1
|
Lear BA, Zhou KQ, Dhillon SK, Lear CA, Bennet L, Gunn AJ. Preventive, rescue and reparative neuroprotective strategies for the fetus and neonate. Semin Fetal Neonatal Med 2024; 29:101542. [PMID: 39472238 DOI: 10.1016/j.siny.2024.101542] [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: 12/13/2024]
Abstract
Neonatal encephalopathy remains a major contributor to death and disability around the world. Acute hypoxia-ischaemia before, during or after birth creates a series of events that can lead to neonatal brain injury. Understanding the evolution of injury underpinned the development of therapeutic hypothermia. This review discusses the determinants of injury, including maturity, the pattern of exposure to HI, impaired placental function, often associated with fetal growth restriction and in the long-term, socio-economic deprivation. Chorioamnionitis has been associated with the presence of NE, but it is important to note that experimentally, inflammation can either sensitize to greater neural injury after HI or alleviate injury, depending on its precise timing. As fetal surveillance tools improve it is likely that improved detection of specific pathways will offer future opportunities for preventive and reparative interventions in utero and after birth.
Collapse
Affiliation(s)
- Benjamin A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Kelly Q Zhou
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Simerdeep K Dhillon
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Christopher A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
2
|
Kirby ED, Andrushko JW, Boyd LA, Koschutnig K, D'Arcy RCN. Sex differences in patterns of white matter neuroplasticity after balance training in young adults. Front Hum Neurosci 2024; 18:1432830. [PMID: 39257696 PMCID: PMC11383771 DOI: 10.3389/fnhum.2024.1432830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction In past work we demonstrated different patterns of white matter (WM) plasticity in females versus males associated with learning a lab-based unilateral motor skill. However, this work was completed in neurologically intact older adults. The current manuscript sought to replicate and expand upon these WM findings in two ways: (1) we investigated biological sex differences in neurologically intact young adults, and (2) participants learned a dynamic full-body balance task. Methods 24 participants (14 female, 10 male) participated in the balance training intervention, and 28 were matched controls (16 female, 12 male). Correlational tractography was used to analyze changes in WM from pre- to post-training. Results Both females and males demonstrated skill acquisition, yet there were significant differences in measures of WM between females and males. These data support a growing body of evidence suggesting that females exhibit increased WM neuroplasticity changes relative to males despite comparable changes in motor behavior (e.g., balance). Discussion The biological sex differences reported here may represent an important factor to consider in both basic research (e.g., collapsing across females and males) as well as future clinical studies of neuroplasticity associated with motor function (e.g., tailored rehabilitation approaches).
Collapse
Affiliation(s)
- Eric D Kirby
- BrainNet, Health and Technology District, Surrey, BC, Canada
- Faculty of Individualized Interdisciplinary Studies, Simon Fraser University, Burnaby, BC, Canada
- Faculty of Science, Simon Fraser University, Burnaby, BC, Canada
| | - Justin W Andrushko
- Djavad Mowafaghian Center for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Sport, Exercise and Rehabilitation, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, Tyne and Wear, United Kingdom
- Brain Behavior Laboratory, Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lara A Boyd
- Djavad Mowafaghian Center for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Brain Behavior Laboratory, Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Karl Koschutnig
- Institute of Psychology, BioTechMed Graz, University of Graz, Graz, Austria
| | - Ryan C N D'Arcy
- BrainNet, Health and Technology District, Surrey, BC, Canada
- Djavad Mowafaghian Center for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Faculty of Applied Sciences, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
3
|
Piao H, Ishikawa H, Kobayashi T, Kitajo K, Yamaguchi A, Koga K, Shozu M. Progesterone improves motor coordination impairments caused by postnatal hypoxic-ischemic brain insult in neonatal male rats. Pediatr Neonatol 2024:S1875-9572(24)00133-5. [PMID: 39181832 DOI: 10.1016/j.pedneo.2024.03.012] [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: 11/15/2023] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Hypoxic-ischemic (HI) insult in infants induces brain injury and results in motor coordination impairments associated with cerebral palsy; however, preventive measures for HI brain injury in preterm infants remain unclear. We investigated the impact of progesterone (P4) in a rat HI insult model that mimics HI brain injury in preterm infants. METHODS Neonatal male rats with their right common carotid artery coagulated were exposed to a 1-h hypoxia (6% oxygen) on postnatal day (PND) 3. P4 (0.2 mg) was subcutaneously administered daily from PND4-12. Motor coordination function and muscular strength were evaluated on PND50 using rotarod and grip strength tests, respectively. Brain histology was evaluated via immunohistochemistry using anti-NeuN, anti-Olig2, and anti-IbaI antibodies on PND15 and PND50. RESULTS In male rats, P4 significantly improved the latency-to-fall off on the rotarod test in the insult rats to the levels of the sham-operation rats. Neither the insult nor P4 administration impacted the grip strength results. No significant differences were observed in the number of neurons, oligodendrocyte progenitor cells (OPCs), and microglia in the motor and somatosensory area of the cortex between the insult and insult followed by P4-administered rats on PND50. The number of OPCs in the corpus callosum was significantly increased in the ipsilateral side compared with the contralateral side of the insult in the P4-administered rats, indicating that P4 facilitates recruitment of OPCs to the corpus callosum. HI insult accelerated neuronal differentiation in rats on PND15, which was abrogated in the P4-administerd group, suggesting that P4 suppresses transient neuronal differentiation caused by the insult. CONCLUSION P4 administration restored motor coordination impairments caused by postnatal HI insult in male rats. The insult timing corresponds to that of human preterm infants, indicating P4's potential for protecting HI brain injury in preterm male infants.
Collapse
Affiliation(s)
- Hongying Piao
- Department of Obstetrics and Gynecology, Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroshi Ishikawa
- Department of Obstetrics and Gynecology, Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Tatsuya Kobayashi
- Department of Obstetrics and Gynecology, Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan; Department of Regulatory Science, Research Promotion Unit, School of Medical Science, Fujita Health University, Aichi, Japan; Fujita Health University Haneda Clinic, Tokyo, Japan
| | - Keiko Kitajo
- Department of Functional Anatomy, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Yamaguchi
- Department of Functional Anatomy, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kaori Koga
- Department of Obstetrics and Gynecology, Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Makio Shozu
- Department of Obstetrics and Gynecology, Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan; Evolution and Reproductive Biology, Medical Mycology Research Center, Chiba University, Chiba, Japan
| |
Collapse
|
4
|
Fieß A, Hartmann A, Mildenberger E, Urschitz MS, Laspas P, Schultheis A, Stoffelns B, Pfeiffer N, Gißler S, Schuster AK. Sex-Specific Differences in the Relationship Between Prematurity and Ocular Geometry. Invest Ophthalmol Vis Sci 2024; 65:23. [PMID: 38874964 PMCID: PMC11182371 DOI: 10.1167/iovs.65.6.23] [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: 12/15/2023] [Accepted: 03/19/2024] [Indexed: 06/15/2024] Open
Abstract
Purpose To explore differences in the relationship between gestational age (GA) and birth weight (BW) percentile and ocular geometry between males and females. Methods The Gutenberg Prematurity Eye Study involved a prospective ophthalmic examination of adults, aged 18 to 52 years, who were born preterm or at term, in Germany. The associations between GA and BW percentile on the main outcome measures were evaluated by uni- and multivariable linear regression analyses. The main outcome measures were central corneal thickness, corneal radius, anterior chamber depth, lens thickness, posterior segment length, and central foveal thickness. Potential sex-specific differences and an effect modification by sex were analyzed. Results This study involved 438 participants (245 females, 193 males) with an average age of 28.6 ± 8.7 years. In female participants, central foveal thickness was negatively associated with a higher GA (B = -2.99; P < 0.001). Similarly, male participants also demonstrated a negative association between central foveal thickness and GA (B = -4.27; P < 0.001). The multivariable model with effect modification revealed that the central foveal thickness was thicker with lower GA. There was an association between the effect modification of GA with sex and central foveal thickness, demonstrating a more pronounced effect of GA on central foveal thickness in male participants (B = 1.29; P = 0.04). Conclusions This study identified a sex-specific correlation between lower GA and thicker central foveal thickness, suggesting differences in the developmental trajectory of this biometric parameter concerning GA. A thicker central foveal thickness might affect the visual acuity of individuals born preterm in adulthood, with a more pronounced impact in males and a potential predisposition to age-related diseases later in life. Sex did not influence the association of GA or BW percentile to other ocular geometric parameters.
Collapse
Affiliation(s)
- Achim Fieß
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alica Hartmann
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Eva Mildenberger
- Division of Neonatology, Department of Pediatrics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael S. Urschitz
- Division of Pediatric Epidemiology, Institute for Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Panagiotis Laspas
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anna Schultheis
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Bernhard Stoffelns
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sandra Gißler
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alexander K. Schuster
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
5
|
Nour Eldine M, Alhousseini M, Nour-Eldine W, Noureldine H, Vakharia KV, Krafft PR, Noureldine MHA. The Role of Oxidative Stress in the Progression of Secondary Brain Injury Following Germinal Matrix Hemorrhage. Transl Stroke Res 2024; 15:647-658. [PMID: 36930383 DOI: 10.1007/s12975-023-01147-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/18/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023]
Abstract
Germinal matrix hemorrhage (GMH) can be a fatal condition responsible for the death of 1.7% of all neonates in the USA. The majority of GMH survivors develop long-term sequalae with debilitating comorbidities. Higher grade GMH is associated with higher mortality rates and higher prevalence of comorbidities. The pathophysiology of GMH can be broken down into two main titles: faulty hemodynamic autoregulation and structural weakness at the level of tissues and cells. Prematurity is the most significant risk factor for GMH, and it predisposes to both major pathophysiological mechanisms of the condition. Secondary brain injury is an important determinant of survival and comorbidities following GMH. Mechanisms of brain injury secondary to GMH include apoptosis, necrosis, neuroinflammation, and oxidative stress. This review will have a special focus on the mechanisms of oxidative stress following GMH, including but not limited to inflammation, mitochondrial reactive oxygen species, glutamate toxicity, and hemoglobin metabolic products. In addition, this review will explore treatment options of GMH, especially targeted therapy.
Collapse
Affiliation(s)
- Mariam Nour Eldine
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | | | - Wared Nour-Eldine
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Hussein Noureldine
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Kunal V Vakharia
- Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, USA
| | - Paul R Krafft
- Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, USA
| | - Mohammad Hassan A Noureldine
- Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, Florida, USA.
| |
Collapse
|
6
|
Van Steenwinckel J, Bokobza C, Laforge M, Shearer IK, Miron VE, Rua R, Matta SM, Hill‐Yardin EL, Fleiss B, Gressens P. Key roles of glial cells in the encephalopathy of prematurity. Glia 2024; 72:475-503. [PMID: 37909340 PMCID: PMC10952406 DOI: 10.1002/glia.24474] [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: 07/19/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 11/03/2023]
Abstract
Across the globe, approximately one in 10 babies are born preterm, that is, before 37 weeks of a typical 40 weeks of gestation. Up to 50% of preterm born infants develop brain injury, encephalopathy of prematurity (EoP), that substantially increases their risk for developing lifelong defects in motor skills and domains of learning, memory, emotional regulation, and cognition. We are still severely limited in our abilities to prevent or predict preterm birth. No longer just the "support cells," we now clearly understand that during development glia are key for building a healthy brain. Glial dysfunction is a hallmark of EoP, notably, microgliosis, astrogliosis, and oligodendrocyte injury. Our knowledge of glial biology during development is exponentially expanding but hasn't developed sufficiently for development of effective neuroregenerative therapies. This review summarizes the current state of knowledge for the roles of glia in infants with EoP and its animal models, and a description of known glial-cell interactions in the context of EoP, such as the roles for border-associated macrophages. The field of perinatal medicine is relatively small but has worked passionately to improve our understanding of the etiology of EoP coupled with detailed mechanistic studies of pre-clinical and human cohorts. A primary finding from this review is that expanding our collaborations with computational biologists, working together to understand the complexity of glial subtypes, glial maturation, and the impacts of EoP in the short and long term will be key to the design of therapies that improve outcomes.
Collapse
Affiliation(s)
| | - Cindy Bokobza
- NeuroDiderot, INSERMUniversité Paris CitéParisFrance
| | | | - Isabelle K. Shearer
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Veronique E. Miron
- Barlo Multiple Sclerosis CentreSt. Michael's HospitalTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
- College of Medicine and Veterinary MedicineThe Dementia Research Institute at The University of EdinburghEdinburghUK
| | - Rejane Rua
- CNRS, INSERM, Centre d'Immunologie de Marseille‐Luminy (CIML), Turing Centre for Living SystemsAix‐Marseille UniversityMarseilleFrance
| | - Samantha M. Matta
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Elisa L. Hill‐Yardin
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | - Bobbi Fleiss
- NeuroDiderot, INSERMUniversité Paris CitéParisFrance
- School of Health and Biomedical SciencesSTEM College, RMIT UniversityBundooraVictoriaAustralia
| | | |
Collapse
|
7
|
McLeod RM, Rosenkrantz TS, Fitch RH. Antenatal Magnesium Sulfate Benefits Female Preterm Infants but Results in Poor Male Outcomes. Pharmaceuticals (Basel) 2024; 17:218. [PMID: 38399433 PMCID: PMC10892166 DOI: 10.3390/ph17020218] [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: 01/12/2024] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Magnesium sulfate (MagSul) is used clinically to prevent eclamptic seizures during pregnancy and as a tocolytic for preterm labor. More recently, it has been implicated as offering neural protection in utero for at-risk infants. However, evidence is mixed. Some studies found that MagSul reduced the incidence of cerebral palsy (CP) but did not improve other measures of neurologic function. Others did not find any improvement in outcomes. Inconsistencies in the literature may reflect the fact that sex effects are largely ignored, despite evidence that MagSul shows sex effects in animal models of neonatal brain injury. The current study used retrospective infant data to assess differences in developmental outcomes as a function of sex and MagSul treatment. We found that on 18-month neurodevelopmental cognitive and language measures, preterm males treated with MagSul (n = 209) had significantly worse scores than their untreated counterparts (n = 135; p < 0.05). Female preterm infants treated with MagSul (n = 220), on the other hand, showed a cognitive benefit relative to untreated females (n = 123; p < 0.05). No significant effects of MagSul were seen among females on language (p > 0.05). These results have tremendous implications for risk-benefit considerations in the ongoing use of MagSul and may explain why benefits have been hard to identify in clinical trials when sex is not considered.
Collapse
Affiliation(s)
- Ruth M. McLeod
- Department of Psychology, College of the Holy Cross, Worcester, MA 01610, USA
| | - Ted S. Rosenkrantz
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA;
| | - R. Holly Fitch
- Department of Psychological Sciences, Behavioral Neuroscience Division, University of Connecticut, Storrs, CT 06269, USA;
| |
Collapse
|
8
|
Beldarrain G, Chillida M, Hilario E, Herrero de la Parte B, Álvarez A, Alonso-Alconada D. URB447 Is Neuroprotective in Both Male and Female Rats after Neonatal Hypoxia-Ischemia and Enhances Neurogenesis in Females. Int J Mol Sci 2024; 25:1607. [PMID: 38338884 PMCID: PMC10855747 DOI: 10.3390/ijms25031607] [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: 12/15/2023] [Revised: 01/15/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
The need for new and effective treatments for neonates suffering from hypoxia-ischemia is urgent, as the only implemented therapy in clinics is therapeutic hypothermia, only effective in 50% of cases. Cannabinoids may modulate neuronal development and brain plasticity, but further investigation is needed to better describe their implication as a neurorestorative therapy after neonatal HI. The cannabinoid URB447, a CB1 antagonist/CB2 agonist, has previously been shown to reduce brain injury after HI, but it is not clear whether sex may affect its neuroprotective and/or neurorestorative effect. Here, URB447 strongly reduced brain infarct, improved neuropathological score, and augmented proliferative capacity and neurogenic response in the damaged hemisphere. When analyzing these effects by sex, URB447 ameliorated brain damage in both males and females, and enhanced cell proliferation and the number of neuroblasts only in females, thus suggesting a neuroprotective effect in males and a double neuroprotective/neurorestorative effect in females.
Collapse
Affiliation(s)
- Gorane Beldarrain
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Marc Chillida
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Enrique Hilario
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Borja Herrero de la Parte
- Department of Surgery and Radiology and Physical Medicine, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Antonia Álvarez
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Daniel Alonso-Alconada
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| |
Collapse
|
9
|
Chanana V, Hackett M, Deveci N, Aycan N, Ozaydin B, Cagatay NS, Hanalioglu D, Kintner DB, Corcoran K, Yapici S, Camci F, Eickhoff J, Frick KM, Ferrazzano P, Levine JE, Cengiz P. TrkB-mediated sustained neuroprotection is sex-specific and Erα-dependent in adult mice following neonatal hypoxia ischemia. Biol Sex Differ 2024; 15:1. [PMID: 38178264 PMCID: PMC10765746 DOI: 10.1186/s13293-023-00573-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of life-long neurological morbidities that result in learning and memory impairments. Evidence suggests that male neonates are more susceptible to the detrimental effects of HI, yet the mechanisms mediating these sex-specific responses to neural injury in neonates remain poorly understood. We previously tested the effects of treatment with a small molecule agonist of the tyrosine kinase B receptor (TrkB), 7,8-dihydroxyflavone (DHF) following neonatal HI and determined that females, but not males exhibit increased phosphorylation of TrkB and reduced apoptosis in their hippocampi. Moreover, these female-specific effects of the TrkB agonist were found to be dependent upon the expression of Erα. These findings demonstrated that TrkB activation in the presence of Erα comprises one pathway by which neuroprotection may be conferred in a female-specific manner. The goal of this study was to determine the role of Erα-dependent TrkB-mediated neuroprotection in memory and anxiety in young adult mice exposed to HI during the neonatal period. METHODS In this study, we used a unilateral hypoxic ischemic (HI) mouse model. Erα+/+ or Erα-/- mice were subjected to HI on postnatal day (P) 9 and mice were treated with either vehicle control or the TrkB agonist, DHF, for 7 days following HI. When mice reached young adulthood, we used the novel object recognition, novel object location and open field tests to assess long-term memory and anxiety-like behavior. The brains were then assessed for tissue damage using immunohistochemistry. RESULTS Neonatal DHF treatment prevented HI-induced decrements in recognition and location memory in adulthood in females, but not in males. This protective effect was absent in female mice lacking Erα. The female-specific improved recognition and location memory outcomes in adulthood conferred by DHF therapy after neonatal HI tended to be or were Erα-dependent, respectively. Interestingly, DHF triggered anxiety-like behavior in both sexes only in the mice that lacked Erα. When we assessed the severity of injury, we found that DHF therapy did not decrease the percent tissue loss in proportion to functional recovery. We additionally observed that the presence of Erα significantly reduced overall HI-associated mortality in both sexes. CONCLUSIONS These observations provide evidence for a therapeutic role for DHF in which TrkB-mediated sustained recovery of recognition and location memories in females are Erα-associated and dependent, respectively. However, the beneficial effects of DHF therapy did not include reduction of gross tissue loss but may be derived from the enhanced functioning of residual tissues in a cell-specific manner.
Collapse
Affiliation(s)
- Vishal Chanana
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Margaret Hackett
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Nazli Deveci
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Nur Aycan
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
| | - Burak Ozaydin
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Nur Sena Cagatay
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Damla Hanalioglu
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
| | - Douglas B Kintner
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Karson Corcoran
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Sefer Yapici
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Furkan Camci
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
| | - Jens Eickhoff
- Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Peter Ferrazzano
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jon E Levine
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Pelin Cengiz
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA.
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
10
|
Kelly LA, Branagan A, Semova G, Molloy EJ. Sex differences in neonatal brain injury and inflammation. Front Immunol 2023; 14:1243364. [PMID: 37954620 PMCID: PMC10634351 DOI: 10.3389/fimmu.2023.1243364] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/17/2023] [Indexed: 11/14/2023] Open
Abstract
Neonatal brain injury and associated inflammation is more common in males. There is a well-recognised difference in incidence and outcome of neonatal encephalopathy according to sex with a pronounced male disadvantage. Neurodevelopmental differences manifest from an early age in infancy with females having a lower incidence of developmental delay and learning difficulties in comparison with males and male sex has consistently been identified as a risk factor for cerebral palsy in epidemiological studies. Important neurobiological differences exist between the sexes with respect to neuronal injury which are especially pronounced in preterm neonates. There are many potential reasons for these sex differences including genetic, immunological and hormonal differences but there are limited studies of neonatal immune response. Animal models with induced neonatal hypoxia have shown various sex differences including an upregulated immune response and increased microglial activation in males. Male sex is recognized to be a risk factor for neonatal hypoxic ischemic encephalopathy (HIE) during the perinatal period and this review discusses in detail the sex differences in brain injury in preterm and term neonates and some of the potential new therapies with possible sex affects.
Collapse
Affiliation(s)
- Lynne A. Kelly
- Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland
- Paediatrics, Trinity Translational Medicine Institute (TTMI), Dublin, Ireland
- Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
| | - Aoife Branagan
- Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland
- Paediatrics, Trinity Translational Medicine Institute (TTMI), Dublin, Ireland
- Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
- Coombe Women and Infants University Hospital Dublin, Dublin, Ireland
| | - Gergana Semova
- Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland
- Paediatrics, Trinity Translational Medicine Institute (TTMI), Dublin, Ireland
- Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
| | - Eleanor J. Molloy
- Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland
- Paediatrics, Trinity Translational Medicine Institute (TTMI), Dublin, Ireland
- Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
- Coombe Women and Infants University Hospital Dublin, Dublin, Ireland
- Neonatology, Children’s Health Ireland (CHI) at Crumlin, Dublin, Ireland
- Neonatology and Neurodisability, Children’s Health Ireland (CHI) at Tallaght, Dublin, Ireland
| |
Collapse
|
11
|
Sewell EK, Shankaran S, Natarajan G, Laptook A, Das A, McDonald SA, Hamrick S, Baack M, Rysavy M, Higgins RD, Chalak L, Patel RM. Evaluation of heterogeneity in effect of therapeutic hypothermia by sex among infants with neonatal encephalopathy. Pediatr Res 2023; 94:1380-1384. [PMID: 37012412 PMCID: PMC10843889 DOI: 10.1038/s41390-023-02586-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Our objective was to examine heterogeneity in the effect of therapeutic hypothermia by sex in infants with moderate or severe neonatal encephalopathy. METHODS We conducted a post hoc analysis of the Induced Hypothermia trial, which included infants born at gestational ages ≥36 weeks, admitted at ≤6 postnatal hours with evidence of severe acidosis or perinatal complications and moderate or severe neonatal encephalopathy. Multivariate modified Poisson regression models were used to compare the treatment effect of whole-body hypothermia versus control, with an evaluation of interaction by sex, on the primary outcome of death or moderate or severe disability at 18-22 months of corrected age. RESULTS A total of 101 infants (51 male, 50 female) were randomly assigned to hypothermia treatment and 104 infants (64 male, 40 female) to control. The primary outcome occurred in 45% of the hypothermia group and 63% of the control group (RR 0.73; 95% CI 0.56, 0.94). There was no significant difference (interaction P = 0.50) in the treatment effect of hypothermia on the primary outcome between females (RR 0.79; 95% CI 0.54, 1.17) compared to males (RR 0.63; 95% CI 0.44, 0.91). CONCLUSION We found no evidence that sex influences the treatment effect of hypothermia in infants with moderate or severe neonatal encephalopathy. IMPACT Preclinical evidence suggests a differential effect in response to cooling treatment of hypoxic-ischemic injury between males and females. We found no evidence of heterogeneity in the treatment effect of whole-body hypothermia by sex in this post hoc subgroup analysis of infants with moderate or severe neonatal encephalopathy from the National Institute of Child Health and Human Development Neonatal Research Network Induced Hypothermia trial.
Collapse
Affiliation(s)
- Elizabeth K Sewell
- Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.
| | | | | | | | - Abhik Das
- RTI International, Rockville, MD, USA
| | | | - Shannon Hamrick
- Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Michelle Baack
- University of South Dakota - Sanford School of Medicine, Sioux Falls, SD, USA
| | - Matthew Rysavy
- University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Lina Chalak
- University of Texas Southwestern, Dallas, TX, USA
| | - Ravi Mangal Patel
- Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| |
Collapse
|
12
|
Chanana V, Hackett M, Deveci N, Aycan N, Ozaydin B, Cagatay NS, Hanalioglu D, Kintner DB, Corcoran K, Yapici S, Camci F, Eickhoff J, Frick KM, Ferrazano P, Levine JE, Cengiz P. TrkB-mediated sustained neuroprotection is sex-specific and ERα dependent in adult mice following neonatal hypoxia ischemia. RESEARCH SQUARE 2023:rs.3.rs-3325405. [PMID: 37720039 PMCID: PMC10503864 DOI: 10.21203/rs.3.rs-3325405/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Background Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of life-long neurological morbidities that result in learning and memory impairments. Evidence suggests that male neonates are more susceptible to the detrimental effects of HI, yet the mechanisms mediating these sex-specific responses to neural injury in neonates remain poorly understood. We previously tested the effects of treatment with a small molecule agonist of the tyrosine kinase B receptor (TrkB), 7,8-dihydroxyflavone (DHF) following neonatal HI and determined that females, but not males exhibit increased phosphorylation of TrkB and reduced apoptosis in their hippocampi. Moreover, these female-specific effects of the TrkB agonist were found to be dependent upon the expression of ERα. These findings demonstrated that TrkB activation in the presence of ERα comprises one pathway by which neuroprotection may be conferred in a female-specific manner. The goal of this study was to determine the role of ERα-dependent TrkB-mediated neuroprotection in memory and anxiety in young adult mice exposed to HI during the neonatal period. Methods In this study we used a unilateral hypoxic ischemic (HI) mouse model. ERα+/+ or ERα-/- mice were subjected to HI on postnatal day (P) 9 and mice were treated with either vehicle control or the TrkB agonist, DHF, for seven days following HI. When mice reached young adulthood, we used the novel object recognition, novel object location and open field tests to assess long-term memory and anxiety like behavior. The brains were then assessed for tissue damage using immunohistochemistry. Results Neonatal DHF treatment prevented HI-induced decrements in recognition and location memory in adulthood in females, but not in males. This protective effect was absent in female mice lacking ERα. Thus, the female-specific and ERα-dependent neuroprotection conferred by DHF therapy after neonatal HI was associated with improved learning and memory outcomes in adulthood. Interestingly, DHF triggered anxiety like behavior in both sexes only in the mice that lacked ERα. When we assessed the severity of injury, we found that DHF therapy did not decrease the percent tissue loss in proportion to functional recovery. We additionally observed that the presence of ERα significantly reduced overall HI-associated mortality in both sexes. Conclusions These observations provide evidence for a therapeutic role for DHF in which sustained recovery of memory in females is TrkB-mediated and ERα-dependent. However, the beneficial effects of DHF therapy did not include reduction of gross tissue loss but may be derived from the enhanced functioning of residual tissues in a cell-specific manner.
Collapse
Affiliation(s)
- Vishal Chanana
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Margaret Hackett
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Nazli Deveci
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Nur Aycan
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - Burak Ozaydin
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Nur Sena Cagatay
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Damla Hanalioglu
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - Douglas B. Kintner
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Karson Corcoran
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Sefer Yapici
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Furkan Camci
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - Jens Eickhoff
- Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison, WI, US
| | - Karyn M. Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Peter Ferrazano
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jon E. Levine
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Pelin Cengiz
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
13
|
Korf JM, McCullough LD, Caretti V. A narrative review on treatment strategies for neonatal hypoxic ischemic encephalopathy. Transl Pediatr 2023; 12:1552-1571. [PMID: 37692539 PMCID: PMC10485647 DOI: 10.21037/tp-23-253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023] Open
Abstract
Background and Objective Hypoxic-ischemic encephalopathy (HIE) is a leading cause of death and disability worldwide. Therapeutic hypothermia (TH) represents a significant achievement in the translation of scientific research to clinical application, but it is currently the only neuroprotective treatment for HIE. This review aims to revisit the use of TH for HIE and its longitudinal impact on patient outcomes to readers new to the field of HIE. We discuss how emerging therapies address the broader pathophysiology of injury progression in the neonatal brain days to years after HIE. Methods We included full articles and book chapters published in English on PubMed with references to "hypoxic ischemic encephalopathy", "birth asphyxia", "therapeutic hypothermia", or "neonatal encephalopathy". We limited our review to outcomes on term infants and to new therapeutics that are in the second phase of clinical trials. Key Content and Findings Despite the use of TH for HIE, mortality remains high. Analysis of longitudinal studies reveals a high incidence of ongoing disability even with the implementation of TH. New therapeutics addressing the secondary phase and the less understood tertiary phase of brain injury are in clinical trials as adjunctive treatments to TH to support additional neurological repair and regeneration. Conclusions TH successfully improves outcomes after HIE, and it continues to be optimized. Larger studies are needed to understand its use in mild cases of HIE and if certain factors, such as sex, affect long term outcomes. TH primarily acts in the initial phases of injury, while new pharmaceutical therapies target additional injury pathways into the tertiary phases of injury. This may allow for more effective approaches to treatment and improvement of long-term functional outcomes after HIE.
Collapse
Affiliation(s)
- Janelle M. Korf
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX, USA
| | - Louise D. McCullough
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX, USA
| | - Viola Caretti
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX, USA
- Department of Pediatrics, Section of Pediatric Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
14
|
Migliori C, Braga M, Siragusa V, Villa MC, Luzi L. The impact of gender medicine on neonatology: the disadvantage of being male: a narrative review. Ital J Pediatr 2023; 49:65. [PMID: 37280693 DOI: 10.1186/s13052-023-01447-2] [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: 10/12/2022] [Accepted: 03/20/2023] [Indexed: 06/08/2023] Open
Abstract
This narrative non-systematic review addresses the sex-specific differences observed both in prenatal period and, subsequently, in early childhood. Indeed, gender influences the type of birth and related complications. The risk of preterm birth, perinatal diseases, and differences on efficacy for pharmacological and non-pharmacological therapies, as well as prevention programs, will be evaluated. Although male newborns get more disadvantages, the physiological changes during growth and factors like social, demographic, and behavioural reverse this prevalence for some diseases. Therefore, given the primary role of genetics in gender differences, further studies specifically targeted neonatal sex-differences will be needed to streamline medical care and improve prevention programs.
Collapse
Affiliation(s)
- Claudio Migliori
- Department of Neonatology, Ospedale San Giuseppe MultiMedica, 20123, Milan, Italy.
| | - Marta Braga
- Department of Neonatology, Ospedale San Giuseppe MultiMedica, 20123, Milan, Italy
| | - Virginia Siragusa
- Department of Neonatology, Ospedale San Giuseppe MultiMedica, 20123, Milan, Italy
| | - Maria Cristina Villa
- Department of Neonatology, Ospedale San Giuseppe MultiMedica, 20123, Milan, Italy
| | - Livio Luzi
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, 20099, Sesto San Giovanni, Milan, Italy
| |
Collapse
|
15
|
Saadat A, Blackwell A, Kaszowski C, Pallera H, Owens D, Lattanzio F, Shah T. Therapeutic hypothermia demonstrates sex-dependent improvements in motor function in a rat model of neonatal hypoxic ischemic encephalopathy. Behav Brain Res 2023; 437:114119. [PMID: 36162642 DOI: 10.1016/j.bbr.2022.114119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/02/2022] [Accepted: 09/18/2022] [Indexed: 11/25/2022]
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) is a neurological disease caused by restricted oxygen and blood flow to the brain at or around the time of birth. Long term cognitive and motor sequelae are common and demonstrate sexual dimorphism in animal studies. Therapeutic hypothermia (TH) is the standard of care for HIE, but provides incomplete neuroprotection. Using the Vannucci model of neonatal HIE, term-equivalent 11-day old rat pups were subjected to mild-moderate hypoxic-ischemic injury (HII), and a subset of animals were treated with TH. Sex-dependent neuroprotection was measured with gross and fine motor control assays, and functional deficits detected with these assays were correlated to injury in specific brain structures. At the equivalent of human adolescence and adulthood (P51-89), accelerod and beam walking tests were used to assess gross motor function, and string-pulling and food handling tests were used to assess fine motor function. At necropsy (P94-97), brain lesions were primarily focused to the posterior cerebrum and characterized by variable reduction in cortical, thalamic and hippocampal regions and glial scarring. Gross motor impairment was detected in male rats with untreated and TH-treated HIE in the accelerod test, but beam walk test data was confounded by the lower body mass of untreated male rats. HIE animals of both sexes demonstrated deficit in the forelimb contralateral to ischemic surgery, observed as unilaterally impaired food handling behaviors, and in string pulling as decreased string contacts and increased in bracing behavior. However, kinematic analyses revealed sex-specific decreases in peak speeds in string reaching and pulling movements. In both sexes, treatment with TH improved body mass, some measures of contralateral forelimb impairment, and the severity of brain lesions to levels not different to Sham surgery rats. Unique differences in behavior following TH were observed in female rats, who took longer to consume food items but traversed beams and approached strings faster than untreated and Sham females. Future use of these motor assays may unravel the subtle, sex-specific differences in HIE outcomes and in developing a customized therapeutic approach to neonatal brain injury.
Collapse
Affiliation(s)
- Angela Saadat
- Neonatal Brain Institute, Children's Specialty Group, USA.
| | - Ashley Blackwell
- Center for Integrative Neuroinflammatory and Inflammatory Diseases, USA; Dept. Radiation Oncology, Eastern Virginia Medical School, USA
| | | | - Haree Pallera
- Neonatal Brain Institute, Children's Specialty Group, USA
| | - Daley Owens
- Neonatal Brain Institute, Children's Specialty Group, USA
| | - Frank Lattanzio
- Dept. Physiological Sciences, Eastern Virginia Medical School, USA
| | - Tushar Shah
- Neonatal Brain Institute, Children's Specialty Group, USA
| |
Collapse
|
16
|
McLeod RM, Rosenkrantz TS, Fitch RH, Koski RR. Sex Differences in Microglia Activation in a Rodent Model of Preterm Hypoxic Ischemic Injury with Caffeine Treatment. Biomedicines 2023; 11:biomedicines11010185. [PMID: 36672692 PMCID: PMC9855625 DOI: 10.3390/biomedicines11010185] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
Preterm infants are often treated with caffeine as a respiratory stimulant. However, follow-up data shows caffeine may also have neuroprotective potential. There are several theories as to how caffeine might protect the brain, but none have been proven. This study looked at caffeine effects on microglial activation in rodent brains post hypoxic ischemic (HI) injury. Rat pups underwent either sham or HI surgery on P6, followed by treatment with either caffeine or saline. Forty-eight hours post-injury, brains were collected and underwent paraffin embedding and sectioning followed by immunofluorescence staining. Ionized calcium binding adaptor molecule 1 (Iba-1) was used to label microglia, and 4',6-diamindino-2-phenylindole (DAPI) was used to label DNA. Cell size measurements of microglia were obtained to gauge microglia activation, and chromatin condensation (DAPI optical density) was used as an index of neuronal cell death. Results suggest that caffeine does offer protective effects, based on significantly increased levels of cell death in HI-saline animals not seen in caffeine-treated HI males and females. However, the mechanism of action may be different. Male HI animals showed marginally reduced microglial activation following caffeine treatment, whereas females did not. Results indicate that though caffeine may act protectively in both sexes by reducing cell death, the benefits may be mediated by different mechanisms.
Collapse
Affiliation(s)
- Ruth Mae McLeod
- Behavioral Neuroscience Division, Department of Psychological Sciences, University of Connecticut, Storrs, CT 06269, USA
- Correspondence:
| | - Ted S. Rosenkrantz
- Department of Pediatrics, University of Connecticut Health Center and Connecticut Children’s Hospital, Farmington, CT 06030, USA
| | - Roslyn Holly Fitch
- Behavioral Neuroscience Division, Department of Psychological Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Rachel R. Koski
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA
| |
Collapse
|
17
|
Long-term neurocognitive functioning of children treated with propranolol or atenolol for infantile hemangioma. Eur J Pediatr 2023; 182:757-767. [PMID: 36478294 PMCID: PMC9899165 DOI: 10.1007/s00431-022-04674-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/13/2022] [Accepted: 10/22/2022] [Indexed: 12/13/2022]
Abstract
UNLABELLED The purpose of this study was to compare long-term neurocognitive functioning (working memory, processing speed, and attention) between children who had been treated with either propranolol or atenolol for infantile hemangioma during infancy. All eligible children (n = 158) aged 6 years or older and treated with propranolol or atenolol as infants were invited to participate in this two-center cross-sectional study. The primary outcome was the Wechsler Intelligence Scale for Children-V Cognitive Proficiency Index (CPI), a measure of working memory, processing speed, and attention. Secondary outcomes were general intelligence, auditory, visuospatial, and narrative memory, as well as executive functioning and sleep. A total of 105 children, of whom 36 had been treated with propranolol (age 6.0-11.8 years, follow-up time 1.6-9.7 years, 19% male) and 69 had been treated with atenolol (age 6.9-9.7 years, follow-up time 4.5-8.4 years, 19% male), were analyzed. The CPI and other neurocognitive outcomes did not differ between the propranolol and atenolol groups and were in line with general population test norms. Post hoc analyses revealed lower CPI scores for males, both compared to participating females (10.3 IQ points, medium effect size) and compared to matched test norms (12.4 IQ points, medium effect size). CONCLUSIONS Long-term neurocognitive functioning did not differ between children treated with propranolol and those treated with atenolol for IH. Overall, propranolol and atenolol appear to be safe treatments for IH regarding long-term neurocognitive functioning. The substantially lower CPI scores in males warrant further investigation. TRIAL REGISTRATION Netherlands Trial Register, NL7703 https://www.trialregister.nl/trial/7703 What is Known: • Infants with infantile hemangioma are effectively treated with propranolol or atenolol. • Parents and professionals are concerned about long-term neurocognitive effects. WHAT IS NEW • No long-term (≥ 6 years) differences in neurocognitive functioning were found between children treated with propranolol or atenolol. • Males treated with beta-blockers had substantially lower IQ scores than treated females and males from the general population, which is a matter of concern and should be considered when evaluating the risk/benefit ratio in less severe forms of infantile hemangioma.
Collapse
|
18
|
Intrauterine Inflammation Leads to Select Sex- and Age-Specific Behavior and Molecular Differences in Mice. Int J Mol Sci 2022; 24:ijms24010032. [PMID: 36613475 PMCID: PMC9819857 DOI: 10.3390/ijms24010032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/30/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Sex-specific differences in behavior have been observed in anxiety and learning in children exposed to prenatal inflammation; however, whether these behaviors manifest differently by age is unknown. This study assesses possible behavioral changes due to in utero inflammation as a function of age in neonatal, juvenile, and adult animals and presents potential molecular targets for observed differences. CD-1 timed pregnant dams were injected in utero with lipopolysaccharide (LPS, 50 μg/animal) or saline at embryonic day 15. No differences in stress responses were measured by neonatal ultrasonic vocalizations between LPS- and saline-exposed groups of either sex. By contrast, prenatal inflammation caused a male-specific increase in anxiety in mature but not juvenile animals. Juvenile LPS-exposed females had decreased movement in open field testing that was not present in adult animals. We additionally observed improved memory retrieval after in utero LPS in the juvenile animals of both sexes, which in males may be related to a perseverative phenotype. However, there was an impairment of long-term memory in only adult LPS-exposed females. Finally, gene expression analyses revealed that LPS induced sex-specific changes in genes involved in hippocampal neurogenesis. In conclusion, intrauterine inflammation has age- and sex-specific effects on anxiety and learning that may correlate to sex-specific disruption of gene expression associated with neurogenesis in the hippocampus.
Collapse
|
19
|
N-Acetylcysteine Administration Attenuates Sensorimotor Impairments Following Neonatal Hypoxic-Ischemic Brain Injury in Rats. Int J Mol Sci 2022; 23:ijms232416175. [PMID: 36555816 PMCID: PMC9783020 DOI: 10.3390/ijms232416175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hypoxic ischemic (HI) brain injury that occurs during neonatal period has been correlated with severe neuronal damage, behavioral deficits and infant mortality. Previous evidence indicates that N-acetylcysteine (NAC), a compound with antioxidant action, exerts a potential neuroprotective effect in various neurological disorders including injury induced by brain ischemia. The aim of the present study was to investigate the role of NAC as a potential therapeutic agent in a rat model of neonatal HI brain injury and explore its long-term behavioral effects. To this end, NAC (50 mg/kg/dose, i.p.) was administered prior to and instantly after HI, in order to evaluate hippocampal and cerebral cortex damage as well as long-term functional outcome. Immunohistochemistry was used to detect inducible nitric oxide synthase (iNOS) expression. The results revealed that NAC significantly alleviated sensorimotor deficits and this effect was maintained up to adulthood. These improvements in functional outcome were associated with a significant decrease in the severity of brain damage. Moreover, NAC decreased the short-term expression of iNOS, a finding implying that iNOS activity may be suppressed and that through this action NAC may exert its therapeutic action against neonatal HI brain injury.
Collapse
|
20
|
Li Y, Wisnowski JL, Chalak L, Mathur AM, McKinstry RC, Licona G, Mayock DE, Chang T, Van Meurs KP, Wu TW, Ahmad KA, Cornet MC, Rao R, Scheffler A, Wu YW. Mild hypoxic-ischemic encephalopathy (HIE): timing and pattern of MRI brain injury. Pediatr Res 2022; 92:1731-1736. [PMID: 35354930 PMCID: PMC9771796 DOI: 10.1038/s41390-022-02026-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/06/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Mild hypoxic-ischemic encephalopathy (HIE) is increasingly recognized as a risk factor for neonatal brain injury. We examined the timing and pattern of brain injury in mild HIE. METHODS This retrospective cohort study includes infants with mild HIE treated at 9 hospitals. Neonatal brain MRIs were scored by 2 reviewers using a validated classification system, with discrepancies resolved by consensus. Severity and timing of MRI brain injury (i.e., acute, subacute, chronic) was scored on the subset of MRIs that were performed at or before 8 days of age. RESULTS Of 142 infants with mild HIE, 87 (61%) had injury on MRI at median age 5 (IQR 4-6) days. Watershed (23%), deep gray (20%) and punctate white matter (18%) injury were most common. Among the 125 (88%) infants who received a brain MRI at ≤8 days, mild (44%) injury was more common than moderate (11%) or severe (4%) injury. Subacute (37%) lesions were more commonly observed than acute (32%) or chronic lesions (1%). CONCLUSION Subacute brain injury is common in newborn infants with mild HIE. Novel neuroprotective treatments for mild HIE will ideally target both subacute and acute injury mechanisms. IMPACT Almost two-thirds of infants with mild HIE have evidence of brain injury on MRI obtained in the early neonatal period. Subacute brain injury was seen in 37% of infants with mild HIE. Neuroprotective treatments for mild HIE will ideally target both acute and subacute injury mechanisms.
Collapse
Affiliation(s)
- Yi Li
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - Jessica L Wisnowski
- Department of Radiology and Pediatrics, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Lina Chalak
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amit M Mathur
- Division of Neonatal Perinatal Medicine, Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Robert C McKinstry
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Genesis Licona
- Division of Neonatology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Dennis E Mayock
- Division of Neonatology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Taeun Chang
- Department of Neurology, Children's National Hospital, George Washington School of Medicine & Health Sciences, Washington, DC, USA
| | - Krisa P Van Meurs
- Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tai-Wei Wu
- Division of Neonatology, Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kaashif A Ahmad
- Pediatrix Medical Group of San Antonio, San Antonio, TX, USA
| | - Marie-Coralie Cornet
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Rakesh Rao
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Aaron Scheffler
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Yvonne W Wu
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
21
|
Koehn LM, Nguyen K, Chen X, Santoso A, Tucker R, Lim YP, Stonestreet BS. Effects of Three Different Doses of Inter-Alpha Inhibitor Proteins on Severe Hypoxia-Ischemia-Related Brain Injury in Neonatal Rats. Int J Mol Sci 2022; 23:13473. [PMID: 36362257 PMCID: PMC9655902 DOI: 10.3390/ijms232113473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/30/2024] Open
Abstract
Hypoxia-ischemia (HI)-related brain injury is an important cause of morbidity and long-standing disability in newborns. We have previously shown that human plasma-derived inter-alpha inhibitor proteins (hIAIPs) attenuate HI-related brain injury in neonatal rats. The optimal dose of hIAIPs for their neuroprotective effects and improvement in behavioral outcomes remains to be determined. We examined the efficacy of 30, 60, or 90 mg/kg of hIAIPs administered to neonatal rats after exposure to HI for 2 h. Postnatal day 7 (P7) Wistar rats were exposed to either sham-surgery or unilateral HI (right carotid artery ligation, 2 h of 8% O2) brain injury. A placebo, 30, 60, or 90 mg/kg of hIAIPs were injected intraperitoneally at 0, 24 and 48 h after HI (n = 9-10/sex). We carried out the following behavioral analyses: P8 (righting reflex), P9 (negative geotaxis) and P10 (open-field task). Rats were humanely killed on P10 and their brains were stained with cresyl violet. Male extension/contraction responses and female righting reflex times were higher in the HI placebo groups than the sham groups. Female open-field exploration was lower in the HI placebo group than the sham group. hIAIPs attenuated these behavioral deficits. However, the magnitude of the responses did not vary by hIAIP dose. hIAIPs reduced male brain infarct volumes in a manner that correlated with improved behavioral outcomes. Increasing the hIAIP dose from 30 to 90 mg/kg did not further accentuate the hIAIP-related decreases in infarct volumes. We conclude that larger doses of hIAIPs did not provide additional benefits over the 30 mg/kg dose for behavior tasks or reductions in infarct volumes in neonatal rats after exposure to severe HI.
Collapse
Affiliation(s)
- Liam M. Koehn
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Kevin Nguyen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Xiaodi Chen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | | | - Richard Tucker
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02905, USA
| | - Barbara S. Stonestreet
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| |
Collapse
|
22
|
Li T, Sun Y, Zhang S, Xu Y, Li K, Xie C, Wang Y, Wang Y, Cao J, Wang X, Penninger JM, Kroemer G, Blomgren K, Zhu C. AIF Overexpression Aggravates Oxidative Stress in Neonatal Male Mice After Hypoxia-Ischemia Injury. Mol Neurobiol 2022; 59:6613-6631. [PMID: 35974295 PMCID: PMC9525408 DOI: 10.1007/s12035-022-02987-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
Abstract
There are sex differences in the severity, mechanisms, and outcomes of neonatal hypoxia-ischemia (HI) brain injury, and apoptosis-inducing factor (AIF) may play a critical role in this discrepancy. Based on previous findings that AIF overexpression aggravates neonatal HI brain injury, we further investigated potential sex differences in the severity and molecular mechanisms underlying the injury using mice that overexpress AIF from homozygous transgenes. We found that the male sex significantly aggravated AIF-driven brain damage, as indicated by the injury volume in the gray matter (2.25 times greater in males) and by the lost volume of subcortical white matter (1.71 greater in males) after HI. As compared to females, male mice exhibited more severe brain injury, correlating with reduced antioxidant capacities, more pronounced protein carbonylation and nitration, and increased neuronal cell death. Under physiological conditions (without HI), the doublecortin-positive area in the dentate gyrus of females was 1.15 times larger than in males, indicating that AIF upregulation effectively promoted neurogenesis in females in the long term. We also found that AIF stimulated carbohydrate metabolism in young males. Altogether, these findings corroborate earlier studies and further demonstrate that AIF is involved in oxidative stress, which contributes to the sex-specific differences observed in neonatal HI brain injury.
Collapse
Affiliation(s)
- Tao Li
- Henan Children's Neurodevelopment Engineering Research Center, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Yanyan Sun
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Department of Human Anatomy, School of Basic Medicine and Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450052, China
| | - Shan Zhang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Kenan Li
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Cuicui Xie
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Yong Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Yafeng Wang
- Henan Children's Neurodevelopment Engineering Research Center, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medicine and Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.,Centre of Perinatal Medicine and Health, Institute of Clinical Science, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Josef M Penninger
- Institute of Molecular Biotechnology, Austrian Academy of Sciences, 1030, Vienna, Austria.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Guido Kroemer
- Centre de Recherche Des Cordeliers, Equipe Labellisée Par La Ligue Contre Le Cancer, Inserm U1138, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, Paris, France.,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.,Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Klas Blomgren
- Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China. .,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, 40530, Gothenburg, Sweden. .,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
23
|
Mike JK, Wu KY, White Y, Pathipati P, Ndjamen B, Hutchings RS, Losser C, Vento C, Arellano K, Vanhatalo O, Ostrin S, Windsor C, Ha J, Alhassen Z, Goudy BD, Vali P, Lakshminrusimha S, Gobburu JVS, Long-Boyle J, Chen P, Wu YW, Fineman JR, Ferriero DM, Maltepe E. Defining longer term outcomes in an ovine model of moderate perinatal hypoxia-ischemia. Dev Neurosci 2022; 44:277-294. [PMID: 35588703 DOI: 10.1159/000525150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 04/06/2022] [Indexed: 11/19/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is the leading cause of neonatal morbidity and mortality worldwide. Approximately 1 million infants born with HIE each year survive with cerebral palsy (CP) and/or serious cognitive disabilities. While infants born with mild and severe HIE frequently result in predictable outcomes, infants born with moderate HIE exhibit variable outcomes that are highly unpredictable. Here, we describe an umbilical cord occlusion (UCO) model of moderate HIE with a 6-day follow-up. Near term lambs (n=27) are resuscitated after the induction of 5 minutes of asystole. Following recovery, lambs are assessed to define neurodevelopmental outcomes. At the end of this period, lambs are euthanized, and brains harvested for histological analysis. Compared with prior models that typically follow lambs for 3 days, the observation of neurobehavioral outcomes for 6 days enables identification of animals that recover significant neurological function. Approximately 35 % of lambs exhibited severe motor deficits throughout the entirety of the 6-day course and, in the most severely affected lambs, developed spastic diparesis similar to that observed in infants who survive severe neonatal HIE (severe, UCOs). Importantly, and similar to outcomes in human neonates, while initially developing significant acidosis and encephalopathy, the remainder of the lambs in this model recovered normal motor activity and exhibited normal neurodevelopmental outcomes by 6 days of life (improved, UCOi). The UCOs group exhibited gliosis and inflammation in both white and gray matter, oligodendrocyte loss, and neuronal loss and cellular death in the hippocampus and cingulate cortex. While the UCOi group exhibited more cellular death and gliosis in the parasagittal cortex and demonstrated more preserved white matter markers, along with reduced markers of inflammation and lower cellular death and neuronal loss in Ca3 of the hippocampus compared with UCOs lambs. Our large animal model of moderate HIE with prolonged follow-up will help further define pathophysiologic drivers of brain injury while enabling identification of predictive biomarkers that correlate with disease outcomes and ultimately help support development of therapeutic approaches to this challenging clinical scenario.
Collapse
Affiliation(s)
- Jana Krystofova Mike
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Katherine Y Wu
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Yasmine White
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Praneeti Pathipati
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Blaise Ndjamen
- Histology and Microscopy Core, Gladstone Institutes University of California San Francisco, San Francisco, California, USA
| | - Rachel S Hutchings
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Courtney Losser
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Christian Vento
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Kimberly Arellano
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Oona Vanhatalo
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Samuel Ostrin
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Christine Windsor
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Janica Ha
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Ziad Alhassen
- Department of Pediatrics, University of California Davis, Davis, California, USA
| | - Brian D Goudy
- Department of Pediatrics, University of California Davis, Davis, California, USA
| | - Payam Vali
- Department of Pediatrics, University of California Davis, Davis, California, USA
| | | | - Jogarao V S Gobburu
- School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
- Initiative for Pediatric Drug and Device Development, San Francisco, California, USA
| | - Janel Long-Boyle
- Initiative for Pediatric Drug and Device Development, San Francisco, California, USA
- School of Pharmacy, University of California San Francisco, San Francisco, California, USA
| | - Peggy Chen
- Department of Pediatrics, University of California Davis, Davis, California, USA
| | - Yvonne W Wu
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Initiative for Pediatric Drug and Device Development, San Francisco, California, USA
| | - Donna M Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Emin Maltepe
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Initiative for Pediatric Drug and Device Development, San Francisco, California, USA
- Department of Biomedical Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
24
|
[Sex differences in clinical outcomes of extremely preterm infants/extremely low birth weight infants: a propensity score matching study]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:514-520. [PMID: 35644191 PMCID: PMC9154374 DOI: 10.7499/j.issn.1008-8830.2201049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVES To study the effect of sex on the clinical outcome of extremely preterm infants (EPIs)/extremely low birth weight infants (ELBWIs) by propensity score matching. METHODS A retrospective analysis was performed for the medical data of 731 EPIs or ELBWIs who were admitted from January 1, 2011 to December 31, 2020. These infants were divided into two groups: male and female. A propensity score matching analysis was performed at a ratio of 1:1. The matching variables included gestational age, birth weight, percentage of withdrawal from active treatment, percentage of small-for-gestational-age infant, percentage of use of pulmonary surfactant, percentage of 1-minute Apgar score ≤3, percentage of mechanical ventilation, duration of mechanical ventilation, percentage of antenatal use of inadequate glucocorticoids, and percentage of hypertensive disorders in pregnancy. The two groups were compared in the incidence rate of main complications during hospitalization and the rate of survival at discharge. RESULTS Before matching, compared with the female group, the male group had significantly higher incidence rates of neonatal respiratory distress syndrome, bronchopulmonary dysplasia (BPD), severe intraventricular hemorrhage, periventricular leukomalacia, necrotizing enterocolitis, and patent ductus arteriosus (P<0.05), while after matching, the male group only had a significantly higher incidence rate of BPD than the female group (P<0.05). There was no significant difference in the rate of survival at discharge between the two groups before and after matching (P>0.05). CONCLUSIONS Male EPIs/ELBWIs have a higher risk of BPD than female EPIs/ELBWIs, but male and female EPIs/ELBWIs tend to have similar outcomes.
Collapse
|
25
|
McCombe PA. The role of sex and pregnancy in multiple sclerosis: what do we know and what should we do? Expert Rev Neurother 2022; 22:377-392. [PMID: 35354378 DOI: 10.1080/14737175.2022.2060079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is more prevalent in women than in men. The sex of the patient, and pregnancy, are reported to be associated with the clinical features of MS. The mechanism of this is unclear. AREAS COVERED This review summarizes data about sex differences in MS and the role of pregnancy. Possible mechanisms for the effects of sex and pregnancy are summarized, and practical suggestions for addressing these issues are provided. EXPERT OPINION There is considerable interdependence of the variables that are associated with MS. Men have a worse outcome of MS, and this could be due to the same factors that lead to greater incidence of neurodegenerative disease in men. The possible role of parity on the long-term outcome of MS is of interest. Future studies that look at the mechanisms of the effects of the sex of the patient on the outcome of MS are required. However, there are some actions that can be taken without further research. We can concentrate on public health measures that address the modifiable risk factors for MS and ensure that disease is controlled in women who intend to become pregnant and use appropriate disease modifying agents during pregnancy.
Collapse
Affiliation(s)
- Pamela A McCombe
- The University of Queensland, Centre for Clinical Research, Royal Brisbane and Women's Hospital, Herston, Australia
| |
Collapse
|
26
|
Lee JW, Profant M, Wang C. Metabolic Sex Dimorphism of the Brain at the Gene, Cell, and Tissue Level. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:212-220. [PMID: 35017210 DOI: 10.4049/jimmunol.2100853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/09/2021] [Indexed: 12/21/2022]
Abstract
The palpable observation in the sex bias of disease prevalence in the CNS has fascinated scientists for several generations. Brain sex dimorphism has been visualized by imaging and analytical tools at the tissue, cellular, and molecular levels. Recent work highlighted the specificity of such sex bias in the brain and its subregions, offering a unique lens through which disease pathogenesis can be investigated. The brain is the largest consumer of energy in the body and provides a unique metabolic environment for diverse lineages of cells. Immune cells are increasingly recognized as an integral part of brain physiology, and their function depends on metabolic homeostasis. This review focuses on metabolic sex dimorphism in brain tissue, resident, and infiltrating immune cells. In this context, we highlight the relevance of recent advances in metabolomics and RNA sequencing technologies at the single cell resolution and the development of novel computational approaches.
Collapse
Affiliation(s)
- Jun Won Lee
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; and
| | - Martin Profant
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; and.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Chao Wang
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; and .,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Wang B, Kulikowicz E, Koehler RC, Yang ZJ. Neuroprotection in the Striatum of Hypoxic-Ischemic Piglets by Simultaneous Inhibition of Dopamine D1 and Adenosine A2A Receptors. Neonatology 2022; 119:354-360. [PMID: 35477141 PMCID: PMC9117515 DOI: 10.1159/000524207] [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: 09/28/2021] [Accepted: 03/21/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Striatal neurons of term newborns are highly vulnerable to hypoxia-ischemia (H-I). In a piglet model of H-I, a dopamine D1 receptor antagonist and an adenosine A2A receptor antagonist alone preferentially protect striatonigral and striatopallidal neurons, respectively. Here, we tested the hypothesis whether the combined treatment with SCH23390, a D1 receptor antagonist, and SCH58261, an A2A receptor antagonist, is more efficacious than individual D1 and A2A receptor antagonist treatment. METHODS Anesthetized newborn piglets were subjected to sham operation (n = 6) or 40 min of hypoxia and 7 min of airway occlusion. At 5 min of reoxygenation, piglets received the vehicle, SCH23390, SCH58261, or the combined treatment (n = 9 in each group). At 4 days of recovery, the number of viable neurons in the entire putamen was estimated by unbiased stereology. RESULTS Stereological results showed that sham-operated piglets had an estimated 2.9 × 106 neurons in the putamen, and the number of viable neurons in hypoxic-ischemic piglets was significantly reduced by 80% to 0.6 × 106/putamen. Treatment with SCH23390, SCH58261, and the combination increased the numbers of viable neurons to 1.4 × 106/putamen, 1.4 × 106/putamen, and 2.1 × 106/putamen, respectively. Notably, the combined treatment improved neuroprotection compared to individual therapy. CONCLUSION We conclude that simultaneous inhibition of dopamine D1 receptors and adenosine A2A receptors saves more neurons than individual treatment in the highly vulnerable putamen of a large-animal neonatal H-I model.
Collapse
Affiliation(s)
- Bing Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ewa Kulikowicz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zeng-Jin Yang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Sheikh A, Meng X, Kao JPY, Kanold PO. Neonatal Hypoxia-Ischemia Causes Persistent Intracortical Circuit Changes in Layer 4 of Rat Auditory Cortex. Cereb Cortex 2021; 32:2575-2589. [PMID: 34729599 DOI: 10.1093/cercor/bhab365] [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: 05/12/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 11/12/2022] Open
Abstract
The connection between early brain injury and subsequent development of disorders is unknown. Neonatal hypoxia-ischemia (HI) alters circuits associated with subplate neurons (SPNs). SPNs are among the first maturing cortical neurons, project to thalamorecipient layer 4 (L4), and are required for the development of thalamocortical connections. Thus, early HI might influence L4 and such influence might persist. We investigated functional circuits to L4 neurons in neonatal rat HI models of different severities (mild and moderate) shortly after injury and at adolescence. We used laser-scanning photostimulation in slices of auditory cortex during P5-10 and P18-23. Mild injuries did not initially (P6/P7) alter the convergence of excitatory inputs from L2/3, but hyperconnectivity emerged by P8-10. Inputs from L4 showed initial hypoconnectivity which resolved by P8-10. Moderate injuries resulted in initial hypoconnectivity from both layers which resolved by P8-10 and led to persistent strengthening of connections. Inhibitory inputs to L4 cells showed similar changes. Functional changes were mirrored by reduced dendritic complexity. We also observed a persistent increase in similarity of L4 circuits, suggesting that HI interferes with developmental circuit refinement and diversification. Altogether, our results show that neonatal HI injuries lead to persistent changes in intracortical connections.
Collapse
Affiliation(s)
- Aminah Sheikh
- Department of Biology, University of Maryland, College Park, MD 20742, USA.,Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD 20742, USA
| | - Xiangying Meng
- Department of Biology, University of Maryland, College Park, MD 20742, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joseph P Y Kao
- Center for Biomedical Engineering and Technology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Patrick O Kanold
- Department of Biology, University of Maryland, College Park, MD 20742, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
29
|
Rocha R, Andrade L, Alves T, Sá S, Pereira PA, Dulce Madeira M, Cardoso A. Behavioral and brain morphological analysis of non-inflammatory and inflammatory rat models of preterm brain injury. Neurobiol Learn Mem 2021; 185:107540. [PMID: 34673263 DOI: 10.1016/j.nlm.2021.107540] [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: 05/15/2021] [Revised: 09/21/2021] [Accepted: 10/07/2021] [Indexed: 11/18/2022]
Abstract
Investigations using preclinical models of preterm birth have much contributed, together with human neuropathological studies, for advances in our understanding of preterm brain injury. Here, we evaluated whether the neurodevelopmental and behavioral consequences of preterm birth induced by a non-inflammatory model of preterm birth using mifepristone would differ from those after inflammatory prenatal transient hypoxia-ischemia (TSHI) model. Pregnant Wistar rats were either injected with mifepristone, and pups were delivered on embryonic day 21 (ED21 group), or laparotomized on the 18th day of gestation for 60 min of uterine arteries occlusion. Rat pups were tested postnatally for characterization of developmental milestones and, after weaning, they were behaviorally tested for anxiety and for spatial learning and memory. One month later, brains were processed for quantification of doublecortin (DCX)- and neuropeptide Y (NPY)-immunoreactive cells, and cholinergic varicosities in the hippocampus. ED21 rats did not differ from controls with respect to neonatal developmental milestones, anxiety, learning and memory functions, and neurochemical parameters. Conversely, in TSHI rats the development of neonatal reflexes was delayed, the levels of anxiety were reduced, and spatial learning and memory was impaired; in the hippocampus, the total number of DCX and NPY cells was increased, and the density of cholinergic varicosities was reduced. With these results we suggest that a preterm birth, in a non-inflammatory prenatal environment, does not significantly change neonatal development and adult neurologic outcome. On other hand, prenatal hypoxia and ischemia (inflammation) modifies developmental trajectory, learning and memory, neurogenesis, and NPY GABAergic and cholinergic brain systems.
Collapse
Affiliation(s)
- Ruben Rocha
- Department of Biomedicine - Unit of Anatomy, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center of Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal; Pediatric Neurology Department, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, 4050-651 Porto, Portugal; Pediatric Emergency Department, Centro Hospitalar Universitário S. João, 4200-319 Porto, Portugal
| | - Leonardo Andrade
- Department of Biomedicine - Unit of Anatomy, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Tânia Alves
- Department of Biomedicine - Unit of Anatomy, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Susana Sá
- Department of Biomedicine - Unit of Anatomy, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center of Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
| | - Pedro A Pereira
- Department of Biomedicine - Unit of Anatomy, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center of Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
| | - M Dulce Madeira
- Department of Biomedicine - Unit of Anatomy, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center of Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
| | - Armando Cardoso
- Department of Biomedicine - Unit of Anatomy, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Center of Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal.
| |
Collapse
|
30
|
Pharmacokinetic/Pharmacodynamic Modelling of Allopurinol, its Active Metabolite Oxypurinol, and Biomarkers Hypoxanthine, Xanthine and Uric Acid in Hypoxic-Ischemic Encephalopathy Neonates. Clin Pharmacokinet 2021; 61:321-333. [PMID: 34617261 PMCID: PMC8813842 DOI: 10.1007/s40262-021-01068-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2021] [Indexed: 12/04/2022]
Abstract
Background Allopurinol, an xanthine oxidase (XO) inhibitor, is a promising intervention that may provide neuroprotection for neonates with hypoxic-ischemic encephalopathy (HIE). Currently, a double-blind, placebo-controlled study (ALBINO, NCT03162653) is investigating the neuroprotective effect of allopurinol in HIE neonates. Objective The aim of the current study was to establish the pharmacokinetics (PK) of allopurinol and oxypurinol, and the pharmacodynamics (PD) of both compounds on hypoxanthine, xanthine, and uric acid in HIE neonates. The dosage used and the effect of allopurinol in this population, either or not undergoing therapeutic hypothermia (TH), were evaluated. Methods Forty-six neonates from the ALBINO study and two historical clinical studies were included. All doses were administered on the first day of life. In the ALBINO study (n = 20), neonates received a first dose of allopurinol 20 mg/kg, and, in the case of TH (n = 13), a second dose of allopurinol 10 mg/kg. In the historical cohorts (n = 26), neonates (all without TH) received two doses of allopurinol 20 mg/kg in total. Allopurinol and oxypurinol population PK, and their effects on inhibiting conversions of hypoxanthine and xanthine to uric acid, were assessed using nonlinear mixed-effects modelling. Results Allopurinol and oxypurinol PK were described by two sequential one-compartment models with an autoinhibition effect on allopurinol metabolism by oxypurinol. For allopurinol, clearance (CL) was 0.83 L/h (95% confidence interval [CI] 0.62–1.09) and volume of distribution (Vd) was 2.43 L (95% CI 2.25–2.63). For metabolite oxypurinol, CL and Vd relative to a formation fraction (fm) were 0.26 L/h (95% CI 0.23–0.3) and 11 L (95% CI 9.9–12.2), respectively. No difference in allopurinol and oxypurinol CL was found between TH and non-TH patients. The effect of allopurinol and oxypurinol on XO inhibition was described by a turnover model of hypoxanthine with sequential metabolites xanthine and uric acid. The combined allopurinol and oxypurinol concentration at the half-maximal XO inhibition was 0.36 mg/L (95% CI 0.31–0.42). Conclusion The PK and PD of allopurinol, oxypurinol, hypoxanthine, xanthine, and uric acid in neonates with HIE were described. The dosing regimen applied in the ALBINO trial leads to the targeted XO inhibition in neonates treated with or without TH. Supplementary Information The online version contains supplementary material available at 10.1007/s40262-021-01068-0.
Collapse
|
31
|
Umbilical cord blood therapy modulates neonatal hypoxic ischemic brain injury in both females and males. Sci Rep 2021; 11:15788. [PMID: 34349144 PMCID: PMC8338979 DOI: 10.1038/s41598-021-95035-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Preclinical and clinical studies have shown that sex is a significant risk factor for perinatal morbidity and mortality, with males being more susceptible to neonatal hypoxic ischemic (HI) brain injury. No study has investigated sexual dimorphism in the efficacy of umbilical cord blood (UCB) cell therapy. HI injury was induced in postnatal day 10 (PND10) rat pups using the Rice-Vannucci method of carotid artery ligation. Pups received 3 doses of UCB cells (PND11, 13, 20) and underwent behavioural testing. On PND50, brains were collected for immunohistochemical analysis. Behavioural and neuropathological outcomes were assessed for sex differences. HI brain injury resulted in a significant decrease in brain weight and increase in tissue loss in females and males. Females and males also exhibited significant cell death, region-specific neuron loss and long-term behavioural deficits. Females had significantly smaller brains overall compared to males and males had significantly reduced neuron numbers in the cortex compared to females. UCB administration improved multiple aspects of neuropathology and functional outcomes in males and females. Females and males both exhibited injury following HI. This is the first preclinical evidence that UCB is an appropriate treatment for neonatal brain injury in both female and male neonates.
Collapse
|
32
|
Chen B, Zhai Q, Ooi K, Cao Y, Qiao Z. Risk Factors for Hydrocephalus in Neonatal Purulent Meningitis: A Single-Center Retrospective Analysis. J Child Neurol 2021; 36:491-497. [PMID: 33393419 DOI: 10.1177/0883073820978032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Hydrocephalus is a potentially lethal complication of neonatal purulent meningitis. Early detection of hydrocephalus helps to determine optimal treatment, improve prognosis, and reduce financial burden. We aimed to analyze the risk factors for hydrocephalus in neonates with purulent meningitis and discuss the characteristics of the disease. METHODS The records of neonatal purulent meningitis admitted to the Children Hospital of Fudan University from January 2013 to September 2019 were retrospectively included in the study cohort. The data of clinical, laboratory, and cranial magnetic resonance images (MRIs) were collected and analyzed (except discharge data) by univariate analysis, and P values <.05 were further analyzed by multivariate logistic regression. RESULTS A total of 197 children who met the inclusion criteria were enrolled in the study cohort. Overall, 39.6% (78/197) of the patients had positive pathogen cultures, and 60.4% (119/197) of patients had clinical diagnosis of meningitis with negative pathogen cultures. Among 197 children, 67 of them experienced hydrocephalus, and the factors that were significantly associated with hydrocephalus in multivariate analysis were female sex, cerebrospinal fluid glucose <2 mmol/L, periventricular leukomalacia, punctate white matter lesions, and pyogenic intraventricular empyema. Children with hydrocephalus had a lower cure rate of meningitis (31.3% vs 75.4%), and poor discharge outcomes. In addition, they had longer length of hospital stay and higher hospital cost. CONCLUSIONS Female sex, cerebrospinal fluid glucose <2 mmol/L, periventricular leukomalacia, punctate white matter lesions, and pyogenic intraventricular empyema were identified as risk factors for hydrocephalus in neonatal purulent meningitis. Children with hydrocephalus had poor discharge outcomes and increased financial burden on their families.
Collapse
Affiliation(s)
- Bin Chen
- Department of Radiology, 145601Children Hospital of Fudan University, Shanghai, China
| | - Qian Zhai
- Department of Neonatology, 145601Children Hospital of Fudan University, Shanghai, China
| | - Kokwin Ooi
- 12478Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun Cao
- Department of Neonatology, 145601Children Hospital of Fudan University, Shanghai, China
| | - Zhongwei Qiao
- Department of Radiology, 145601Children Hospital of Fudan University, Shanghai, China
| |
Collapse
|
33
|
Why the Hips Remain Stable When the Spine Strays: A Deeper Analysis of the Relationship Between Hip Displacement and Severe Scoliosis in Patients With Cerebral Palsy. J Pediatr Orthop 2021; 41:261-266. [PMID: 33825716 DOI: 10.1097/bpo.0000000000001765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Many patients with spastic quadriplegic cerebral palsy (CP) and severe scoliosis develop hip displacement, whereas others do not. We investigated demographic characteristics, risk factors for CP, and imaging findings associated with nondisplaced hips in patients with CP and severe scoliosis. METHODS We retrospectively analyzed records of 229 patients with spastic quadriplegic CP and severe scoliosis who presented for treatment at our US academic tertiary care hospital between August 2005 and September 2015. Demographic characteristics, risk factors for CP, and brain magnetic resonance imaging (MRI) findings were documented. Patients were classified as Gross Motor Function Classification System (GMFCS) level 4 or higher, with 58% at GMFCS level 5.3. Displaced hips (n=181 patients) were defined as a migration percentage of ≥30% or previous surgery for hip displacement/adductor contractures. Patients who did not meet these criteria were classified as nondisplaced (n=48 patients). We used univariate analysis and multivariate logistic regression to determine associations between patient factors and hip displacement (alpha=0.05). RESULTS Patients born at term (≥37 wk) had 2.5 times the odds [95% confidence interval (CI): 1.3-5.0] of having nondisplaced hips compared with patients born prematurely. Females had 2.0 times the odds (95% CI: 1.0-3.9) of having nondisplaced hips compared with males. Patients with normal brain MRI findings had 9.6 times the odds (95% CI: 2.3-41) of having nondisplaced hips compared with patients with abnormal findings. Hip displacement was not associated with race (P>0.05). CONCLUSIONS Gestational age 37 weeks or above, female sex, and normal brain MRI findings are independently associated with nondisplaced hips in patients with spastic quadriplegic CP and severe scoliosis. These findings direct attention to characteristics that may place patients at greater risk of displacement. Future work may influence preventative screening practices and improve patient counseling regarding the risk of hip displacement. LEVEL OF EVIDENCE Level III-retrospective comparative study.
Collapse
|
34
|
Drastichova Z, Rudajev V, Pallag G, Novotny J. Proteome profiling of different rat brain regions reveals the modulatory effect of prolonged maternal separation on proteins involved in cell death-related processes. Biol Res 2021; 54:4. [PMID: 33557947 PMCID: PMC7871601 DOI: 10.1186/s40659-021-00327-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/25/2021] [Indexed: 01/08/2023] Open
Abstract
Background Early-life stress in the form of maternal separation can be associated with alterations in offspring neurodevelopment and brain functioning. Here, we aimed to investigate the potential impact of prolonged maternal separation on proteomic profiling of prefrontal cortex, hippocampus and cerebellum of juvenile and young adult rats. A special attention was devoted to proteins involved in the process of cell death and redox state maintenance. Methods Long-Evans pups were separated from their mothers for 3 h daily over the first 3 weeks of life (during days 2–21 of age). Brain tissue samples collected from juvenile (22-day-old) and young adult (90-day-old) rats were used for label-free quantitative (LFQ) proteomic analysis. In parallel, selected oxidative stress markers and apoptosis-related proteins were assessed biochemically and by Western blot, respectively. Results In total, 5526 proteins were detected in our proteomic analysis of rat brain tissue. Approximately one tenth of them (586 proteins) represented those involved in cell death processes or regulation of oxidative stress balance. Prolonged maternal separation caused changes in less than half of these proteins (271). The observed alterations in protein expression levels were age-, sex- and brain region-dependent. Interestingly, the proteins detected by mass spectrometry that are known to be involved in the maintenance of redox state were not markedly altered. Accordingly, we did not observe any significant differences between selected oxidative stress markers, such as the levels of hydrogen peroxide, reduced glutathione, protein carbonylation and lipid peroxidation in brain samples from rats that underwent maternal separation and from the corresponding controls. On the other hand, a number of changes were found in cell death-associated proteins, mainly in those involved in the apoptotic and autophagic pathways. However, there were no detectable alterations in the levels of cleaved products of caspases or Bcl-2 family members. Taken together, these data indicate that the apoptotic and autophagic cell death pathways were not activated by maternal separation either in adolescent or young adult rats. Conclusion Prolonged maternal separation can distinctly modulate expression profiles of proteins associated with cell death pathways in prefrontal cortex, hippocampus and cerebellum of juvenile rats and the consequences of early-life stress may last into adulthood and likely participate in variations in stress reactivity. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-021-00327-5.
Collapse
Affiliation(s)
- Zdenka Drastichova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Vladimir Rudajev
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Gergely Pallag
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jiri Novotny
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic.
| |
Collapse
|
35
|
Sanches EF, Dos Santos TM, Odorcyk F, Untertriefallner H, Rezena E, Hoeper E, Avila T, Martini AP, Venturin GT, da Costa JC, Greggio S, Netto CA, Wyse AT. Pregnancy swimming prevents early brain mitochondrial dysfunction and causes sex-related long-term neuroprotection following neonatal hypoxia-ischemia in rats. Exp Neurol 2021; 339:113623. [PMID: 33529673 DOI: 10.1016/j.expneurol.2021.113623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 10/22/2022]
Abstract
Neonatal hypoxia-ischemia (HI) is a major cause of cognitive impairments in infants. Antenatal strategies improving the intrauterine environment can have high impact decreasing pregnancy-derived intercurrences. Physical exercise alters the mother-fetus unity and has been shown to prevent the energetic challenge imposed by HI. This study aimed to reveal neuroprotective mechanisms afforded by pregnancy swimming on early metabolic failure and late cognitive damage, considering animals' sex as a variable. Pregnant Wistar rats were submitted to daily swimming exercise (20' in a tank filled with 32 °C water) during pregnancy. Neonatal HI was performed in male and female pups at postnatal day 7. Electron chain transport, mitochondrial mass and function and ROS formation were assessed in the right brain hemisphere 24 h after HI. From PND45, reference and working spatial memory were tested in the Morris water maze. MicroPET-FDG images were acquired 24 h after injury (PND8) and at PND60, following behavioral analysis. HI induced early energetic failure, decreased enzymatic activity in electron transport chain, increased production of ROS in cortex and hippocampus as well as caused brain glucose metabolism dysfunction and late cognitive impairments. Maternal swimming was able to prevent mitochondrial dysfunction and to improve spatial memory. The intergenerational effects of swimming were sex-specific, since male rats were benefited most. In conclusion, maternal swimming was able to affect the mitochondrial response to HI in the offspring's brains, preserving its function and preventing cognitive damage in a sex-dependent manner, adding relevant information on maternal exercise neuroprotection and highlighting the importance of mitochondria as a therapeutic target for HI neuropathology.
Collapse
Affiliation(s)
- E F Sanches
- Biochemistry Post-graduation Program, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Brazil; Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - T M Dos Santos
- Biochemistry Post-graduation Program, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Brazil; Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - F Odorcyk
- Biochemistry Post-graduation Program, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Brazil; Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - H Untertriefallner
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - E Rezena
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - E Hoeper
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - T Avila
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - A P Martini
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - G T Venturin
- Preclinical Research Center, Brain Institute of Rio Grande do Sul (BraIns), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - J C da Costa
- Preclinical Research Center, Brain Institute of Rio Grande do Sul (BraIns), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - S Greggio
- Preclinical Research Center, Brain Institute of Rio Grande do Sul (BraIns), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - C A Netto
- Biochemistry Post-graduation Program, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Brazil; Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - A T Wyse
- Biochemistry Post-graduation Program, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Brazil; Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
36
|
Arteaga Cabeza O, Zhang Z, Smith Khoury E, Sheldon RA, Sharma A, Zhang F, Slusher BS, Kannan RM, Kannan S, Ferriero DM. Neuroprotective effects of a dendrimer-based glutamate carboxypeptidase inhibitor on superoxide dismutase transgenic mice after neonatal hypoxic-ischemic brain injury. Neurobiol Dis 2020; 148:105201. [PMID: 33271328 PMCID: PMC8351403 DOI: 10.1016/j.nbd.2020.105201] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/28/2020] [Accepted: 11/23/2020] [Indexed: 01/05/2023] Open
Abstract
The result of a deprivation of oxygen and glucose to the brain, hypoxic-ischemic encephalopathy (HIE), remains the most common cause of death and disability in human neonates globally and is mediated by glutamate toxicity and inflammation. We have previously shown that the enzyme glutamate carboxypeptidase (GCPII) is overexpressed in activated microglia in the presence of inflammation in fetal/newborn rabbit brain. We assessed the therapeutic utility of a GCPII enzyme inhibitor called 2-(3-Mercaptopropyl) pentanedioic acid (2MPPA) attached to a dendrimer (D-2MPPA), in order to target activated microglia in an experimental neonatal hypoxia-ischemia (HI) model using superoxide dismutase transgenic (SOD) mice that are often more injured after hypoxia-ischemia than wildtype animals. SOD overexpressing and wild type (WT) mice underwent permanent ligation of the left common carotid artery followed by 50 min of asphyxiation (10% O2) to induce HI injury on postnatal day 9 (P9). Cy5-labeled dendrimers were administered to the mice at 6 h, 24 h or 72 h after HI and brains were evaluated by immunofluorescence analysis 24 h after the injection to visualize microglial localization and uptake over time. Expression of GCPII enzyme was analyzed in microglia 24 h after the HI injury. The expression of pro- and anti-inflammatory cytokines were analyzed 24 h and 72 h post-HI. Brain damage was analyzed histologically 7 days post-HI in the three randomly assigned groups: control (C); hypoxic-ischemic (HI); and HI mice who received a single dose of D-2MPPA 6 h post-HI (HI+D-2MPPA). First, we found that GCPII was overexpressed in activated microglia 24 h after HI in the SOD overexpressing mice. Also, there was an increase in microglial activation 24 h after HI in the ipsilateral hippocampus which was most visible in the SOD+HI group. Dendrimers were mostly taken up by microglia by 24 h post-HI; uptake was more prominent in the SOD+HI mice than in the WT+HI. The inflammatory profile showed significant increase in expression of KC/GRO following injury in SOD mice compared to WT at 24 and 72 h. A greater and significant decrease in KC/GRO was seen in the SOD mice following treatment with D-2MPPA. Seven days after HI, D-2MPPA treatment decreased brain injury in the SOD+HI group, but not in WT+HI. This reduced damage was mainly seen in hippocampus and cortex. Our data indicate that the best time point to administer D-2MPPA is 6 h post-HI in order to suppress the expression of GCPII by 24 h after the damage since dendrimer localization in microglia is seen as early as 6 h with the peak of GCPII upregulation in activated microglia seen at 24 h post-HI. Ultimately, treatment with D-2MPPA at 6 h post-HI leads to a decrease in inflammatory profiles by 24 h and reduction in brain injury in the SOD overexpressing mice.
Collapse
Affiliation(s)
- O Arteaga Cabeza
- Departments of Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Z Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - E Smith Khoury
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - R A Sheldon
- Departments of Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA; Departments of Newborn Brain Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - A Sharma
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - F Zhang
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - B S Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - R M Kannan
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - S Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - D M Ferriero
- Departments of Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA; Departments of Neurology, University of California San Francisco, San Francisco, CA 94158, USA; Departments of Newborn Brain Research Institute, University of California San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
37
|
White M, Connor KL. In Utero HIV Exposure and the Early Nutritional Environment Influence Infant Neurodevelopment: Findings from an Evidenced Review and Meta-Analysis. Nutrients 2020; 12:nu12113375. [PMID: 33147767 PMCID: PMC7692402 DOI: 10.3390/nu12113375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/22/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022] Open
Abstract
The developing brain is especially vulnerable to infection and suboptimal nutrition during the pre- and early postnatal periods. Exposure to maternal human immunodeficiency virus (HIV) infection and antiretroviral therapies (ART) in utero and during breastfeeding can adversely influence infant (neuro) developmental trajectories. How early life nutrition may be optimised to improve neurodevelopmental outcomes for infants who are HIV-exposed has not been well characterised. We conducted an up-to-date evidence review and meta-analysis on the influence of HIV exposure in utero and during breastfeeding, and early life nutrition, on infant neurodevelopmental outcomes before age three. We report that exposure to maternal HIV infection may adversely influence expressive language development, in particular, and these effects may be detectable within the first three years of life. Further, while male infants may be especially vulnerable to HIV exposure, few studies overall reported sex-comparisons, and whether there are sex-dependent effects of HIV exposure on neurodevelopment remains a critical knowledge gap to fill. Lastly, early life nutrition interventions, including daily maternal multivitamin supplementation during the perinatal period, may improve neurodevelopmental outcomes for infants who are HIV-exposed. Our findings suggest that the early nutritional environment may be leveraged to improve early neurodevelopmental trajectories in infants who have been exposed to HIV in utero. A clear understanding of how this environment should be optimised is key for developing targeted nutrition interventions during critical developmental periods in order to mitigate adverse outcomes later in life and should be a priority of future research.
Collapse
|
38
|
Madaan P, Chand P, Linn K, Wanigasinghe J, Lhamu Mynak M, Poudel P, Riikonen R, Kumar A, Dhir P, Negi S, Sahu JK. Management practices for West syndrome in South Asia: A survey study and meta-analysis. Epilepsia Open 2020; 5:461-474. [PMID: 32913954 PMCID: PMC7469760 DOI: 10.1002/epi4.12419] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/24/2020] [Accepted: 07/08/2020] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVES Considering the dearth of literature on West syndrome (WS) from South Asian countries, this study aimed to evaluate the management practices in South Asia by an online survey and meta-analysis. METHODS An online questionnaire was sent to 223 pediatric neurologists/pediatricians in India, Pakistan, Myanmar, Sri Lanka, Bhutan, Nepal, and Bangladesh. Their responses were evaluated and supplemented by a meta-analysis. RESULTS Of 125 responses received (response rate: 56%), around 60% of responders observed male preponderance and an approximate lead-time-to-treatment (LTTT) of 4-12 weeks. The commonest etiology observed was a static structural insult (88.6% of responders). Most commonly used first-line drug (country-wise) was as follows: India-adrenocorticotropin hormone (ACTH, 50%); Pakistan-oral steroids (45.5%); Myanmar, Sri Lanka, and Nepal-oral steroids (94.4%); Bangladesh-ACTH (2/2); Bhutan-vigabatrin (3/5). ACTH and vigabatrin are not available in Myanmar and Nepal. The most commonly used regime for ACTH was maximal-dose-at-initiation-regime in India, Sri Lanka, and Bangladesh and gradually escalating-regime in Pakistan. Maximum dose of prednisolone was variable-most common response from India: 3-4 mg/kg/d; Pakistan, Bhutan, and Bangladesh: 2 mg/kg/d; Sri Lanka, Nepal, and Myanmar: 5-8 mg/kg/d or 60 mg/d. The total duration of hormonal therapy (including tapering) ranged from 4 to 12 weeks (67/91). Most responders considered cessation of spasms for four weeks as complete response (54/111) and advised electroencephalography (EEG; 104/123) to check for hypsarrhythmia resolution. Difficult access to pediatric EEG in Bhutan and Nepal is concerning. More than 95% of responders felt a need for more awareness. The meta-analysis supported the preponderance of male gender (68%; confidence interval [CI]: 64%-73%), structural etiology(80%; CI 73%-86%), longer LTTT (2.4 months; CI 2.1-2.6 months), and low response rate to hormonal therapy(18% and 28% for ACTH and oral steroids respectively) in WS in South Asia. SIGNIFICANCE This study highlights the practices and challenges in the management of WS in South Asia. These include a preponderance of male gender and structural etiology, a longer LTTT, difficult access to pediatric EEG, nonavailability of ACTH and vigabatrin in some countries, and low effectiveness of hormonal therapy in this region.
Collapse
Affiliation(s)
- Priyanka Madaan
- Pediatric Neurology UnitDepartment of PediatricsPostgraduate Institute of Medical Education and ResearchChandigarhIndia
| | | | - Kyaw Linn
- Pediatric Neurology UnitYangon Children HospitalYangonMyanmar
| | | | - Mimi Lhamu Mynak
- Department of PediatricsJigme Dorji Wangchuck National Referral HospitalThimphuBhutan
| | - Prakash Poudel
- Department of PediatricsB.P. Koirala Institute of Health SciencesDharanNepal
| | - Raili Riikonen
- Child NeurologyChildren's HospitalUniversity of Eastern Finland and Kuopio University HospitalKuopioFinland
| | - Amit Kumar
- Department of NeurologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Pooja Dhir
- Pediatric Neurology UnitDepartment of PediatricsPostgraduate Institute of Medical Education and ResearchChandigarhIndia
| | - Sandeep Negi
- Pediatric Neurology UnitDepartment of PediatricsPostgraduate Institute of Medical Education and ResearchChandigarhIndia
| | - Jitendra Kumar Sahu
- Pediatric Neurology UnitDepartment of PediatricsPostgraduate Institute of Medical Education and ResearchChandigarhIndia
| |
Collapse
|
39
|
Le Dieu-Lugon B, Dupré N, Legouez L, Leroux P, Gonzalez BJ, Marret S, Leroux-Nicollet I, Cleren C. Why considering sexual differences is necessary when studying encephalopathy of prematurity through rodent models. Eur J Neurosci 2019; 52:2560-2574. [PMID: 31885096 DOI: 10.1111/ejn.14664] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/25/2019] [Accepted: 12/05/2019] [Indexed: 12/01/2022]
Abstract
Preterm birth is a high-risk factor for the development of gray and white matter abnormalities, referred to as "encephalopathy of prematurity," that may lead to life-long motor, cognitive, and behavioral impairments. The prevalence and clinical outcomes of encephalopathy of prematurity differ between sexes, and elucidating the underlying biological basis has become a high-priority challenge. Human studies are often limited to assessment of brain region volumes by MRI, which does not provide much information about the underlying mechanisms of lesions related to very preterm birth. However, models using KO mice or pharmacological manipulations in rodents allow relevant observations to help clarify the mechanisms of injury sustaining sex-differential vulnerability. This review focuses on data obtained from mice aged P1-P5 or rats aged P3 when submitted to cerebral damage such as hypoxia-ischemia, as their brain lesions share similarities with lesion patterns occurring in very preterm human brain, before 32 gestational weeks. We first report data on the mechanisms underlying the development of sexual brain dimorphism in rodent, focusing on the hippocampus. In the second part, we describe sex specificities of rodent models of encephalopathy of prematurity (RMEP), focusing on mechanisms underlying differences in hippocampal vulnerability. Finally, we discuss the relevance of these RMEP. Together, this review highlights the need to systematically search for potential effects of sex when studying the mechanisms underlying deficits in RMEP in order to design effective sex-specific medical interventions in human preterms.
Collapse
Affiliation(s)
- Bérénice Le Dieu-Lugon
- Normandy Centre for Genomic and Personalized Medicine, UNIROUEN, Inserm U1245 Team 4, Normandy University, Rouen, France
| | - Nicolas Dupré
- Normandy Centre for Genomic and Personalized Medicine, UNIROUEN, Inserm U1245 Team 4, Normandy University, Rouen, France
| | - Lou Legouez
- Normandy Centre for Genomic and Personalized Medicine, UNIROUEN, Inserm U1245 Team 4, Normandy University, Rouen, France
| | - Philippe Leroux
- Normandy Centre for Genomic and Personalized Medicine, UNIROUEN, Inserm U1245 Team 4, Normandy University, Rouen, France
| | - Bruno J Gonzalez
- Normandy Centre for Genomic and Personalized Medicine, UNIROUEN, Inserm U1245 Team 4, Normandy University, Rouen, France
| | - Stéphane Marret
- Normandy Centre for Genomic and Personalized Medicine, UNIROUEN, Inserm U1245 Team 4, Normandy University, Rouen, France.,Department of Neonatal Paediatrics and Intensive Care, Rouen University Hospital, Rouen, France
| | - Isabelle Leroux-Nicollet
- Normandy Centre for Genomic and Personalized Medicine, UNIROUEN, Inserm U1245 Team 4, Normandy University, Rouen, France
| | - Carine Cleren
- Normandy Centre for Genomic and Personalized Medicine, UNIROUEN, Inserm U1245 Team 4, Normandy University, Rouen, France
| |
Collapse
|