Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Front Neurosci ; 15: 744840, 2021.
Article in English | MEDLINE | ID: mdl-34630028

ABSTRACT

This comprehensive review focuses on our current understanding of the proposed physiological and pathological functions of extracellular vesicles (EVs) in the developing brain. Furthermore, since EVs have attracted great interest as potential novel cell-free therapeutics, we discuss advances in the knowledge of stem cell- and astrocyte-derived EVs in relation to their potential for protection and repair following perinatal brain injury. This review identified 13 peer-reviewed studies evaluating the efficacy of EVs in animal models of perinatal brain injury; 12/13 utilized mesenchymal stem cell-derived EVs (MSC-EVs) and 1/13 utilized astrocyte-derived EVs. Animal model, method of EV isolation and size, route, timing, and dose administered varied between studies. Notwithstanding, EV treatment either improved and/or preserved perinatal brain structures both macroscopically and microscopically. Additionally, EV treatment modulated inflammatory responses and improved brain function. Collectively this suggests EVs can ameliorate, or repair damage associated with perinatal brain injury. These findings warrant further investigation to identify the optimal cell numbers, source, and dosage regimens of EVs, including long-term effects on functional outcomes.

2.
Int J Mol Sci ; 22(12)2021 Jun 19.
Article in English | MEDLINE | ID: mdl-34205464

ABSTRACT

BACKGROUND: Toll-like receptor (TLR) agonists are key immunomodulatory factors that can markedly ameliorate or exacerbate hypoxic-ischemic brain injury. We recently demonstrated that central infusion of the TLR7 agonist Gardiquimod (GDQ) following asphyxia was highly neuroprotective after 3 days but not 7 days of recovery. We hypothesize that this apparent transient neuroprotection is associated with modulation of seizure-genic processes and hemodynamic control. METHODS: Fetuses received sham asphyxia or asphyxia induced by umbilical cord occlusion (20.9 ± 0.5 min) and were monitored continuously for 7 days. GDQ 3.34 mg or vehicle were infused intracerebroventricularly from 1 to 4 h after asphyxia. RESULTS: GDQ infusion was associated with sustained moderate hypertension that resolved after 72 h recovery. Electrophysiologically, GDQ infusion was associated with reduced number and burden of postasphyxial seizures in the first 18 h of recovery (p < 0.05). Subsequently, GDQ was associated with induction of slow rhythmic epileptiform discharges (EDs) from 72 to 96 h of recovery (p < 0.05 vs asphyxia + vehicle). The total burden of EDs was associated with reduced numbers of neurons in the caudate nucleus (r2 = 0.61, p < 0.05) and CA1/2 hippocampal region (r2 = 0.66, p < 0.05). CONCLUSION: These data demonstrate that TLR7 activation by GDQ modulated blood pressure and suppressed seizures in the early phase of postasphyxial recovery, with subsequent prolonged induction of epileptiform activity. Speculatively, this may reflect delayed loss of early protection or contribute to differential neuronal survival in subcortical regions.


Subject(s)
Aminoquinolines/therapeutic use , Hypoxia-Ischemia, Brain/drug therapy , Imidazoles/therapeutic use , Seizures/prevention & control , Toll-Like Receptor 7/agonists , Aminoquinolines/pharmacology , Animals , Blood Pressure/drug effects , Drug Evaluation, Preclinical , Electroencephalography , Female , Fetal Therapies/methods , Hypoxia-Ischemia, Brain/complications , Imidazoles/pharmacology , Pregnancy , Premature Birth , Seizures/etiology , Sheep
3.
Stem Cells Transl Med ; 10(3): 427-440, 2021 03.
Article in English | MEDLINE | ID: mdl-33103374

ABSTRACT

There is increasing evidence that administration of many types of stem cells, including human amnion epithelial cells (hAECs), can reduce hypoxic-ischemic injury, including in the perinatal brain. However, the therapeutic window for single dose treatment is not known. We compared the effects of early and delayed intracerebroventricular administration of hAECs in fetal sheep at 0.7 gestation on brain injury induced by 25 minutes of complete umbilical cord occlusion (UCO) or sham occlusion. Fetuses received either 1 × 106 hAECs or vehicle alone, as an infusion over 1 hour, either 2 or 24 hours after UCO. Fetuses were killed for brain histology at 7 days post-UCO. hAEC infusion at both 2 and 24 hours had dramatic anti-inflammatory and anti-gliotic effects, including significantly attenuating the increase in microglia after UCO in the white and gray matter and the number of astrocytes in the white matter. Both protocols partially improved myelination, but had no effect on total or immature/mature numbers of oligodendrocytes. Neuronal survival in the hippocampus was increased by hAEC infusion at either 2 or 24 hours, whereas only hAECs at 24 hours were associated with improved neuronal survival in the striatum and thalamus. Neither protocol improved recovery of electroencephalographic (EEG) power. These data suggest that a single infusion of hAECs is anti-inflammatory, anti-gliotic, and neuroprotective in preterm fetal sheep when given up to 24 hours after hypoxia-ischemia, but was associated with limited white matter protection after 7 days recovery and no improvement in the recovery of EEG power.


Subject(s)
Amnion , Gliosis , Hypoxia, Brain/therapy , Stem Cell Transplantation , Amnion/cytology , Animals , Epithelial Cells/transplantation , Female , Fetus , Gliosis/therapy , Humans , Pregnancy , Sheep , Stem Cells , Umbilical Cord
4.
Sci Rep ; 10(1): 6904, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32327682

ABSTRACT

In the preterm brain, accumulating evidence suggests toll-like receptors (TLRs) are key mediators of the downstream inflammatory pathways triggered by hypoxia-ischemia (HI), which have the potential to exacerbate or ameliorate injury. Recently we demonstrated that central acute administration of the TLR7 agonist Gardiquimod (GDQ) confers neuroprotection in the preterm fetal sheep at 3 days post-asphyxial recovery. However, it is unknown whether GDQ can afford long-term protection. To address this, we examined the long-term effects of GDQ. Briefly, fetal sheep (0.7 gestation) received sham asphyxia or asphyxia induced by umbilical cord occlusion, and were studied for 7 days recovery. Intracerebroventricular (ICV) infusion of GDQ (total dose 3.34 mg) or vehicle was performed from 1-4 hours after asphyxia. GDQ was associated with a robust increase in concentration of tumor necrosis factor-(TNF)-α in the fetal plasma, and interleukin-(IL)-10 in both the fetal plasma and cerebrospinal fluid. GDQ did not significantly change the number of total and immature/mature oligodendrocytes within the periventricular and intragyral white matter. No changes were observed in astroglial and microglial numbers and proliferating cells in both white matter regions. GDQ increased neuronal survival in the CA4 region of the hippocampus, but was associated with exacerbated neuronal injury within the caudate nucleus. In conclusion, our data suggest delayed acute ICV administration of GDQ after severe HI in the developing brain may not support long-term neuroprotection.


Subject(s)
Aminoquinolines/administration & dosage , Aminoquinolines/therapeutic use , Asphyxia/embryology , Brain/pathology , Fetus/pathology , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Premature Birth/drug therapy , Toll-Like Receptor 7/agonists , Aminoquinolines/pharmacology , Animals , Apoptosis/drug effects , Arterial Pressure/drug effects , Asphyxia/blood , Asphyxia/cerebrospinal fluid , Asphyxia/physiopathology , Blood Gas Analysis , Body Weight/drug effects , Brain/drug effects , Caspase 3/metabolism , Cell Polarity/drug effects , Cell Proliferation/drug effects , Cytokines/blood , Cytokines/cerebrospinal fluid , Female , Fetus/drug effects , Heart Rate/drug effects , Hydrogen-Ion Concentration , Imidazoles/pharmacology , Injections, Intraventricular , Male , Metabolome/drug effects , Neurons/drug effects , Neurons/pathology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Organ Size/drug effects , Premature Birth/blood , Premature Birth/cerebrospinal fluid , Premature Birth/physiopathology , Sheep , Time Factors , Umbilical Cord/pathology
5.
J Neuroinflammation ; 17(1): 92, 2020 Mar 23.
Article in English | MEDLINE | ID: mdl-32293473

ABSTRACT

BACKGROUND: Increased circulating levels of tumor necrosis factor (TNF) are associated with greater risk of impaired neurodevelopment after preterm birth. In this study, we tested the hypothesis that systemic TNF inhibition, using the soluble TNF receptor Etanercept, would attenuate neuroinflammation in preterm fetal sheep exposed to lipopolysaccharide (LPS). METHODS: Chronically instrumented preterm fetal sheep at 0.7 of gestation were randomly assigned to receive saline (control; n = 7), LPS infusion (100 ng/kg i.v. over 24 h then 250 ng/kg/24 h for 96 h plus 1 µg LPS boluses at 48, 72, and 96 h, to induce inflammation; n = 8) or LPS plus two i.v. infusions of Etanercept (2 doses, 5 mg/kg infused over 30 min, 48 h apart) started immediately before LPS-exposure (n = 8). Sheep were killed 10 days after starting infusions, for histology. RESULTS: LPS boluses were associated with increased circulating TNF, interleukin (IL)-6 and IL-10, electroencephalogram (EEG) suppression, hypotension, tachycardia, and increased carotid artery perfusion (P < 0.05 vs. control). In the periventricular and intragyral white matter, LPS exposure increased gliosis, TNF-positive cells, total oligodendrocytes, and cell proliferation (P < 0.05 vs control), but did not affect myelin expression or numbers of neurons in the cortex and subcortical regions. Etanercept delayed the rise in circulating IL-6, prolonged the increase in IL-10 (P < 0.05 vs. LPS), and attenuated EEG suppression, hypotension, and tachycardia after LPS boluses. Histologically, Etanercept normalized LPS-induced gliosis, and increase in TNF-positive cells, proliferation, and total oligodendrocytes. CONCLUSION: TNF inhibition markedly attenuated white matter gliosis but did not affect mature oligodendrocytes after prolonged systemic inflammation in preterm fetal sheep. Further studies of long-term brain maturation are now needed.


Subject(s)
Gliosis/drug therapy , Inflammation Mediators/antagonists & inhibitors , Premature Birth/drug therapy , Tumor Necrosis Factor Inhibitors/administration & dosage , Tumor Necrosis Factor-alpha/antagonists & inhibitors , White Matter/drug effects , Animals , Etanercept/administration & dosage , Female , Fetus , Gliosis/metabolism , Inflammation Mediators/metabolism , Infusions, Intravenous , Pregnancy , Premature Birth/metabolism , Sheep , Tumor Necrosis Factor-alpha/metabolism , White Matter/metabolism
6.
J Physiol ; 598(5): 999-1015, 2020 03.
Article in English | MEDLINE | ID: mdl-31912503

ABSTRACT

KEY POINTS: Recombinant human erythropoietin (rEpo) is neuroprotective in immature animals, but it is unclear whether the combination of high-dose rEpo therapy with therapeutic hypothermia can further improve outcomes. Hypothermia and rEpo independently improved neuronal survival, with greater improvement with hypothermia, and similarly reduced numbers of caspase-3 positive cells and reactive microglia after 7 days recovery. Hypothermia, but not rEpo, was associated with markedly improved EEG power, whereas both interventions improved recovery of EEG frequency. There was no significant improvement in any outcome after combined rEpo and hypothermia compared with hypothermia alone, and of concern, the combination was associated with increased numbers of cortical caspase-3-positive cells compared with ischaemia-hypothermia. These data suggest that the mechanisms of neuroprotection with hypothermia and rEpo overlap and, thus, high-dose rEpo infusion does not appear to be an effective adjunct therapy for therapeutic hypothermia. ABSTRACT: Therapeutic hypothermia for hypoxic-ischaemic encephalopathy (HIE) provides incomplete neuroprotection. Recombinant human erythropoietin (rEpo) is neuroprotective in immature animals, but it is unclear whether adjunct rEpo therapy with therapeutic hypothermia can further improve outcomes. Near-term fetal sheep received sham-ischaemia (n = 9) or global cerebral ischaemia for 30 min (ischaemia-vehicle, n = 8), followed by intravenous infusion of rEpo (ischaemia-Epo, n = 8; 5000 U/kg loading dose, then 833.3 U/kg/h), cerebral hypothermia (ischaemia-hypothermia, n = 8), or rEpo plus hypothermia (ischaemia-Epo-hypothermia, n = 8), from 3 to 72 h post ischaemia. Fetal brains were collected 7 days after cerebral ischaemia. Cerebral ischaemia was associated with severe neuronal loss and microglial induction in the parasagittal cortex and subcortical regions. Hypothermia reduced overall neuronal loss, cortical caspase-3 and reactive microglia in the striatum and cortex, with greater recovery of electroencephalographic (EEG) power and spectral edge (SEF) from 48 h onwards. rEpo independently improved neuronal survival in the parasagittal cortex, hippocampal CA4 and thalamus, and reduced cortical caspase-3 and activated microglia in striatal and cortical areas, with greater SEF from 120 h onwards. However, ischaemia-Epo-hypothermia did not further improve outcomes compared with ischaemia-hypothermia and was associated with increased numbers of cortical caspase-3-positive cells. These findings suggest that although delayed, prolonged treatment with both hypothermia and rEpo are independently neuroprotective, they have overlapping anti-inflammatory and anti-apoptotic mechanisms, such that the delayed, high-dose rEpo infusion for 3 days did not materially augment neuroprotection with therapeutic hypothermia.


Subject(s)
Erythropoietin , Hypothermia, Induced , Hypoxia-Ischemia, Brain , Animals , Electroencephalography , Fetus , Hypoxia-Ischemia, Brain/therapy , Sheep
7.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R369-R378, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31913689

ABSTRACT

Activation of Toll-like receptors (TLRs) after hypoxic-ischemic brain injury can exacerbate injury but also alleviate cell loss, as recently demonstrated with the TLR7 agonist Gardiquimod (GDQ). However, TLR agonists also modulate vascular function and neuronal excitability. Thus, we examined the effects of TLR7 activation with GDQ on cardiovascular function and seizures after asphyxia in preterm fetal sheep at 0.7 gestation (104 days, term ∼147 days). Fetuses received sham asphyxia or asphyxia induced by umbilical cord occlusion for 25 min or asphyxia followed by a continuous intracerebroventricular infusion of 3.34 mg of GDQ from 1 to 4 h after asphyxia. Fetuses were monitored continuously for 72 h postasphyxia. GDQ treatment was associated with sustained, moderate hypertension for 72 h (P < 0.05), with a transient increase in heart rate. Electroencephalographic (EEG) power was suppressed for the entire postasphyxial period in both groups, whereas EEG spectral edge transiently increased during the GDQ infusion compared with asphyxia alone (P < 0.05), with higher ß- and lower δ-EEG frequencies (P < 0.05). This increase in EEG frequency was not related to epileptiform activity. After the GDQ infusion, there was earlier onset of high-amplitude stereotypic evolving seizures, with increased numbers of seizures and seizure burden (P < 0.05). Hemodynamic function and seizure activity are important indices of preterm wellbeing. These data highlight the importance of physiological monitoring during preclinical testing of potential neuroprotective strategies.


Subject(s)
Aminoquinolines/toxicity , Asphyxia Neonatorum/drug therapy , Hypertension/chemically induced , Imidazoles/toxicity , Neuroprotective Agents/toxicity , Premature Birth , Seizures/chemically induced , Tachycardia/chemically induced , Toll-Like Receptor 7/agonists , Animals , Animals, Newborn , Asphyxia Neonatorum/physiopathology , Blood Pressure/drug effects , Brain Waves/drug effects , Disease Models, Animal , Gestational Age , Heart Rate/drug effects , Hypertension/physiopathology , Risk Assessment , Seizures/physiopathology , Sheep, Domestic , Signal Transduction , Tachycardia/physiopathology , Time Factors
8.
Sci Rep ; 9(1): 9562, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31267031

ABSTRACT

Preterm brain injury is highly associated with inflammation, which is likely related in part to sterile responses to hypoxia-ischemia. We have recently shown that neuroprotection with inflammatory pre-conditioning in the immature brain is associated with induction of toll-like receptor 7 (TLR7). We therefore tested the hypothesis that central administration of a synthetic TLR7 agonist, gardiquimod (GDQ), after severe hypoxia-ischemia in preterm-equivalent fetal sheep would improve white and gray matter recovery. Fetal sheep at 0.7 of gestation received sham asphyxia or asphyxia induced by umbilical cord occlusion for 25 minutes, followed by a continuous intracerebroventricular infusion of GDQ or vehicle from 1 to 4 hours (total dose 1.8 mg/kg). Sheep were killed 72 hours after asphyxia for histology. GDQ significantly improved survival of immature and mature oligodendrocytes (2',3'-cyclic-nucleotide 3'-phosphodiesterase, CNPase) and total oligodendrocytes (oligodendrocyte transcription factor 2, Olig-2) within the periventricular and intragyral white matter. There were reduced numbers of cells showing cleaved caspase-3 positive apoptosis and astrogliosis (glial fibrillary acidic protein, GFAP) in both white matter regions. Neuronal survival was increased in the dentate gyrus, caudate and medial thalamic nucleus. Central infusion of GDQ was associated with a robust increase in fetal plasma concentrations of the anti-inflammatory cytokines, interferon-ß (IFN-ß) and interleukin-10 (IL-10), with no significant change in the concentration of the pro-inflammatory cytokine, tumor necrosis factor-α (TNF-α). In conclusion, delayed administration of the TLR7 agonist, GDQ, after severe hypoxia-ischemia in the developing brain markedly ameliorated white and gray matter damage, in association with upregulation of anti-inflammatory cytokines. These data strongly support the hypothesis that modulation of secondary inflammation may be a viable therapeutic target for injury of the preterm brain.


Subject(s)
Asphyxia/metabolism , Gray Matter/drug effects , Gray Matter/metabolism , Neuroprotective Agents/administration & dosage , Premature Birth/metabolism , Toll-Like Receptor 7/agonists , White Matter/drug effects , White Matter/metabolism , Animals , Biomarkers , Blood Gas Analysis , Caspase 3/metabolism , Cell Count , Cell Proliferation , Cytokines/blood , Cytokines/metabolism , Electroencephalography , Hypoxia-Ischemia, Brain/etiology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/prevention & control , Immunohistochemistry , Infusions, Intraventricular , Microglia/drug effects , Microglia/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Sheep , Time Factors
9.
Front Physiol ; 10: 227, 2019.
Article in English | MEDLINE | ID: mdl-30984006

ABSTRACT

In human beings the immature brain is highly plastic and depending on the stage of gestation is particularly vulnerable to a range of insults that if sufficiently severe, can result in long-term motor, cognitive and behavioral impairment. With improved neonatal care, the incidence of major motor deficits such as cerebral palsy has declined with prematurity. Unfortunately, however, milder forms of injury characterized by diffuse non-cystic white matter lesions within the periventricular region and surrounding white matter, involving loss of oligodendrocyte progenitors and subsequent axonal hypomyelination as the brain matures have not. Existing therapeutic options for treatment of preterm infants have proved inadequate, partly owing to an incomplete understanding of underlying post-injury cellular and molecular changes that lead to poor neurodevelopmental outcomes. This has reinforced the need to improve our understanding of brain plasticity, explore novel solutions for the development of protective strategies, and identify biomarkers. Compelling evidence exists supporting the involvement of microRNAs (miRNAs), a class of small non-coding RNAs, as important post-transcriptional regulators of gene expression with functions including cell fate specification and plasticity of synaptic connections. Importantly, miRNAs are differentially expressed following brain injury, and can be packaged within exosomes/extracellular vesicles, which play a pivotal role in assuring their intercellular communication and passage across the blood-brain barrier. Indeed, an increasing number of investigations have examined the roles of specific miRNAs following injury and regeneration and it is apparent that this field of research could potentially identify protective therapeutic strategies to ameliorate perinatal brain injury. In this review, we discuss the most recent findings of some important miRNAs in relation to the development of the brain, their dysregulation, functions and regulatory roles following brain injury, and discuss how these can be targeted either as biomarkers of injury or neuroprotective agents.

10.
J Cereb Blood Flow Metab ; 39(2): 223-239, 2019 02.
Article in English | MEDLINE | ID: mdl-28895475

ABSTRACT

Perinatal hypoxic-ischemic (HI) brain injury remains highly associated with neurodevelopmental disability after preterm birth. There is increasing evidence that disability is linked with impaired white matter maturation, but there is no specific treatment. In this study, we evaluated whether, in preterm fetal sheep, delayed intranasal infusion of human amnion epithelial cells (hAECs) given 1, 3 and 10 days after severe HI, induced by umbilical cord occlusion for 25 min, can restore white matter maturation or reduce delayed cell loss. After 21 days recovery, asphyxia was associated with reduced electroencephalographic (EEG) maturation, brain weight and cortical area, impaired maturation of oligodendrocytes (OLs), no significant loss of total OLs but a marked reduction in immature/mature OLs and reduced myelination. Intranasal infusion of hAECs was associated with improved brain weight and restoration of immature/mature OLs and fractional area of myelin basic protein, with reduced microglia and astrogliosis. Cortical EEG frequency distribution was partially improved, with reduced loss of cortical area, and attenuated cleaved-caspase-3 expression and microgliosis. Neuronal survival in deep grey matter nuclei was improved, with reduced microglia, astrogliosis and cleaved-caspase-3-positive apoptosis. These findings suggest that delayed intranasal hAEC administration has potential to alleviate chronic dysmaturation after perinatal HI.


Subject(s)
Amnion , Asphyxia Neonatorum , Cerebral Cortex , Epithelial Cells/transplantation , Neurons , Animals , Animals, Newborn , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/pathology , Asphyxia Neonatorum/physiopathology , Asphyxia Neonatorum/therapy , Caspase 3/metabolism , Cell Survival , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Disease Models, Animal , Electroencephalography , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Heterografts , Humans , Neurons/metabolism , Neurons/pathology , Sheep
11.
Front Biosci (Landmark Ed) ; 23(12): 2204-2226, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29772556

ABSTRACT

Brain damage resulting from perinatal hypoxia-ischemia evolves slowly over time. While a small number of brain cells may die during a sufficiently profound period of hypoxia-ischemia, many will show initial recovery during a "latent" phase characterized by actively suppressed neural metabolism and activity. Critically, this transient recovery may be followed after ~6 hours by a phase of secondary deterioration, with delayed seizures, failure of mitochondrial function, cytotoxic edema, and bulk cell death over ~72 hours. This is followed by a tertiary phase of remodeling and recovery. Understanding the mechanisms of injury that occur during each phase may allow for the development of more targeted treatments. This review discusses the mechanisms of injury that occur during the primary, latent, secondary and tertiary phases of injury and potential treatments that target one or more of these phases. Treatment during the latent phase has the greatest potential to prevent injury. In the secondary phase of injury, anticonvulsants can attenuate seizures but show limited neuroprotection. By contrast, there is increasing preclinical evidence that neurorestorative therapies may improve long-term outcomes.


Subject(s)
Brain Injuries/prevention & control , Fetal Diseases/prevention & control , Hypoxia-Ischemia, Brain/therapy , Perinatal Care/methods , Animals , Brain Injuries/etiology , Brain Injuries/physiopathology , Female , Fetal Diseases/etiology , Fetal Diseases/physiopathology , Humans , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/physiopathology , Pregnancy , Time Factors
12.
Dev Med Child Neurol ; 60(2): 126-133, 2018 02.
Article in English | MEDLINE | ID: mdl-29194585

ABSTRACT

Children surviving preterm birth have a high risk of disability, particularly cognitive and learning problems. There is extensive clinical and experimental evidence that disability is now primarily related to dysmaturation of white and gray matter, defined by failure of oligodendrocyte maturation and neuronal dendritic arborization, rather than cell death alone. The etiology of this dysmaturation is multifactorial, with contributions from hypoxia-ischemia, infection/inflammation and barotrauma. Intriguingly, these factors can interact to both increase and decrease damage. In this review we summarize preclinical and clinical evidence that all of these factors trigger secondary or chronic inflammation and gliosis. Thus, we hypothesize that these shared pathological features play a key role in a final common pathway that leads to the impaired neural maturation and connectivity and cognitive/motor impairments that are commonly observed in infants born preterm. This raises the possibility that secondary or chronic inflammation may be a viable therapeutic target for delayed interventions to improve neurodevelopmental outcomes after preterm birth. WHAT THIS PAPER ADDS: Hypoxia-ischemia, infection/inflammation, and barotrauma/volutrauma all contribute to preterm brain injury. Multiple different triggers of preterm brain injury are associated with central nervous system dysmaturation. Secondary brain inflammation may be a viable target to improve neurodevelopment after preterm birth.


Subject(s)
Hypoxia-Ischemia, Brain/physiopathology , Infant, Premature, Diseases/physiopathology , Inflammation/etiology , Animals , Humans
13.
Hormones (Athens) ; 16(3): 282-290, 2017 Jul.
Article in English | MEDLINE | ID: mdl-29278514

ABSTRACT

OBJECTIVE: To investigate whether maternal serum concentrations of placental growth hormone (GH-V), insulin-like growth factor (IGF) 1 and 2, and IGF binding proteins (IGFBP) 1 and 3 were altered in pregnancies complicated by gestational diabetes mellitus (GDM). METHOD: In a nested case-control study, GDM cases (n=28) and matched controls (n=28) were selected from the Screening for Pregnancy Endpoints (SCOPE) biobank in Auckland, New Zealand. Maternal serum hormone concentrations at 20 weeks of gestation were determined by enzyme-linked immunosorbent assay (ELISA). RESULTS: There was no significant difference in maternal serum GH-V concentration in the GDM group compared to the control group (1.64 ± 0.11 ng/ml vs. 1.38 ± 0.10 ng/ml, p=0.079). However, GDM cases who delivered large for gestational age (LGA) babies had significantly higher serum GH-V concentrations compared to non-diabetic control cases. Maternal IGF-1 concentrations in GDM pregnancies were significantly higher than in controls (275.7 ± 11.5 ng/ml vs. 218.5 ± 11.1 ng/ml, p <0.001). Maternal IGFBP-1 concentrations were significantly lower in GDM pregnancies than in controls (41.04 ± 3.42 ng/ml vs. 67.58 ± 6.17 ng/ml, p <0.001). There were no significant differences in serum IGF-2 and IGFBP-3 concentrations between groups. CONCLUSION: Concentrations of IGF-1 and IGFBP-1 in maternal serum were altered in GDM pregnancies compared to controls, suggesting that the IGF axis plays a role in the development of this condition. GH-V may be associated with macrosomia as increased maternal GH-V was observed in GDM cases who delivered LGA babies.


Subject(s)
Diabetes, Gestational/blood , Insulin-Like Growth Factor Binding Proteins/blood , Placenta Growth Factor/blood , Pregnancy Trimester, Second/blood , Somatomedins/metabolism , Adult , Case-Control Studies , Female , Humans , Pregnancy
14.
Pregnancy Hypertens ; 10: 149-154, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29153669

ABSTRACT

OBJECTIVE: To investigate whether maternal serum concentrations of placental growth hormone (GH-V), insulin-like growth factor (IGF) 1 and 2, and IGF binding proteins (IGFBP) 1 and 3 were altered in pregnancies complicated by later preeclampsia (PE). STUDY DESIGN: In a nested case-control study, PE cases (n=71) and matched controls (n=71) were selected from the Screening for Pregnancy Endpoints (SCOPE) biobank in Auckland, New Zealand. Maternal serum hormone concentrations at 20weeks of gestation were determined by ELISA. RESULTS: We found that maternal serum GH-V concentration at 20weeks of gestation was unaltered in the PE group, compared to the control group (median, 1.78ng/ml vs. 1.65ng/ml, p=0.884). Maternal IGF-1 and IGFBP-3 concentrations and the IGF-1/IGFBP-3 ratio in PE pregnancies were significantly higher than in controls (median, 253.1ng/ml vs. 204.3ng/ml, p<0.0001; 8535ng/ml vs. 7711ng/ml, p=0.0023; 0.032vs. 0.026, p<0.0001, respectively), whereas maternal IGFBP-1 concentration was significantly lower in PE pregnancies than in controls (median, 34.85ng/ml vs. 48.92ng/ml, p=0.0006). CONCLUSION: Our findings suggest a potential role of IGFs and IGFBPs in the prediction of pregnancies complicated by PE. However, the maternal serum concentration of GH-V at 20weeks' gestation is unlikely to be useful in the early prediction of PE.


Subject(s)
Biomarkers/blood , Pre-Eclampsia/metabolism , Pregnancy Trimester, Second , Adult , Enzyme-Linked Immunosorbent Assay , Female , Humans , Insulin-Like Growth Factor Binding Protein 1/blood , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/metabolism , Placental Hormones/blood , Pre-Eclampsia/blood , Pre-Eclampsia/diagnosis , Predictive Value of Tests , Pregnancy , Prenatal Diagnosis
15.
J Cereb Blood Flow Metab ; 37(3): 1080-1094, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27207167

ABSTRACT

Perinatal asphyxia in preterm infants remains a significant contributor to abnormal long-term neurodevelopmental outcomes. Recombinant human erythropoietin has potent non-haematopoietic neuroprotective properties, but there is limited evidence for protection in the preterm brain. Preterm (0.7 gestation) fetal sheep received sham asphyxia (sham occlusion) or asphyxia induced by umbilical cord occlusion for 25 min, followed by an intravenous infusion of vehicle (occlusion-vehicle) or recombinant human erythropoietin (occlusion-Epo, 5000 international units by slow push, then 832.5 IU/h), starting 30 min after asphyxia and continued until 72 h. Recombinant human erythropoietin reduced neuronal loss and numbers of caspase-3-positive cells in the striatal caudate nucleus, CA3 and dentate gyrus of the hippocampus, and thalamic medial nucleus ( P < 0.05 vs. occlusion-vehicle). In the white matter tracts, recombinant human erythropoietin increased total, but not immature/mature oligodendrocytes ( P < 0.05 vs. occlusion-vehicle), with increased cell proliferation and reduced induction of activated caspase-3, microglia and astrocytes ( P < 0.05). Finally, occlusion-Epo reduced seizure burden, with more rapid recovery of electroencephalogram power, spectral edge frequency, and carotid blood flow. In summary, prolonged infusion of recombinant human erythropoietin after severe asphyxia in preterm fetal sheep was partially neuroprotective and improved electrophysiological and cerebrovascular recovery, in association with reduced apoptosis and inflammation.


Subject(s)
Asphyxia/drug therapy , Erythropoietin/administration & dosage , Gray Matter/drug effects , White Matter/drug effects , Animals , Electroencephalography/drug effects , Erythropoietin/pharmacology , Female , Humans , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Pregnancy , Sheep
16.
Endocrine ; 53(3): 839-47, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26979526

ABSTRACT

Previously we reported that prenatal undernutrition (UN) leads to a dysregulation of appetite suppression through alterations in hypothalamic neuropeptide gene expression. In the current study, we expand our observations and investigate neuroendocrine transcriptional responses and central leptin sensitivity within the arcuate nucleus of rats exposed to prenatal UN or a postnatal high-fat diet (HF). Pregnant Wistar rats were fed a standard chow diet either ad libitum (AD) or at 30 % of AD intake throughout gestation (UN) resulting in either control or intrauterine growth-restricted female offspring. At weaning, AD offspring were fed either a chow (C) or a HF (30 % fat wt/wt) diet ad libitum for the remainder of the study, whereas UN offspring were fed a chow diet only. At ~142 days, AD and UN offspring received either recombinant rat leptin (L) or saline (S) subcutaneously for 14 days. Prenatal UN had a significant effect on hypothalamic NPY (P < 0.0001), AgRP (P < 0.01) and ObRb (P < 0.02) mRNA expression compared to AD chow-fed offspring. A postnatal HF diet had a significant effect on AgRP mRNA expression (P < 0.001), compared to AD chow-fed offspring, but no effect on NPY and ObRb expression. Leptin treatment, in both UN and HF offspring, was ineffective in reducing NPY and AgRP mRNA expression, and had no effect on ObRb expression. These findings suggest that prenatal UN and a postnatal HF diet lead to differential neuroendocrine gene expression in the hypothalamic arcuate nuclei and reduced sensitivity to leptin's anorexigenic effects.


Subject(s)
Agouti-Related Protein/genetics , Arcuate Nucleus of Hypothalamus/metabolism , Gene Expression , Malnutrition/metabolism , Neuropeptide Y/genetics , Obesity/metabolism , Prenatal Exposure Delayed Effects/metabolism , Agouti-Related Protein/metabolism , Animals , Diet, High-Fat , Female , Leptin/pharmacology , Male , Malnutrition/genetics , Neuropeptide Y/metabolism , Obesity/genetics , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Rats , Rats, Wistar
17.
Growth Factors ; 34(5-6): 203-209, 2016 12.
Article in English | MEDLINE | ID: mdl-28122472

ABSTRACT

To investigate the relationship between maternal serum concentrations of placental growth hormone (GH-V), insulin-like growth factor (IGF)-1 and 2, IGF binding proteins (IGFBP)-1 and 3 and birth weight in appropriate-for-gestational-age (AGA), large-for-gestational-age (LGA) and small-for-gestational-age (SGA) cases in a nested case-control study. Maternal serum samples were selected from the Screening for Pregnancy Endpoints (SCOPE) biobank in Auckland, New Zealand. Serum hormone concentrations were determined by ELISA. We found that maternal serum GH-V concentrations at 20 weeks of gestation in LGA pregnancies were significantly higher than in AGA and SGA pregnancies. Maternal GH-V concentrations were positively correlated to birth weights and customized birth weight centiles, while IGFBP-1 concentrations were inversely related to birth weights and customized birth weight centiles. Our findings suggest that maternal serum GH-V and IGFBP-1 concentrations at 20 weeks' gestation are associated with fetal growth.


Subject(s)
Birth Weight , Growth Hormone/blood , Placental Hormones/blood , Adult , Case-Control Studies , Female , Fetal Development , Humans , Infant, Low Birth Weight/blood , Infant, Newborn , Insulin-Like Growth Factor Binding Protein 1/blood , Insulin-Like Growth Factor I/metabolism , Pregnancy
18.
Dev Neurosci ; 37(6): 497-514, 2015.
Article in English | MEDLINE | ID: mdl-26184807

ABSTRACT

Acute exposure to subclinical infection modulates subsequent hypoxia-ischemia (HI) injury in a time-dependent manner, likely by cross-talk through Toll-like receptors (TLRs), but the specific pathways are unclear in the preterm-equivalent brain. In the present study, we tested the hypothesis that repeated low-dose exposure to lipopolysaccharide (LPS) before acute ischemia would be associated with induction of specific TLRs that are potentially neuroprotective. Fetal sheep at 0.65 gestation (term is ∼145 days) received intravenous boluses of low-dose LPS for 5 days (day 1, 50 ng/kg; days 2-5, 100 ng/kg) or the same volume of saline. Either 4 or 24 h after the last bolus of LPS, complete carotid occlusion was induced for 22 min. Five days after LPS, brains were collected. Pretreatment with LPS for 5 days decreased cellular apoptosis, microglial activation and reactive astrogliosis in response to HI injury induced 24 but not 4 h after the last dose of LPS. This was associated with upregulation of TLR4, TLR7 and IFN-ß mRNA, and increased fetal plasma IFN-ß concentrations. The association of reduced white matter apoptosis and astrogliosis after repeated low-dose LPS finishing 24 h but not 4 h before cerebral ischemia, with central and peripheral induction of IFN-ß, suggests the possibility that IFN-ß may be an important mediator of endogenous neuroprotection in the developing brain.


Subject(s)
Apoptosis/drug effects , Brain/embryology , Hypoxia-Ischemia, Brain/immunology , Lipopolysaccharides/pharmacology , Prenatal Injuries/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 7/immunology , Animals , Brain/immunology , Brain Injuries/immunology , Brain Injuries/prevention & control , Disease Models, Animal , Fetus/drug effects , Fetus/immunology , Interferon-beta/blood , Sheep, Domestic
19.
Am J Physiol Regul Integr Comp Physiol ; 307(4): R387-95, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24944248

ABSTRACT

Perinatal exposure to infection is highly associated with adverse outcomes. Experimentally, acute, severe exposure to gram-negative bacterial lipopolysaccharide (LPS) is associated with increased fetal heart rate variability (FHRV). It is unknown whether FHRV is affected by subclinical infection with or without acute exacerbations. We therefore tested the hypothesis that FHRV would be associated with hypotension after acute on chronic exposure to LPS. Chronically instrumented fetal sheep at 0.7 gestation were exposed to a continuous low-dose LPS infusion (n = 12, 100 ng/kg over 24 h, followed by 250 ng·kg(-1)·24 h(-1) for a further 96 h) or the same volume of saline (n = 10). Boluses of either 1 µg LPS or saline were given at 48, 72, and 96 h. Low-dose infusion was not associated with hemodynamic or FHRV changes. The first LPS bolus was associated with tachycardia and suppression of nuchal electromyographic activity in all fetuses. Seven of twelve fetuses developed hypotension (a fall in mean arterial blood pressure ≥5 mmHg). FHRV was transiently increased only at the onset of hypotension, in association with increased cytokine induction and electroencephalogram suppression. FHRV then fell before the nadir of hypotension, with transient suppression of short-term FHRV. After the second LPS bolus, the hypotension group showed a biphasic pattern of a transient increase in FHRV followed by more prolonged suppression. These findings suggest that infection-related hypotension in the preterm fetus mediates the transient increase in FHRV and that repeated exposure to LPS leads to progressive loss of FHRV.


Subject(s)
Bradycardia/physiopathology , Fetal Heart/physiopathology , Heart Rate, Fetal , Hypotension/physiopathology , Lipopolysaccharides , Sepsis/physiopathology , Tachycardia/physiopathology , Animals , Arterial Pressure , Biomarkers/blood , Blood Gas Analysis , Blood Glucose/metabolism , Bradycardia/chemically induced , Bradycardia/diagnosis , Circadian Rhythm , Disease Models, Animal , Electrocardiography , Electroencephalography , Electromyography , Female , Fetal Blood/metabolism , Fetal Monitoring/methods , Gestational Age , Hydrogen-Ion Concentration , Hypotension/chemically induced , Hypotension/diagnosis , Lactic Acid/blood , Pregnancy , Sepsis/chemically induced , Sepsis/diagnosis , Sheep , Tachycardia/chemically induced , Tachycardia/diagnosis , Time Factors
20.
J Neuroinflammation ; 11: 89, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24886663

ABSTRACT

BACKGROUND: Perinatal asphyxia and exposure to intrauterine infection are associated with impaired neurodevelopment in preterm infants. Acute exposure to non-injurious infection and/or inflammation can either protect or sensitize the brain to subsequent hypoxia-ischemia. However, the effects of subacute infection and/or inflammation are unclear. In this study we tested the hypothesis that acute-on-chronic exposure to lipopolysaccharide (LPS) would exacerbate white matter injury after subsequent asphyxia in preterm fetal sheep. METHODS: Fetal sheep at 0.7 gestational age received a continuous LPS infusion at 100 ng/kg for 24 hours, then 250 ng/kg/24 hours for 96 hours, plus 1 µg boluses of LPS at 48, 72, and 96 hours or the same volume of saline. Four hours after the last bolus, complete umbilical cord occlusion or sham occlusion was induced for 15 minutes. Sheep were sacrificed 10 days after the start of infusions. RESULTS: LPS exposure was associated with induction of microglia and astrocytes and loss of total and immature and mature oligodendrocytes (n = 9) compared to sham controls (n = 9). Umbilical cord occlusion with saline infusions was associated with induction of microglia, astrogliosis, and loss of immature and mature oligodendrocytes (n = 9). LPS exposure before asphyxia (n = 8) was associated with significantly reduced microglial activation and astrogliosis and improved numbers of immature and mature oligodendrocytes compared to either LPS exposure or asphyxia alone. CONCLUSIONS: Contrary to our initial hypothesis, the combination of acute-on-chronic LPS with subsequent asphyxia reduced neuroinflammation and white matter injury compared with either intervention alone.


Subject(s)
Asphyxia Neonatorum/physiopathology , Inflammation/prevention & control , Leukoencephalopathies/prevention & control , Prenatal Exposure Delayed Effects/physiopathology , Animals , Disease Models, Animal , Embryo, Mammalian , Endotoxins/toxicity , Female , Inflammation/etiology , Leukoencephalopathies/etiology , Leukoencephalopathies/pathology , Lipopolysaccharides/toxicity , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Sheep , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...