1
|
Sancerni T, Montel V, Dereumetz J, Cochon L, Coq JO, Bastide B, Canu MH. Enduring effects of acute prenatal ischemia in rat soleus muscle, and protective role of erythropoietin. J Muscle Res Cell Motil 2025; 46:23-34. [PMID: 39549147 DOI: 10.1007/s10974-024-09684-6] [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: 09/30/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
Motor disorders are considered to originate mainly from brain lesions. Placental dysfunction or maternal exposure to a persistently hypoxic environment is a major cause of further motor disorders such as cerebral palsy. Our main goal was to determine the long-term effects of mild intrauterine acute ischemic stress on rat soleus myofibres and whether erythropoietin treatment could prevent these changes. Rat embryos were subjected to ischemic stress at embryonic day E17. They then received an intraperitoneal erythropoietin injection at postnatal days 1-5. Soleus muscles were collected at postnatal day 28. Prenatal ischemic stress durably affected muscle structure, as indicated by the greater fiber cross-sectional area (+ 18%) and the greater number of mature vessels (i.e. vessels with mature endothelial cells) per myofibres (+ 43%), and muscle biochemistry, as shown by changes in signaling pathways involved in protein synthesis/degradation balance (-81% for 4EBP1; -58% for AKT) and Hif1α expression levels (+ 95%). Erythropoietin injection in ischemic pups had a weak protective effect: it increased muscle mass (+ 25% with respect to ischemic pups) and partially prevented the increase in muscle degradation pathways and mature vascularization, whereas it exacerbated the decrease in synthesis pathways. Hence, erythropoietin treatment after acute ischemic stress contributes to muscle adaptation to ischemic conditions.
Collapse
Affiliation(s)
- Tiphaine Sancerni
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France
| | - Valérie Montel
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France
| | - Julie Dereumetz
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France
| | - Laetitia Cochon
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France
| | - Jacques-Olivier Coq
- Institut des Sciences du Mouvement (ISM), Team 'Plasticité des Systèmes Nerveux et Musculaires', UMR 7287 CNRS, Aix-Marseille Université Faculté des Sports, Marseille Cedex 09, F-13288, France
| | - Bruno Bastide
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France
| | - Marie-Hélène Canu
- Univ. Lille, Univ Artois, Univ Littoral Côte d'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR7369, Lille, F-59000, France.
| |
Collapse
|
2
|
Vetrovoy O, Potapova S, Stratilov V, Tyulkova E. Comparative Analysis of the Effects of Maternal Hypoxia and Placental Ischemia on HIF1-Dependent Metabolism and the Glucocorticoid System in the Embryonic and Newborn Rat Brain. Int J Mol Sci 2024; 25:13342. [PMID: 39769106 PMCID: PMC11727977 DOI: 10.3390/ijms252413342] [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: 10/18/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Prenatal hypoxia, often accompanied by maternal glucocorticoid stress, can predispose offspring to neurological disorders in adulthood. If placental ischemia (PI) primarily reduces fetal oxygen supply, the maternal hypoxia (MH) model also elicits a pronounced fetal glucocorticoid exposure. Here, we compared MH and PI in rats to distinguish their unique and overlapping effects on embryonic and newborn brain development. We analyzed glucocorticoid transport into the developing brain, glucocorticoid receptor (GR) expression, and GR-dependent transcription, along with key enzymes regulating glucocorticoid metabolism in maternal (MP) and fetal placentas (FP) and in the brain. Additionally, we examined hypoxia-inducible factor 1-alpha (HIF1α) and its downstream genes, as well as glycolysis and the pentose phosphate pathway, both associated with the transport of substrates essential for glucocorticoid synthesis and degradation. Both MH and PI induced HIF1-dependent metabolic alterations, enhancing glycolysis and transiently disrupting redox homeostasis. However, only MH caused a maternal glucocorticoid surge that altered early fetal brain glucocorticoid responsiveness. Over time, these differences may lead to distinct long-term outcomes in neuronal structure and function. This work clarifies the individual contributions of hypoxic and glucocorticoid stresses to fetal brain development, suggesting that combining the MH and PI models could provide valuable insights for future investigations into the mechanisms underlying developmental brain pathologies, including non-heritable psychoneurological and neurodegenerative disorders.
Collapse
Affiliation(s)
- Oleg Vetrovoy
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Makarova emb. 6, 199034 Saint-Petersburg, Russia (V.S.)
| | | | | | | |
Collapse
|
3
|
Khalki H, Lacerda DC, Karoutchi C, Delcour M, Dupuis O, Kochmann M, Brezun J, Dupont E, Amin M, Darnaudéry M, Canu M, Barbe MF, Coq J. Early movement restriction impairs the development of sensorimotor integration, motor skills and memory in rats: Towards a preclinical model of developmental coordination disorder? Eur J Neurosci 2024; 60:6830-6850. [PMID: 39523702 PMCID: PMC11612839 DOI: 10.1111/ejn.16594] [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: 11/06/2023] [Revised: 07/04/2024] [Accepted: 07/30/2024] [Indexed: 11/16/2024]
Abstract
Children with neurodevelopmental disorders, such as developmental coordination disorder (DCD), exhibit gross to fine sensorimotor impairments, reduced physical activity and interactions with the environment and people. This disorder co-exists with cognitive deficits, executive dysfunctions and learning impairments. Previously, we demonstrated in rats that limited amounts and atypical patterns of movements and somatosensory feedback during early movement restriction manifested in adulthood as degraded postural and locomotor abilities, and musculoskeletal histopathology, including muscle atrophy, hyperexcitability within sensorimotor circuitry and maladaptive cortical plasticity, leading to functional disorganization of the primary somatosensory and motor cortices in the absence of cortical histopathology. In this study, we asked how this developmental sensorimotor restriction (SMR) started to impact the integration of multisensory information and the emergence of sensorimotor reflexes in rats. We also questioned the enduring impact of SMR on motor activities, pain and memory. SMR led to deficits in the emergence of swimming and sensorimotor reflexes, the development of pain and altered locomotor patterns and posture with toe-walking, adult motor performance and night spontaneous activity. In addition, SMR induced exploratory hyperactivity, short-term impairments in object-recognition tasks and long-term deficits in object-location tasks. SMR rats displayed minor alterations in histological features of the hippocampus, entorhinal, perirhinal and postrhinal cortices yet no obvious changes in the prefrontal cortex. Taken all together, these results show similarities with the symptoms observed in children with DCD, although further exploration seems required to postulate whether developmental SMR corresponds to a rat model of DCD.
Collapse
Affiliation(s)
- Hanane Khalki
- Aix Marseille UniversitéMarseilleFrance
- B2DRN, Polydisciplinary Faculty of Béni MellalSultan Moulay Slimane UniversityMorocco
| | - Diego Cabral Lacerda
- Aix Marseille UniversitéMarseilleFrance
- Post Graduate Program in NutritionFederal University of PernambucoRecifePernambucoBrazil
- Present address:
Studies in Nutrition and Phenotypic Plasticity Unit, Department of NutritionFederal University of PernambucoRecifeBrazil
| | - Corane Karoutchi
- Aix Marseille UniversitéMarseilleFrance
- Centre National de la Recherche Scientifique (CNRS)UMR7289 Institut de Neurosciences de la Timone (INT)MarseilleFrance
- Present address:
Centre d'Investigation Clinique (CIC 1407), Hôpital Cardiologique Louis PradelBronFrance
| | - Maxime Delcour
- Aix Marseille UniversitéMarseilleFrance
- Present address:
Cégep de Saint LaurentMontréalCanada
| | - Orlane Dupuis
- Univ Lille, Univ Artois, Univ Littoral Côte d'Opale, ULR 7369, URePSSS – Unité de Recherche Pluridisciplinaire Sport Santé SociétéLilleFrance
| | - Marine Kochmann
- Aix Marseille UniversitéMarseilleFrance
- Centre National de la Recherche Scientifique (CNRS)UMR7289 Institut de Neurosciences de la Timone (INT)MarseilleFrance
| | - Jean‐Michel Brezun
- Aix Marseille UniversitéMarseilleFrance
- Centre National de la Recherche Scientifique (CNRS), UMR7287 Institut des Sciences du Mouvement (ISM)MarseilleFrance
| | - Erwan Dupont
- Univ Lille, Univ Artois, Univ Littoral Côte d'Opale, ULR 7369, URePSSS – Unité de Recherche Pluridisciplinaire Sport Santé SociétéLilleFrance
| | - Mamta Amin
- Aging + Cardiovascular Discovery Center, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
| | - Muriel Darnaudéry
- Institut National de Recherche pour l'agriculture, l'Alimentation et l'Environnement (INRAE), UMR1286 NutriNeuroUniversité de BordeauxBordeauxFrance
| | - Marie‐Hélène Canu
- Univ Lille, Univ Artois, Univ Littoral Côte d'Opale, ULR 7369, URePSSS – Unité de Recherche Pluridisciplinaire Sport Santé SociétéLilleFrance
| | - Mary F. Barbe
- Aging + Cardiovascular Discovery Center, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPAUSA
| | - Jacques‐Olivier Coq
- Aix Marseille UniversitéMarseilleFrance
- Centre National de la Recherche Scientifique (CNRS)UMR7289 Institut de Neurosciences de la Timone (INT)MarseilleFrance
- Centre National de la Recherche Scientifique (CNRS), UMR7287 Institut des Sciences du Mouvement (ISM)MarseilleFrance
| |
Collapse
|
4
|
Ertürk E, Işık Ü, Şirin FB. Analysis of Serum VEGF, IGF-1, and HIF-1α Levels in ADHD. J Atten Disord 2024; 28:58-65. [PMID: 37700676 DOI: 10.1177/10870547231197211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
OBJECTIVE In recent years, it has been emphasized that various growth factors that affect neurogenesis may lead to ADHD. In this study, we aimed to investigate the role of VEGF, IGF-1, and HIF-1α growth factors in the etiopathogenesis of ADHD. METHOD Levels of VEGF, IGF-1, and HIF-1α were compared between 40 ADHD children and 40 healthy children, aged 7 to 13 years. RESULT VEGF, IGF-1, and HIF-1α levels did not significantly differ between the groups. There was a negative correlation between serum VEGF levels and the parent-rated T-DSM-IV-S (AD) subscale. There was a positive correlation between serum IGF-1 levels and the parent-rated T-DSM-IV-S (AD) subscale, and SDQ (ES) subscale. CONCLUSION Given our limitations and the fact that some of our findings differ from those of other studies, it is evident that this area requires additional research with larger samples.
Collapse
Affiliation(s)
- Emre Ertürk
- Süleyman Demirel University, Isparta, Turkey
| | | | | |
Collapse
|
5
|
Tsuji M, Tanaka N, Koike H, Sato Y, Shimoyama Y, Itoh A. Various Organ Damages in Rats with Fetal Growth Restriction and Their Slight Attenuation by Bifidobacterium breve Supplementation. Life (Basel) 2023; 13:2005. [PMID: 37895387 PMCID: PMC10607936 DOI: 10.3390/life13102005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/14/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Children with fetal growth restriction (FGR) and its resultant low birthweight (LBW) are at a higher risk of developing various health problems later in life, including renal diseases, metabolic syndrome, and sarcopenia. The mechanism through which LBW caused by intrauterine hypoperfusion leads to these health problems has not been properly investigated. Oral supplementation with probiotics is expected to reduce these risks in children. In the present study, rat pups born with FGR-LBW after mild intrauterine hypoperfusion were supplemented with either Bifidobacterium breve (B. breve) or a vehicle from postnatal day 1 (P1) to P21. Splanchnic organs and skeletal muscles were evaluated at six weeks of age. Compared with the sham group, the LBW-vehicle group presented significant changes as follows: overgrowth from infancy to childhood; lighter weight of the liver, kidneys, and gastrocnemius and plantaris muscles; reduced height of villi in the ileum; and increased depth of crypts in the jejunum. Some of these changes were milder in the LBW-B.breve group. In conclusion, this rat model could be useful for investigating the mechanisms of how FGR-LBW leads to future health problems and for developing interventions for these problems. Supplementation with B. breve in early life may modestly attenuate these problems.
Collapse
Affiliation(s)
- Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women’s University, Kyoto 605-8501, Japan
| | - Nao Tanaka
- Department of Food and Nutrition, Kyoto Women’s University, Kyoto 605-8501, Japan
| | - Hitomi Koike
- Department of Food and Nutrition, Kyoto Women’s University, Kyoto 605-8501, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya 466-8560, Japan;
| | - Yoshie Shimoyama
- Department of Pathology, Nagoya University Hospital, Nagoya 466-8560, Japan
| | - Ayaka Itoh
- Department of Food and Nutrition, Kyoto Women’s University, Kyoto 605-8501, Japan
| |
Collapse
|
6
|
Tsuji M, Mukai T, Sato Y, Azuma Y, Yamamoto S, Cayetanot F, Bodineau L, Onoda A, Nagamura-Inoue T, Coq JO. Umbilical cord-derived mesenchymal stromal cell therapy to prevent the development of neurodevelopmental disorders related to low birth weight. Sci Rep 2023; 13:3841. [PMID: 36882440 PMCID: PMC9992354 DOI: 10.1038/s41598-023-30817-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/01/2023] [Indexed: 03/09/2023] Open
Abstract
Low birth weight (LBW) increases the risk of neurodevelopmental disorders (NDDs) such as attention-deficit/hyperactive disorder and autism spectrum disorder, as well as cerebral palsy, for which no prophylactic measure exists. Neuroinflammation in fetuses and neonates plays a major pathogenic role in NDDs. Meanwhile, umbilical cord-derived mesenchymal stromal cells (UC-MSCs) exhibit immunomodulatory properties. Therefore, we hypothesized that systemic administration of UC-MSCs in the early postnatal period may attenuate neuroinflammation and thereby prevent the emergence of NDDs. The LBW pups born to dams subjected to mild intrauterine hypoperfusion exhibited a significantly lesser decrease in the monosynaptic response with increased frequency of stimulation to the spinal cord preparation from postnatal day 4 (P4) to P6, suggesting hyperexcitability, which was improved by intravenous administration of human UC-MSCs (1 × 105 cells) on P1. Three-chamber sociability tests at adolescence revealed that only LBW males exhibited disturbed sociability, which tended to be ameliorated by UC-MSC treatment. Other parameters, including those determined via open-field tests, were not significantly improved by UC-MSC treatment. Serum or cerebrospinal fluid levels of pro-inflammatory cytokines were not elevated in the LBW pups, and UC-MSC treatment did not decrease these levels. In conclusion, although UC-MSC treatment prevents hyperexcitability in LBW pups, beneficial effects for NDDs are marginal.
Collapse
Affiliation(s)
- Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshi-cho, Imakumano, Higashiyama-ku, Kyoto, 605-8501, Japan.
| | - Takeo Mukai
- Department of Cell Processing and Transfusion, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Yasue Azuma
- Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshi-cho, Imakumano, Higashiyama-ku, Kyoto, 605-8501, Japan
| | - Saki Yamamoto
- Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshi-cho, Imakumano, Higashiyama-ku, Kyoto, 605-8501, Japan
| | - Florence Cayetanot
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Laurence Bodineau
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Atsuto Onoda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jacques-Olivier Coq
- Centre National de la Recherche Scientifique (CNRS), Institut des Sciences du Mouvement (ISM) UMR7287, Aix Marseille Université, 163 avenue de Luminy, CC 910, 13288, Marseille Cedex 09, France.
| |
Collapse
|
7
|
Ortiz M, Loidl F, Vázquez‐Borsetti P. Transition to extrauterine life and the modeling of perinatal asphyxia in rats. WIREs Mech Dis 2022; 14:e1568. [DOI: 10.1002/wsbm.1568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 05/11/2022] [Accepted: 05/14/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Mauro Ortiz
- Universidad de Buenos Aires Buenos Aires Argentina
| | - Fabián Loidl
- Consejo Nacional de Investigaciones Científicas y Técnicas Buenos Aires Argentina
| | | |
Collapse
|
8
|
Itoh A, Tanaka N, Fukunaga S, Nakano-Doi A, Matsuyama T, Nakagomi T, Tsuji M. Bifidobacterium breve during infancy attenuates mobility in low birthweight rats. Pediatr Int 2022; 64:e15209. [PMID: 35938576 DOI: 10.1111/ped.15209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/25/2022] [Accepted: 04/05/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Children with low birthweight (LBW) have a higher risk for developing attention-deficit/hyperactivity disorder, for which no prophylactic measure exists. The gut microbiota in infants with LBW is different from that in infants with normal birthweight and is associated with attention-deficit/hyperactivity disorder. Oral supplementation with Bifidobacterium has several health benefits, such as suppressing inflammation. METHODS We examined the effect of gavage supplementation with Bifidobacterium breve M-16V from postnatal days 1-21 in a rat model of intrauterine hypoperfusion. RESULTS The open-field test at 5 weeks of age (equivalent to human pubertal age) showed that rats in the LBW-vehicle group were marginally hyperactive compared with rats in the sham group, while rats in the LBW-B.breve group were significantly hypoactive compared with rats in the LBW-vehicle group. The gut microbiota in the LBW-vehicle group exhibited a profile significantly different from that in the sham group, whereas the gut microbiota in the LBW-B.breve group did not exhibit a significant difference from that in the sham group. Anatomical/histological evaluation at 6 weeks of age demonstrated that the brain weight and the cerebral areas on coronal sections were reduced in the LBW groups compared with the sham group. Probiotic supplementation did not ameliorate these morphological brain anomalies in LBW animals. The percentage of Iba-1+ cells in the brain was not different among the LBW-B.breve, LBW-vehicle, and sham groups. CONCLUSION Bifidobacterium breve supplementation during early life is suggested to have the potential to help children with LBW attenuate hypermobility in adolescence.
Collapse
Affiliation(s)
- Ayaka Itoh
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Nao Tanaka
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Sachiko Fukunaga
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Akiko Nakano-Doi
- Laboratory of Neurogenesis and CNS Repair, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan.,Department of Therapeutic Progress in Brain Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan
| | - Tomohiro Matsuyama
- Department of Therapeutic Progress in Brain Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan
| | - Takayuki Nakagomi
- Laboratory of Neurogenesis and CNS Repair, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan.,Department of Therapeutic Progress in Brain Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| |
Collapse
|
9
|
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
|
10
|
Yin S, Meng Y, Liu C, Wang Y. MIUH Inhibits the Hippocampal Neuron Growth in Fetal Rat by Affecting the PTEN Pathway. Neurochem Res 2021; 46:2046-2055. [PMID: 34003417 DOI: 10.1007/s11064-021-03342-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 04/09/2021] [Accepted: 05/12/2021] [Indexed: 11/29/2022]
Abstract
Mild intrauterine hypoperfusion (MIUH) can induce placental dysfunction and lead to long-term changes during the process of brain development. A better understanding of the mechanism of MIUH will help in the development of new neuroprotective strategies for the placental chamber. To better understand the mechanism of the effect of MIUH on the neural development of offspring, we constructed a model of MIUH in pregnant rats. The proliferation, apoptosis, and autophagy of hippocampal neurons in fetal rats were studied via flow cytometry, immunofluorescence staining, JC-1 staining, western blotting, and real-time polymerase chain reaction at different time points (6, 24, 48, and 72 h). The results showed that MIUH significantly inhibited the proliferation of hippocampal neurons and promoted their apoptosis and autophagy. Simultaneously, MIUH could promote PTEN expression and affect the PTEN signaling pathway. bpV, an inhibitor of PTEN, could restore the inhibition of hippocampal nerve cell growth caused by MIUH. MIUH may inhibit neuronal proliferation and promote neuronal apoptosis and autophagy by regulating the PTEN signaling pathway.
Collapse
Affiliation(s)
- Shaowei Yin
- Department of Gynaecology and Obstetrics, Shengjing Hospital of China Medical University, China Medical University, Shenyang, 110004, China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Shenyang, 110004, China
| | - Yilin Meng
- Department of Gynaecology and Obstetrics, Shengjing Hospital of China Medical University, China Medical University, Shenyang, 110004, China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Shenyang, 110004, China
| | - Caixia Liu
- Department of Gynaecology and Obstetrics, Shengjing Hospital of China Medical University, China Medical University, Shenyang, 110004, China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Shenyang, 110004, China
| | - Yuan Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
11
|
Spinillo A, Dominoni M, Caporali C, Olivieri I, La Piana R, Longo S, Cesari S, Fiandrino G, Orcesi S, Gardella B. Placental Histological Features and Neurodevelopmental Outcomes at Two Years in Very-Low-Birth-Weight Infants. Pediatr Neurol 2021; 120:63-70. [PMID: 34022751 DOI: 10.1016/j.pediatrneurol.2021.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/11/2021] [Accepted: 04/11/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND We evaluated the rates of placental pathologic lesions and their relationship with two-year neurodevelopmental outcomes in very-low-birth-weight (VLBW) infants. METHODS This is a cohort observational study comprising 595 VLBW infants during 2007 to 2015. Neurodevelopmental assessment was carried out at 24 months corrected age. RESULTS In univariate analysis the rates of survival with normal neurodevelopmental outcomes were lower in pregnancies with severe histologic chorioamnionitis (38 of 43, 88.4% when compared with 305 of 450, 67.8%), severe maternal vascular malperfusion (MVM) (17 of 37, 45.9% when compared with 326/492, 66.3%), and intravillous hemorrhage (37 of 82, 45.1% when compared with 306 of 449, 68.1%). In logistic models, severe MVM (adjusted odds ratio [adj. OR] = 0.45, 95% confidence interval [CI] = 0.22 to 0.92), severe fetal vascular malperfusion (FVM) (adj. OR = 0.46, 95% CI = 0.22 to 0.45), and intravillous hemorrhage (adj. OR = 0.38, 95% CI = 0.22 to 0.62) were associated with lower rates of infant survival with normal neurodevelopmental outcome. FVM (adj. OR = 0.46, 95% CI = 0.21 to 0.97) and intravillous hemorrhage (adj. OR = 0.37, 95% CI = 0.22 to 0.62) were also the only placental lesions that were independent predictors of a lower rate of intact survival in stepwise analysis for prognostic factors of the entire cohort. CONCLUSIONS Placental pathologic findings such as severe MVM, FVM, and intravillous hemorrhage are significant predictors of neonatal survival and subsequent adverse neurodevelopmental outcomes. Data on the placental pathology could be useful in the neurodevelopmental follow-up of VLBW infants.
Collapse
Affiliation(s)
- Arsenio Spinillo
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy; Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Mattia Dominoni
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Camilla Caporali
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Ivana Olivieri
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Roberta La Piana
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Stefania Longo
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Stefania Cesari
- Department of Pathology, IRCCS Foundation Policlinico San Matteo, Pavia, Italy
| | - Giacomo Fiandrino
- Department of Pathology, IRCCS Foundation Policlinico San Matteo, Pavia, Italy
| | - Simona Orcesi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Barbara Gardella
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo and University of Pavia, Pavia, Italy; Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy.
| |
Collapse
|
12
|
Ahn SY, Jie H, Jung WB, Jeong JH, Ko S, Im GH, Park WS, Lee JH, Chang YS, Chung S. Stem cell restores thalamocortical plasticity to rescue cognitive deficit in neonatal intraventricular hemorrhage. Exp Neurol 2021; 342:113736. [PMID: 33945790 DOI: 10.1016/j.expneurol.2021.113736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/07/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
Severe neonatal intraventricular hemorrhage (IVH) patients incur long-term neurologic deficits such as cognitive disabilities. Recently, the intraventricular transplantation of allogeneic human umbilical cord blood-derived mesenchymal stem cells (MSCs) has drawn attention as a therapeutic potential to treat severe IVH. However, its pathological synaptic mechanism is still elusive. We here demonstrated that the integration of the somatosensory input was significantly distorted by suppressing feed-forward inhibition (FFI) at the thalamocortical (TC) inputs in the barrel cortices of neonatal rats with IVH by using BOLD-fMRI signal and brain slice patch-clamp technique. This is induced by the suppression of Hebbian plasticity via an increase in tumor necrosis factor-α expression during the critical period, which can be effectively reversed by the transplantation of MSCs. Furthermore, we showed that MSC transplantation successfully rescued IVH-induced learning deficits in the sensory-guided decision-making in correlation with TC FFI in the layer 4 barrel cortex.
Collapse
Affiliation(s)
- So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Hyesoo Jie
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Won-Beom Jung
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 86364, Republic of Korea; Department of Global Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji-Hyun Jeong
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sukjin Ko
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Geun Ho Im
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 86364, Republic of Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Jung Hee Lee
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 86364, Republic of Korea; Department of Global Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea; Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea.
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea.
| | - Seungsoo Chung
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
13
|
Pla L, Illa M, Loreiro C, Lopez MC, Vázquez-Aristizabal P, Kühne BA, Barenys M, Eixarch E, Gratacós E. Structural Brain Changes during the Neonatal Period in a Rabbit Model of Intrauterine Growth Restriction. Dev Neurosci 2021; 42:217-229. [PMID: 33677448 DOI: 10.1159/000512948] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) is associated with abnormal neurodevelopment, but the associated structural brain changes are poorly documented. The aim of this study was to describe in an animal model the brain changes at the cellular level in the gray and white matter induced by IUGR during the neonatal period. METHODS The IUGR model was surgically induced in pregnant rabbits by ligating 40-50% of the uteroplacental vessels in 1 horn, whereas the uteroplacental vessels of the contralateral horn were not ligated. After 5 days, IUGR animals from the ligated horn and controls from the nonligated were delivered. On the day of delivery, perinatal data and placentas were collected. On postnatal day 1, functional changes were first evaluated, and thereafter, neuronal arborization in the frontal cortex and density of pre-oligodendrocytes, astrocytes, and microglia in the corpus callosum were evaluated. RESULTS Higher stillbirth in IUGR fetuses together with a reduced birth weight as compared to controls was evidenced. IUGR animals showed poorer functional results, an altered neuronal arborization pattern, and a decrease in the pre-oligodendrocytes, with no differences in microglia and astrocyte densities. CONCLUSIONS Overall, in the rabbit model used, IUGR is related to functional and brain changes evidenced already at birth, including changes in the neuronal arborization and abnormal oligodendrocyte maturation.
Collapse
Affiliation(s)
- Laura Pla
- BCNatal
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain
| | - Miriam Illa
- BCNatal
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain, .,Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain,
| | - Carla Loreiro
- BCNatal
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mari Carmen Lopez
- BCNatal
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain
| | - Paula Vázquez-Aristizabal
- BCNatal
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain
| | - Britta Anna Kühne
- BCNatal
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain.,GRET, INSA-UB and Toxicology Unit, Pharmacology, Toxicology and Therapeutical Chemistry Department, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Marta Barenys
- GRET, INSA-UB and Toxicology Unit, Pharmacology, Toxicology and Therapeutical Chemistry Department, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Elisenda Eixarch
- BCNatal
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Eduard Gratacós
- BCNatal
- Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, Spain.,Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Center for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| |
Collapse
|
14
|
Ogawa Y, Tanaka E, Sato Y, Tsuji M. Brain damage caused by neonatal hypoxia-ischemia and the effects of hypothermia in severe combined immunodeficient (SCID) mice. Exp Neurol 2020; 337:113577. [PMID: 33359474 DOI: 10.1016/j.expneurol.2020.113577] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 10/22/2022]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a major cause of brain damage in newborns. Although therapeutic hypothermia has been shown to be neuroprotective against neonatal HIE in clinical trials, its effect is not satisfactory. Cell-based therapies have attracted much attention as novel treatments for HIE. Preclinical studies on a variety of human cell transplantation methods have been performed in immunodeficient/immunosuppressed animals, such as severe combined immunodeficient (SCID) mice, which lack functional T and B lymphocytes. The detailed characteristics of neonatal HIE in SCID mice, however, have not been delineated. In preclinical studies, novel therapies for neonatal HIE should be evaluated in combination with hypothermia, which has become a standard treatment for neonatal HIE. However, the effects of hypothermia in SCID mice have not been delineated. In the present study, we compared neonatal hypoxic-ischemic (HI) brain damage in SCID mice and wild-type mice treated with or without hypothermia. Male and female mouse pups were subjected to HI insult induced by unilateral common carotid artery ligation combined with systemic hypoxia on postnatal day 12. In the first 4 h after HI insult, body temperature was maintained at 36 °C for the normothermia groups or 32 °C for the hypothermia groups. The severity of brain damage in SCID mice did not differ from that in wild-type mice based on most evaluations, i.e., cerebral blood flow, hemiparesis, muscle strength, spontaneous activity, cerebral hemispheric volume, neuropathological injury, and serum cytokine levels, although spleen weight, brain weight, leukocyte counts and the levels of some cytokines in the peripheral blood were different between genotypes. The effects of hypothermia in SCID mice were comparable to those in wild-type mice based on most evaluations. Taken together, these findings indicate that SCID mice can be used as an appropriate preclinical model for cell therapies for neonatal HIE.
Collapse
Affiliation(s)
- Yuko Ogawa
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Japan; Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan.
| | - Emi Tanaka
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Japan; Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan.
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal - Neonatal Care, Nagoya University Hospital, Nagoya, Japan.
| | - Masahiro Tsuji
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Japan; Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan.
| |
Collapse
|
15
|
Choi MS, Chung YY, Kim DJ, Kim ST, Jun YH. Immunoreactivity of MAPK Signaling in a Rat Model of Intrauterine Growth Retardation Induced by Uterine Artery Ligation. In Vivo 2020; 34:649-657. [PMID: 32111765 DOI: 10.21873/invivo.11819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 12/21/2019] [Accepted: 01/05/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Intrauterine growth retardation (IUGR) causes very low birth weight and is related to the morbidity and mortality of the newborn. In our previous study, expression of brain-derived neurotrophic factor (BDNF) was found reduced in the cerebral cortex and dentate gyrus of fetuses with IUGR. BDNF protected cortical neurons against hypoxic injury via activation of the extracellular signal-related kinase (ERK) pathway. The aim of the current study was to observe the immunoreactivity of ERK in mature neurons and proliferating cells. MATERIALS AND METHODS Uterine artery ligation was performed at 17 days of gestation (dg). Rat fetuses were obtained at 21 dg using cesarean section. Fetuses were designated either to the growth retardation (GR) group when removed from the horn with uterine artery ligation, or to the control group when removed from the other horn with the untied artery. Immunohistochemistry was performed with primary antibodies on paraffin-embedded forebrain sections. RESULTS The density and proportion of cells expressing PCNA, ERK, and phosphate ERK in the subventricular zone (SVZ) was not different between the control and GR group. The density and proportion of NeuN- and phosphate ERK-positive cells in the cerebral parietal cortex was lower in the GR group, compared to the control group. CONCLUSION Although IUGR had no effect on the proliferation of cells in the SVZ, it reduced neuronal survival in the cerebral parietal cortex, which was associated with the decrease of pERK-positive cell density and proportion in the cerebral cortex.
Collapse
Affiliation(s)
- Min Seon Choi
- Department of Pediatrics, Chosun University Hospital, Gwang-ju, Republic of Korea
| | - Yoon Young Chung
- Department of Anatomy, School of Medicine, Chosun University, Gwang-ju, Republic of Korea
| | - Dong-Joon Kim
- Department of Anesthesiology and Pain Medicine, Chosun University Hospital, Gwang-ju, Republic of Korea
| | - Seong Taeck Kim
- Department of Ophthalmology, Chosun University Hospital, Gwang-ju, Republic of Korea
| | - Yong Hyun Jun
- Department of Anatomy, School of Medicine, Chosun University, Gwang-ju, Republic of Korea
| |
Collapse
|
16
|
From cerebral palsy to developmental coordination disorder: Development of preclinical rat models corresponding to recent epidemiological changes. Ann Phys Rehabil Med 2020; 63:422-430. [DOI: 10.1016/j.rehab.2019.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 02/05/2023]
|
17
|
Hamdy N, Eide S, Sun HS, Feng ZP. Animal models for neonatal brain injury induced by hypoxic ischemic conditions in rodents. Exp Neurol 2020; 334:113457. [PMID: 32889009 DOI: 10.1016/j.expneurol.2020.113457] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023]
Abstract
Neonatal hypoxia-ischemia and resulting encephalopathies are of significant concern. Intrapartum asphyxia is a leading cause of neonatal death globally. Among surviving infants, there remains a high incidence of hypoxic-ischemic encephalopathy due to neonatal hypoxic-ischemic brain injury, manifesting as mild conditions including attention deficit hyperactivity disorder, and debilitating disorders such as cerebral palsy. Various animal models of neonatal hypoxic brain injury have been implemented to explore cellular and molecular mechanisms, assess the potential of novel therapeutic strategies, and characterize the functional and behavioural correlates of injury. Each of the animal models has individual advantages and limitations. The present review looks at several widely-used and alternative rodent models of neonatal hypoxia and hypoxia-ischemia; it highlights their strengths and limitations, and their potential for continued and improved use.
Collapse
Affiliation(s)
- Nancy Hamdy
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sarah Eide
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
18
|
Kitase Y, Sato Y, Arai S, Onoda A, Ueda K, Go S, Mimatsu H, Jabary M, Suzuki T, Ito M, Saito A, Hirakawa A, Mukai T, Nagamura-Inoue T, Takahashi Y, Tsuji M, Hayakawa M. Establishment of a Novel Fetal Growth Restriction Model and Development of a Stem-Cell Therapy Using Umbilical Cord-Derived Mesenchymal Stromal Cells. Front Cell Neurosci 2020; 14:212. [PMID: 32848614 PMCID: PMC7401876 DOI: 10.3389/fncel.2020.00212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 06/16/2020] [Indexed: 11/13/2022] Open
Abstract
Fetal growth restriction (FGR) is a major complication of prenatal ischemic/hypoxic exposure and affects 5%-10% of pregnancies. It causes various disorders, including neurodevelopmental disabilities due to chronic hypoxia, circulatory failure, and malnutrition via the placenta, and there is no established treatment. Therefore, the development of treatments is an urgent task. We aimed to develop a new FGR rat model with a gradual restrictive load of uterus/placental blood flow and to evaluate the treatment effect of the administration of umbilical cord-derived mesenchymal stromal cells (UC-MSCs). To create the FGR rat model, we used ameroid constrictors that had titanium on the outer wall and were composed of C-shaped casein with a notch and center hole inside that gradually narrowed upon absorbing water. The ameroid constrictors were attached to bilateral ovarian/uterine arteries on the 17th day of pregnancy to induce chronic mild ischemia, which led to FGR with over 20% bodyweight reduction. After the intravenous administration of 1 × 105 UC-MSCs, we confirmed a significant improvement in the UC-MSC group in a negative geotaxis test at 1 week after birth and a rotarod treadmill test at 5 months old. In the immunobiological evaluation, the total number of neurons counted via the stereological counting method was significantly higher in the UC-MSC group than in the vehicle-treated group. These results indicate that the UC-MSCs exerted a treatment effect for neurological impairment in the FGR rats.
Collapse
Affiliation(s)
- Yuma Kitase
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.,Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Sakiko Arai
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.,Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsuto Onoda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.,Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi, Japan
| | - Kazuto Ueda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Shoji Go
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Haruka Mimatsu
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.,Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mahboba Jabary
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.,Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshihiko Suzuki
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Miharu Ito
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Akiko Saito
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Akihiro Hirakawa
- Clinical Research Center, Division of Biostatistics and Data Science, Medical and Dental University, Tokyo, Japan
| | - Takeo Mukai
- Department of Cell Processing and Transfusion, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Tsuji
- Department of Food and Nutrition, Faculty of Home Economics, Kyoto Women's University, Kyoto, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
19
|
Tsuji M, Ohshima M, Yamamoto Y, Saito S, Hattori Y, Tanaka E, Taguchi A, Ihara M, Ogawa Y. Cilostazol, a Phosphodiesterase 3 Inhibitor, Moderately Attenuates Behaviors Depending on Sex in the Ts65Dn Mouse Model of Down Syndrome. Front Aging Neurosci 2020; 12:106. [PMID: 32372946 PMCID: PMC7186592 DOI: 10.3389/fnagi.2020.00106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/30/2020] [Indexed: 11/16/2022] Open
Abstract
People with Down syndrome, which is a trisomy of chromosome 21, exhibit intellectual disability from infancy and neuropathology similar to Alzheimer’s disease, such as amyloid plaques, from an early age. Recently, we showed that cilostazol, a selective inhibitor of phosphodiesterase (PDE) 3, promotes the clearance of amyloid β and rescues cognitive deficits in a mouse model of Alzheimer’s disease. The objective of the present study was to examine whether cilostazol improves behaviors in the most widely used animal model of Down syndrome, i.e., Ts65Dn mice. Mice were supplemented with cilostazol from the fetal period until young adulthood. Supplementation significantly ameliorated novel-object recognition in Ts65Dn females and partially ameliorated sensorimotor function as determined by the rotarod test in Ts65Dn females and hyperactive locomotion in Ts65Dn males. Cilostazol supplementation significantly shortened swimming distance in Ts65Dn males in the Morris water maze test, suggesting that the drug improved cognitive function, although it did not shorten swimming duration, which was due to decreased swimming speed. Thus, this study suggests that early supplementation with cilostazol partially rescues behavioral abnormalities seen in Down syndrome and indicates that the effects are sex-dependent.
Collapse
Affiliation(s)
- Masahiro Tsuji
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Makiko Ohshima
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yumi Yamamoto
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Satoshi Saito
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan.,Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yorito Hattori
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Emi Tanaka
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yuko Ogawa
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| |
Collapse
|
20
|
Yin SW, Wang Y, Meng YL, Liu CX. Effects of mild intrauterine hypoperfusion in the second trimester on memory and learning function in rat offspring. Neural Regen Res 2020; 15:2082-2088. [PMID: 32394966 PMCID: PMC7716030 DOI: 10.4103/1673-5374.282268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Mild intrauterine hypoperfusion (MIUH) is a serious pathological event that affects the growth and development of fetuses and offspring. MIUH can lead to growth restriction, low birth weight, neurodevelopmental disorders, and other adverse clinical outcomes. To study the effects of MIUH on learning and memory function in offspring, a model of MIUH was established by placing a coil (length 2.5 mm, diameter 0.24 mm) on the uterine artery and ovarian uterine artery of Sprague-Dawley rats in the second trimester of pregnancy (day 17). Next, 120 mg/kg lithium chloride (the MIUH + Li group) or normal saline (the MIUH group) was injected intraperitoneally into these rats. In addition, 120 mg/kg lithium chloride (the Li group) or normal saline (the SHAM group) was injected intraperitoneally into pregnant rats without coil placement. The Morris water maze was used to detect changes in learning and memory ability in the offspring at 4 weeks after birth. In the MIUH group, the escape latency and journey length before reaching the platform were both increased, and the number of times that the platform was crossed and the activity time in the target quadrant within 90 seconds were both decreased compared with the SHAM group. Immunofluorescence double staining and western blot assays demonstrated that hippocampal nestin and Ki67 (both cell-proliferation-related proteins) expression was significantly downregulated in the MIUH group compared with the SHAM group. Furthermore, western blot assays were conducted to investigate changes in related signaling pathway proteins in the brains of offspring rats, and revealed that glycogen synthase kinase 3β (GSK3β) expression was upregulated and β-catenin expression was downregulated in the MIUH group compared with the SHAM group. In addition, compared with the MIUH group, the expression levels of p-GSK3β and β-catenin were upregulated in the MIUH + Li group. These results suggest that MIUH may affect learning and memory function in rat offspring by regulating the GSK3β signaling pathway. The experimental procedures were approved by Animal Ethics Committee of Shengjing Hospital of China Medical University (approval No. 2018PS07K) in June 2018.
Collapse
Affiliation(s)
- Shao-Wei Yin
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Shenyang, Liaoning Province, China
| | - Yuan Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yi-Lin Meng
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Shenyang, Liaoning Province, China
| | - Cai-Xia Liu
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Shenyang, Liaoning Province, China
| |
Collapse
|
21
|
Pregnolato S, Chakkarapani E, Isles AR, Luyt K. Glutamate Transport and Preterm Brain Injury. Front Physiol 2019; 10:417. [PMID: 31068830 PMCID: PMC6491644 DOI: 10.3389/fphys.2019.00417] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/27/2019] [Indexed: 12/19/2022] Open
Abstract
Preterm birth complications are the leading cause of child death worldwide and a top global health priority. Among the survivors, the risk of life-long disabilities is high, including cerebral palsy and impairment of movement, cognition, and behavior. Understanding the molecular mechanisms of preterm brain injuries is at the core of future healthcare improvements. Glutamate excitotoxicity is a key mechanism in preterm brain injury, whereby the accumulation of extracellular glutamate damages the delicate immature oligodendrocytes and neurons, leading to the typical patterns of injury seen in the periventricular white matter. Glutamate excitotoxicity is thought to be induced by an interaction between environmental triggers of injury in the perinatal period, particularly cerebral hypoxia-ischemia and infection/inflammation, and developmental and genetic vulnerabilities. To avoid extracellular build-up of glutamate, the brain relies on rapid uptake by sodium-dependent glutamate transporters. Astrocytic excitatory amino acid transporter 2 (EAAT2) is responsible for up to 95% of glutamate clearance, and several lines of evidence suggest that it is essential for brain functioning. While in the adult EAAT2 is predominantly expressed by astrocytes, EAAT2 is transiently upregulated in the immature oligodendrocytes and selected neuronal populations during mid-late gestation, at the peak time for preterm brain injury. This developmental upregulation may interact with perinatal hypoxia-ischemia and infection/inflammation and contribute to the selective vulnerability of the immature oligodendrocytes and neurons in the preterm brain. Disruption of EAAT2 may involve not only altered expression but also impaired function with reversal of transport direction. Importantly, elevated EAAT2 levels have been found in the reactive astrocytes and macrophages of human infant post-mortem brains with severe white matter injury (cystic periventricular leukomalacia), potentially suggesting an adaptive mechanism against excitotoxicity. Interestingly, EAAT2 is suppressed in animal models of acute hypoxic-ischemic brain injury at term, pointing to an important and complex role in newborn brain injuries. Enhancement of EAAT2 expression and transport function is gathering attention as a potential therapeutic approach for a variety of adult disorders and awaits exploration in the context of the preterm brain injuries.
Collapse
Affiliation(s)
- Silvia Pregnolato
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Elavazhagan Chakkarapani
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Anthony R Isles
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Karen Luyt
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
22
|
Tadalafil treatment in mice for preeclampsia with fetal growth restriction has neuro-benefic effects in offspring through modulating prenatal hypoxic conditions. Sci Rep 2019; 9:234. [PMID: 30659198 PMCID: PMC6338749 DOI: 10.1038/s41598-018-36084-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 11/14/2018] [Indexed: 01/08/2023] Open
Abstract
We have demonstrated that tadalafil facilitates fetal growth in mice with L-NG-nitroarginine methyl ester (L-NAME)-induced preeclampsia (PE) with fetal growth restriction (FGR). Tadalafil is a selective phosphodiesterase 5 inhibitor that dilates the maternal blood sinuses in the placenta, thereby facilitating the growth of the fetus. The purpose of this study was to investigate the effects of tadalafil treatment for PE and FGR on the developing brain in FGR offspring using an L-NAME-induced mouse model of PE with FGR. A control group of dams received carboxymethylcellulose (CMC). L-NAME-treated groups received L-NAME dissolved in CMC from 11 days post coitum (d.p.c.). The L-NAME-treated dams were divided into two subgroups 14 d.p.c. One subgroup continued to receive L-NAME. The other subgroup received L-NAME with tadalafil suspended in CMC. Tadalafil treatment for PE with FGR reduced the expression of hypoxia-inducible factor-2α in the placenta and in the brain of the FGR fetus. Moreover, tadalafil treatment in utero shows improved synaptogenesis and myelination in FGR offspring on postnatal day 15 (P15) and P30. These results suggest that tadalafil treatment for PE with FGR not only facilitates fetal growth, but also has neuroprotective effects on the developing brain of FGR offspring through modulating prenatal hypoxic conditions.
Collapse
|
23
|
Paz Levy D, Wainstock T, Sheiner E, Sergienko R, Landau D, Walfisch A. Maternal recurrent pregnancy loss is associated with an increased risk for long-term neurological morbidity in offspring. Dev Med Child Neurol 2019; 61:91-97. [PMID: 30058166 DOI: 10.1111/dmcn.13976] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2018] [Indexed: 12/24/2022]
Abstract
AIM To determine whether maternal history of recurrent pregnancy loss (RPL) impacts on the long-term neurological health of the offspring. METHOD A population-based cohort analysis was performed, including all singletons born between 1991 and 2014 at a single regional tertiary centre. Neurological morbidity up to age 18 years (including cognitive, motor, and psychiatric disorders) in children born to mothers with and without a history of RPL was compared. Cumulative neurological morbidity incidence was compared with survival curves and a Weibull multivariable survival model to control for follow-up time and relevant confounders. RESULTS A total of 242 187 deliveries met the inclusion criteria during the study period, 5% (n=12 182) of which were in mothers with RPL. Epilepsy and developmental disorders were significantly more common in the group with RPL (0.95 vs 0.74/1000 person-years [p=0.009] and 0.22 vs 0.09/1000 person-years [p<0.001]). The survival curves demonstrated significantly higher cumulative incidences of epilepsy and developmental disorders in the group with RPL. The multivariable model exhibited an independent association between maternal RPL and childhood epilepsy (adjusted hazard ratio 1.23; 95% confidence interval 1.01-1.50) and developmental disorders in the offspring (adjusted hazard ratio 2.41; 95% confidence interval 1.60-3.64). INTERPRETATION A history of maternal RPL appears to be independently associated with long-term neurological morbidity of the offspring. WHAT THIS PAPER ADDS Hospitalizations for epilepsy and developmental disorders are significantly more common among children of mothers with a history of recurrent pregnancy loss.
Collapse
Affiliation(s)
- Dorit Paz Levy
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tamar Wainstock
- Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eyal Sheiner
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ruslan Sergienko
- Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Daniella Landau
- Department of Neonatology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Asnat Walfisch
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
24
|
Wright JL, Chu HX, Kagan BJ, Ermine CM, Kauhausen JA, Parish CL, Sobey CG, Thompson LH. Local Injection of Endothelin-1 in the Early Neonatal Rat Brain Models Ischemic Damage Associated with Motor Impairment and Diffuse Loss in Brain Volume. Neuroscience 2018; 393:110-122. [PMID: 30300704 DOI: 10.1016/j.neuroscience.2018.09.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/23/2018] [Accepted: 09/28/2018] [Indexed: 11/16/2022]
Abstract
Cerebral palsy is an irreversible movement disorder resulting from cerebral damage sustained during prenatal or neonatal brain development. As survival outcomes for preterm injury improve, there is increasing need to model ischemic injury at earlier neonatal time-points to better understand the subsequent pathological consequences. Here we demonstrate a novel neonatal ischemic model using focal administration of the potent vasoconstrictor peptide, endothelin-1 (ET-1), in newborn rats. The functional and histopathological outcomes compare favourably to those reported following the widely used hypoxic ischemia (HI) model. These include a robust motor deficit sustained into adulthood and recapitulation of hallmark features of preterm human brain injury, including atrophy of subcortical white matter and periventricular fiber bundles. Compared to procedures involving carotid artery manipulation and periods of hypoxia, the ET-1 ischemia model represents a rapid and technically simplified model more amenable to larger cohorts and with the potential to direct the locus of ischemic damage to specific brain areas.
Collapse
Affiliation(s)
- Jordan L Wright
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
| | - Hannah X Chu
- Biomedicine Discovery Institute and Department of Pharmocology, Monash University, Melbourne, VIC, Australia
| | - Brett J Kagan
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Charlotte M Ermine
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Jessica A Kauhausen
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Christopher G Sobey
- Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC, Australia
| | - Lachlan H Thompson
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
| |
Collapse
|
25
|
Coq JO, Delcour M, Ogawa Y, Peyronnet J, Castets F, Turle-Lorenzo N, Montel V, Bodineau L, Cardot P, Brocard C, Liabeuf S, Bastide B, Canu MH, Tsuji M, Cayetanot F. Mild Intrauterine Hypoperfusion Leads to Lumbar and Cortical Hyperexcitability, Spasticity, and Muscle Dysfunctions in Rats: Implications for Prematurity. Front Neurol 2018; 9:423. [PMID: 29973904 PMCID: PMC6020763 DOI: 10.3389/fneur.2018.00423] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/22/2018] [Indexed: 12/22/2022] Open
Abstract
Intrauterine ischemia-hypoxia is detrimental to the developing brain and leads to white matter injury (WMI), encephalopathy of prematurity (EP), and often to cerebral palsy (CP), but the related pathophysiological mechanisms remain unclear. In prior studies, we used mild intrauterine hypoperfusion (MIUH) in rats to successfully reproduce the diversity of clinical signs of EP, and some CP symptoms. Briefly, MIUH led to inflammatory processes, diffuse gray and WMI, minor locomotor deficits, musculoskeletal pathologies, neuroanatomical and functional disorganization of the primary somatosensory and motor cortices, delayed sensorimotor reflexes, spontaneous hyperactivity, deficits in sensory information processing, memory and learning impairments. In the present study, we investigated the early and long-lasting mechanisms of pathophysiology that may be responsible for the various symptoms induced by MIUH. We found early hyperreflexia, spasticity and reduced expression of KCC2 (a chloride cotransporter that regulates chloride homeostasis and cell excitability). Adult MIUH rats exhibited changes in muscle contractile properties and phenotype, enduring hyperreflexia and spasticity, as well as hyperexcitability in the sensorimotor cortex. Taken together, these results show that reduced expression of KCC2, lumbar hyperreflexia, spasticity, altered properties of the soleus muscle, as well as cortical hyperexcitability may likely interplay into a self-perpetuating cycle, leading to the emergence, and persistence of neurodevelopmental disorders (NDD) in EP and CP, such as sensorimotor impairments, and probably hyperactivity, attention, and learning disorders.
Collapse
Affiliation(s)
- Jacques-Olivier Coq
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, UMR 7289, Aix Marseille Université, Marseille, France.,Centre National de la Recherche Scientifique, Neurosciences Intégratives et Adaptatives, UMR 7260, Aix Marseille Université, Marseille, France
| | - Maxime Delcour
- Centre National de la Recherche Scientifique, Neurosciences Intégratives et Adaptatives, UMR 7260, Aix Marseille Université, Marseille, France
| | - Yuko Ogawa
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Julie Peyronnet
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, UMR 7289, Aix Marseille Université, Marseille, France
| | - Francis Castets
- Centre National de la Recherche Scientifique, Institut de Biologie du Développement de Marseille, UMR 7288, Aix-Marseille Université, Marseille, France
| | - Nathalie Turle-Lorenzo
- FR 3512 Fédération 3C, Aix Marseille Université - Centre National de la Recherche Scientifique, Marseille, France
| | - Valérie Montel
- EA 7369 ≪Activité Physique, Muscle et Santé≫ - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Université de Lille, Lille, France
| | - Laurence Bodineau
- Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Phillipe Cardot
- Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Cécile Brocard
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, UMR 7289, Aix Marseille Université, Marseille, France
| | - Sylvie Liabeuf
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, UMR 7289, Aix Marseille Université, Marseille, France
| | - Bruno Bastide
- EA 7369 ≪Activité Physique, Muscle et Santé≫ - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Université de Lille, Lille, France
| | - Marie-Hélène Canu
- EA 7369 ≪Activité Physique, Muscle et Santé≫ - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Université de Lille, Lille, France
| | - Masahiro Tsuji
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Florence Cayetanot
- Centre National de la Recherche Scientifique, Institut de Neurosciences de la Timone, UMR 7289, Aix Marseille Université, Marseille, France.,Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| |
Collapse
|
26
|
Tsuji M, Coq JO, Ogawa Y, Yamamoto Y, Ohshima M. A Rat Model of Mild Intrauterine Hypoperfusion with Microcoil Stenosis. J Vis Exp 2018. [PMID: 29364276 DOI: 10.3791/56723] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Intrauterine hypoperfusion/ischemia is one of the major causes of intrauterine/fetal growth restriction, preterm birth, and low birth weight. Most studies of this phenomenon have been performed in either models with severe intrauterine ischemia or models with gradient degree of intrauterine hypoperfusion. No study has been performed in a model on uniform mild intrauterine hypoperfusion (MIUH). Two models have been used for studies of MIUH: a model based on suture ligation of either side of the arterial arcade formed with the uterine and ovarian arteries, and a transient model based on clipping the bilateral ovarian arteries and aorta having patency. Those two rodent models of MIUH have some limitations, e.g., not all fetuses are subjected to MIUH, depending on their position in the uterine horn. In our MIUH model, all fetuses are subjected to a comparable level of intrauterine hypoperfusion. MIUH was achieved by mild stenosis of all four arteries feeding the uterus, i.e., the bilateral uterine and ovarian arteries. Arterial stenosis was induced by metal microcoils wrapped around the feeding arteries. Producing arterial stenosis with microcoils allowed us to control, optimize, and reproduce decreased blood flow with very little inter-animal variability and a low mortality rate, thus enabling accurate evaluation. When microcoils with an inner diameter of 0.24 mm were used, the blood flow in both the placenta and fetus was mildly decreased (approximately 30% from the pre-stenosis level in the placenta). The offspring of our MIUH model clearly demonstrates long-lasting alterations in neurological, neuroanatomical and behavioral test results.
Collapse
Affiliation(s)
- Masahiro Tsuji
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center;
| | | | - Yuko Ogawa
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center
| | - Yumi Yamamoto
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center
| | - Makiko Ohshima
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center
| |
Collapse
|
27
|
|
28
|
Ruff CA, Faulkner SD, Rumajogee P, Beldick S, Foltz W, Corrigan J, Basilious A, Jiang S, Thiyagalingam S, Yager JY, Fehlings MG. The extent of intrauterine growth restriction determines the severity of cerebral injury and neurobehavioural deficits in rodents. PLoS One 2017; 12:e0184653. [PMID: 28934247 PMCID: PMC5608203 DOI: 10.1371/journal.pone.0184653] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 08/28/2017] [Indexed: 12/16/2022] Open
Abstract
Background Cerebral Palsy (CP) is the most common physical pediatric neurodevelopmental disorder and spastic diplegic injury is its most frequent subtype. CP results in substantial neuromotor and cognitive impairments that have significant socioeconomic impact. Despite this, its underlying pathophysiological mechanisms and etiology remain incompletely understood. Furthermore, there is a need for clinically relevant injury models, which a) reflect the heterogeneity of the condition and b) can be used to evaluate new translational therapies. To address these key knowledge gaps, we characterized a chronic placental insufficiency (PI) model, using bilateral uterine artery ligation (BUAL) of dams. This injury model results in intrauterine growth restriction (IUGR) in pups, and animals recapitulate the human phenotype both in terms of neurobehavioural and anatomical deficits. Methods Effects of BUAL were studied using luxol fast blue (LFB)/hematoxylin & eosin (H&E) staining, immunohistochemistry, quantitative Magnetic Resonance Imaging (MRI), and Catwalk neurobehavioural tests. Results Neuroanatomical analysis revealed regional ventricular enlargement and corpus callosum thinning in IUGR animals, which was correlated with the extent of growth restriction. Olig2 staining revealed reductions in oligodendrocyte density in white and grey matter structures, including the corpus callosum, optic chiasm, and nucleus accumbens. The caudate nucleus, along with other brain structures such as the optic chiasm, internal capsule, septofimbrial and lateral septal nuclei, exhibited reduced size in animals with IUGR. The size of the pretectal nucleus was reduced only in moderately injured animals. MAG/NF200 staining demonstrated reduced myelination and axonal counts in the corpus callosum of IUGR animals. NeuN staining revealed changes in neuronal density in the hippocampus and in the thickness of hippocampal CA2 and CA3 regions. Diffusion weighted imaging (DWI) revealed regional white and grey matter changes at 3 weeks of age. Furthermore, neurobehavioural testing demonstrated neuromotor impairments in animals with IUGR in paw intensities, swing speed, relative print positions, and phase dispersions. Conclusions We have characterized a rodent model of IUGR and have demonstrated that the neuroanatomical and neurobehavioural deficits mirror the severity of the IUGR injury. This model has the potential to be applied to examine the pathobiology of and potential therapeutic strategies for IUGR-related brain injury. Thus, this work has potential translational relevance for the study of CP.
Collapse
Affiliation(s)
- Crystal A. Ruff
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Stuart D. Faulkner
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Prakasham Rumajogee
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Stephanie Beldick
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Warren Foltz
- STARR facility, Toronto Medical Discovery Tower, Toronto, Ontario, Canada
| | - Jennifer Corrigan
- Section of Pediatric Neurosciences, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Alfred Basilious
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shangjun Jiang
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shanojan Thiyagalingam
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jerome Y. Yager
- Section of Pediatric Neurosciences, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Michael G. Fehlings
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|