MO (C.B., M.P., D.M., D.D.L., J.M.S.)
| | - David D Limbrick
- From the Department of Neurological Surgery, Washington University in St Louis, MO (C.B., M.P., D.M., D.D.L., J.M.S.)
| | - Jennifer M Strahle
- From the Department of Neurological Surgery, Washington University in St Louis, MO (C.B., M.P., D.M., D.D.L., J.M.S.)
| |
Collapse
|
32
|
Goulding DS, Vogel RC, Pandya CD, Shula C, Gensel JC, Mangano FT, Goto J, Miller BA. Neonatal hydrocephalus leads to white matter neuroinflammation and injury in the corpus callosum of Ccdc39 hydrocephalic mice. J Neurosurg Pediatr 2020; 25:476-483. [PMID: 32032950 PMCID: PMC7415550 DOI: 10.3171/2019.12.peds19625] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/05/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The authors sought to determine if hydrocephalus caused a proinflammatory state within white matter as is seen in many other forms of neonatal brain injury. Common causes of hydrocephalus (such as trauma, infection, and hemorrhage) are inflammatory insults themselves and therefore confound understanding of how hydrocephalus itself affects neuroinflammation. Recently, a novel animal model of hydrocephalus due to a genetic mutation in the Ccdc39 gene has been developed in mice. In this model, ciliary dysfunction leads to early-onset ventriculomegaly, astrogliosis, and reduced myelination. Because this model of hydrocephalus is not caused by an antecedent proinflammatory insult, it was utilized to study the effect of hydrocephalus on inflammation within the white matter of the corpus callosum. METHODS A Meso Scale Discovery assay was used to measure levels of proinflammatory cytokines in whole brain from animals with and without hydrocephalus. Immunohistochemistry was used to measure macrophage activation and NG2 expression within the white matter of the corpus callosum in animals with and without hydrocephalus. RESULTS In this model of hydrocephalus, levels of cytokines throughout the brain revealed a more robust increase in classic proinflammatory cytokines (interleukin [IL]-1β, CXCL1) than in immunomodulatory cytokines (IL-10). Increased numbers of macrophages were found within the corpus callosum. These macrophages were polarized toward a proinflammatory phenotype as assessed by higher levels of CD86, a marker of proinflammatory macrophages, compared to CD206, a marker for antiinflammatory macrophages. There was extensive structural damage to the corpus callosum of animals with hydrocephalus, and an increase in NG2-positive cells. CONCLUSIONS Hydrocephalus without an antecedent proinflammatory insult induces inflammation and tissue injury in white matter. Future studies with this model will be useful to better understand the effects of hydrocephalus on neuroinflammation and progenitor cell development. Antiinflammatory therapy for diseases that cause hydrocephalus may be a powerful strategy to reduce tissue damage.
Collapse
Affiliation(s)
- Danielle S. Goulding
- Department of Neurosurgery, University of Kentucky,
Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of
Kentucky, Lexington, Kentucky
| | - R. Caleb Vogel
- Department of Neurosurgery, University of Kentucky,
Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of
Kentucky, Lexington, Kentucky
| | - Chirayu D. Pandya
- Department of Neurosurgery, University of Kentucky,
Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of
Kentucky, Lexington, Kentucky
| | - Crystal Shula
- Division of Pediatric Neurosurgery, Cincinnati
Children’s Hospital Medical Center, Cincinnati, Ohio
| | - John C. Gensel
- Spinal Cord and Brain Injury Research Center, University of
Kentucky, Lexington, Kentucky
- Department of Physiology, University of Kentucky,
Lexington, Kentucky
| | - Francesco T. Mangano
- Division of Pediatric Neurosurgery, Cincinnati
Children’s Hospital Medical Center, Cincinnati, Ohio
| | - June Goto
- Division of Pediatric Neurosurgery, Cincinnati
Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Brandon A. Miller
- Department of Neurosurgery, University of Kentucky,
Lexington, Kentucky
- Spinal Cord and Brain Injury Research Center, University of
Kentucky, Lexington, Kentucky
| |
Collapse
|
33
|
Formation and Detection of Highly Oxidized Hemoglobin Forms in Biological Fluids during Hemolytic Conditions. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8929020. [PMID: 32377310 PMCID: PMC7196973 DOI: 10.1155/2020/8929020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
Hemolytic diseases are characterized by an accelerated breakdown of red blood cells (RBCs) and the release of hemoglobin (Hb). Following, RBC lysis Hb oxidation occurs with the formation of different redox states of Hb (metHb and ferrylHb) and the release of heme. ferrylHb is unstable and decomposes to metHb with the concomitant formation of globin radicals and eventually covalently crosslinked Hb multimers. The goal of the present study was to determine the concentrations of the different redox states of Hb in biological samples during hemolytic conditions. We used plasma and urine samples of mice with intravascular hemolysis and human cerebrospinal fluid (CSF) samples following intraventricular hemorrhage. Because ferrylHb is highly unstable, we also addressed the fate of this species. metHb and free heme time-dependently accumulate in plasma and CSF samples following intravascular hemolysis and intraventricular hemorrhage, respectively. ferrylHb is hardly detectable in the biological samples during hemolytic conditions. Under in vitro conditions, ferrylHb decomposes quickly to metHb, which process is associated with the formation of covalently crosslinked Hb multimers. We detected these covalently crosslinked Hb multimers in plasma, urine, and CSF samples during hemolytic conditions. Because globin modification is specific for these Hb forms, we propose to call this heterogeneous form of Hb produced during ferrylHb decomposition as globin-modified oxidized Hb (gmoxHb). Understanding the formation and the contribution of gmoxHb species to the pathogenesis of hemolytic conditions could have therapeutic implications in the treatment of hemolytic diseases.
Collapse
|
34
|
Garland P, Morton MJ, Haskins W, Zolnourian A, Durnford A, Gaastra B, Toombs J, Heslegrave AJ, More J, Okemefuna AI, Teeling JL, Graversen JH, Zetterberg H, Moestrup SK, Bulters DO, Galea I. Haemoglobin causes neuronal damage in vivo which is preventable by haptoglobin. Brain Commun 2020; 2:fcz053. [PMID: 32346673 PMCID: PMC7188517 DOI: 10.1093/braincomms/fcz053] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After subarachnoid haemorrhage, prolonged exposure to toxic extracellular haemoglobin occurs in the brain. Here, we investigate the role of haemoglobin neurotoxicity in vivo and its prevention. In humans after subarachnoid haemorrhage, haemoglobin in cerebrospinal fluid was associated with neurofilament light chain, a marker of neuronal damage. Most haemoglobin was not complexed with haptoglobin, an endogenous haemoglobin scavenger present at very low concentration in the brain. Exogenously added haptoglobin bound most uncomplexed haemoglobin, in the first 2 weeks after human subarachnoid haemorrhage, indicating a wide therapeutic window. In mice, the behavioural, vascular, cellular and molecular changes seen after human subarachnoid haemorrhage were recapitulated by modelling a single aspect of subarachnoid haemorrhage: prolonged intrathecal exposure to haemoglobin. Haemoglobin-induced behavioural deficits and astrocytic, microglial and synaptic changes were attenuated by haptoglobin. Haptoglobin treatment did not attenuate large-vessel vasospasm, yet improved clinical outcome by restricting diffusion of haemoglobin into the parenchyma and reducing small-vessel vasospasm. In summary, haemoglobin toxicity is of clinical importance and preventable by haptoglobin, independent of large-vessel vasospasm.
Collapse
Affiliation(s)
- Patrick Garland
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Matthew J Morton
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - William Haskins
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Ardalan Zolnourian
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Andrew Durnford
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Ben Gaastra
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Jamie Toombs
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK.,Department of Neurodegenerative Disease, Institute of Neurology, London, WC1N 3BG, UK
| | - Amanda J Heslegrave
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK.,Department of Neurodegenerative Disease, Institute of Neurology, London, WC1N 3BG, UK
| | - John More
- Research & Development Department, Bio Products Laboratory Limited, Elstree, Hertfordshire, WD6 3BX, UK
| | - Azubuike I Okemefuna
- Research & Development Department, Bio Products Laboratory Limited, Elstree, Hertfordshire, WD6 3BX, UK
| | - Jessica L Teeling
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO16 6YD, UK
| | - Jonas H Graversen
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Henrik Zetterberg
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK.,Department of Neurodegenerative Disease, Institute of Neurology, London, WC1N 3BG, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mo¨ lndal, S-431 80, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mo¨ lndal, S-431 80, Sweden
| | - Soren K Moestrup
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark.,Department of Clinical Biochemistry, Aarhus University Hospital, 8200 Aarhus N, Denmark.,Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Diederik O Bulters
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK.,Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| |
Collapse
|
35
|
Intracerebral Hemorrhage: Blood Components and Neurotoxicity. Brain Sci 2019; 9:brainsci9110316. [PMID: 31717522 PMCID: PMC6896063 DOI: 10.3390/brainsci9110316] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/30/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke which is associated with the highest mortality and morbidity rates of all strokes. Although it is a major public health problem, there is no effective treatment for ICH. As a consequence of ICH, various blood components accumulate in the brain parenchyma and are responsible for much of the secondary brain damage and ICH-induced neurological deficits. Therefore, the strategies that could attenuate the blood component-induced neurotoxicity and improve hematoma resolution are highly needed. The present article provides an overview of blood-induced brain injury after ICH and emphasizes the need to conduct further studies elucidating the mechanisms of hematoma resolution after ICH.
Collapse
|
36
|
Thomas AJ, Ogilvy CS, Griessenauer CJ, Hanafy KA. Macrophage CD163 expression in cerebrospinal fluid: association with subarachnoid hemorrhage outcome. J Neurosurg 2019; 131:47-53. [PMID: 30028262 DOI: 10.3171/2018.2.jns172828] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/16/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Even though heme-induced cerebral inflammation contributes to many of the adverse sequelae seen in patients with subarachnoid hemorrhage (SAH), little is known about the mechanism; mouse models have shown a critical role for macrophages/microglia. Macrophage CD163 is a hemoglobin scavenger receptor involved in blood clearance after SAH. The authors hypothesized that the modified Fisher score is independently associated with cerebrospinal fluid (CSF) macrophage CD163 expression on postictal day 1, and that CSF macrophage CD163 expression is associated with 1-month neurological outcome. METHODS CSF macrophages from 21 SAH and 28 unruptured aneurysm patients (control) were analyzed for CD163 expression using flow cytometry and confocal microscopy on postictal day 1. Significant associations with modified Fisher scale grades or modified Rankin Scale scores were determined using linear regression and a matched case control analysis. RESULTS CSF macrophage CD163 expression was significantly increased in SAH patients compared with controls (p < 0.001). The modified Fisher scale (mF) grades (β = 0.407, p = 0.005) and CSF bilirubin concentrations (β = 0.311, p = 0.015) were positively and independently associated with CSF macrophage CD163 expression when the analysis was controlled for age and sex. CSF macrophages from an SAH patient with a high mF grade had increased co-localization of CD163 and glycophorin A (CD235a, an erythrocyte marker) compared with those from an SAH patient with a low mF grade. The controls had no co-localization. CSF macrophage CD163 expression (p = 0.003) was inversely associated with 1-month neurological outcome, when SAH patients were matched based on mF grade. CONCLUSIONS This early study suggests that CSF macrophage CD163 expression, as measured by flow cytometry, may have some neuroprotective function given its inverse association with outcome and provides unique insights into the neuroinflammatory process after SAH.
Collapse
Affiliation(s)
| | | | | | - Khalid A Hanafy
- 2Department of Neurology, and
- 3Division of Neurointensive Care Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
37
|
Romantsik O, Agyemang AA, Sveinsdóttir S, Rutardóttir S, Holmqvist B, Cinthio M, Mörgelin M, Gumus G, Karlsson H, Hansson SR, Åkerström B, Ley D, Gram M. The heme and radical scavenger α 1-microglobulin (A1M) confers early protection of the immature brain following preterm intraventricular hemorrhage. J Neuroinflammation 2019; 16:122. [PMID: 31174551 PMCID: PMC6554963 DOI: 10.1186/s12974-019-1486-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/25/2019] [Indexed: 12/31/2022] Open
Abstract
Background Germinal matrix intraventricular hemorrhage (GM-IVH) is associated with cerebro-cerebellar damage in very preterm infants, leading to neurodevelopmental impairment. Penetration, from the intraventricular space, of extravasated red blood cells and extracellular hemoglobin (Hb), to the periventricular parenchyma and the cerebellum has been shown to be causal in the development of brain injury following GM-IVH. Furthermore, the damage has been described to be associated with the cytotoxic nature of extracellular Hb-metabolites. To date, there is no therapy available to prevent infants from developing either hydrocephalus or serious neurological disability. Mechanisms previously described to cause brain damage following GM-IVH, i.e., oxidative stress and Hb-metabolite toxicity, suggest that the free radical and heme scavenger α1-microglobulin (A1M) may constitute a potential neuroprotective intervention. Methods Using a preterm rabbit pup model of IVH, where IVH was induced shortly after birth in pups delivered by cesarean section at E29 (3 days prior to term), we investigated the brain distribution of recombinant A1M (rA1M) following intracerebroventricular (i.c.v.) administration at 24 h post-IVH induction. Further, short-term functional protection of i.c.v.-administered human A1M (hA1M) following IVH in the preterm rabbit pup model was evaluated. Results Following i.c.v. administration, rA1M was distributed in periventricular white matter regions, throughout the fore- and midbrain and extending to the cerebellum. The regional distribution of rA1M was accompanied by a high co-existence of positive staining for extracellular Hb. Administration of i.c.v.-injected hA1M was associated with decreased structural tissue and mitochondrial damage and with reduced mRNA expression for proinflammatory and inflammatory signaling-related genes induced by IVH in periventricular brain tissue. Conclusions The results of this study indicate that rA1M/hA1M is a potential candidate for neuroprotective treatment following preterm IVH. Electronic supplementary material The online version of this article (10.1186/s12974-019-1486-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Olga Romantsik
- Pediatrics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | | | | | | | - Magnus Cinthio
- Department of Electrical Measurements, Lund University, Lund, Sweden
| | - Mattias Mörgelin
- Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Gulcin Gumus
- Fetal i+D Fetal Medicine Research Center, BCNatal Barcelona Center for Maternal-Fetal and Neonatal Medicine, University of Barcelona, Barcelona, Spain
| | | | - Stefan R Hansson
- Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Bo Åkerström
- Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - David Ley
- Pediatrics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Magnus Gram
- Pediatrics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden. .,Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden. .,A1M Pharma AB, Lund, Sweden.
| |
Collapse
|
38
|
Guo X, Qi X, Li H, Duan Z, Wei Y, Zhang F, Tian M, Ma L, You C. Deferoxamine Alleviates Iron Overload and Brain Injury in a Rat Model of Brainstem Hemorrhage. World Neurosurg 2019; 128:e895-e904. [PMID: 31082547 DOI: 10.1016/j.wneu.2019.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Brainstem hemorrhage (BSH) is the most dangerous and devastating subtype of intracerebral hemorrhage and is associated with high morbidity and mortality. However, to date, no effective prevention methods or specific therapies have been available to improve its clinical outcomes. We preliminarily explored the efficacy of deferoxamine (DFO), a clinical chelator known for its iron-scavenging activities, in a rat model of BSH induced with collagenase infusion. METHODS DFO or saline was administrated 6 hours after BSH induction and then every 12 hours for ≤7 days. The survival curve of the rats was created, and the neurological scores were examined on days 1, 3, and 7 after BSH. The rats were sacrificed after 1, 3, and 7 days of DFO treatment for histological examination and immunohistochemistry. RESULTS The results showed that administration of DFO delayed erythrocytes lysis, reduced iron deposition, reduced reactive oxygen species generation, reduced heme oxygenase-1 expression, and alleviated brain injury such as neuron degeneration and myelin sheath injury. However, DFO did not improve the survival rate and neurobehavioral outcomes in this model. CONCLUSIONS Administration of DFO had limited therapeutic effects on collagenase-induced brainstem hemorrhage in rats. Some potential explanations were proposed, and more preclinical work is required to clarify the controversial curative effect of DFO in ICH.
Collapse
Affiliation(s)
- Xi Guo
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Qi
- Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhongxin Duan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Wei
- Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fan Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Meng Tian
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China; West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Lu Ma
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China; West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
39
|
Wang Y, Wu Y, Li T, Wang X, Zhu C. Iron Metabolism and Brain Development in Premature Infants. Front Physiol 2019; 10:463. [PMID: 31105583 PMCID: PMC6494966 DOI: 10.3389/fphys.2019.00463] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
Iron is important for a remarkable array of essential functions during brain development, and it needs to be provided in adequate amounts, especially to preterm infants. In this review article, we provide an overview of iron metabolism and homeostasis at the cellular level, as well as its regulation at the mRNA translation level, and we emphasize the importance of iron for brain development in fetal and early life in preterm infants. We also review the risk factors for disrupted iron metabolism that lead to high risk of developing iron deficiency and subsequent adverse effects on neurodevelopment in preterm infants. At the other extreme, iron overload, which is usually caused by excess iron supplementation in iron-replete preterm infants, might negatively impact brain development or even induce brain injury. Maintaining the balance of iron during the fetal and neonatal periods is important, and thus iron status should be monitored routinely and evaluated thoroughly during the neonatal period or before discharge of preterm infants so that iron supplementation can be individualized.
Collapse
Affiliation(s)
- Yafeng Wang
- Department of Neonatology (NICU), Children’s Hospital Affiliated Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Clinical Neuroscience, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Yanan Wu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tao Li
- Department of Neonatology (NICU), Children’s Hospital Affiliated Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Clinical Neuroscience, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Physiology, Sahlgrenska Academy, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Clinical Neuroscience, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
40
|
Impaired hippocampal development and outcomes in very preterm infants with perinatal brain injury. NEUROIMAGE-CLINICAL 2019; 22:101787. [PMID: 30991622 PMCID: PMC6446074 DOI: 10.1016/j.nicl.2019.101787] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 03/12/2019] [Accepted: 03/16/2019] [Indexed: 12/16/2022]
Abstract
Preterm infants are at high risk for brain injury during the perinatal period. Intraventricular hemorrhage and periventricular leukomalacia, the two most common patterns of brain injury in prematurely-born children, are associated with poor neurodevelopmental outcomes. The hippocampus is known to be critical for learning and memory; however, it remains unknown how these forms of brain injury affect hippocampal growth and how the resulting alterations in hippocampal development relate to childhood outcomes. To investigate these relationships, hippocampal segmentations were performed on term equivalent MRI scans from 55 full-term infants, 85 very preterm infants (born ≤32 weeks gestation) with no to mild brain injury and 73 very preterm infants with brain injury (e.g., grade III/IV intraventricular hemorrhage, post-hemorrhagic hydrocephalus, cystic periventricular leukomalacia). Infants then underwent standardized neurodevelopmental testing using the Bayley Scales of Infant and Toddler Development, 3rd edition at age 2 years, corrected for prematurity. To delineate the effects of brain injury on early hippocampal development, hippocampal volumes were compared across groups and associations between neonatal volumes and neurodevelopmental outcomes at age 2 years were explored. Very preterm infants with brain injury had smaller hippocampal volumes at term equivalent age compared to term and very preterm infants with no to mild injury, with the smallest hippocampi among those with grade III/IV intraventricular hemorrhage and post-hemorrhagic hydrocephalus. Further, larger ventricle size was associated with smaller hippocampal size. Smaller hippocampal volumes were related to worse motor performance at age 2 years across all groups. In addition, smaller hippocampal volumes in infants with brain injury were correlated with impaired cognitive scores at age 2 years, a relationship specific to this group. Consistent with our preclinical findings, these findings demonstrate that perinatal brain injury is associated with hippocampal size in preterm infants, with smaller volumes related to domain-specific neurodevelopmental impairments in this high-risk clinical population. Perinatal brain injury is related to smaller hippocampal volumes in preterm infants Infants with high-grade intraventricular hemorrhage have smallest hippocampi Larger ventricular size is related to smaller hippocampal volumes in hydrocephalus Smaller hippocampi are related to worse cognitive outcomes in brain injured infants Smaller hippocampal volumes associated with worse motor performance across groups
Collapse
|
41
|
Nicaise C, Marneffe C, Bouchat J, Gilloteaux J. Osmotic Demyelination: From an Oligodendrocyte to an Astrocyte Perspective. Int J Mol Sci 2019; 20:E1124. [PMID: 30841618 PMCID: PMC6429405 DOI: 10.3390/ijms20051124] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 12/15/2022] Open
Abstract
Osmotic demyelination syndrome (ODS) is a disorder of the central myelin that is often associated with a precipitous rise of serum sodium. Remarkably, while the myelin and oligodendrocytes of specific brain areas degenerate during the disease, neighboring neurons and axons appear unspoiled, and neuroinflammation appears only once demyelination is well established. In addition to blood‒brain barrier breakdown and microglia activation, astrocyte death is among one of the earliest events during ODS pathology. This review will focus on various aspects of biochemical, molecular and cellular aspects of oligodendrocyte and astrocyte changes in ODS-susceptible brain regions, with an emphasis on the crosstalk between those two glial cells. Emerging evidence pointing to the initiating role of astrocytes in region-specific degeneration are discussed.
Collapse
Affiliation(s)
| | - Catherine Marneffe
- Laboratory of Glia Biology (VIB-KU Leuven Center for Brain & Disease Research), Department of Neuroscience, KU Leuven, 3000 Leuven, Belgium.
| | - Joanna Bouchat
- URPhyM-NARILIS, Université de Namur, 5000 Namur, Belgium.
| | - Jacques Gilloteaux
- URPhyM-NARILIS, Université de Namur, 5000 Namur, Belgium.
- Department of Anatomical Sciences, St George's University School of Medicine, Newcastle upon Tyne NE1 8ST, UK.
| |
Collapse
|
42
|
Huang J, Jiang Q. Dexmedetomidine Protects Against Neurological Dysfunction in a Mouse Intracerebral Hemorrhage Model by Inhibiting Mitochondrial Dysfunction-Derived Oxidative Stress. J Stroke Cerebrovasc Dis 2019; 28:1281-1289. [PMID: 30797643 DOI: 10.1016/j.jstrokecerebrovasdis.2019.01.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/09/2019] [Accepted: 01/19/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a subtype of stroke with high disability and mortality. Dexmedetomidine (Dex) has been shown to provide neuroprotection in several neurological diseases. The aim of present study was to investigate the effects of Dex on ICH-induced neurological deficits and brain injury and the underlying mechanisms. METHODS ICH mouse model was established by intracerebral injection of autologous blood, followed by Dex or vehicle treatment. Neurological function, brain water content, neuronal activity, and oxidative parameters were determined. The protein expressions of peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), uncoupling protein 2, and manganese-dependent superoxide dismutase were examined by western blotting. RESULTS Dex administration significantly inhibited ICH-induced the memory impairment, dyskinesia, brain edema, and neuron loss. In addition, ICH-induced the increase in brain oxidative stress level was markedly attenuated after Dex treatment, as evidenced by increased glutathione peroxidase and superoxide dismutase levels and reduced malondialdehyde and nitric oxide levels. Compared with vehicle-treated ICH mice, Dex-treated ICH mice showed significantly decreased intracellular reactive oxygen species (ROS) and mitochondrial ROS (mROS) production in brain, but had no effects on the increased nicotinamide-adenine dinucleotide phosphate oxidase activity. However, stimulation of mROS abrogated the inhibitory effects of Dex on neurological deficits and oxidative stress. The decrease in production of adenosine triphosphate and the expressions of PGC-1α, uncoupling protein 2, and manganese-dependent superoxide dismutase induced by ICH was restored by Dex treatment. CONCLUSIONS Our results reveal that Dex improves ICH-induced neurological deficits and brain injury by inhibiting PGC-1α pathway inactivation and mitochondrial dysfunction-derived oxidative stress.
Collapse
Affiliation(s)
- Jing Huang
- Department of Anesthesiology, Changzhou No.2 People's Hospital, The Affiliated Hospital to Nanjing Medical University, Changzhou, Jiangsu, China
| | - Qiang Jiang
- Department of Anesthesiology, Changzhou No.2 People's Hospital, The Affiliated Hospital to Nanjing Medical University, Changzhou, Jiangsu, China.
| |
Collapse
|
43
|
Liu C, Liang MC, Soong TW. Nitric Oxide, Iron and Neurodegeneration. Front Neurosci 2019; 13:114. [PMID: 30833886 PMCID: PMC6388708 DOI: 10.3389/fnins.2019.00114] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/30/2019] [Indexed: 12/25/2022] Open
Abstract
Iron is a crucial cofactor for several physiological functions in the brain including transport of oxygen, DNA synthesis, mitochondrial respiration, synthesis of myelin, and neurotransmitter metabolism. If iron concentration exceeds the capacity of cellular sequestration, excessive labile iron will be harmful by generating oxidative stress that leads to cell death. In patients suffering from Parkinson disease, the total amount of iron in the substantia nigra was reported to increase with disease severity. High concentrations of iron were also found in the amyloid plaques and neurofibrillary tangles of human Alzheimer disease brains. Besides iron, nitric oxide (NO) produced in high concentration has been associated with neurodegeneration. NO is produced as a co-product when the enzyme NO synthase converts L-arginine to citrulline, and NO has a role to support normal physiological functions. When NO is produced in a high concentration under pathological conditions such as inflammation, aberrantly S-nitrosylated proteins can initiate neurodegeneration. Interestingly, NO is closely related with iron homeostasis. Firstly, it regulates iron-related gene expression through a system involving iron regulatory protein and its cognate iron responsive element (IRP-IRE). Secondly, it modified the function of iron-related protein directly via S-nitrosylation. In this review, we examine the recent advances about the potential role of dysregulated iron homeostasis in neurodegeneration, with an emphasis on AD and PD, and we discuss iron chelation as a potential therapy. This review also highlights the changes in iron homeostasis caused by NO. An understanding of these mechanisms will help us formulate strategies to reverse or ameliorate iron-related neurodegeneration in diseases such as AD and PD.
Collapse
Affiliation(s)
- Chao Liu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- Neurobiology/Ageing Program, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - Mui Cheng Liang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology/Ageing Program, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology/Ageing Program, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- National Neuroscience Institute, Singapore, Singapore
| |
Collapse
|
44
|
Leijser LM, de Vries LS. Preterm brain injury: Germinal matrix-intraventricular hemorrhage and post-hemorrhagic ventricular dilatation. HANDBOOK OF CLINICAL NEUROLOGY 2019; 162:173-199. [PMID: 31324310 DOI: 10.1016/b978-0-444-64029-1.00008-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Germinal matrix hemorrhage and intraventricular hemorrhages (GMH-IVH) remain a common and clinically significant problem in preterm infants, particularly extremely preterm infants. A large GMH-IVH is often complicated by posthemorrhagic ventricular dilation (PHVD) or parenchymal hemorrhagic infarction and is associated with an increased risk of adverse neurologic sequelae. The widespread use of cranial ultrasonography since the early 1980s has shown a gradual decrease in the incidence of GMH-IVH and has helped with the identification of antenatal and perinatal risk factors and timing of the lesion. The increased use of magnetic resonance imaging (MRI) has contributed to more detailed visualization of the site and extent of the GMH-IVH. In addition, MRI has contributed to the awareness of associated white matter changes as well as associated cerebellar hemorrhages. Although GMH-IVH and PHVD still cannot be prevented, cerebrospinal fluid drainage initiated in the early stage of PHVD development seems to be associated with a better neurodevelopmental outcome. Further studies are underway to improve treatment strategies for PHVD and to potentially prevent and repair GMH-IVH and PHVD and associated brain injury. This chapter discusses the pathogenesis, incidence, risk factors, and management, including preventive measures, of GHM-IVH and PHVD.
Collapse
Affiliation(s)
- Lara M Leijser
- Department of Pediatrics, Section of Neonatology, University of Calgary, Cumming School of Medicine, Calgary, Canada
| | - Linda S de Vries
- Department of Neonatology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
45
|
Ko HR, Ahn SY, Chang YS, Hwang I, Yun T, Sung DK, Sung SI, Park WS, Ahn JY. Human UCB-MSCs treatment upon intraventricular hemorrhage contributes to attenuate hippocampal neuron loss and circuit damage through BDNF-CREB signaling. Stem Cell Res Ther 2018; 9:326. [PMID: 30463591 PMCID: PMC6249960 DOI: 10.1186/s13287-018-1052-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/02/2018] [Accepted: 10/17/2018] [Indexed: 12/15/2022] Open
Abstract
Background Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) have been shown to prevent brain damage and improve neurocognition following intraventricular hemorrhage (IVH). However, the molecular mechanisms underlying the effects of hUCB-MSCs are still elusive. Thus, as the hippocampus is essential for learning, memory, and cognitive functions and is intimately involved in the ventricular system, making it a potential site of IVH-induced injury, we determined the molecular basis of the effects of hUCB-derived MSCs on hippocampal neurogenesis and the recovery of hippocampal neural circuits after IVH in a rodent model. Methods We inflicted severe IVH injury on postnatal day 4 (P4) in rats. After confirmation of successful induction of IVH using MRI (P5), intracerebroventricular administration of MSCs (ICV-MSC) was performed at 2 days post-injury (P6). For hippocampal synaptic determination, a rat entorhinal-hippocampus (EH) organotypic slice co-culture (OSC) was performed using day 3 post-IVH brains (P7) with or without ICV-MSCs. A similar strategy of experiments was applied to those rats receiving hUCB-MSC transfected with BDNF-Si-RNA for knockdown of BDNF or scrambled siRNA controls after IVH. The molecular mechanism of the MSCs effects on neurogenesis and the attenuation of neuron death was determined by evaluation of BDNF-TrkB-Akt-CREB signaling axis. Results We showed that treatment with hUCB-MSCs attenuated neuronal loss and promoted neurogenesis in the hippocampus, an area highly vulnerable to IVH-induced brain injury. hUCB-MSCs activate BDNF-TrkB receptor signaling, eliciting intracellular activation of Akt and/or Erk and subsequent phosphorylation of CREB, which is responsible for promoting rat BDNF transcription. In addition to the beneficial effects of neuroprotection and neurogenesis, hUCB-MSCs also contribute to the restoration of impaired synaptic circuits in the hippocampus and improve neurocognitive functions in IVH-injured neonatal rat through BDNF-TrkB-CREB signaling axis activation. Conclusions Our data suggest that hUCB-MSCs possess therapeutic potential for treating neuronal loss and neurocognitive dysfunction in IVH through the activation of intracellular TrkB-CREB signaling that is invoked by hUCB-MSC-secreted BDNF. Electronic supplementary material The online version of this article (10.1186/s13287-018-1052-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyo Rim Ko
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea
| | - Inwoo Hwang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Taegwan Yun
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Dong Kyung Sung
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea. .,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea.
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea. .,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea. .,Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea. .,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, 06351, South Korea.
| |
Collapse
|
46
|
Zhang H, Zhabyeyev P, Wang S, Oudit GY. Role of iron metabolism in heart failure: From iron deficiency to iron overload. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1925-1937. [PMID: 31109456 DOI: 10.1016/j.bbadis.2018.08.030] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/25/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022]
Abstract
Iron metabolism is a balancing act, and biological systems have evolved exquisite regulatory mechanisms to maintain iron homeostasis. Iron metabolism disorders are widespread health problems on a global scale and range from iron deficiency to iron-overload. Both types of iron disorders are linked to heart failure. Iron play a fundamental role in mitochondrial function and various enzyme functions and iron deficiency has a particular negative impact on mitochondria function. Given the high-energy demand of the heart, iron deficiency has a particularly negative impact on heart function and exacerbates heart failure. Iron-overload can result from excessive gut absorption of iron or frequent use of blood transfusions and is typically seen in patients with congenital anemias, sickle cell anemia and beta-thalassemia major, or in patients with primary hemochromatosis. This review provides an overview of normal iron metabolism, mechanisms underlying development of iron disorders in relation to heart failure, including iron-overload cardiomyopathy, and clinical perspective on the treatment options for iron metabolism disorders.
Collapse
Affiliation(s)
- Hao Zhang
- Division of Cardiology, Department of Medicine, Canada; Mazankowski Alberta Heart Institute, Canada
| | - Pavel Zhabyeyev
- Division of Cardiology, Department of Medicine, Canada; Mazankowski Alberta Heart Institute, Canada
| | - Shaohua Wang
- Mazankowski Alberta Heart Institute, Canada; Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Canada; Mazankowski Alberta Heart Institute, Canada.
| |
Collapse
|
47
|
Koschnitzky JE, Keep RF, Limbrick DD, McAllister JP, Morris JA, Strahle J, Yung YC. Opportunities in posthemorrhagic hydrocephalus research: outcomes of the Hydrocephalus Association Posthemorrhagic Hydrocephalus Workshop. Fluids Barriers CNS 2018; 15:11. [PMID: 29587767 PMCID: PMC5870202 DOI: 10.1186/s12987-018-0096-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/09/2018] [Indexed: 12/19/2022] Open
Abstract
The Hydrocephalus Association Posthemorrhagic Hydrocephalus Workshop was held on July 25 and 26, 2016 at the National Institutes of Health. The workshop brought together a diverse group of researchers including pediatric neurosurgeons, neurologists, and neuropsychologists with scientists in the fields of brain injury and development, cerebrospinal and interstitial fluid dynamics, and the blood-brain and blood-CSF barriers. The goals of the workshop were to identify areas of opportunity in posthemorrhagic hydrocephalus research and encourage scientific collaboration across a diverse set of fields. This report details the major themes discussed during the workshop and research opportunities identified for posthemorrhagic hydrocephalus. The primary areas include (1) preventing intraventricular hemorrhage, (2) stopping primary and secondary brain damage, (3) preventing hydrocephalus, (4) repairing brain damage, and (5) improving neurodevelopment outcomes in posthemorrhagic hydrocephalus.
Collapse
Affiliation(s)
| | - Richard F. Keep
- University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109 USA
| | - David D. Limbrick
- Washington University in St. Louis School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110 USA
| | - James P. McAllister
- Washington University in St. Louis School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110 USA
| | - Jill A. Morris
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Neuroscience Center, 6001 Executive Blvd, NSC Rm 2112, Bethesda, MD 20892 USA
| | - Jennifer Strahle
- Washington University in St. Louis School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110 USA
| | - Yun C. Yung
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Rd., Building 7, La Jolla, CA 92037 USA
| |
Collapse
|
48
|
Wang G, Wang L, Sun XG, Tang J. Haematoma scavenging in intracerebral haemorrhage: from mechanisms to the clinic. J Cell Mol Med 2017; 22:768-777. [PMID: 29278306 PMCID: PMC5783832 DOI: 10.1111/jcmm.13441] [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] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 09/14/2017] [Indexed: 01/22/2023] Open
Abstract
The products of erythrocyte lyses, haemoglobin (Hb) and haem, are recognized as neurotoxins and the main contributors to delayed cerebral oedema and tissue damage after intracerebral haemorrhage (ICH). Finding a means to efficiently promote absorption of the haemolytic products (Hb and haem) around the bleeding area in the brain through stimulating the function of the body's own garbage cleaning system is a novel clinical challenge and critical for functional recovery after ICH. In this review, available information of the brain injury mechanisms underlying ICH and endogenous haematoma scavenging system is provided. Meanwhile, potential intervention strategies are discussed. Intracerebral blood itself has ‘toxic’ effects beyond its volume effect after ICH. Haptoglobin–Hb–CD163 as well as haemopexin–haem–LRP1 is believed to be the most important endogenous scavenging pathway which participates in blood components resolution following ICH. PPARγ–Nrf2 activates the aforementioned clearance pathway and then accelerates haematoma clearance. Meanwhile, the scavenger receptors as novel targets for therapeutic interventions to treat ICH are also highlighted.
Collapse
Affiliation(s)
- Gaiqing Wang
- The second Hospital of Shanxi Medical University, Tai Yuan, China
| | - Li Wang
- The second Hospital of Shanxi Medical University, Tai Yuan, China
| | - Xin-Gang Sun
- The second Hospital of Shanxi Medical University, Tai Yuan, China
| | - Jiping Tang
- Department of Physiology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
49
|
Garton T, Hua Y, Xiang J, Xi G, Keep RF. Challenges for intraventricular hemorrhage research and emerging therapeutic targets. Expert Opin Ther Targets 2017; 21:1111-1122. [PMID: 29067856 PMCID: PMC6097191 DOI: 10.1080/14728222.2017.1397628] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Intraventricular hemorrhage (IVH) affects both premature infants and adults. In both demographics, it has high mortality and morbidity. There is no FDA approved therapy that improves neurological outcome in either population highlighting the need for additional focus on therapeutic targets and treatments emerging from preclinical studies. Areas covered: IVH induces both initial injury linked to the physical effects of the blood (mass effect) and secondary injury linked to the brain response to the hemorrhage. Preclinical studies have identified multiple secondary injury mechanisms following IVH, and particularly the role of blood components (e.g. hemoglobin, iron, thrombin). This review, with an emphasis on pre-clinical IVH research, highlights therapeutic targets and treatments that may be of use in prevention, acute care, or repair of damage. Expert opinion: An IVH is a potentially devastating event. Progress has been made in elucidating injury mechanisms, but this has still to translate to the clinic. Some pathways involved in injury also have beneficial effects (coagulation cascade/inflammation). A greater understanding of the downstream pathways involved in those pathways may allow therapeutic development. Iron chelation (deferoxamine) is in clinical trial for intracerebral hemorrhage and preclinical data suggest it may be a potential treatment for IVH.
Collapse
Affiliation(s)
- Thomas Garton
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Ya Hua
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Jianming Xiang
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Guohua Xi
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| | - Richard F Keep
- a Department of Neurosurgery , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
50
|
Haptoglobin Genotype and Outcome after Subarachnoid Haemorrhage: New Insights from a Meta-Analysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6747940. [PMID: 29104730 PMCID: PMC5634574 DOI: 10.1155/2017/6747940] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/10/2017] [Accepted: 07/25/2017] [Indexed: 01/01/2023]
Abstract
Haptoglobin (Hp) is a plasma protein involved in clearing extracellular haemoglobin and regulating inflammation; it exists as two genetic variants (Hp1 and Hp2). In a meta-analysis of six published studies, we confirm that Hp genotype affects short-term outcome (cerebral vasospasm and/or delayed cerebral ischemia) after subarachnoid haemorrhage (SAH) but not long-term outcome (Glasgow Outcome Score and modified Rankin Scale between one and three months). A closer examination of the heterozygous group revealed that the short-term outcome of Hp2-1 individuals clustered with that of Hp1-1 and not Hp2-2, suggesting that the presence of one Hp1 allele was sufficient to confer protection. Since the presence of the Hp dimer is the only common feature between Hp1-1 and Hp2-1 individuals, the absence of this Hp moiety is most likely to underlie vasospasm in Hp2-2 individuals. These results have implications for prognosis after SAH and will inform further research into Hp-based mechanism of action and treatment.
Collapse
|