1
|
Fox Á, Doyle E, Reynolds A, Geary M, Cuzzilla R, Hayes B. Placental histology for infants with hypoxic ischaemic encephalopathy compared with healthy controls: a case-control study. BMC Pediatr 2025; 25:294. [PMID: 40229702 PMCID: PMC11995492 DOI: 10.1186/s12887-025-05652-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND The role of the placenta in the development of hypoxic ischaemic encephalopathy (HIE) remains undefined. There is limited research comparing placental histology for infants with HIE and healthy controls. This is limiting our ability to understand its role in HIE. This study hypothesised that placental pathology is more common in infants with HIE compared with healthy infants and aimed to report the differences in placental histology between infants with HIE and healthy controls. METHODS A case-control study of infants with moderate andsevere HIE and healthy controls at a single tertiary Neonatal Intensive Care Unit. Placental histology was reviewed by one perinatal histopathologist using consensus guidelines. RESULTS Seventy-four cases and 98 controls were included. Cases had a higher incidence of pathology, including fetal vascular malperfusion, histological chorioamnionitis and delayed villous maturation. CONCLUSION This study demonstrates a higher incidence of placental pathology for infants born with HIE suggesting that the placenta is an important factor in the pathogenesis of HIE. Further research is required to delineate this relationship.
Collapse
Affiliation(s)
- Áine Fox
- Royal College of Surgeons (RCSI), Dublin, Ireland.
- Department of Neonatology, Rotunda Hospital, Dublin, Ireland.
| | - Emma Doyle
- Department of Pathology, Rotunda Hospital, Dublin, Ireland
| | | | - Michael Geary
- Royal College of Surgeons (RCSI), Dublin, Ireland
- Department of Obstetrics, Rotunda Hospital, Dublin, Ireland
| | - Rocco Cuzzilla
- Royal College of Surgeons (RCSI), Dublin, Ireland
- Newborn Research and Neonatal Services, The Royal Women's Hospital, Melbourne, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne, Melbourne, Australia
| | - Breda Hayes
- Royal College of Surgeons (RCSI), Dublin, Ireland
- Department of Neonatology, Rotunda Hospital, Dublin, Ireland
| |
Collapse
|
2
|
Nunes RR, Durán-Carabali LE, Ribeiro NH, Sirena DH, Tassinari ID, Netto CA, Paz AH, de Fraga LS. Impact of peripheral immune cells in experimental neonatal hypoxia-ischemia: A systematic review and meta-analysis. Int Immunopharmacol 2025; 145:113682. [PMID: 39637576 DOI: 10.1016/j.intimp.2024.113682] [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: 08/30/2024] [Revised: 11/10/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
Infiltration of peripheral immune cells into the brain following neonatal hypoxia-ischemia (HI) contributes to increased neuroinflammation and brain injury. However, the specific roles of different immune cell types in neonatal brain injury remain poorly understood. Although existing evidence suggests a potential role for sexual dimorphism in HI outcomes, this aspect has been insufficiently investigated. In this systematic review and meta-analysis, we examined the brain infiltration of peripheral immune cells in rodents of both sexes following neonatal HI. A total of 25 studies were included. Our analysis revealed significant increases in the infiltration of various subtypes of leukocytes after HI, along with increased brain injury, cell death, and neuroinflammation, and reduced neuronal survival. Notably, males exhibited a greater degree of immune cell infiltration and more pronounced neuroinflammation compared to females. These findings suggest that infiltrating leukocytes contribute significantly to the pathophysiology of neonatal HI, with sexually dimorphic responses further influencing the outcomes. It is crucial that future research focuses on elucidating the specific roles of immune cell subtypes to better understand the mechanisms underlying brain damage after HI and identify novel therapeutic targets. Moreover, the observed sex differences highlight the need to consider sex as a key factor when developing strategies for the treatment of neonatal HI.
Collapse
Affiliation(s)
- Ricardo Ribeiro Nunes
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Luz Elena Durán-Carabali
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Departamento de Ciências Fisiológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Nícolas Heller Ribeiro
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Dienifer Hermann Sirena
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Isadora D'Ávila Tassinari
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carlos Alexandre Netto
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Ana Helena Paz
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Luciano Stürmer de Fraga
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.
| |
Collapse
|
3
|
Dhillon SK, Gressens P, Barks J, Gunn AJ. Uncovering the Role of Inflammation with Asphyxia in the Newborn. Clin Perinatol 2024; 51:551-564. [PMID: 39095095 DOI: 10.1016/j.clp.2024.04.012] [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] [Indexed: 08/04/2024]
Abstract
The etiology of perinatal brain injury is multifactorial, but exposure to perinatal hypoxiaischemia (HI) is a major underlying factor. This review discusses the role of exposure to infection/inflammation in the evolution of HI brain injury, changes in immune responsiveness to subsequent inflammatory challenges after HI and modulation of neural outcomes with interaction between perinatal HI and inflammatory insults. The authors critically assess the clinical and preclinical evidence for the neuroprotective efficacy of therapeutic hypothermia and other anti-inflammatory treatments for inflammation-sensitized HI injury.
Collapse
Affiliation(s)
- Simerdeep K Dhillon
- Department of Physiology, The University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Pierre Gressens
- Université Paris Cité, NeuroDiderot, Inserm, F-75019 Paris, France
| | - John Barks
- Department of Pediatrics and Communicable Diseases, The University of Michigan, 2018 MLB, Ann Arbor, MI 48109, USA
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand.
| |
Collapse
|
4
|
Bernis ME, Hakvoort C, Nacarkucuk E, Burkard H, Bremer AS, Zweyer M, Maes E, Grzelak KA, Sabir H. Neuroprotective Effect of Clemastine Improved Oligodendrocyte Proliferation through the MAPK/ERK Pathway in a Neonatal Hypoxia Ischemia Rat Model. Int J Mol Sci 2024; 25:8204. [PMID: 39125778 PMCID: PMC11311837 DOI: 10.3390/ijms25158204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy is the most common cause of long-term disability in term neonates, and white matter injury is the primary cause of cerebral palsy. Therapies that focus on the neuroprotection of myelination and oligodendrocyte proliferation could potentially ameliorate long-lasting neurological impairments after hypoxic-ischemic encephalopathy. Clemastine, a histamine H1 antagonist, has been shown to exert neuroprotective effects in multiple sclerosis and spinal cord injury by promoting oligodendrogenesis and re-myelination. In this study, we demonstrated the neuroprotective effects of clemastine in our rat model of neonatal hypoxic-ischemic brain injury. Animals received a single intraperitoneal injection of either vehicle or clemastine (10 mg/kg) for 6 consecutive days. Our results showed a significant reduction in white matter loss after treatment, with a clear effect of clemastine on oligodendrocytes, showing a significant increase in the number of Olig2+ cells. We characterized the MAPK/ERK pathway as a potential mechanistic pathway underlying the neuroprotective effects of clemastine. Altogether, our results demonstrate that clemastine is a potential compound for the treatment of hypoxic-ischemic encephalopathy, with a clear neuroprotective effect on white matter injury by promoting oligodendrogenesis.
Collapse
Affiliation(s)
- Maria E. Bernis
- Neonatologie und Pädiatrische Intensivmedizin, Eltern-Kind-Zentrum, Universitätsklinikum Bonn, 53127 Bonn, Germany; (M.E.B.); (C.H.); (E.N.); (H.B.); (A.-S.B.); (M.Z.); (E.M.); (K.A.G.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Charlotte Hakvoort
- Neonatologie und Pädiatrische Intensivmedizin, Eltern-Kind-Zentrum, Universitätsklinikum Bonn, 53127 Bonn, Germany; (M.E.B.); (C.H.); (E.N.); (H.B.); (A.-S.B.); (M.Z.); (E.M.); (K.A.G.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Efe Nacarkucuk
- Neonatologie und Pädiatrische Intensivmedizin, Eltern-Kind-Zentrum, Universitätsklinikum Bonn, 53127 Bonn, Germany; (M.E.B.); (C.H.); (E.N.); (H.B.); (A.-S.B.); (M.Z.); (E.M.); (K.A.G.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Hannah Burkard
- Neonatologie und Pädiatrische Intensivmedizin, Eltern-Kind-Zentrum, Universitätsklinikum Bonn, 53127 Bonn, Germany; (M.E.B.); (C.H.); (E.N.); (H.B.); (A.-S.B.); (M.Z.); (E.M.); (K.A.G.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Anna-Sophie Bremer
- Neonatologie und Pädiatrische Intensivmedizin, Eltern-Kind-Zentrum, Universitätsklinikum Bonn, 53127 Bonn, Germany; (M.E.B.); (C.H.); (E.N.); (H.B.); (A.-S.B.); (M.Z.); (E.M.); (K.A.G.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Margit Zweyer
- Neonatologie und Pädiatrische Intensivmedizin, Eltern-Kind-Zentrum, Universitätsklinikum Bonn, 53127 Bonn, Germany; (M.E.B.); (C.H.); (E.N.); (H.B.); (A.-S.B.); (M.Z.); (E.M.); (K.A.G.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Elke Maes
- Neonatologie und Pädiatrische Intensivmedizin, Eltern-Kind-Zentrum, Universitätsklinikum Bonn, 53127 Bonn, Germany; (M.E.B.); (C.H.); (E.N.); (H.B.); (A.-S.B.); (M.Z.); (E.M.); (K.A.G.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Kora A. Grzelak
- Neonatologie und Pädiatrische Intensivmedizin, Eltern-Kind-Zentrum, Universitätsklinikum Bonn, 53127 Bonn, Germany; (M.E.B.); (C.H.); (E.N.); (H.B.); (A.-S.B.); (M.Z.); (E.M.); (K.A.G.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Hemmen Sabir
- Neonatologie und Pädiatrische Intensivmedizin, Eltern-Kind-Zentrum, Universitätsklinikum Bonn, 53127 Bonn, Germany; (M.E.B.); (C.H.); (E.N.); (H.B.); (A.-S.B.); (M.Z.); (E.M.); (K.A.G.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| |
Collapse
|
5
|
De Gregorio C, Gallardo J, Berríos-Cárcamo P, Handy Á, Santapau D, González-Madrid A, Ezquer M, Morales P, Luarte A, Corvalán D, Wyneken Ú, Ezquer F. Methadone directly impairs central nervous system cells in vitro. Sci Rep 2024; 14:16978. [PMID: 39043899 PMCID: PMC11266518 DOI: 10.1038/s41598-024-67860-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Methadone is a synthetic long-acting opioid that is increasingly used in the replacement therapy of opioid-addicted patients, including pregnant women. However, methadone therapy in this population poses challenges, as it induces cognitive and behavioral impairments in infants exposed to this opioid during prenatal development. In animal models, prenatal methadone exposure results in detrimental consequences to the central nervous system, such as: (i) increased neuronal apoptosis; (ii) disruption of oligodendrocyte maturation and increased apoptosis and (iii) increased microglia and astrocyte activation. However, it remains unclear whether these deleterious effects result from a direct effect of methadone on brain cells. Therefore, our goal was to uncover the impact of methadone on single brain cell types in vitro. Primary cultures of rat neurons, oligodendrocytes, microglia, and astrocytes were treated for three days with 10 µM methadone to emulate a chronic administration. Apoptotic neurons were identified by cleaved caspase-3 detection, and synaptic density was assessed by the juxtaposition of presynaptic and postsynaptic markers. Apoptosis of oligodendrocyte precursors was determined by cleaved caspase-3 detection. Oligodendrocyte myelination was assessed by immunofluorescence, while microglia and astrocyte proinflammatory activation were assessed by both immunofluorescence and RT-qPCR. Methadone treatment increased neuronal apoptosis and reduced synaptic density. Furthermore, it led to increased oligodendrocyte apoptosis and a reduction in the myelinating capacity of these cells, and promoted the proinflammatory activation of microglia and astrocytes. We showed that methadone, the most widely used drug in opioid replacement therapy for pregnant women with opioid addiction, directly impairs brain cells in vitro, highlighting the need for developing alternative therapies to address opioid addiction in this population.
Collapse
Affiliation(s)
| | - Javiera Gallardo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Pablo Berríos-Cárcamo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Álex Handy
- Faculty of Natural Sciences, Mathematics, and Environment, Universidad Tecnológica Metropolitana, Santiago, Chile
| | - Daniela Santapau
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Antonia González-Madrid
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Marcelo Ezquer
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile
| | - Paola Morales
- Program of Molecular and Clinical Pharmacology, ICBM, Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Alejandro Luarte
- Neuroscience Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Daniela Corvalán
- Neuroscience Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Úrsula Wyneken
- Neuroscience Program, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Avenida Plaza 680, Santiago, Chile.
- Research Center for the Development of Novel Therapeutics Alternatives for Alcohol Use Disorders, Santiago, Chile.
| |
Collapse
|
6
|
Fang M, Liu J, Zhang Z, Li Y, Zhu J, Lin Z. Chloroquine Protects Hypoxia/Ischemia-Induced Neonatal Brain Injury in Rats by Mitigating Blood-Brain Barrier Disruption. ACS Chem Neurosci 2023; 14:1764-1773. [PMID: 37116216 DOI: 10.1021/acschemneuro.2c00650] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Neonatal hypoxic-ischemic (H/I) brain damage (HIBD) is a devastating condition for which there are presently no effective therapeutic strategies against its severe neurological deficits in neonates and young children. Traditionally, H/I induces the compromise of the blood-brain barrier (BBB), which causes neuronal cell death, eventually resulting in brain secondary injury. In addition to neonatal HIBD, chloroquine (CQ) has been proved to exert a protective effect on BBB disruption in several brain injury models. The main purpose of this research was to study whether CQ protects the BBB from H/I insult and confers beneficial neuroprotection in the neonatal Rice-Vannucci rat model. Herein, we reported that CQ administration significantly reduced brain damage and improved behavioral dysplasia after H/I injury. Moreover, we demonstrated the protective effects of CQ on BBB integrity, evidenced by ameliorating brain edema and Evans blue extravasation, inhibiting the degeneration of the tight junction and adherens junction proteins, and improving pericyte survival in neonatal rats after HIBD. These findings indicated that CQ administration protected the BBB against H/I injury, thereby ameliorating brain damage and promoting neurofunctional recovery. Collectively, our data demonstrated that CQ played a crucial role in BBB integrity after neonatal H/I injury, which sheds light on the development of therapeutic agents to treat HIBD.
Collapse
Affiliation(s)
- Mingchu Fang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang 325027, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang 325027, China
| | - Jian Liu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhiwei Zhang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yueqi Li
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianghu Zhu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang 325027, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang 325027, China
| | - Zhenlang Lin
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang 325027, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
7
|
Temporal Characterization of Microglia-Associated Pro- and Anti-Inflammatory Genes in a Neonatal Inflammation-Sensitized Hypoxic-Ischemic Brain Injury Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2479626. [PMID: 35281473 PMCID: PMC8906938 DOI: 10.1155/2022/2479626] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/22/2021] [Accepted: 01/27/2022] [Indexed: 02/06/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) mainly affects preterm and term newborns, leading to a high risk of brain damage. Coexisting infection/inflammation and birth asphyxia are key factors associated with intracerebral increase of proinflammatory cytokines linked to HIE. Microglia are key mediators of inflammation during perinatal brain injury, characterized by their phenotypic plasticity, which may facilitate their participation in both the progression and resolution of injury-induced inflammation. The purpose of this study was to investigate the temporal expression of genes associated with pro- and anti-inflammatory cytokines as well as the nucleotide-binding domain, leucine-rich repeat protein (NLRP-3) inflammasome from microglia cells. For this purpose, we used our established neonatal rat model of inflammation-sensitized hypoxic-ischemic (HI) brain injury in seven-day-old rats. We assessed gene expression profiles of 11 cytokines and for NLRP-3 using real-time PCR from sorted CD11b/c microglia of brain samples at different time points (3.5 h after LPS injection and 0, 5, 24, 48, and 72 hours post HI) following different treatments: vehicle, E. coli lipopolysaccharide (LPS), vehicle/HI, and LPS/HI. Our results showed that microglia are early key mediators of the inflammatory response and exacerbate the inflammatory response following HI, polarizing into a predominant proinflammatory M1 phenotype in the early hours post HI. The brains only exposed to HI showed a delay in the expression of proinflammatory cytokines. We also demonstrated that NLRP-3 plays a role in the inflammatory resolution with a high expression after HI insult. The combination of both, a preinfection/inflammation condition and hypoxia-ischemia, resulted in a higher proinflammatory cytokine storm, highlighting the significant contribution of acute inflammation sensitizing prior to a hypoxic insult on the severity of perinatal brain damage.
Collapse
|
8
|
Tetorou K, Sisa C, Iqbal A, Dhillon K, Hristova M. Current Therapies for Neonatal Hypoxic-Ischaemic and Infection-Sensitised Hypoxic-Ischaemic Brain Damage. Front Synaptic Neurosci 2021; 13:709301. [PMID: 34504417 PMCID: PMC8421799 DOI: 10.3389/fnsyn.2021.709301] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Neonatal hypoxic-ischaemic brain damage is a leading cause of child mortality and morbidity, including cerebral palsy, epilepsy, and cognitive disabilities. The majority of neonatal hypoxic-ischaemic cases arise as a result of impaired cerebral perfusion to the foetus attributed to uterine, placental, or umbilical cord compromise prior to or during delivery. Bacterial infection is a factor contributing to the damage and is recorded in more than half of preterm births. Exposure to infection exacerbates neuronal hypoxic-ischaemic damage thus leading to a phenomenon called infection-sensitised hypoxic-ischaemic brain injury. Models of neonatal hypoxia-ischaemia (HI) have been developed in different animals. Both human and animal studies show that the developmental stage and the severity of the HI insult affect the selective regional vulnerability of the brain to damage, as well as the subsequent clinical manifestations. Therapeutic hypothermia (TH) is the only clinically approved treatment for neonatal HI. However, the number of HI infants needed to treat with TH for one to be saved from death or disability at age of 18-22 months, is approximately 6-7, which highlights the need for additional or alternative treatments to replace TH or increase its efficiency. In this review we discuss the mechanisms of HI injury to the immature brain and the new experimental treatments studied for neonatal HI and infection-sensitised neonatal HI.
Collapse
Affiliation(s)
| | | | | | | | - Mariya Hristova
- Perinatal Brain Repair Group, Department of Maternal and Fetal Medicine, UCL Institute for Women’s Health, London, United Kingdom
| |
Collapse
|
9
|
Sävman K, Wang W, Rafati AH, Svedin P, Nair S, Golubinskaya V, Ardalan M, Brown KL, Karlsson-Bengtsson A, Mallard C. Galectin-3 modulates microglia inflammation in vitro but not neonatal brain injury in vivo under inflammatory conditions. Dev Neurosci 2021; 43:296-311. [PMID: 34130282 DOI: 10.1159/000517687] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/24/2021] [Indexed: 11/19/2022] Open
Abstract
Microglia may contribute to injury but may also have neuroprotective properties. Galectin-3 has immunomodulatory properties that may affect the microglia phenotype and subsequent development of injury. Galectin-3 contributes to experimental hypoxic-ischemic (HI) injury in the neonatal brain, but it is unclear if galectin-3 has similar effects on infectious and sterile inflammation. Thus, we investigated the effect of galectin-3 on microglia in vitro under normal as well as infectious and sterile inflammatory conditions, and the effect of galectin-3 on neonatal brain injury following an infectious challenge in vivo. Conditions mimicking infectious or sterile inflammation were evaluated in primary microglia cell cultures from newborn mice, using LPS (10 ng/mL) and TNF-α (100 ng/mL). The response to galectin-3 was tested alone or together with LPS or TNF-α. Supernatants were collected 24 h after treatment and analyzed for 23 inflammatory mediators including pro- and anti-inflammatory cytokines and chemokines using multiplex protein analysis, as well as ELISA for MCP-1 and insulin-like growth factor (IGF)-1. Phosphorylation of proteins (AKT, ERK1/2, IκB-α, JNK, and p38) was determined in microglia cells. Neonatal brain injury was induced by a combination of LPS and HI (LPS + HI) in postnatal day 9 transgenic mice lacking functional galectin-3 and wild-type controls. LPS and TNF-α induced pro-inflammatory (9/11 vs. 9/10) and anti-inflammatory (6/6 vs. 2/6) cytokines, as well as chemokines (6/6 vs. 4/6) in a similar manner, except generally lower amplitude of the TNF-α-induced response. Galectin-3 alone had no effect on any of the proteins analyzed. Galectin-3 reduced the LPS- and TNF-α-induced microglia response for cytokines, chemokines, and phosphorylation of IκB-α. LPS decreased baseline IGF-1 levels, and the levels were restored by galectin-3. Brain injury or microglia response after LPS + HI was not affected by galectin-3 deficiency. Galectin-3 has no independent effect on microglia but modulates inflammatory activation in vitro. The effect was similar under infectious and sterile inflammatory conditions, suggesting that galectin-3 regulates inflammation not just by binding to LPS or toll-like receptor-4. Galectin-3 restores IGF-1 levels reduced by LPS-induced inflammation, suggesting a potential protective effect on infectious injury. However, galectin-3 deficiency did not affect microglia activation and was not beneficial in an injury model encompassing an infectious challenge.
Collapse
Affiliation(s)
- Karin Sävman
- Department of Pediatrics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Neonatology, The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Wei Wang
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ali Hoseinpoor Rafati
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Svedin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Syam Nair
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Veronika Golubinskaya
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryam Ardalan
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kelly L Brown
- Department of Pediatrics, University of British Columbia and the British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Anna Karlsson-Bengtsson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
10
|
Tapia-Bustos A, Lespay-Rebolledo C, Vío V, Pérez-Lobos R, Casanova-Ortiz E, Ezquer F, Herrera-Marschitz M, Morales P. Neonatal Mesenchymal Stem Cell Treatment Improves Myelination Impaired by Global Perinatal Asphyxia in Rats. Int J Mol Sci 2021; 22:ijms22063275. [PMID: 33806988 PMCID: PMC8004671 DOI: 10.3390/ijms22063275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/07/2021] [Accepted: 03/15/2021] [Indexed: 01/09/2023] Open
Abstract
The effect of perinatal asphyxia (PA) on oligodendrocyte (OL), neuroinflammation, and cell viability was evaluated in telencephalon of rats at postnatal day (P)1, 7, and 14, a period characterized by a spur of neuronal networking, evaluating the effect of mesenchymal stem cell (MSCs)-treatment. The issue was investigated with a rat model of global PA, mimicking a clinical risk occurring under labor. PA was induced by immersing fetus-containing uterine horns into a water bath for 21 min (AS), using sibling-caesarean-delivered fetuses (CS) as controls. Two hours after delivery, AS and CS neonates were injected with either 5 μL of vehicle (10% plasma) or 5 × 104 MSCs into the lateral ventricle. Samples were assayed for myelin-basic protein (MBP) levels; Olig-1/Olig-2 transcriptional factors; Gglial phenotype; neuroinflammation, and delayed cell death. The main effects were observed at P7, including: (i) A decrease of MBP-immunoreactivity in external capsule, corpus callosum, cingulum, but not in fimbriae of hippocampus; (ii) an increase of Olig-1-mRNA levels; (iii) an increase of IL-6-mRNA, but not in protein levels; (iv) an increase in cell death, including OLs; and (v) MSCs treatment prevented the effect of PA on myelination, OLs number, and cell death. The present findings show that PA induces regional- and developmental-dependent changes on myelination and OLs maturation. Neonatal MSCs treatment improves survival of mature OLs and myelination in telencephalic white matter.
Collapse
Affiliation(s)
- Andrea Tapia-Bustos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
- Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago 8370149, Chile
| | - Carolyne Lespay-Rebolledo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Valentina Vío
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Ronald Pérez-Lobos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Emmanuel Casanova-Ortiz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago 7710162, Chile;
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
- Correspondence: (M.H.-M.); (P.M.); Tel.: +56-229786788 (M.H.-M. & P.M.)
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Correspondence: (M.H.-M.); (P.M.); Tel.: +56-229786788 (M.H.-M. & P.M.)
| |
Collapse
|
11
|
Holloway RK, Ireland G, Sullivan G, Becher JC, Smith C, Boardman JP, Gressens P, Miron VE. Microglial inflammasome activation drives developmental white matter injury. Glia 2021; 69:1268-1280. [PMID: 33417729 PMCID: PMC8607465 DOI: 10.1002/glia.23963] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/15/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022]
Abstract
Injury to the developing brain during the perinatal period often causes hypomyelination, leading to clinical deficits for which there is an unmet therapeutic need. Dysregulation of inflammation and microglia have been implicated, yet the molecular mechanisms linking these to hypomyelination are unclear. Using human infant cerebrospinal fluid (CSF) and postmortem tissue, we found that microglial activation of the pro-inflammatory molecular complex the NLRP3 inflammasome is associated with pathology. By developing a novel mouse brain explant model of microglial inflammasome activation, we demonstrate that blocking the inflammasome rescues myelination. In human and mouse, we discovered a link between the inflammasome product IL1β and increased levels of follistatin, an endogenous inhibitor of activin-A. Follistatin treatment was sufficient to reduce myelination, whereas myelination was rescued in injured explants upon follistatin neutralization or supplementation with exogenous activin-A. Our data reveal that inflammasome activation in microglia drives hypomyelination and identifies novel therapeutic strategies to reinstate myelination following developmental injury.
Collapse
Affiliation(s)
- Rebecca K Holloway
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Graeme Ireland
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Gemma Sullivan
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Julie-Clare Becher
- Simpson Centre for Reproductive Health, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, Centre for Comparative Pathology, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - James P Boardman
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Pierre Gressens
- Department of Perinatal Imaging and Health, Rayne's Institute, King's College London, London, UK.,PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
12
|
Scher MS. "The First Thousand Days" Define a Fetal/Neonatal Neurology Program. Front Pediatr 2021; 9:683138. [PMID: 34408995 PMCID: PMC8365757 DOI: 10.3389/fped.2021.683138] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/27/2021] [Indexed: 01/11/2023] Open
Abstract
Gene-environment interactions begin at conception to influence maternal/placental/fetal triads, neonates, and children with short- and long-term effects on brain development. Life-long developmental neuroplasticity more likely results during critical/sensitive periods of brain maturation over these first 1,000 days. A fetal/neonatal program (FNNP) applying this perspective better identifies trimester-specific mechanisms affecting the maternal/placental/fetal (MPF) triad, expressed as brain malformations and destructive lesions. Maladaptive MPF triad interactions impair progenitor neuronal/glial populations within transient embryonic/fetal brain structures by processes such as maternal immune activation. Destructive fetal brain lesions later in pregnancy result from ischemic placental syndromes associated with the great obstetrical syndromes. Trimester-specific MPF triad diseases may negatively impact labor and delivery outcomes. Neonatal neurocritical care addresses the symptomatic minority who express the great neonatal neurological syndromes: encephalopathy, seizures, stroke, and encephalopathy of prematurity. The asymptomatic majority present with neurologic disorders before 2 years of age without prior detection. The developmental principle of ontogenetic adaptation helps guide the diagnostic process during the first 1,000 days to identify more phenotypes using systems-biology analyses. This strategy will foster innovative interdisciplinary diagnostic/therapeutic pathways, educational curricula, and research agenda among multiple FNNP. Effective early-life diagnostic/therapeutic programs will help reduce neurologic disease burden across the lifespan and successive generations.
Collapse
Affiliation(s)
- Mark S Scher
- Division of Pediatric Neurology, Department of Pediatrics, Fetal/Neonatal Neurology Program, Emeritus Scholar Tenured Full Professor in Pediatrics and Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
13
|
Vázquez-Borsetti P, Acuña A, Soliño M, López-Costa JJ, Kargieman L, Loidl FC. Deep hypothermia prevents striatal alterations produced by perinatal asphyxia: Implications for the prevention of dyskinesia and psychosis. J Comp Neurol 2020; 528:2679-2694. [PMID: 32301107 DOI: 10.1002/cne.24925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/23/2022]
Abstract
GABAergic medium spiny neurons are the main neuronal population in the striatum. Calbindin is preferentially expressed in medium spiny neurons involved in the indirect pathway. The aim of the present work is to analyze the effect of perinatal asphyxia on different subpopulations of GABAergic neurons in the striatum and to assess the outcome of deep therapeutic hypothermia. The uterus of pregnant rats was removed by cesarean section and the fetuses were exposed to hypoxia by immersion in water (19 min) at 37°C (perinatal asphyxia). The hypothermic group was exposed to 10°C during 30 min after perinatal asphyxia. The rats were euthanized at the age of one month (adolescent/adult rats), their brains were dissected out and coronal sections were immunolabeled for calbindin, calretinin, NeuN, and reelin. Reelin+ cells showed no staining in the striatum besides subventricular zone. The perinatal asphyxia (PA) group showed a significant decrease in calbindin neurons and a paradoxical increase in neurons estimated by NeuN staining. Moreover, calretinin+ cells, a specific subpopulation of GABAergic neurons, showed an increase caused by PA. Deep hypothermia reversed most of these alterations probably by protecting calbindin neurons. Similarly, there was a reduction of the diameter of the anterior commissure produced by the asphyxia that was prevented by hypothermic treatment.
Collapse
Affiliation(s)
- Pablo Vázquez-Borsetti
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", UBA-CONICET, Buenos Aires, Argentina
| | - Andrés Acuña
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", UBA-CONICET, Buenos Aires, Argentina
| | - Manuel Soliño
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", UBA-CONICET, Buenos Aires, Argentina
| | - Juan José López-Costa
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", UBA-CONICET, Buenos Aires, Argentina
| | - Lucila Kargieman
- IFIBYNE (UBA-CONICET) Instituto de Fisiología, Biología Molecular y Neurociencias-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fabián César Loidl
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", UBA-CONICET, Buenos Aires, Argentina
| |
Collapse
|
14
|
Farfán N, Carril J, Redel M, Zamorano M, Araya M, Monzón E, Alvarado R, Contreras N, Tapia-Bustos A, Quintanilla ME, Ezquer F, Valdés JL, Israel Y, Herrera-Marschitz M, Morales P. Intranasal Administration of Mesenchymal Stem Cell Secretome Reduces Hippocampal Oxidative Stress, Neuroinflammation and Cell Death, Improving the Behavioral Outcome Following Perinatal Asphyxia. Int J Mol Sci 2020; 21:ijms21207800. [PMID: 33096871 PMCID: PMC7589575 DOI: 10.3390/ijms21207800] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Perinatal Asphyxia (PA) is a leading cause of motor and neuropsychiatric disability associated with sustained oxidative stress, neuroinflammation, and cell death, affecting brain development. Based on a rat model of global PA, we investigated the neuroprotective effect of intranasally administered secretome, derived from human adipose mesenchymal stem cells (MSC-S), preconditioned with either deferoxamine (an hypoxia-mimetic) or TNF-α+IFN-γ (pro-inflammatory cytokines). PA was generated by immersing fetus-containing uterine horns in a water bath at 37 °C for 21 min. Thereafter, 16 μL of MSC-S (containing 6 μg of protein derived from 2 × 105 preconditioned-MSC), or vehicle, were intranasally administered 2 h after birth to asphyxia-exposed and control rats, evaluated at postnatal day (P) 7. Alternatively, pups received a dose of either preconditioned MSC-S or vehicle, both at 2 h and P7, and were evaluated at P14, P30, and P60. The preconditioned MSC-S treatment (i) reversed asphyxia-induced oxidative stress in the hippocampus (oxidized/reduced glutathione); (ii) increased antioxidative Nuclear Erythroid 2-Related Factor 2 (NRF2) translocation; (iii) increased NQO1 antioxidant protein; (iv) reduced neuroinflammation (decreasing nuclearNF-κB/p65 levels and microglial reactivity); (v) decreased cleaved-caspase-3 cell-death; (vi) improved righting reflex, negative geotaxis, cliff aversion, locomotor activity, anxiety, motor coordination, and recognition memory. Overall, the study demonstrates that intranasal administration of preconditioned MSC-S is a novel therapeutic strategy that prevents the long-term effects of perinatal asphyxia.
Collapse
Affiliation(s)
- Nancy Farfán
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Jaime Carril
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Martina Redel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Marta Zamorano
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Maureen Araya
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Estephania Monzón
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Raúl Alvarado
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Norton Contreras
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (N.C.); (J.L.V.)
| | - Andrea Tapia-Bustos
- School of Pharmacy, Faculty of Medicine, Universidad Andres Bello, Santiago 8370149, Chile;
| | - María Elena Quintanilla
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - José Luis Valdés
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (N.C.); (J.L.V.)
| | - Yedy Israel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine University of Chile, Santiago 8380453, Chile; (N.F.); (J.C.); (M.R.); (M.Z.); (M.A.); (E.M.); (R.A.); (M.E.Q.); (Y.I.); (M.H.-M.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (N.C.); (J.L.V.)
- Correspondence: ; Tel.: +56-229786788
| |
Collapse
|
15
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Cochrane Database Syst Rev 2020; 8:CD013202. [PMID: 32813884 PMCID: PMC7438027 DOI: 10.1002/14651858.cd013202.pub2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypoxic-ischaemic encephalopathy (HIE) is a leading cause of mortality and long-term neurological sequelae, affecting thousands of children worldwide. Current therapies to treat HIE are limited to cooling. Stem cell-based therapies offer a potential therapeutic approach to repair or regenerate injured brain tissue. These preclinical findings have now culminated in ongoing human neonatal trials. OBJECTIVES To determine the efficacy and safety of stem cell-based interventions for the treatment of hypoxic-ischaemic encephalopathy (HIE) in newborn infants. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2020, Issue 5), MEDLINE via PubMed (1966 to 8 June 2020), Embase (1980 to 8 June 2020), and CINAHL (1982 to 8 June 2020). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials. SELECTION CRITERIA Randomised controlled trials, quasi-randomised controlled trials and cluster trials comparing 1) stem cell-based interventions (any type) compared to control (placebo or no treatment); 2) use of mesenchymal stem/stromal cells (MSCs) of type (e.g. number of doses or passages) or source (e.g. autologous versus allogeneic, or bone marrow versus cord) versus MSCs of other type or source; 3) use of stem cell-based interventions other than MSCs of type (e.g. mononuclear cells, oligodendrocyte progenitor cells, neural stem cells, hematopoietic stem cells, and inducible pluripotent stem cells) or source (e.g. autologous versus allogeneic, or bone marrow versus cord) versus stem cell-based interventions other than MSCs of other type or source; or 4) MSCs versus stem cell-based interventions other than MSCs. DATA COLLECTION AND ANALYSIS For each of the included trials, two authors independently planned to extract data (e.g. number of participants, birth weight, gestational age, type and source of MSCs or other stem cell-based interventions) and assess the risk of bias (e.g. adequacy of randomisation, blinding, completeness of follow-up). The primary outcomes considered in this review are all-cause neonatal mortality, major neurodevelopmental disability, death or major neurodevelopmental disability assessed at 18 to 24 months of age. We planned to use the GRADE approach to assess the quality of evidence. MAIN RESULTS Our search strategy yielded 616 references. Two review authors independently assessed all references for inclusion. We did not find any completed studies for inclusion. Fifteen RCTs are currently registered and ongoing. We describe the three studies we excluded. AUTHORS' CONCLUSIONS There is currently no evidence from randomised trials that assesses the benefit or harms of stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants.
Collapse
Affiliation(s)
- Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Lund University, Skåne University Hospital, Lund, Sweden
| | - Olga Romantsik
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Alvaro Moreira
- Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David Ley
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Bernard Thébaud
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Canada
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell Research, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
16
|
Scher MS. Neurologic outcome after fetal inflammatory response syndrome: Trimester-specific considerations. Semin Fetal Neonatal Med 2020; 25:101137. [PMID: 33158496 DOI: 10.1016/j.siny.2020.101137] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Clinical signs and neuroimaging patterns associated with the fetal inflammatory response syndrome (FIRS) worsen or mimic the clinical repertoire after intrapartum hypoxic-ischemic encephalopathy (HIE) during labor and/or parturition. Diagnostic considerations expressed as neonatal encephalopathy (NE) must consider chronic as well as acute factors associated with FIRS. Trimester-specific factors adversely alter the interactions of the maternal/placental/fetal (MPF) triad and influence the postnatal phenotype of FIRS. Anticipatory guidance for families by clinicians caring for survivors with FIRS, as well as researchers, must consider acute and chronic effects that influence neurologic outcome. Novel neurotherapeutic interventions must include prenatal preventive as well as peripartum/postnatal rescue and repair strategies to effectively reduce the presence and severity of sequelae from FIRS.
Collapse
Affiliation(s)
- Mark S Scher
- Emeritus Full Professor of Pediatrics and Neurology, Rainbow Babies and Children's Hospital/MacDonald Hospital for Women, University Hospitals Cleveland Medical Center, Case Western Reserve University, School of Medicine, 11100 Euclid Avenue Cleveland, Ohio, 44106, USA.
| |
Collapse
|
17
|
McNamara NB, Miron VE. Microglia in developing white matter and perinatal brain injury. Neurosci Lett 2019; 714:134539. [PMID: 31614181 DOI: 10.1016/j.neulet.2019.134539] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/13/2022]
Abstract
Perinatal brain injury (PBI) to the developing white matter results in hypomyelination of axons and can cause long-term motor and cognitive deficits e.g. cerebral palsy. There are currently no approved therapies aimed at repairing the white matter following insult, underscoring the need to investigate the mechanisms underlying the pathogenesis of PBI. Microglia have been strongly implicated, but their function and heterogeneity in this context remain poorly understood, posing a barrier to the development of microglia-targeted therapies for white matter repair following PBI. In this review, we discuss the roles of microglia in normal white matter development and in PBI, and potential drug strategies to influence microglial responses in this setting.
Collapse
Affiliation(s)
- Niamh B McNamara
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
18
|
C1 Esterase Inhibitor Reduces BBB Leakage and Apoptosis in the Hypoxic Developing Mouse Brain. Neuromolecular Med 2019; 22:31-44. [DOI: 10.1007/s12017-019-08560-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022]
|
19
|
Martinello KA, Meehan C, Avdic-Belltheus A, Lingam I, Ragab S, Hristova M, Tann CJ, Peebles D, Hagberg H, Wolfs TGAM, Klein N, Tachtsidis I, Golay X, Kramer BW, Fleiss B, Gressens P, Robertson NJ. Acute LPS sensitization and continuous infusion exacerbates hypoxic brain injury in a piglet model of neonatal encephalopathy. Sci Rep 2019; 9:10184. [PMID: 31308390 PMCID: PMC6629658 DOI: 10.1038/s41598-019-46488-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 06/29/2019] [Indexed: 12/12/2022] Open
Abstract
Co-existing infection/inflammation and birth asphyxia potentiate the risk of developing neonatal encephalopathy (NE) and adverse outcome. In a newborn piglet model we assessed the effect of E. coli lipopolysaccharide (LPS) infusion started 4 h prior to and continued for 48 h after hypoxia on brain cell death and systemic haematological changes compared to LPS and hypoxia alone. LPS sensitized hypoxia resulted in an increase in mortality and in brain cell death (TUNEL positive cells) throughout the whole brain, and in the internal capsule, periventricular white matter and sensorimotor cortex. LPS alone did not increase brain cell death at 48 h, despite evidence of neuroinflammation, including the greatest increases in microglial proliferation, reactive astrocytosis and cleavage of caspase-3. LPS exposure caused splenic hypertrophy and platelet count suppression. The combination of LPS and hypoxia resulted in the highest and most sustained systemic white cell count increase. These findings highlight the significant contribution of acute inflammation sensitization prior to an asphyxial insult on NE illness severity.
Collapse
Affiliation(s)
- Kathryn A Martinello
- Institute for Women's Health, University College London, London, United Kingdom
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Christopher Meehan
- Institute for Women's Health, University College London, London, United Kingdom
| | | | - Ingran Lingam
- Institute for Women's Health, University College London, London, United Kingdom
| | - Sara Ragab
- Institute for Women's Health, University College London, London, United Kingdom
| | - Mariya Hristova
- Institute for Women's Health, University College London, London, United Kingdom
| | - Cally J Tann
- Institute for Women's Health, University College London, London, United Kingdom
- Maternal, Adolescent, Reproductive and Child Health Centre, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Donald Peebles
- Institute for Women's Health, University College London, London, United Kingdom
| | - Henrik Hagberg
- Centre of Perinatal Medicine & Health, Department of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Centre for the Developing Brain, Department of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Tim G A M Wolfs
- Department of Paediatrics, University of Maastricht, Maastricht, Netherlands
| | - Nigel Klein
- Infection, Inflammation and Rheumatology, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Ilias Tachtsidis
- Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Xavier Golay
- Institute of Neurology, University College London, London, United Kingdom
| | - Boris W Kramer
- Department of Paediatrics, University of Maastricht, Maastricht, Netherlands
| | - Bobbi Fleiss
- Centre for the Developing Brain, Department of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Pierre Gressens
- Centre for the Developing Brain, Department of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Nicola J Robertson
- Institute for Women's Health, University College London, London, United Kingdom.
- Division of Neonatology, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
20
|
Fragopoulou AF, Qian Y, Heijtz RD, Forssberg H. Can Neonatal Systemic Inflammation and Hypoxia Yield a Cerebral Palsy-Like Phenotype in Periadolescent Mice? Mol Neurobiol 2019; 56:6883-6900. [PMID: 30941732 PMCID: PMC6728419 DOI: 10.1007/s12035-019-1548-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 03/12/2019] [Indexed: 12/16/2022]
Abstract
Cerebral palsy (CP) is one of the most common childhood-onset motor disabilities, attributed to injuries of the immature brain in the foetal or early postnatal period. The underlying mechanisms are poorly understood, rendering prevention and treatment strategies challenging. The aim of the present study was to establish a mouse model of CP for preclinical assessment of new interventions. For this purpose, we explored the impact of a double neonatal insult (i.e. systemic inflammation combined with hypoxia) on behavioural and cellular outcomes relevant to CP during the prepubertal to adolescent period of mice. Pups were subjected to intraperitoneal lipopolysaccharide (LPS) injections from postnatal day (P) 3 to P6 followed by hypoxia at P7. Gene expression analysis at P6 revealed a strong inflammatory response in a brain region-dependent manner. A comprehensive battery of behavioural assessments performed between P24 and P47 showed impaired limb placement and coordination when walking on a horizontal ladder in both males and females. Exposed males also displayed impaired performance on a forelimb skilled reaching task, altered gait pattern and increased exploratory activity. Exposed females showed a reduction in grip strength and traits of anxiety-like behaviour. These behavioural alterations were not associated with gross morphological changes, white matter lesions or chronic inflammation in the brain. Our results indicate that the neonatal double-hit with LPS and hypoxia can induce subtle long-lasting deficits in motor learning and fine motor skills, which partly reflect the symptoms of children with CP who have mild gross and fine motor impairments.
Collapse
Affiliation(s)
- Adamantia F Fragopoulou
- Department of Neuroscience, Biomedicum, Karolinska Institutet, 171 77, Stockholm, Sweden. .,Department of Women's and Children's Health, Karolinska Institutet, 171 76, Stockholm, Sweden.
| | - Yu Qian
- Department of Neuroscience, Biomedicum, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Rochellys Diaz Heijtz
- Department of Neuroscience, Biomedicum, Karolinska Institutet, 171 77, Stockholm, Sweden.,INSERM U1239, University of Rouen Normandy, 76130, Mont-Saint-Aignan, France
| | - Hans Forssberg
- Department of Women's and Children's Health, Karolinska Institutet, 171 76, Stockholm, Sweden.
| |
Collapse
|
21
|
Xu MY, Wang YF, Wei PJ, Gao YQ, Zhang WT. Hypoxic preconditioning improves long-term functional outcomes after neonatal hypoxia-ischemic injury by restoring white matter integrity and brain development. CNS Neurosci Ther 2019; 25:734-747. [PMID: 30689302 PMCID: PMC6515700 DOI: 10.1111/cns.13102] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/16/2022] Open
Abstract
Aims Neonatal hypoxia–ischemia (H/I) results in gray and white matter injury, characterized by neuronal loss, failure of neural network formation, retarded myelin formation, and abnormal accumulation of oligodendrocyte progenitor cells (OPCs). These changes lead to severe neurological deficits and mortality. Sublethal hypoxic preconditioning (HPC) can protect the developing brain against H/I. However, limited evidence is available concerning its effect on white matter injury. Methods In this study, P6 neonatal Sprague‐Dawley rats were subjected to normoxic (21% O2) or HPC (7.8% O2) for 3 hours followed 24 hours later by H/I brain injury. Neurological deficits were assessed by gait, righting reflex, foot fault, and Morris water maze tests. Compound action potential of the corpus callosum was recorded 35 days after surgery, and the correlation between axon myelination and neurological function was determined. Results Hypoxic preconditioning significantly attenuated H/I brain injury at 7 days and remarkably improved both sensorimotor and cognitive functional performances up to 35 days after H/I. HPC‐afforded improvement in long‐term neurological outcomes was attributable, at least in part, to restoration of the differentiation and maturation capacity in oligodendrocyte progenitor cells, amelioration of microglia/macrophage activation and neuroinflammation, and continuation of brain development after H/I. Conclusions Hypoxic preconditioning restores white matter repair, development, and functional integrity in developing brain after H/I brain injury.
Collapse
Affiliation(s)
- Ming-Yue Xu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Yang-Fan Wang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Peng-Ju Wei
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Yan-Qin Gao
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Wen-Ting Zhang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Scher MS. Fetal neurology: Principles and practice with a life-course perspective. HANDBOOK OF CLINICAL NEUROLOGY 2019; 162:1-29. [PMID: 31324306 DOI: 10.1016/b978-0-444-64029-1.00001-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Clinical service, educational, and research components of a fetal/neonatal neurology program are anchored by the disciplines of developmental origins of health and disease and life-course science as programmatic principles. Prenatal participation provides perspectives on maternal, fetal, and placental contributions to health or disease for fetal and subsequent neonatal neurology consultations. This program also provides an early-life diagnostic perspective for neurologic specialties concerned with brain health and disease throughout childhood and adulthood. Animal models and birth cohort studies have demonstrated how the science of epigenetics helps to understand gene-environment interactions to better predict brain health or disease. Fetal neurology consultations provide important diagnostic contributions during critical or sensitive periods of brain development when future neurotherapeutic interventions will maximize adaptive neuroplasticity. Age-specific normative neuroinformatics databases that employ computer-based strategies to integrate clinical/demographic, neuroimaging, neurophysiologic, and genetic datasets will more accurately identify either symptomatic patients or those at risk for brain disorders who would benefit from preventive, rescue, or reparative treatment choices throughout the life span.
Collapse
Affiliation(s)
- Mark S Scher
- Division of Pediatric Neurology, Case Western Reserve University, Cleveland, OH, United States.
| |
Collapse
|
23
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Hippokratia 2018. [DOI: 10.1002/14651858.cd013202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Matteo Bruschettini
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
- Skåne University Hospital; Cochrane Sweden; Wigerthuset, Remissgatan 4, first floor room 11-221 Lund Sweden 22185
| | - Olga Romantsik
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Alvaro Moreira
- University of Texas Health Science Center at San Antonio; Pediatrics, Division of Neonatology; San Antonio Texas USA
| | - David Ley
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Bernard Thébaud
- Children's Hospital of Eastern Ontario; Department of Pediatrics; Ottawa ON Canada
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research; Ottawa Canada
- University of Ottawa; Department of Cellular and Molecular Medicine; Ottawa Canada
| |
Collapse
|
24
|
Wagenaar N, Nijboer CH, van Bel F. Repair of neonatal brain injury: bringing stem cell-based therapy into clinical practice. Dev Med Child Neurol 2017; 59:997-1003. [PMID: 28786482 DOI: 10.1111/dmcn.13528] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/07/2017] [Indexed: 12/15/2022]
Abstract
Hypoxic-ischaemic brain injury is one of most important causes of neonatal mortality and long-term neurological morbidity in infants born at term. At present, only hypothermia in infants with perinatal hypoxic-ischaemic encephalopathy has shown benefit as a neuroprotective strategy. Otherwise, current treatment options for neonatal brain injury mainly focus on controlling (associated) symptoms. Regeneration of the injured neonatal brain with stem cell-based therapies is emerging and experimental results are promising. At present, increasing efforts are made to bring stem cell-based therapies to the clinic. Among all progenitor cell types, mesenchymal stromal (stem) cells seem to be most promising for human use given their neuroregenerative properties and favourable safety profile. This review summarizes the actual state, potential hurdles and possibilities of stem cell-based therapy for neonatal brain injury in the clinical setting. An early version of this paper was presented at the Groningen Early Intervention Meeting which was held in April 2016.
Collapse
Affiliation(s)
- Nienke Wagenaar
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Cora H Nijboer
- Laboratory of NeuroImmunology and Developmental Origins of Disease, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Frank van Bel
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| |
Collapse
|
25
|
Oppliger B, Joerger-Messerli M, Mueller M, Reinhart U, Schneider P, Surbek DV, Schoeberlein A. Intranasal Delivery of Umbilical Cord-Derived Mesenchymal Stem Cells Preserves Myelination in Perinatal Brain Damage. Stem Cells Dev 2017; 25:1234-42. [PMID: 27392671 DOI: 10.1089/scd.2016.0027] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Preterm white matter injury (WMI) is an important cause for long-term disability. Stem cell transplantation has been proposed as a novel therapeutic approach. However, intracerebral transplantation is not feasible for clinical purpose in newborns. Intranasal delivery of cells to the brain might be a promising, noninvasive therapeutic approach to restore the damaged brain. Therefore, our goal is to study the remyelinating potential of human Wharton's jelly mesenchymal stem cells (hWJ-MSCs) after intranasal delivery. Wistar rat pups, previously brain-damaged by a combined hypoxic-ischemic and inflammatory insult, received hWJ-MSC (150,000 cells in 3 μL) that were intranasally delivered twice to each nostril (600,000 cells total). WMI was assessed by immunohistochemistry and western blot for myelination, astrogliosis, and microgliosis. The expression of preoligodendrocyte markers, and neurotrophic factors, was analyzed by real-time polymerase chain reaction. Animals treated with intranasally delivered hWJ-MSC showed increased myelination and decreased gliosis compared to untreated animals. hWJ-MSC may, therefore, modulate the activation of microglia and astrocytes, resulting in a change of the brain microenvironment, which facilitates the maturation of oligodendrocyte lineage cells. This is the first study to show that intranasal delivery of hWJ-MSC in rats prevented hypomyelination and microgliosis in a model of WMI in the premature rat brain. Further studies should address the dose and frequency of administration.
Collapse
Affiliation(s)
- Byron Oppliger
- 1 Department of Obstetrics and Gynecology, University Hospital Bern , Bern, Switzerland .,2 Laboratory for Prenatal Medicine, Department of Clinical Research, University of Bern , Bern, Switzerland .,3 Graduate School for Biomedical Sciences (GCB), University of Bern , Bern, Switzerland
| | - Marianne Joerger-Messerli
- 1 Department of Obstetrics and Gynecology, University Hospital Bern , Bern, Switzerland .,2 Laboratory for Prenatal Medicine, Department of Clinical Research, University of Bern , Bern, Switzerland
| | - Martin Mueller
- 1 Department of Obstetrics and Gynecology, University Hospital Bern , Bern, Switzerland .,2 Laboratory for Prenatal Medicine, Department of Clinical Research, University of Bern , Bern, Switzerland .,4 Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine , New Haven, Connecticut
| | - Ursula Reinhart
- 1 Department of Obstetrics and Gynecology, University Hospital Bern , Bern, Switzerland .,2 Laboratory for Prenatal Medicine, Department of Clinical Research, University of Bern , Bern, Switzerland
| | - Philipp Schneider
- 1 Department of Obstetrics and Gynecology, University Hospital Bern , Bern, Switzerland .,2 Laboratory for Prenatal Medicine, Department of Clinical Research, University of Bern , Bern, Switzerland
| | - Daniel V Surbek
- 1 Department of Obstetrics and Gynecology, University Hospital Bern , Bern, Switzerland .,2 Laboratory for Prenatal Medicine, Department of Clinical Research, University of Bern , Bern, Switzerland
| | - Andreina Schoeberlein
- 1 Department of Obstetrics and Gynecology, University Hospital Bern , Bern, Switzerland .,2 Laboratory for Prenatal Medicine, Department of Clinical Research, University of Bern , Bern, Switzerland
| |
Collapse
|
26
|
Gussenhoven R, Ophelders DRMG, Kemp MW, Payne MS, Spiller OB, Beeton ML, Stock SJ, Cillero-Pastor B, Barré FPY, Heeren RMA, Kessels L, Stevens B, Rutten BP, Kallapur SG, Jobe AH, Kramer BW, Wolfs TGAM. The Paradoxical Effects of Chronic Intra-Amniotic Ureaplasma parvum Exposure on Ovine Fetal Brain Development. Dev Neurosci 2017; 39:472-486. [PMID: 28848098 DOI: 10.1159/000479021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/24/2017] [Indexed: 01/23/2023] Open
Abstract
Chorioamnionitis is associated with adverse neurodevelopmental outcomes in preterm infants. Ureaplasma spp. are the microorganisms most frequently isolated from the amniotic fluid of women diagnosed with chorioamnionitis. However, controversy remains concerning the role of Ureaplasma spp. in the pathogenesis of neonatal brain injury. We hypothesize that reexposure to an inflammatory trigger during the perinatal period might be responsible for the variation in brain outcomes of preterms following Ureaplasma-driven chorioamnionitis. To investigate these clinical scenarios, we performed a detailed multimodal study in which ovine neurodevelopmental outcomes were assessed following chronic intra-amniotic Ureaplasma parvum (UP) infection either alone or combined with subsequent lipopolysaccharide (LPS) exposure. We show that chronic intra-amniotic UP exposure during the second trimester provoked a decrease in astrocytes, increased oligodendrocyte numbers, and elevated 5-methylcytosine levels. In contrast, short-term LPS exposure before preterm birth induced increased microglial activation, myelin loss, elevation of 5-hydroxymethylcytosine levels, and lipid profile changes. These LPS-induced changes were prevented by chronic preexposure to UP (preconditioning). These data indicate that chronic UP exposure has dual effects on preterm brain development in utero. On the one hand, prolonged UP exposure causes detrimental cerebral changes that may predispose to adverse postnatal clinical outcomes. On the other, chronic intra-amniotic UP exposure preconditions the brain against a second inflammatory hit. This study demonstrates that microbial interactions and the timing and duration of the inflammatory insults determine the effects on the fetal brain. Therefore, this study helps to understand the complex and diverse postnatal neurological outcomes following UP driven chorioamnionitis.
Collapse
Affiliation(s)
- Ruth Gussenhoven
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
A Controversial Medicolegal Issue: Timing the Onset of Perinatal Hypoxic-Ischemic Brain Injury. Mediators Inflamm 2017; 2017:6024959. [PMID: 28883688 PMCID: PMC5572618 DOI: 10.1155/2017/6024959] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/18/2017] [Indexed: 12/11/2022] Open
Abstract
Perinatal hypoxic-ischemic brain injury, as a result of chronic, subacute, and acute insults, represents the pathological consequence of fetal distress and birth or perinatal asphyxia, that is, “nonreassuring fetal status.” Hypoxic-ischemic injury (HII) is typically characterized by an early phase of damage, followed by a delayed inflammatory local response, in an apoptosis-necrosis continuum. In the early phase, the cytotoxic edema and eventual acute lysis take place; with reperfusion, additional damage should be assigned to excitotoxicity and oxidative stress. Finally, a later phase involves all the inflammatory activity and long-term neural tissue repairing and remodeling. In this model mechanism, loss of mitochondrial function is supposed to be the hallmark of secondary injury progression, and autophagy which is lysosome-mediated play a role in enhancing brain injury. Early-induced molecules driven by hypoxia, as chaperonins HSPs and ORP150, besides common markers for inflammatory responses, have predictive value in timing the onset of neonatal HII; on the other hand, clinical biomarkers for HII diagnosis, as CK-BB, LDH, S-100beta, and NSE, could be useful to predict outcomes.
Collapse
|
28
|
Ziemka-Nalecz M, Janowska J, Strojek L, Jaworska J, Zalewska T, Frontczak-Baniewicz M, Sypecka J. Impact of neonatal hypoxia-ischaemia on oligodendrocyte survival, maturation and myelinating potential. J Cell Mol Med 2017; 22:207-222. [PMID: 28782169 PMCID: PMC5742723 DOI: 10.1111/jcmm.13309] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/15/2017] [Indexed: 02/06/2023] Open
Abstract
Hypoxic-ischaemic episodes experienced at the perinatal period commonly lead to a development of neurological disabilities and cognitive impairments in neonates or later in childhood. Clinical symptoms often are associated with the observed alterations in white matter in the brains of diseased children, suggesting contribution of triggered oligodendrocyte/myelin pathology to the resulting disorders. To date, the processes initiated by perinatal asphyxia remain unclear, hampering the ability to develop preventions. To address the issue, the effects of temporal hypoxia-ischaemia on survival, proliferation and the myelinating potential of oligodendrocytes were evaluated ex vivo using cultures of hippocampal organotypic slices and in vivo in rat model of perinatal asphyxia. The potential engagement of gelatinases in oligodendrocyte maturation was assessed as well. The results pointed to a significant decrease in the number of oligodendrocyte progenitor cells (OPCs), which is compensated for to a certain extent by the increased rate of OPC proliferation. Oligodendrocyte maturation seemed however to be significantly altered. An ultrastructural examination of selected brain regions performed several weeks after the insult showed however that the process of developing central nervous system myelination proceeds efficiently resulting in enwrapping the majority of axons in compact myelin. The increased angiogenesis in response to neonatal hypoxic-ischaemic insult was also noticed. In conclusion, the study shows that hypoxic-ischaemic episodes experienced during the most active period of nervous system development might be efficiently compensated for by the oligodendroglial cell response triggered by the insult. The main obstacle seems to be the inflammatory process modulating the local microenvironment.
Collapse
Affiliation(s)
- Malgorzata Ziemka-Nalecz
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Justyna Janowska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Lukasz Strojek
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Jaworska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
29
|
Ziemka-Nalecz M, Jaworska J, Zalewska T. Insights Into the Neuroinflammatory Responses After Neonatal Hypoxia-Ischemia. J Neuropathol Exp Neurol 2017; 76:644-654. [DOI: 10.1093/jnen/nlx046] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
30
|
Elsayed YN, Fraser D. Integrated Evaluation of Neonatal Hemodynamics, Part 2: Systematic Bedside Assessment. Neonatal Netw 2017; 35:192-203. [PMID: 27461198 DOI: 10.1891/0730-0832.35.4.192] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Intact hemodynamics results when there is adequate oxygen uptake by the respiratory system, normal cardiac output, sufficient oxygen-carrying capacity of blood, and intact autoregulatory mechanisms to maintain enough oxygenation for normal end-organ function. The current routine monitoring of cardiovascular dynamics in sick preterm and term infants has been based on incomplete evaluation and relies on nonspecific and sometimes misleading clinical markers such as blood pressure. A thorough understanding of perinatal and neonatal cardiovascular, respiratory, oxygen, and other specific end-organ physiology is also mandatory for proper targeted interpretation.
Collapse
Affiliation(s)
- Yasser N Elsayed
- University of Manitoba, Faculty of Medicine, Winnipeg, MB R3E0L8 Canada
| | | |
Collapse
|
31
|
Braccioli L, Heijnen CJ, Coffer PJ, Nijboer CH. Delayed administration of neural stem cells after hypoxia-ischemia reduces sensorimotor deficits, cerebral lesion size, and neuroinflammation in neonatal mice. Pediatr Res 2017; 81:127-135. [PMID: 27632779 DOI: 10.1038/pr.2016.172] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/07/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Hypoxic-ischemic (HI) encephalopathy causes mortality and severe morbidity in neonates. Treatments with a therapeutic window >6 h are currently not available. Here, we explored whether delayed transplantation of allogenic neural stem cells (NSCs) at 10 d after HI could be a tool to repair HI brain injury and improve behavioral impairments. METHODS HI was induced in 9-d-old mice. Animals received NSCs or vehicle intracranially in the hippocampus at 10 d post-HI. Sensorimotor performance was assessed by cylinder rearing test. Lesion size, synaptic integrity, and fate of injected NSCs were determined by immuno-stainings. Neuroinflammation was studied by immuno-stainings of brain sections, primary glial cultures, and TNFα ELISA. RESULTS NSC transplantation at 10 d post-insult induced long-term improvement of motor performance and synaptic integrity, and reduced lesion size compared to vehicle-treatment. HI-induced neuroinflammation was reduced after NSC treatment, at least partially by factors secreted by NSCs. Injected NSCs migrated toward and localized at the damaged hippocampus. Transplanted NSCs differentiated toward the neuronal lineage and formed a niche with endogenous precursors. CONCLUSION Our study provides evidence of the efficacy of NSC transplantation late after HI as a tool to reduce neonatal HI brain injury through regeneration of the lesion.
Collapse
Affiliation(s)
- Luca Braccioli
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paul J Coffer
- Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
32
|
Tuor UI, Sule M, Qiao M. Combined damage produced by multiple mild cerebral insults assessed using MRI in neonatal rats. Neurosci Lett 2016; 634:13-18. [PMID: 27702627 DOI: 10.1016/j.neulet.2016.09.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/21/2016] [Accepted: 09/30/2016] [Indexed: 10/20/2022]
Abstract
PURPOSE To determine whether damage to neonatal brain is exacerbated with multiple mild cerebral insults as detected with MRI and corroborated using histology. MATERIALS AND METHODS The combined brain injury produced by multiple procedures was compared in neonatal rats having: Sham surgery at P5, Sham surgery at P5 plus a diffuse mild transient unilateral cerebral hypoxia ischemia (HI) at P7, HI alone, and a minor photothrombotic (PT) stroke at P5 followed by HI. MRI after the ischemic insults was followed by final histology. RESULTS PT produced lesions with increased T2 and decreased apparent diffusion coefficient for water (ADC) but no significant effects of a second HI. However, near the PT lesion/parietal cortex there were patchy areas of enhanced T2 and decreased ADC in 6/9, 3/8 and 0/8 animals in the PT+HI, Sham+HI and HI groups, respectively (P<0.05). Patches corresponded histologically to increased vacuolation and cell death and were more pronounced in the PT+HI and Sham+PT groups than the HI group. CONCLUSION The extent of damage produced by a minor neonatal stroke followed by a diffuse HI two days later results in heterogeneous enhancement of T2, ADC and histological injury near the lesion. Surgical procedures including mechanical head manipulation followed by HI also produced some enhanced heterogeneity of hypoxic-ischemic injury affirming the need for sham controls.
Collapse
Affiliation(s)
- Ursula I Tuor
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, 3280 Hospital Dr, NW, Calgary, T2N 2T8, Canada.
| | - Manasi Sule
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, 3280 Hospital Dr, NW, Calgary, T2N 2T8, Canada
| | - Min Qiao
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, 3280 Hospital Dr, NW, Calgary, T2N 2T8, Canada
| |
Collapse
|
33
|
Yao L, Lu P, Ling EA. Melatonin Suppresses Toll Like Receptor 4-Dependent Caspase-3 Signaling Activation Coupled with Reduced Production of Proinflammatory Mediators in Hypoxic Microglia. PLoS One 2016; 11:e0166010. [PMID: 27812200 PMCID: PMC5094586 DOI: 10.1371/journal.pone.0166010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/21/2016] [Indexed: 12/14/2022] Open
Abstract
Microglia activation and associated inflammatory response play pivotal roles in the pathogenesis of different neurodegenerative diseases including neonatal hypoxic brain injury. Here we show that caspase3 expression was upregulated in activated microglia after hypoxic exposure, and remarkably, the cell viability remained unaffected alluding to the possibility of a non-apoptotic role of caspase3 in activated microglia. Chemical inhibition of caspase3 suppressed microglia activation as evident by an obvious reduction in expression of proinflammatory mediators and NF-κB signaling activation. Hypoxia induced caspase3 activation was TLR4 dependent as supported by the fact that caspase3 activation was hindered in cells with TLR4 knockdown. Interestingly, melatonin treatment significantly suppressed caspase3 activation. More importantly, melatonin also inhibited the increase in TLR4 protein and mRNA expression in hypoxic microglia. Inhibition of TLR4 expression by melatonin was also found in microglia of postnatal rats subjected to hypoxic exposure. Taken together, it is concluded that melatonin could inhibit TLR4 expression in hypoxic microglia followed by suppression of caspase3 activation leading to decrease in production of proinflammatory mediators.
Collapse
Affiliation(s)
- Linli Yao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pengfei Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
34
|
Vázquez-Borsetti P, Peña E, Rico C, Noto M, Miller N, Cohon D, Acosta JM, Ibarra M, Loidl FC. Perinatal Asphyxia Reduces the Number of Reelin Neurons in the Prelimbic Cortex and Deteriorates Social Interaction in Rats. Dev Neurosci 2016; 38:241-250. [DOI: 10.1159/000448244] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/08/2016] [Indexed: 11/19/2022] Open
Abstract
Obstetrical complications of perinatal asphyxia (PA) can often induce lesions that, in the long-term, manifest as schizophrenia. A deterioration of the medial prefrontal cortex (mPFC) and a reduction in the number of GABAergic neurons are commonly observed in the pathophysiology of schizophrenia. In this study, we investigated the link between PA, reelin and calbindin diminution and psychiatric diseases that involve social interaction deficits. This was achieved by observing the effect of 19 min of asphyxia on both subpopulations of GABAergic neurons. PA was produced by water immersion of fetus-containing uterus horns removed by cesarean section from ready-to-deliver rats. PA generated a significant and specific decrease in the number of reelin-secreting neurons in mPFC layer VI [F(2, 6) = 8.716, p = 0.016; PA vs. vaginal controls (VC), p = 0.03, and PA vs. cesarean controls (CC), p = 0.022]. This reduction reached approximately 60% on average. Changes in the percentage of reelin neurons including all the cortex layers did not achieve a significant outcome but a trend: CC % 10.61 ± 1.34; PA % 8.64 ± 1.71 [F(2, 6) = 1.299, p = 0.33]. In the case of calbindin, there was a significant decrease in cell density in the PA group [2-way repeated-measures ANOVA, F(1, 4) = 13.03, p = 0.0226]. The multiple-comparisons test showed significant differences in the superficial aspect of layer II (Sidak test for multiple comparisons CC vs. PA at 200 µm: p = 0.003). A small, but significant difference could be seen when the distance from the pia mater to the start of layer VI was analyzed (CC mean ± SEM = 768.9 ± 8.382; PA mean ± SEM = 669.3 ± 17.75; p = 0.036). Rats exposed to PA showed deterioration in social interactions, which manifested as a decrease in play soliciting. In this model, which involved severe/moderate asphyxia, we did not find significant changes in locomotive activity or anxiety indicators in the open field task. The loss of reelin neurons could be conducive to the shrinkage of the prelimbic cortex through the reduction in neuropil and the deterioration of the function of this structure.
Collapse
|
35
|
Ophelders DRMG, Gussenhoven R, Lammens M, Küsters B, Kemp MW, Newnham JP, Payne MS, Kallapur SG, Jobe AH, Zimmermann LJ, Kramer BW, Wolfs TGAM. Neuroinflammation and structural injury of the fetal ovine brain following intra-amniotic Candida albicans exposure. J Neuroinflammation 2016; 13:29. [PMID: 26842664 PMCID: PMC4739103 DOI: 10.1186/s12974-016-0492-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 01/24/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Intra-amniotic Candida albicans (C. Albicans) infection is associated with preterm birth and high morbidity and mortality rates. Survivors are prone to adverse neurodevelopmental outcomes. The mechanisms leading to these adverse neonatal brain outcomes remain largely unknown. To better understand the mechanisms underlying C. albicans-induced fetal brain injury, we studied immunological responses and structural changes of the fetal brain in a well-established translational ovine model of intra-amniotic C. albicans infection. In addition, we tested whether these potential adverse outcomes of the fetal brain were improved in utero by antifungal treatment with fluconazole. METHODS Pregnant ewes received an intra-amniotic injection of 10(7) colony-forming units C. albicans or saline (controls) at 3 or 5 days before preterm delivery at 0.8 of gestation (term ~ 150 days). Fetal intra-amniotic/intra-peritoneal injections of fluconazole or saline (controls) were administered 2 days after C. albicans exposure. Post mortem analyses for fungal burden, peripheral immune activation, neuroinflammation, and white matter/neuronal injury were performed to determine the effects of intra-amniotic C. albicans and fluconazole treatment. RESULTS Intra-amniotic exposure to C. albicans caused a severe systemic inflammatory response, illustrated by a robust increase of plasma interleukin-6 concentrations. Cerebrospinal fluid cultures were positive for C. albicans in the majority of the 3-day C. albicans-exposed animals whereas no positive cultures were present in the 5-day C. albicans-exposed and fluconazole-treated animals. Although C. albicans was not detected in the brain parenchyma, a neuroinflammatory response in the hippocampus and white matter was seen which was characterized by increased microglial and astrocyte activation. These neuroinflammatory changes were accompanied by structural white matter injury. Intra-amniotic fluconazole reduced fetal mortality but did not attenuate neuroinflammation and white matter injury. CONCLUSIONS Intra-amniotic C. albicans exposure provoked acute systemic and neuroinflammatory responses with concomitant white matter injury. Fluconazole treatment prevented systemic inflammation without attenuating cerebral inflammation and injury.
Collapse
Affiliation(s)
- Daan R M G Ophelders
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands. .,School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands.
| | - Ruth Gussenhoven
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands. .,School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands.
| | - Martin Lammens
- Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium.
| | - Benno Küsters
- Department of Pathology, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands.
| | - Matthew W Kemp
- School of Women's and Infants' Health, The University of Western Australia (M550), 35 Stirling Highway, Crawley, WA, 6009, Australia.
| | - John P Newnham
- School of Women's and Infants' Health, The University of Western Australia (M550), 35 Stirling Highway, Crawley, WA, 6009, Australia.
| | - Matthew S Payne
- School of Women's and Infants' Health, The University of Western Australia (M550), 35 Stirling Highway, Crawley, WA, 6009, Australia.
| | - Suhas G Kallapur
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45208, USA.
| | - Allan H Jobe
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45208, USA.
| | - Luc J Zimmermann
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands. .,School of Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands.
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands. .,School of Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands. .,School of Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands.
| | - Tim G A M Wolfs
- Department of Pediatrics, Maastricht University Medical Center, PO box 5800, Maastricht, 6202 AZ, The Netherlands. .,School of Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands.
| |
Collapse
|
36
|
van Tilborg E, Heijnen CJ, Benders MJ, van Bel F, Fleiss B, Gressens P, Nijboer CH. Impaired oligodendrocyte maturation in preterm infants: Potential therapeutic targets. Prog Neurobiol 2015; 136:28-49. [PMID: 26655283 DOI: 10.1016/j.pneurobio.2015.11.002] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 11/02/2015] [Accepted: 11/18/2015] [Indexed: 12/20/2022]
Abstract
Preterm birth is an evolving challenge in neonatal health care. Despite declining mortality rates among extremely premature neonates, morbidity rates remain very high. Currently, perinatal diffuse white matter injury (WMI) is the most commonly observed type of brain injury in preterm infants and has become an important research area. Diffuse WMI is associated with impaired cognitive, sensory and psychological functioning and is increasingly being recognized as a risk factor for autism-spectrum disorders, ADHD, and other psychological disturbances. No treatment options are currently available for diffuse WMI and the underlying pathophysiological mechanisms are far from being completely understood. Preterm birth is associated with maternal inflammation, perinatal infections and disrupted oxygen supply which can affect the cerebral microenvironment by causing activation of microglia, astrogliosis, excitotoxicity, and oxidative stress. This intricate interplay of events negatively influences oligodendrocyte development, causing arrested oligodendrocyte maturation or oligodendrocyte cell death, which ultimately results in myelination failure in the developing white matter. This review discusses the current state in perinatal WMI research, ranging from a clinical perspective to basic molecular pathophysiology. The complex regulation of oligodendrocyte development in healthy and pathological conditions is described, with a specific focus on signaling cascades that may play a role in WMI. Furthermore, emerging concepts in the field of WMI and issues regarding currently available animal models are put forward. Novel insights into the molecular mechanisms underlying impeded oligodendrocyte maturation in diffuse WMI may aid the development of novel treatment options which are desperately needed to improve the quality-of-life of preterm neonates.
Collapse
Affiliation(s)
- Erik van Tilborg
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manon J Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank van Bel
- Department of Neonatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bobbi Fleiss
- Inserm, Paris U1141, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS, Paris 1141, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Pierre Gressens
- Inserm, Paris U1141, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS, Paris 1141, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|