1
|
Xu L, Jin X, Lu Y, Zheng B, Zheng Z, Chen L, Zhu H. Increased PLAGL1 Gene Methylation in Cord Blood is Positively Correlated with Brain Injury in Chorioamniotic Preterm Infants. Biochem Genet 2025; 63:1361-1380. [PMID: 38564096 DOI: 10.1007/s10528-024-10762-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024]
Abstract
The study aims to explore the epigenetic mechanisms of neurodevelopmental impairment accompanied in chorioamniotic preterm infants. Our study included 16 full-term infants and 69 preterm infants. The methylation status of the pleomorphic adenoma gene-like 1 (PLAGL1) gene in the cord blood was determined by pyrosequencing. Brain B-ultrasonography and magnetic resonance imaging (MRI) were performed to diagnose brain injury. The activity of candidate fragments of PLAGL1 and the effect of methylation on PLAGL1 activity were evaluated by double luciferase reporter assay. The data showed that there were no differences in the methylation levels of each Cytosine-phosphate-Guanine (CpG) site of PLAGL1 between full-term and preterm infants. Within preterm infants, the methylation levels of the CpG2, CpG3, CpG4, and CpG5 sites were increased in the chorioamnionitis group compared with the no chorioamnionitis group. The areas under curves (AUCs) of the receiver operating characteristic (ROC) curves of CpG2, CpG3, CpG4, and CpG5 were 0.656, 0.653, 0.670, and 0.712, respectively. Meanwhile, the methylation level of the CpG2 site was increased in preterm babies with brain injury compared with those without brain injury, and the AUC of CpG2 was 0.648, with a sensitivity of 75.9% and a specificity of 50.0%. A double luciferase reporter assay revealed that PLAGL1 fragments had enhancer-like activity and that the methylated form of PLAGL1 weakened this activity. Thus, PLAGL1 hypermethylation in chorioamniotic preterm infants is positively correlated with brain injury. Our results suggest a potential use for PLAGL1 methylation as a biomarker in the diagnosis of brain injury.
Collapse
Affiliation(s)
- Limin Xu
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, People's Republic of China.
| | - Xiamin Jin
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, People's Republic of China
| | - Younan Lu
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, People's Republic of China
| | - Bangxu Zheng
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, People's Republic of China
| | - Zhoushu Zheng
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, People's Republic of China
| | - Lili Chen
- Ningbo Women and Children's Hospital, Ningbo, Zhejiang, People's Republic of China
| | - Huaqiang Zhu
- Zhejiang Pharmaceutical University, Ningbo, Zhejiang, People's Republic of China.
| |
Collapse
|
2
|
Serdar M, Walther KA, Gallert M, Kempe K, Obst S, Labusek N, Herrmann R, Herz J, Felderhoff-Müser U, Bendix I. Prenatal inflammation exacerbates hyperoxia-induced neonatal brain injury. J Neuroinflammation 2025; 22:57. [PMID: 40022130 PMCID: PMC11871844 DOI: 10.1186/s12974-025-03389-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Premature born infants are at high risk to develop white matter injury (WMI). Hyperoxia and perinatal inflammation are main risk factors for preterm birth and associated brain injury. To date the majority of experimental studies have focused on isolated insults. However, clinically, WMI injury is a multifactorial disorder caused by a variety of triggers. To establish a clinically relevant rodent model of WMI, we combined prenatal inflammation with postnatal hyperoxia to investigate individual, and additive or synergistic effects on inflammatory processes, myelination and grey matter development. METHODS At embryonic day 20, pregnant Wistar rat dams received either a single intraperitoneal injection of 100 µg/ kg lipopolysaccharide (LPS) or sodium chloride. Offspring were either exposed to hyperoxia (80% O2) or normoxia (21% O2) from postnatal day 3 to 5. Animals were sacrificed immediately after hyperoxia or 6 days later, corresponding to term-equivalent age. White and grey matter development and neuroinflammatory responses were investigated at cellular and molecular levels applying immunohistochemistry, western blotting, real time PCR in brain tissues and multiplex protein expression analysis on serum samples. RESULTS Prenatal inflammation combined with postnatal hyperoxia resulted in reduced body weight and length in the offspring, accompanied by increased serum leptin levels at term equivalent age. The altered body parameters, like body weight, were associated with decreased brain volume, thinning of deep cortical layers and hypomyelination. As potential underlying mechanisms, we identified severe myelination deficits and an increased microglia activation associated with elevated inflammatory cytokine expression in brain tissues, while peripheral cytokine levels were reduced. Interestingly, effects on body size were mainly mediated by prenatal LPS, independent of hyperoxia, while oligodendrocyte degeneration was mainly induced by postnatal hyperoxia, independent of prenatal inflammation. However, for the majority of pathological changes, including brain size, myelination deficits, microglia activation and inflammatory cytokine expression, additive or synergistic effects were detected. CONCLUSION Prenatal inflammation combined with postnatal hyperoxia results in aggravated myelination deficits and inflammatory responses compared to single insults, making it an ideal model to improve our understanding of the complex pathophysiology underlying WMI and to evaluate urgently needed therapies.
Collapse
Affiliation(s)
- Meray Serdar
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kay-Anja Walther
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Markus Gallert
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Karina Kempe
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stefanie Obst
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nicole Labusek
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ralf Herrmann
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ursula Felderhoff-Müser
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Ivo Bendix
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
3
|
Kim C, Ufkes S, Guo T, Chau V, Synnes A, Grunau RE, Miller SP. Associations of Bronchopulmonary Dysplasia and Infection with School-Age Brain Development in Children Born Preterm. J Pediatr 2025; 281:114524. [PMID: 40023219 DOI: 10.1016/j.jpeds.2025.114524] [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: 09/27/2024] [Revised: 02/04/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVE To determine the association of bronchopulmonary dysplasia (BPD) and culture-positive infection with neurodevelopment and white matter maturation at 8 years of age in children born preterm. STUDY DESIGN Prospective cohort study of 164 children born at 24-32 weeks of gestation followed to 8 years of age (89 male, median [IQR] age: 8.24 [8.07-8.58] years). At age 8, IQ (Weschler Abbreviated Scale of Intelligence, second Ed), working memory (Wechsler Intelligence Scale for Children, fifth Ed), visual-motor (Beery-Buktenica Developmental Test of Visual-Motor Integration sixth Ed), and motor outcomes (Movement Assessment Battery for Children-2) were assessed. Diffusion tensor imaging and tract-based spatial statistics were used to assess fractional anisotropy (FA). RESULTS Fifty of 164 children had BPD and 72/164 had culture-positive infection. At 8 years, BPD was associated with a 9.8-point decrease in motor (CI -17.9 to -1.8, P = .02) and 6.0-point decrease in visual-motor scores (CI -10.5 to -1.5, P = .009), whereas infection was associated with a 6.3-point decrease in IQ (CI -12.3 to -0.3, P = .04), after adjusting for gestational age and white matter injury volume. BPD was associated with left hemisphere-dominant FA reductions, which were associated with worse motor (P = .000006) and visual-motor (P = .00005) outcomes, whereas infection was associated with bilateral FA reductions, which were associated with lower IQ scores (P = .03) and poorer working memory (P = .01) at 8 years. CONCLUSIONS This cohort study of children born very preterm suggests that BPD and culture-positive infection are distinctly associated with impaired white-matter development and poor neurodevelopmental outcomes. These findings imply neonatal illnesses impact the brain nonuniformly, suggesting opportunities for targeted intervention.
Collapse
Affiliation(s)
- Caroline Kim
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Pediatrics, BC Children's Hospital Research Institute and the University of British Columbia, Vancouver, BC, Canada
| | - Steven Ufkes
- Department of Pediatrics, BC Children's Hospital Research Institute and the University of British Columbia, Vancouver, BC, Canada
| | - Ting Guo
- Neuroscience and Mental Health Program, SickKids Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Vann Chau
- Neuroscience and Mental Health Program, SickKids Research Institute, Hospital for Sick Children, Toronto, ON, Canada; Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Anne Synnes
- Department of Pediatrics, BC Children's Hospital Research Institute and the University of British Columbia, Vancouver, BC, Canada
| | - Ruth E Grunau
- Department of Pediatrics, BC Children's Hospital Research Institute and the University of British Columbia, Vancouver, BC, Canada
| | - Steven P Miller
- Department of Pediatrics, BC Children's Hospital Research Institute and the University of British Columbia, Vancouver, BC, Canada; Neuroscience and Mental Health Program, SickKids Research Institute, Hospital for Sick Children, Toronto, ON, Canada; Department of Pediatrics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Lawrence SM, Wynn JL, Gordon SM. Neonatal bacteremia and sepsis. REMINGTON AND KLEIN'S INFECTIOUS DISEASES OF THE FETUS AND NEWBORN INFANT 2025:183-232.e25. [DOI: 10.1016/b978-0-323-79525-8.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Saito J, Shibasaki J, Yamamoto K, Fujita M, Toyoshima K. Predictive value of serum interleukin-6 for neonatal encephalopathy outcomes. J Neonatal Perinatal Med 2024:19345798241297068. [PMID: 39973533 DOI: 10.1177/19345798241297068] [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: 02/21/2025]
Abstract
Background: Serum interleukin-6 (IL-6) may predict adverse outcomes of neonatal encephalopathy (NE); however, limited data regarding the predictive utility of IL-6 during neurodevelopmental follow-up are available. We aimed to determine the utility of IL-6 for predicting adverse outcomes at 18 to 22 months of age.Methods: Eighty-seven patients with NE who received therapeutic hypothermia were enrolled in this study. Serial serum IL-6 levels during the first 3 postnatal days were collected. Patients were classified into three groups: (1) death, (2) survival with moderate to severe neurodevelopmental disability (NDD) at 18-22 months of age, and (3) survival without NDD (favorable outcome). The predictive ability of IL-6 was determined by the area under the receiver-operating characteristic curve (AUC).Results: Serial IL-6 data of 80 patients with NE were available and showed peak levels on postnatal day 1; these levels gradually decreased toward day 3. By 18-22 months of age, 13 and 17 patients died and experienced moderate to severe NDD without death, respectively. Fifty patients experienced favorable outcomes. Higher IL-6 levels on day 1 predicted the composite adverse outcome (including death and survival with NDD; n = 30; AUC, 0.648). Higher IL-6 levels on day 1 predicted death (n = 13; AUC, 0.799), whereas higher IL-6 levels on day 1 predicted survival with NDD (n = 17; AUC, 0.536).Conclusions: The AUC of IL-6 that predicted survival with NDD was lower than the AUC of IL-6 that predicted death; therefore, IL-6 may have insufficient utility for predicting NDD without death.
Collapse
Affiliation(s)
- Junichi Saito
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Jun Shibasaki
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kouji Yamamoto
- Department of Biostatistics, School of Medicine, Yokohama City University, Yokohama, Japan
| | - Maya Fujita
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Katsuaki Toyoshima
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Japan
| |
Collapse
|
6
|
Saito J, Shibasaki J, Yamamoto K, Fujita M, Toyoshima K. Predictive value of serum interleukin-6 for neonatal encephalopathy outcomes. J Neonatal Perinatal Med 2024; 17:770-778. [PMID: 39365327 DOI: 10.3233/npm-230224] [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: 10/05/2024]
Abstract
Background: Serum interleukin-6 (IL-6) may predict adverse outcomes of neonatal encephalopathy (NE); however, limited data regarding the predictive utility of IL-6 during neurodevelopmental follow-up are available. We aimed to determine the utility of IL-6 for predicting adverse outcomes at 18 to 22 months of age.Methods: Eighty-seven patients with NE who received therapeutic hypothermia were enrolled in this study. Serial serum IL-6 levels during the first 3 postnatal days were collected. Patients were classified into three groups: (1) death, (2) survival with moderate to severe neurodevelopmental disability (NDD) at 18-22 months of age, and (3) survival without NDD (favorable outcome). The predictive ability of IL-6 was determined by the area under the receiver-operating characteristic curve (AUC).Results: Serial IL-6 data of 80 patients with NE were available and showed peak levels on postnatal day 1; these levels gradually decreased toward day 3. By 18-22 months of age, 13 and 17 patients died and experienced moderate to severe NDD without death, respectively. Fifty patients experienced favorable outcomes. Higher IL-6 levels on day 1 predicted the composite adverse outcome (including death and survival with NDD; n = 30; AUC, 0.648). Higher IL-6 levels on day 1 predicted death (n = 13; AUC, 0.799), whereas higher IL-6 levels on day 1 predicted survival with NDD (n = 17; AUC, 0.536).Conclusions: The AUC of IL-6 that predicted survival with NDD was lower than the AUC of IL-6 that predicted death; therefore, IL-6 may have insufficient utility for predicting NDD without death.
Collapse
Affiliation(s)
- Junichi Saito
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Jun Shibasaki
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kouji Yamamoto
- Department of Biostatistics, School of Medicine, Yokohama City University, Yokohama, Japan
| | - Maya Fujita
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Katsuaki Toyoshima
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Japan
| |
Collapse
|
7
|
Bakhtazad S, Ghotbeddin Z, Tabandeh MR, Rahimi K. Alpha-pinene ameliorate behavioral deficit induced by early postnatal hypoxia in the rat: study the inflammatory mechanism. Sci Rep 2024; 14:6416. [PMID: 38494527 PMCID: PMC10944845 DOI: 10.1038/s41598-024-56756-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 03/11/2024] [Indexed: 03/19/2024] Open
Abstract
Neonatal hypoxia has a negative impact on the developing brain during the sensitive period. Inflammation plays a key role in the physiological response to hypoxic stress. Considering the anti-inflammatory properties of alpha-pinene, which has received a lot of attention in recent years, in this research we focused on the impact of alpha-pinene on the behavioral responses and proinflammatory factors in rats subjected to the neonatal hypoxia. This study involved Wistar rats (7-day-old) that were divided into six experimental groups, including a control group, groups receiving different doses of alpha-pinene (5 and 10 mg/kg), a hypoxia group receiving 7% O2 and 93% N2, 90 min duration for 7 days, and groups receiving alpha-pinene 30 min before hypoxia. All injections were done intraperitoneally. The rats were evaluated for proinflammatory factors 24 h after exposure to hypoxia (PND14) and at the end of the behavioral test (PND54). The results showed that hypoxia led to decreased motor activity, coordination, and memory, as well as increased inflammation. However, the rats that received alpha-pinene showed improved behavioral responses and reduced inflammation compared to the hypoxia group (all cases p < 0.05). This suggests that alpha-pinene may have a protective effect via anti-inflammatory properties against the negative impacts of hypoxia on the developing brain.
Collapse
Affiliation(s)
- Sharareh Bakhtazad
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Zohreh Ghotbeddin
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
- Stem Cell and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Mohammad Reza Tabandeh
- Stem Cell and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Kaveh Rahimi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
8
|
Melan N, Pradat P, Godbert I, Pastor-Diez B, Basson E, Picaud JC. Neurodevelopment at 24 months corrected age in extremely preterm infants treated with dexamethasone alternatives during the late postnatal period: a cohort study. Eur J Pediatr 2024; 183:677-687. [PMID: 37955745 PMCID: PMC10912127 DOI: 10.1007/s00431-023-05319-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/18/2023] [Accepted: 10/28/2023] [Indexed: 11/14/2023]
Abstract
The administration of dexamethasone has been associated with suboptimal neurodevelopment. We aimed to compare the development of extremely premature infants treated or not with alternatives to dexamethasone: betamethasone, hydrocortisone hemisuccinate. This retrospective cohort study included infants born before 29 weeks of gestational age, treated or not with late (day ≥ 7) postnatal steroids (betamethasone, hydrocortisone hemisuccinate). The neurodevelopment outcome was evaluated at 24 months corrected age, after adjustment on comorbidities of extreme prematurity. In order to analyse their overall development, data about growth and respiratory outcomes were collected. Among the 192 infants included, 59 (30.7%) received postnatal steroids. Suboptimal neurodevelopment concerned 37/59 (62.7%) postnatal steroid-treated and 43/133 (38.1%; p = 0.002) untreated infants. However, in multivariable analysis, only severe neonatal morbidity (p = 0.007) and male gender (p = 0.027) were associated with suboptimal neurodevelopment outcome at 24 months. Conclusions: Betamethasone or hydrocortisone hemisuccinate treatment was not an independent risk for suboptimal neurological development, growth and respiratory outcomes assessed at 24 months corrected age in extremely premature infants. Registration number: The study was registered on the ClinicalTrials.gov register: NCT05055193. What is Known: • Late postnatal steroids are used to treat bronchopulmonary dysplasia • Meta-analyses warned against the neurological risk of dexamethasone use during neonatal period. Early or late hydrocortisone hemisuccinate has been evaluated in multiple studies, none of which have reported an adverse effect on neurodevelopment at least to 2 years. Data about the use of betamethasone are scarce. What is New: • The risk of suboptimal neurodevelopment was higher among extremely premature infants who received postnatal steroids when compared to those who did not. • Betamethasone and hydrocortisone hemisuccinate treatment was not an independent risk factor for suboptimal neurodevelopment at 24 months corrected age.
Collapse
Affiliation(s)
- Nathalie Melan
- Department of Neonatology, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Pierre Pradat
- Centre for Clinical Research, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Isabelle Godbert
- Department of Neonatology, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Blandine Pastor-Diez
- Department of Neonatology, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Eliane Basson
- Department of Neonatology, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Jean-Charles Picaud
- Department of Neonatology, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France.
- CarMen Laboratory, INSERM, INRA, Université Claude Bernard Lyon 1, Pierre-Bénite, 69310, Lyon, France.
| |
Collapse
|
9
|
Ganesh R, Yadav S, Hurt RT, Mueller MR, Aakre CA, Gilman EA, Grach SL, Overgaard J, Snyder MR, Collins NM, Croghan IT, Badley AD, Razonable RR, Salonen BR. Pro Inflammatory Cytokines Profiles of Patients With Long COVID Differ Between Variant Epochs. J Prim Care Community Health 2024; 15:21501319241254751. [PMID: 38808863 PMCID: PMC11138192 DOI: 10.1177/21501319241254751] [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: 02/20/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Over 30% of patients with COVID-19 have persistent symptoms that last beyond 30 days and referred to as Long COVID. Long COVID has been associated with a persistent elevation in peripheral cytokines including interleukin-6, interleukin-1β, and tumor necrosis factor-α. This study reports cytokine profiles of patients in our clinic across SARS-COV-2 variant epochs. METHODS The clinical cytokine panel was analyzed in patients with Long COVID during periods that were stratified according to variant epoch. The 4 variant epochs were defined as: (1) wild-type through alpha, (2) alpha/beta/gamma, (3) delta, and (4) omicron variants. RESULTS A total of 390 patients had the clinical cytokine panel performed; the median age was 48 years (IQR 38-59) and 62% were female. Distribution by variant was wild-type and alpha, 50% (n = 196); alpha/beta/gamma, 7.9% (n = 31); delta, 18% (n = 72); and omicron, 23% (n = 91). Time to cytokine panel testing was significantly longer for the earlier epochs. Tumor necrosis factor-α (P < .001) and interleukin 1β (P < .001) were significantly more elevated in the earlier epochs (median of 558 days in wild-type through Alpha epoch vs 263 days in omicron epoch, P < .001)). Nucleocapsid antibodies were consistently detected across epochs. DISCUSSION When stratified by variant epoch, patients with early epoch Long COVID had persistently elevated peripheral pro-inflammatory cytokine levels when compared to later epoch Long COVID. Patients with Long COVID have similar clusters of symptoms across epochs, suggesting that the underlying pathology is independent of the peripheral cytokine signature.
Collapse
|
10
|
Gigase FAJ, Smith E, Collins B, Moore K, Snijders GJLJ, Katz D, Bergink V, Perez-Rodriquez MM, De Witte LD. The association between inflammatory markers in blood and cerebrospinal fluid: a systematic review and meta-analysis. Mol Psychiatry 2023; 28:1502-1515. [PMID: 37055513 PMCID: PMC10266485 DOI: 10.1038/s41380-023-01976-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 01/06/2023] [Accepted: 01/19/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND Neuroinflammatory processes have been hypothesized to play a role in the pathogenesis of psychiatric and neurological diseases. Studies on this topic often rely on analysis of inflammatory biomarkers in peripheral blood. Unfortunately, the extent to which these peripheral markers reflect inflammatory processes in the central nervous system (CNS) is unclear. METHODS We performed a systematic review and found 29 studies examining the association between inflammatory marker levels in blood and cerebrospinal (CSF) samples. We performed a random effects meta-analysis of 21 studies (pooled n = 1679 paired samples) that reported the correlation of inflammatory markers in paired blood-CSF samples. RESULTS A qualitative review revealed moderate to high quality of included studies with the majority of studies reporting no significant correlation of inflammatory markers between paired blood-CSF. Meta-analyses revealed a significant low pooled correlation between peripheral and CSF biomarkers (r = 0.21). Meta-analyses of individual cytokines revealed a significant pooled correlation for IL-6 (r = 0.26) and TNFα (r = 0.3) after excluding outlier studies, but not for other cytokines. Sensitivity analyses showed that correlations were highest among participants with a median age above 50 (r = 0.46) and among autoimmune disorder patients (r = 0.35). CONCLUSION This systematic review and meta-analysis revealed poor correlation between peripheral and central inflammatory markers in paired blood-CSF samples, with increased correlations in certain study populations. Based on the current findings, peripheral inflammatory markers are a poor reflection of the neuroinflammatory profile.
Collapse
Affiliation(s)
- Frederieke A J Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
- Department of Clinical and Medical Psychology, Tilburg University, Tilburg, The Netherlands.
- Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Emma Smith
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Brett Collins
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Kendall Moore
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Gijsje J L J Snijders
- Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Daniel Katz
- Department of Anesthesiology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Veerle Bergink
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | | | - Lotje D De Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| |
Collapse
|
11
|
Brégère C, Fisch U, Halbeisen FS, Schneider C, Dittmar T, Stricker S, Aghlmandi S, Guzman R. Doublecortin and Glypican-2 concentrations in the cerebrospinal fluid from infants are developmentally downregulated. PLoS One 2023; 18:e0279343. [PMID: 36800341 PMCID: PMC9937498 DOI: 10.1371/journal.pone.0279343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 12/05/2022] [Indexed: 02/18/2023] Open
Abstract
OBJECTIVE Doublecortin (DCX) and glypican-2 (GPC2) are neurodevelopmental proteins involved in the differentiation of neural stem/progenitor cells (NSPCs) to neurons, and are developmentally downregulated in neurons after birth. In this study, we investigated whether the concentrations of DCX and GPC2 in the cerebrospinal fluid (CSF) from human pediatric patients reflect this developmental process or are associated with cerebral damage or inflammatory markers. METHODS CSF was collected from pediatric patients requiring neurosurgical treatment. The concentrations of DCX, GPC2, neuron-specific enolase (NSE), S100 calcium-binding protein B (S100B), and cytokines (IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-13, IFN-γ, and TNF-⍺) were measured using immunoassays. RESULTS From March 2013 until October 2018, 63 CSF samples were collected from 38 pediatric patients (20 females; 17 patients with repeated measurements); the median term born-adjusted age was 3.27 years [Q1: 0.31, Q3: 7.72]. The median concentration of DCX was 329 pg/ml [Q1: 192.5, Q3: 1179.6] and that of GPC2 was 26 pg/ml [Q1: 13.25, Q3: 149.25]. DCX and GPC2 concentrations independently significantly associated with age, and their concentration declined with advancing age, reaching undetectable levels at 0.3 years for DCX, and plateauing at 1.5 years for GPC2. Both DCX and GPC2 associated with hydrocephalus, NSE, IL-1β, IL-2, IL-8, IL-13. No relationship was found between sex, acute infection, S100B, IL-4, IL-6, IL-10, IFN-γ, TNF-α and DCX or GPC2, respectively. CONCLUSIONS Concentrations of DCX and GPC2 in the CSF from pediatric patients are developmentally downregulated, with the highest concentrations measured at the earliest adjusted age, and reflect a neurodevelopmental stage rather than a particular disease state.
Collapse
Affiliation(s)
- Catherine Brégère
- Department of Biomedicine, Brain Ischemia and Regeneration, University Hospital Basel, Basel, Switzerland
| | - Urs Fisch
- Department of Biomedicine, Brain Ischemia and Regeneration, University Hospital Basel, Basel, Switzerland,Department of Neurology, University Hospital Basel, Basel, Switzerland
| | - Florian Samuel Halbeisen
- Basel Institute for Clinical Epidemiology and Biostatistics, University of Basel, Basel, Switzerland
| | - Christian Schneider
- Division of Pediatric Neurosurgery, University of Basel Children’s Hospital, Basel, Switzerland
| | - Tanja Dittmar
- Department of Biomedicine, Brain Ischemia and Regeneration, University Hospital Basel, Basel, Switzerland
| | - Sarah Stricker
- Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Soheila Aghlmandi
- Basel Institute for Clinical Epidemiology and Biostatistics, University of Basel, Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine, Brain Ischemia and Regeneration, University Hospital Basel, Basel, Switzerland,Division of Pediatric Neurosurgery, University of Basel Children’s Hospital, Basel, Switzerland,Department of Neurosurgery, University Hospital Basel, Basel, Switzerland,Medical Faculty, University of Basel, Basel, Switzerland,* E-mail:
| |
Collapse
|
12
|
Shahub S, Lin KC, Muthukumar S, Prasad S. A Proof-of-Concept Electrochemical Skin Sensor for Simultaneous Measurement of Glial Fibrillary Acidic Protein (GFAP) and Interleukin-6 (IL-6) for Management of Traumatic Brain Injuries. BIOSENSORS 2022; 12:bios12121095. [PMID: 36551062 PMCID: PMC9775589 DOI: 10.3390/bios12121095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 05/28/2023]
Abstract
This work demonstrates the use of a noninvasive, sweat-based dual biomarker electrochemical sensor for continuous, prognostic monitoring of a Traumatic Brain Injury (TBI) with the aim of enhancing patient outcomes and reducing the time to treatment after injury. A multiplexed SWEATSENSER was used for noninvasive continuous monitoring of glial fibrillary acidic protein (GFAP) and Interleukin-6 (IL-6) in a human sweat analog and in human sweat. Electrochemical impedance spectroscopy (EIS) and chronoamperometry (CA) were used to measure the sensor response. The assay chemistry was characterized using Fourier Transform Infrared Spectroscopy (FTIR). The SWEATSENSER was able to detect GFAP and IL-6 in sweat over a dynamic range of 3 log orders for GFAP and 2 log orders for IL-6. The limit of detection (LOD) for GFAP detection in the sweat analog was estimated to be 14 pg/mL using EIS and the LOD for IL-6 was estimated to be 10 pg/mL using EIS. An interference study was performed where the specific signal was significantly higher than the non-specific signal. Finally, the SWEATSENSER was able to distinguish between GFAP and IL-6 in simulated conditions of a TBI in human sweat. This work demonstrates the first proof-of-feasibility of a multiplexed TBI marker combined with cytokine and inflammatory marker detection in passively expressed sweat in a wearable form-factor that can be utilized toward better management of TBIs. This is the first step toward demonstrating a noninvasive enabling technology that can enable baseline tracking of an inflammatory response.
Collapse
Affiliation(s)
- Sarah Shahub
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Kai-Chun Lin
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Sriram Muthukumar
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- EnLiSense LLC, Allen, TX 75013, USA
| | - Shalini Prasad
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
13
|
Stone TW, Clanchy FIL, Huang YS, Chiang NY, Darlington LG, Williams RO. An integrated cytokine and kynurenine network as the basis of neuroimmune communication. Front Neurosci 2022; 16:1002004. [PMID: 36507331 PMCID: PMC9729788 DOI: 10.3389/fnins.2022.1002004] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Two of the molecular families closely associated with mediating communication between the brain and immune system are cytokines and the kynurenine metabolites of tryptophan. Both groups regulate neuron and glial activity in the central nervous system (CNS) and leukocyte function in the immune system, although neither group alone completely explains neuroimmune function, disease occurrence or severity. This essay suggests that the two families perform complementary functions generating an integrated network. The kynurenine pathway determines overall neuronal excitability and plasticity by modulating glutamate receptors and GPR35 activity across the CNS, and regulates general features of immune cell status, surveillance and tolerance which often involves the Aryl Hydrocarbon Receptor (AHR). Equally, cytokines and chemokines define and regulate specific populations of neurons, glia or immune system leukocytes, generating more specific responses within restricted CNS regions or leukocyte populations. In addition, as there is a much larger variety of these compounds, their homing properties enable the superimposition of dynamic variations of cell activity upon local, spatially limited, cell populations. This would in principle allow the targeting of potential treatments to restricted regions of the CNS. The proposed synergistic interface of 'tonic' kynurenine pathway affecting baseline activity and the superimposed 'phasic' cytokine system would constitute an integrated network explaining some features of neuroimmune communication. The concept would broaden the scope for the development of new treatments for disorders involving both the CNS and immune systems, with safer and more effective agents targeted to specific CNS regions.
Collapse
Affiliation(s)
- Trevor W. Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom,*Correspondence: Trevor W. Stone,
| | - Felix I. L. Clanchy
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Yi-Shu Huang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Nien-Yi Chiang
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - L. Gail Darlington
- Department of Internal Medicine, Ashtead Hospital, Ashtead, United Kingdom
| | - Richard O. Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Leifsdottir K, Thelin EP, Lassarén P, Siljehav V, Nilsson P, Eksborg S, Herlenius E. Proteomic profiles in cerebrospinal fluid predicted death and disability in term infants with perinatal asphyxia: A pilot study. Acta Paediatr 2022; 111:961-970. [PMID: 35106835 PMCID: PMC9305740 DOI: 10.1111/apa.16277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/23/2021] [Accepted: 01/31/2022] [Indexed: 11/29/2022]
Abstract
Aim Perinatal asphyxia, resulting in hypoxic‐ischaemic encephalopathy (HIE), has been associated with high mortality rates and severe lifelong neurodevelopmental disabilities. Our aim was to study the association between the proteomic profile in cerebrospinal fluid (CSF) and the degree of HIE and long‐term outcomes. Methods We prospectively enrolled 18‐term born infants with HIE and 10‐term born controls between 2000 and 2004 from the Karolinska University Hospital. An antibody suspension bead array and FlexMap3D analysis was used to characterise 178 unique brain‐derived and inflammation associated proteins in their CSF. Results Increased CSF concentrations of several brain‐specific proteins were observed in the proteome of HIE patients compared with the controls. An upregulation of neuroinflammatory pathways was also noted and this was confirmed by pathway analysis. Principal component analysis revealed a gradient from favourable to unfavourable HIE grades and outcomes. The proteins that provided strong predictors were structural proteins, including myelin basic protein and alpha‐II spectrin. The functional proteins included energy‐related proteins like neuron‐specific enolase and synaptic regulatory proteins. Increased CSF levels of 51 proteins correlated with adverse outcomes in infants with HIE. Conclusion Brain‐specific proteins and neuroinflammatory mediators in CSF may predict HIE degrees and outcomes after perinatal asphyxia.
Collapse
Affiliation(s)
- Kristin Leifsdottir
- Department of Women´s and Children´s Health Karolinska Institutet Stockholm Sweden
- Astrid Lindgren Children’s Hospital Karolinska University Hospital Stockholm Sweden
| | - Eric P Thelin
- Department of Clinical Neuroscience Karolinska Institutet Stockholm Sweden
- Department of Neurology Karolinska University Hospital Stockholm Sweden
| | - Philipp Lassarén
- Department of Clinical Neuroscience Karolinska Institutet Stockholm Sweden
| | - Veronica Siljehav
- Department of Women´s and Children´s Health Karolinska Institutet Stockholm Sweden
- Astrid Lindgren Children’s Hospital Karolinska University Hospital Stockholm Sweden
| | - Peter Nilsson
- Department of Protein Science KTH Royal Institute of Technology SciLifeLab Stockholm Sweden
| | - Staffan Eksborg
- Department of Women´s and Children´s Health Karolinska Institutet Stockholm Sweden
- Astrid Lindgren Children’s Hospital Karolinska University Hospital Stockholm Sweden
| | - Eric Herlenius
- Department of Women´s and Children´s Health Karolinska Institutet Stockholm Sweden
- Astrid Lindgren Children’s Hospital Karolinska University Hospital Stockholm Sweden
| |
Collapse
|
15
|
Jiang JK, Wang C, Yin R, Jiang ZD. Functional status of brainstem auditory pathway in babies born below 30 week gestation with necrotizing enterocolitis. Brain Dev 2022; 44:263-270. [PMID: 35042649 DOI: 10.1016/j.braindev.2021.12.008] [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: 11/04/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Recent studies showed that neonatal necrotizing enterocolitis (NEC) adversely affects the brainstem auditory pathway in babies born at 30-40 week gestation. We compared the functional status of the pathway between babies born below 30 week gestation with NEC and those without NEC for any differences to understand whether NEC also affects the pathway in babies born at a smaller gestation. METHOD Brainstem auditory evoked response was studied at term in NEC babies born below 30 week gestation. The data obtained were compared with age-matched non-NEC babies for any abnormalities, and then compared with previously reported NEC babies born at 30-34 week gestation for any differences. RESULTS Although the latencies of waves I and III did not differ significantly between NEC and non-NEC babies, wave V latency in NEC babies was longer than in non-NEC babies at all click rates used. In particular, I-V interpeak interval, reflecting brainstem conduction time, in NEC babies was significant longer than in non-NEC babies. Wave V amplitude and the V/I amplitude ratios in NEC babies was smaller than in non-NEC babies at some click rates. The I-V interval in our NEC babies born below 30 week gestation was longer than in previously reported NEC babies born at 30-34 week gestation at all click rates. CONCLUSION NEC babies born below 30 week gestation are associated with delayed brainstem conduction time. Functional status of the brainstem auditory pathway in NEC babies born below 30 week gestation is less favorable than that in those with greater gestation.
Collapse
Affiliation(s)
- James Ken Jiang
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Cui Wang
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Rong Yin
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Ze Dong Jiang
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Low-Dose Delta-9-Tetrahydrocannabinol as Beneficial Treatment for Aged APP/PS1 Mice. Int J Mol Sci 2022; 23:ijms23052757. [PMID: 35269905 PMCID: PMC8910894 DOI: 10.3390/ijms23052757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/15/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
Studies on the effective and safe therapeutic dosage of delta-9-tetrahydrocannabinol (THC) for the treatment of Alzheimer’s disease (AD) have been sparse due to the concern about THC’s psychotropic activity. The present study focused on demonstrating the beneficial effect of low-dose THC treatment in preclinical AD models. The effect of THC on amyloid-β (Aβ) production was examined in N2a/AβPPswe cells. An in vivo study was conducted in aged APP/PS1 transgenic mice that received an intraperitoneal injection of THC at 0.02 and 0.2 mg/kg every other day for three months. The in vitro study showed that THC inhibited Aβ aggregation within a safe dose range. Results of the radial arm water maze (RAWM) test demonstrated that treatment with 0.02 and 0.2 mg/kg of THC for three months significantly improved the spatial learning performance of aged APP/PS1 mice in a dose-dependent manner. Results of protein analyses revealed that low-dose THC treatment significantly decreased the expression of Aβ oligomers, phospho-tau and total tau, and increased the expression of Aβ monomers and phospho-GSK-3β (Ser9) in the THC-treated brain tissues. In conclusion, treatment with THC at 0.2 and 0.02 mg/kg improved the spatial learning of aged APP/PS1 mice, suggesting low-dose THC is a safe and effective treatment for AD.
Collapse
|
17
|
Leifsdottir K, Jost K, Siljehav V, Thelin EP, Lassarén P, Nilsson P, Haraldsson Á, Eksborg S, Herlenius E. The cerebrospinal fluid proteome of preterm infants predicts neurodevelopmental outcome. Front Pediatr 2022; 10:921444. [PMID: 35928685 PMCID: PMC9343678 DOI: 10.3389/fped.2022.921444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/29/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Survival rate increases for preterm infants, but long-term neurodevelopmental outcome predictors are lacking. Our primary aim was to determine whether a specific proteomic profile in cerebrospinal fluid (CSF) of preterm infants differs from that of term infants and to identify novel biomarkers of neurodevelopmental outcome in preterm infants. METHODS Twenty-seven preterm infants with median gestational age 27 w + 4 d and ten full-term infants were enrolled prospectively. Protein profiling of CSF were performed utilizing an antibody suspension bead array. The relative levels of 178 unique brain derived proteins and inflammatory mediators, selected from the Human Protein Atlas, were measured. RESULTS The CSF protein profile of preterm infants differed from that of term infants. Increased levels of brain specific proteins that are associated with neurodevelopment and neuroinflammatory pathways made up a distinct protein profile in the preterm infants. The most significant differences were seen in proteins involved in neurodevelopmental regulation and synaptic plasticity, as well as components of the innate immune system. Several proteins correlated with favorable outcome in preterm infants at 18-24 months corrected age. Among the proteins that provided strong predictors of outcome were vascular endothelial growth factor C, Neurocan core protein and seizure protein 6, all highly important in normal brain development. CONCLUSION Our data suggest a vulnerability of the preterm brain to postnatal events and that alterations in protein levels may contribute to unfavorable neurodevelopmental outcome.
Collapse
Affiliation(s)
- Kristin Leifsdottir
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden.,The Children's Hospital of Iceland, Reykjavik, Iceland
| | - Kerstin Jost
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Veronica Siljehav
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Eric P Thelin
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Philipp Lassarén
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Peter Nilsson
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
| | | | - Staffan Eksborg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Eric Herlenius
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
18
|
Lear BA, Lear CA, Dhillon SK, Davidson JO, Bennet L, Gunn AJ. Is late prevention of cerebral palsy in extremely preterm infants plausible? Dev Neurosci 2021; 44:177-185. [PMID: 34937030 DOI: 10.1159/000521618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/16/2021] [Indexed: 11/19/2022] Open
Abstract
Preterm birth continues to be associated with neurodevelopmental problems including cerebral palsy. Cystic white matter injury is still the major neuropathology underlying cerebral palsy, affecting 1-3% of preterm infants. Although rates have gradually fallen over time, the pathogenesis and evolution of cystic white matter injury are still poorly understood. Hypoxia-ischemia (HI) remains an important contributor yet there is no established treatment to prevent injury. Clinically, serial ultrasound and magnetic resonance imaging studies typically show delayed development of cystic lesions 2 to 4 weeks after birth. This raises the important and unresolved question as to whether this represents slow evolution of injury occurring around the time of birth, or repeated injury over many weeks after birth. There is increasing evidence that tertiary injury after HI can contribute to impairment of white and grey matter maturation. In the present review, we discuss preclinical evidence that severe, cystic white matter injury can evolve for many weeks after acute HI and is associated with microglia activity. This suggests the intriguing hypothesis that the tertiary phase of injury is not as subtle as often thought and that there may be a window of therapeutic opportunity for one to two weeks after hypoxic-ischemic injury to prevent delayed cystic lesions and so further reduce the risk of cerebral palsy after preterm birth.
Collapse
Affiliation(s)
- Benjamin A Lear
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Christopher A Lear
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | | | - Joanne O Davidson
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
19
|
Wang B, Zeng H, Liu J, Sun M. Effects of Prenatal Hypoxia on Nervous System Development and Related Diseases. Front Neurosci 2021; 15:755554. [PMID: 34759794 PMCID: PMC8573102 DOI: 10.3389/fnins.2021.755554] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
The fetal origins of adult disease (FOAD) hypothesis, which was proposed by David Barker in the United Kingdom in the late 1980s, posited that adult chronic diseases originated from various adverse stimuli in early fetal development. FOAD is associated with a wide range of adult chronic diseases, including cardiovascular disease, cancer, type 2 diabetes and neurological disorders such as schizophrenia, depression, anxiety, and autism. Intrauterine hypoxia/prenatal hypoxia is one of the most common complications of obstetrics and could lead to alterations in brain structure and function; therefore, it is strongly associated with neurological disorders such as cognitive impairment and anxiety. However, how fetal hypoxia results in neurological disorders remains unclear. According to the existing literature, we have summarized the causes of prenatal hypoxia, the effects of prenatal hypoxia on brain development and behavioral phenotypes, and the possible molecular mechanisms.
Collapse
Affiliation(s)
- Bin Wang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongtao Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingliu Liu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
20
|
Shim SY, Cho SJ, Park EA. Neurodevelopmental Outcomes at 18-24 Months of Corrected Age in Very Low Birth Weight Infants with Late-onset Sepsis. J Korean Med Sci 2021; 36:e205. [PMID: 34490752 PMCID: PMC8422039 DOI: 10.3346/jkms.2021.36.e205] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 07/09/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Preterm infants are prone to sepsis owing to their immature innate immunity and prolonged hospitalization. We aimed to evaluate the association between late-onset sepsis (LOS) during hospitalization and neurodevelopmental delay at 18-24 months of corrected age in very low birth weight infants (VLBWIs), and to ascertain this association when adjusted for perinatal risk factors. METHODS This is a population-based study of VLBWIs born at 23-32 weeks of gestation between January 2014 and December 2017 who were enrolled in the Korean Neonatal Network. Bayley scales of infant development were evaluated at 18-24 months of corrected age in 2,098 infants. To test for LOS as a risk factor for neurodevelopmental delay, multiple logistic regression was used and adjusted for parental education status and clinical variables. RESULTS Blood culture positive LOS was identified in 419 (20.0%) infants. Cognitive and motor delays were found in 392 (18.7%) and 347 (16.5%) infants, respectively. When multivariate analysis was performed, LOS had a significant association with cognitive delay (odds ratio, 1.48; 95% confidence interval, 1.02-2.16), but no association with motor delay in VLBWIs. Both delays were significantly more frequent in cases of intraventricular hemorrhage (IVH) ≥ grade 3, periventricular leukomalacia (PVL), and intrauterine growth restriction (IUGR) and duration of mechanical ventilation. Male sex and necrotizing enterocolitis ≥ grade 2 had an effect on motor delay, whereas paternal college graduation affected cognitive delay. CONCLUSION In VLBWIs with LOS, there is a heightened risk of cognitive delays at 18-24 months of corrected age. Brain injury, such as severe IVH and PVL, duration of mechanical ventilation, and IUGR, were also associated with cognitive and motor delays.
Collapse
Affiliation(s)
- So-Yeon Shim
- Division of Neonatology, Department of Pediatrics, Ewha Womans University College of Medicine, Seoul, Korea.
| | - Su Jin Cho
- Division of Neonatology, Department of Pediatrics, Ewha Womans University College of Medicine, Seoul, Korea
| | - Eun Ae Park
- Division of Neonatology, Department of Pediatrics, Ewha Womans University College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Bruschi M, Petretto A, Cama A, Pavanello M, Bartolucci M, Morana G, Ramenghi LA, Garré ML, Ghiggeri GM, Panfoli I, Candiano G. Potential biomarkers of childhood brain tumor identified by proteomics of cerebrospinal fluid from extraventricular drainage (EVD). Sci Rep 2021; 11:1818. [PMID: 33469081 PMCID: PMC7815722 DOI: 10.1038/s41598-020-80647-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/24/2020] [Indexed: 11/10/2022] Open
Abstract
Brain tumors are the most common solid tumors in childhood. There is the need for biomarkers of residual disease, therapy response and recurrence. Cerebrospinal fluid (CSF) is a source of brain tumor biomarkers. We analyzed the proteome of waste CSF from extraventricular drainage (EVD) from 29 children bearing different brain tumors and 17 controls needing EVD insertion for unrelated causes. 1598 and 1526 proteins were identified by liquid chromatography-coupled tandem mass spectrometry proteomics in CSF control and brain tumor patients, respectively, 263 and 191 proteins being exclusive of either condition. Bioinformatic analysis revealed promising protein biomarkers for the discrimination between control and tumor (TATA-binding protein-associated factor 15 and S100 protein B). Moreover, Thymosin beta-4 (TMSB4X) and CD109, and 14.3.3 and HSP90 alpha could discriminate among other brain tumors and low-grade gliomas plus glyoneuronal tumors/pilocytic astrocytoma, or embryonal tumors/medulloblastoma. Biomarkers were validated by ELISA assay. Our method was able to distinguish among brain tumor vs non-tumor/hemorrhagic conditions (controls) and to differentiate two large classes of brain tumors. Further prospective studies may assess whether the biomarkers proposed by our discovery approach can be identified in other bodily fluids, therefore less invasively, and are useful to guide therapy and predict recurrences.
Collapse
Affiliation(s)
- Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Andrea Petretto
- Core Facilities-Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Armando Cama
- Department of Neurosurgery, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marco Pavanello
- Department of Neurosurgery, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Martina Bartolucci
- Core Facilities-Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Giovanni Morana
- Unit of Neuroradiology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Maria Luisa Garré
- Department of Neuroncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gian Marco Ghiggeri
- UO of Nephrology, Dialysis and Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Isabella Panfoli
- Dipartimento di Farmacia (DIFAR), Università di Genova, V.le Benedetto XV, 3, 16132, Genoa, Italy.
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
22
|
Ghaneifar Z, Yousefi Z, Tajik F, Nikfar B, Ghalibafan F, Abdollahi E, Momtazi-Borojeni AA. The potential therapeutic effects of curcumin on pregnancy complications: Novel insights into reproductive medicine. IUBMB Life 2020; 72:2572-2583. [PMID: 33107698 DOI: 10.1002/iub.2399] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 10/11/2020] [Indexed: 01/13/2023]
Abstract
Pregnancy complications including preeclampsia, preterm birth, intrauterine growth restriction, and gestational diabetes are the main adverse reproductive outcomes. Excessive inflammation and oxidative stress play crucial roles in the pathogenesis of pregnancy disorders. Curcumin, the main polyphenolic compound derived from Curcuma longa, is mainly known by its anti-inflammatory and antioxidant properties. There are in vitro and in vivo reports revealing the preventive and ameliorating effects of curcumin against pregnancy complications. Here, we aimed to seek mechanisms underlying the modulatory effects of curcumin on dysregulated inflammatory and oxidative responses in various pregnancy complications.
Collapse
Affiliation(s)
- Zahra Ghaneifar
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Yousefi
- School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Fatemeh Tajik
- Faculty of medicine, Azad University of Tehran, Tehran, Iran
| | - Banafsheh Nikfar
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghalibafan
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Abdollahi
- Department of Medical Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
23
|
Davidson JO, van den Heuij LG, Fraser M, Wassink G, Miller SL, Lim R, Wallace EM, Jenkin G, Gunn AJ, Bennet L. Window of opportunity for human amnion epithelial stem cells to attenuate astrogliosis after umbilical cord occlusion in preterm fetal sheep. Stem Cells Transl Med 2020; 10:427-440. [PMID: 33103374 PMCID: PMC7900589 DOI: 10.1002/sctm.20-0314] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/22/2020] [Accepted: 10/09/2020] [Indexed: 12/19/2022] Open
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.
Collapse
Affiliation(s)
- Joanne O Davidson
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Lotte G van den Heuij
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Mhoyra Fraser
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Guido Wassink
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Euan M Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Alistair J Gunn
- 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
| |
Collapse
|
24
|
Howarth C, Banerjee J, Leung T, Eaton S, Morris JK, Aladangady N. Cerebral Oxygenation in Preterm Infants With Necrotizing Enterocolitis. Pediatrics 2020; 146:peds.2020-0337. [PMID: 32848028 DOI: 10.1542/peds.2020-0337] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Preterm infants with necrotizing enterocolitis (NEC) are known to have worse neurodevelopmental outcomes, but there is no substantial evidence to support an underlying pathophysiology. We aimed to examine whether cerebral oxygenation differs in those infants who develop NEC compared to cerebral oxygenation in those who do not. METHODS We examined 48 infants <30 weeks' gestation admitted to a tertiary level NICU from October 2016 to May 2018. Infants with birth weight less than or equal to the second percentile, abnormal antenatal dopplers or twin-to-twin-transfusion-syndrome were excluded. Cerebral oximetry measurements were performed by using a near-infrared spectroscopy (NIRS) monitor weekly for 60 minutes, allowing measurement of cerebral tissue oxygenation index from the first week of life to 36 weeks postconceptional age. Weekly clinical status was also recorded. NEC was defined as greater than or equal to Bell stage 2. RESULTS The median birth weight was 884 g (range of 460-1600 g), the median weeks' gestational age was 26 + 3/7 (23 + 0/7 to 29 + 6/7), and 52% were girls. In total, 276 NIRS measurements were completed, and 7 infants developed NEC. NIRS measurements from 1 infant with NEC and 4 infants without NEC who developed hemorrhagic parenchymal infarcts were excluded from analysis. Infants who developed NEC had significantly lower cerebral tissue oxygenation index than those who did not (P = .011), even when adjusted for confounders, including gestational age, birth weight, patent ductus arteriosus, enteral feeds, sex, ethnicity, and hemoglobin. CONCLUSIONS Infants with NEC have significantly lower cerebral tissue oxygenation throughout their neonatal intensive care stay in comparison with those who did not develop NEC. This is a novel finding and could explain their worse neurodevelopmental outcome.
Collapse
Affiliation(s)
- Claire Howarth
- Homerton University Hospital National Health Service Foundation Trust, London, United Kingdom.,Queen Mary University of London, London, United Kingdom
| | - Jayanta Banerjee
- Imperial College Healthcare National Health Service Trust, London, United Kingdom.,Imperial College London, London, United Kingdom
| | - Terence Leung
- Department of Medical Physics and Biomedical Engineering and
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; and
| | - Joan K Morris
- St George's, University of London, London, United Kingdom
| | - Narendra Aladangady
- Homerton University Hospital National Health Service Foundation Trust, London, United Kingdom; .,Queen Mary University of London, London, United Kingdom
| |
Collapse
|
25
|
Anuriev AM, Gorbachev VI. [Hypoxic-ischemic brain damage in premature newborns]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 119:63-69. [PMID: 31825364 DOI: 10.17116/jnevro201911908263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
One of the main causes of cerebral dysfunction in premature newborns is hypoxia. High mortality and lifelong morbidity in these children is a frequent result of neonatal hypoxic brain damage. The article presents some data on the prevalence of neurological diseases that have arisen in the perinatal period, and highlights the key etiological factors leading to hypoxia in both the intranatal and early postnatal periods. The pathogenesis of hypoxic-ischemic brain lesions in premature infants is described in detail. At the same time, more careful consideration is given to the glutathione system, which protects against lipid peroxidation, the glutamate-calcium cascade, and the excitotoxicity mediated by it, as well as the processes of necrosis and apoptosis of nerve cells. The advantages and disadvantages of modern methods for diagnosing cerebral lesions are noted, and the principles of treatment of these disorders are analyzed.
Collapse
Affiliation(s)
- A M Anuriev
- Irkutsk State Medical Academy of Postgraduate Education - Branch Campus of the Russian Medical Academy of Continuing Professional Education, Irkutsk, Russia
| | - V I Gorbachev
- Irkutsk State Medical Academy of Postgraduate Education - Branch Campus of the Russian Medical Academy of Continuing Professional Education, Irkutsk, Russia
| |
Collapse
|
26
|
Sweetman DU, Strickland T, Melo AM, Kelly LA, Onwuneme C, Watson WR, Murphy JFA, Slevin M, Donoghue V, O'Neill A, Molloy EJ. Neonatal Encephalopathy Is Associated With Altered IL-8 and GM-CSF Which Correlates With Outcomes. Front Pediatr 2020; 8:556216. [PMID: 33628760 PMCID: PMC7899044 DOI: 10.3389/fped.2020.556216] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Aim: To investigate the relationship between cytokines associated with innate immune cell activation and brain injury and outcome in infants with NE compared to neonatal controls. Methods: Serum and CSF biomarkers associated with activated neutrophils and monocytes [Interleukin-8 (IL-8) and Granulocyte-Macrophage-Colony-Stimulating-Factor (GM-CSF)] were serially measured using duplex immunoassays on days 1, 3 and 7 in term newborns with NE and controls. Results were compared to grade of encephalopathy, seizures, MRI brain imaging, mortality and Bayley Score of Infant and Toddler Development (Bayley-III) at 2 years of age. Results: Ninety-four infants had serum samples collected with 34 CSF samples. NE Grade II/III was significantly associated with elevated on day 2 serum IL-8. Mortality was best predicted by elevated day 1 IL-8. GM-CSF was initially elevated on day 1 and abnormal MRI imaging was associated with decreased day 2 GM-CSF. Elevated GM-CSF at day of life 6-7 correlated negatively with composite cognitive, language and motor Bayley-III scores at 2 years. Conclusion: Moderate or severe NE and mortality was associated with elevated IL-8. Day 2 GM-CSF could predict abnormal MRI results in NE and Bayley-III. Therefore, these cytokines are altered in NE and may predict early outcomes and further implicate inflammatory processes in NE.
Collapse
Affiliation(s)
- Deirdre U Sweetman
- Neonatology, National Maternity Hospital, Dublin, Ireland.,National Children's Research Centre, Dublin, Ireland.,Paediatrics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tammy Strickland
- Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St James Hospital, Dublin, Ireland
| | - Ashanty M Melo
- Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St James Hospital, Dublin, Ireland
| | - Lynne A Kelly
- National Children's Research Centre, Dublin, Ireland.,Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St James Hospital, Dublin, Ireland
| | - Chike Onwuneme
- Neonatology, National Maternity Hospital, Dublin, Ireland.,UCD School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - William R Watson
- UCD School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - John F A Murphy
- Neonatology, National Maternity Hospital, Dublin, Ireland.,Paediatrics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Marie Slevin
- Neonatology, National Maternity Hospital, Dublin, Ireland
| | - Veronica Donoghue
- Radiology Department, Children's University Hospital, Dublin, Ireland
| | - Amanda O'Neill
- UCD School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Eleanor J Molloy
- National Children's Research Centre, Dublin, Ireland.,Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St James Hospital, Dublin, Ireland.,Childrens University Hospital (CHI) at Tallght, Tallaght University Hospital, Dublin, Ireland.,Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland.,Neonatology, Children's Health Ireland at Crumlin, Dublin, Ireland
| |
Collapse
|
27
|
Pregnolato S, Chakkarapani E, Isles AR, Luyt K. Glutamate Transport and Preterm Brain Injury. Front Physiol 2019; 10:417. [PMID: 31068830 PMCID: PMC6491644 DOI: 10.3389/fphys.2019.00417] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/27/2019] [Indexed: 12/19/2022] Open
Abstract
Preterm birth complications are the leading cause of child death worldwide and a top global health priority. Among the survivors, the risk of life-long disabilities is high, including cerebral palsy and impairment of movement, cognition, and behavior. Understanding the molecular mechanisms of preterm brain injuries is at the core of future healthcare improvements. Glutamate excitotoxicity is a key mechanism in preterm brain injury, whereby the accumulation of extracellular glutamate damages the delicate immature oligodendrocytes and neurons, leading to the typical patterns of injury seen in the periventricular white matter. Glutamate excitotoxicity is thought to be induced by an interaction between environmental triggers of injury in the perinatal period, particularly cerebral hypoxia-ischemia and infection/inflammation, and developmental and genetic vulnerabilities. To avoid extracellular build-up of glutamate, the brain relies on rapid uptake by sodium-dependent glutamate transporters. Astrocytic excitatory amino acid transporter 2 (EAAT2) is responsible for up to 95% of glutamate clearance, and several lines of evidence suggest that it is essential for brain functioning. While in the adult EAAT2 is predominantly expressed by astrocytes, EAAT2 is transiently upregulated in the immature oligodendrocytes and selected neuronal populations during mid-late gestation, at the peak time for preterm brain injury. This developmental upregulation may interact with perinatal hypoxia-ischemia and infection/inflammation and contribute to the selective vulnerability of the immature oligodendrocytes and neurons in the preterm brain. Disruption of EAAT2 may involve not only altered expression but also impaired function with reversal of transport direction. Importantly, elevated EAAT2 levels have been found in the reactive astrocytes and macrophages of human infant post-mortem brains with severe white matter injury (cystic periventricular leukomalacia), potentially suggesting an adaptive mechanism against excitotoxicity. Interestingly, EAAT2 is suppressed in animal models of acute hypoxic-ischemic brain injury at term, pointing to an important and complex role in newborn brain injuries. Enhancement of EAAT2 expression and transport function is gathering attention as a potential therapeutic approach for a variety of adult disorders and awaits exploration in the context of the preterm brain injuries.
Collapse
Affiliation(s)
- Silvia Pregnolato
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Elavazhagan Chakkarapani
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Anthony R Isles
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Karen Luyt
- Department of Neonatal Neurology, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
28
|
Fu G, Zhang W, Dai J, Liu J, Li F, Wu D, Xiao Y, Shah C, Sweeney JA, Wu M, Lui S. Increased Peripheral Interleukin 10 Relate to White Matter Integrity in Schizophrenia. Front Neurosci 2019; 13:52. [PMID: 30792621 PMCID: PMC6374337 DOI: 10.3389/fnins.2019.00052] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/21/2019] [Indexed: 02/05/2023] Open
Abstract
Background: Schizophrenia is characterized by the disruption of microstructural white matter (WM) integrity, while the pathogenesis remains unclear. Inflammation has been associated with the WM pathology in schizophrenia. Interleukin 10 (IL-10) has been proven to be related to schizophrenia in both animal and human models. The aim of this study was to explore whether peripheral IL-10 was associated with microstructural WM integrity in schizophrenia. Methods: A total of 47 patients with schizophrenia (SZ) and 49 healthy controls (HC) underwent diffusion tensor imaging and venous blood sampling. Tract-based spatial statistics was conducted to explore the differences in fractional anisotropy (FA), radial diffusivity (RD), mean diffusivity (MD), and axial diffusivity (AD) between patients and controls. A quantitative chemiluminescence assay was performed to measure peripheral IL-10 levels. General linear regression analysis using a stepwise method was applied to examine the relationship between peripheral IL-10 and diffusion measures. Results: Compared with the HC, peripheral IL-10 levels were higher and a significant reduction of FA and AD, and increase of RD and MD were observed in SZ (corrected p < 0.05). A regression analysis revealed that peripheral IL-10 was negatively correlated with FA in the right posterior thalamic radiation and left inferior fronto-occipital fasciculus, in SZ (β = -0.51, p = 0.01; β = -0.47, p = 0.02, respectively) but not in HC (β = -0.01, p = 0.95; β = -0.003, p = 0.98, respectively), and the differences in regression curves were significant (z = 2.50, p = 0.01; z = 2.37, p = 0.02, respectively). IL-10 was negatively connected with MD in the right parietal arcuate fasciculus (β = -0.40, p = 0.048) and body of the corpus callosum (β = -0.43, p = 0.03) in SZ, while not in HC. The magnitude of correlation in the patient and control group was different (z = 2.48, p = 0.01 and z = 2.61, p < 0.01, respectively). In addition, IL-10 was positively correlated with RD in the right parietal arcuate fasciculus in patients (β = 0.45, p = 0.04) but not in HC (β = 0.26, p = 0.94), but the correlation coefficients were not significant (z = 0.98, p = 0.32). Conclusion: Our findings demonstrated that elevated peripheral IL-10 levels were associated with the disruption of microstructural WM integrity in schizophrenia, supporting the notion that inflammation plays a regulatory role in the pathology of microstructural WM and is associated with schizophrenia.
Collapse
Affiliation(s)
- Gui Fu
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Wenjing Zhang
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Jing Dai
- The Fourth People’s Hospital of Chengdu, Sichuan, China
| | - Jieke Liu
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Fei Li
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Dongsheng Wu
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Yuan Xiao
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Chandan Shah
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - John A. Sweeney
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, United States
| | - Min Wu
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Su Lui
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Plummer EA, Wang Q, Larson-Nath CM, Scheurer JM, Ramel SE. Body composition and cognition in preschool-age children with congenital gastrointestinal anomalies. Early Hum Dev 2019; 129:5-10. [PMID: 30562643 PMCID: PMC6382521 DOI: 10.1016/j.earlhumdev.2018.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/30/2018] [Accepted: 12/02/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Children with congenital gastrointestinal anomalies (CGIAs) experience multiple stressors while hospitalized in neonatal intensive care units during an essential time of growth and development. Early stress and inadequate nutrition are linked to altered growth patterns and later neurodevelopmental delays. In other at-risk populations, improved fat-free mass (FFM) accretion is associated with improved cognitive outcomes. OBJECTIVE To determine if body composition is associated with cognitive function in preschool-age children with CGIAs. STUDY DESIGN An observational study examined body composition and cognition in 34 preschool-age children with CGIAs. Anthropometric measurements and body composition testing via air displacement plethysmography were obtained. Measurements were compared with a reference group of healthy, term-born children. Cognition was measured with the NIH Toolbox Early Childhood Cognition Battery. Linear regression was used to test the association of body composition with cognitive function. RESULTS Compared with the reference group, children with CGIAs had similar anthropometric measurements (weight, height, and body mass index z-scores) and body composition at preschool-age. Processing speed scores were lower than standardized means (p = 0.001). Increased FFM was associated with higher receptive vocabulary scores (p = 0.001), cognitive flexibility scores (p = 0.005), and general cognitive function scores (p = 0.05). CONCLUSIONS At preschool-age, children with CGIAs have similar growth and body composition to their peers. In children with CGIAs, higher FFM was associated with higher cognitive scores. Closer tracking of body composition and interventions aimed at increasing FFM may improve long-term outcomes in this population.
Collapse
Affiliation(s)
- Erin A Plummer
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States.
| | - Qi Wang
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis, MN, United States
| | - Catherine M Larson-Nath
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Johannah M Scheurer
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Sara E Ramel
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
30
|
van den Heuij LG, Fraser M, Miller SL, Jenkin G, Wallace EM, Davidson JO, Lear CA, Lim R, Wassink G, Gunn AJ, Bennet L. Delayed intranasal infusion of human amnion epithelial cells improves white matter maturation after asphyxia in preterm fetal sheep. J Cereb Blood Flow Metab 2019; 39:223-239. [PMID: 28895475 PMCID: PMC6365606 DOI: 10.1177/0271678x17729954] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
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.
Collapse
Affiliation(s)
- Lotte G van den Heuij
- 1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Mhoyra Fraser
- 1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Suzanne L Miller
- 2 The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Graham Jenkin
- 2 The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Euan M Wallace
- 2 The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Joanne O Davidson
- 1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Christopher A Lear
- 1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Rebecca Lim
- 2 The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Guido Wassink
- 1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- 1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- 1 Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
31
|
Abstract
Despite the advances in neonatal intensive care, the preterm brain remains vulnerable to white matter injury (WMI) and disruption of normal brain development (i.e., dysmaturation). Compared to severe cystic WMI encountered in the past decades, contemporary cohorts of preterm neonates experience milder WMIs. More than destructive lesions, disruption of the normal developmental trajectory of cellular elements of the white and the gray matter occurs. In the acute phase, in response to hypoxia-ischemia and/or infection and inflammation, multifocal areas of necrosis within the periventricular white matter involve all cellular elements. Later, chronic WMI is characterized by diffuse WMI with aberrant regeneration of oligodendrocytes, which fail to mature to myelinating oligodendrocytes, leading to myelination disturbances. Complete neuronal degeneration classically accompanies necrotic white matter lesions, while altered neurogenesis, represented by a reduction of the dendritic arbor and synapse formation, is observed in response to diffuse WMI. Neuroimaging studies now provide more insight in assessing both injury and dysmaturation of both gray and white matter. Preterm brain injury remains an important cause of neurodevelopmental disabilities, which are still observed in up to 50% of the preterm survivors and take the form of a complex combination of motor, cognitive, and behavioral concerns.
Collapse
Affiliation(s)
- Juliane Schneider
- Department of Woman-Mother-Child, Clinic of Neonatology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | - Steven P Miller
- Division of Neurology and Centre for Brain and Mental Health, Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
32
|
Nalivaeva NN, Turner AJ, Zhuravin IA. Role of Prenatal Hypoxia in Brain Development, Cognitive Functions, and Neurodegeneration. Front Neurosci 2018; 12:825. [PMID: 30510498 PMCID: PMC6254649 DOI: 10.3389/fnins.2018.00825] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/22/2018] [Indexed: 12/15/2022] Open
Abstract
This review focuses on the role of prenatal hypoxia in the development of brain functions in the postnatal period and subsequent increased risk of neurodegenerative disorders in later life. Accumulating evidence suggests that prenatal hypoxia in critical periods of brain formation results in significant changes in development of cognitive functions at various stages of postnatal life which correlate with morphological changes in brain structures involved in learning and memory. Prenatal hypoxia also leads to a decrease in brain adaptive potential and plasticity due to the disturbance in the process of formation of new contacts between cells and propagation of neuronal stimuli, especially in the cortex and hippocampus. On the other hand, prenatal hypoxia has a significant impact on expression and processing of a variety of genes involved in normal brain function and their epigenetic regulation. This results in changes in the patterns of mRNA and protein expression and their post-translational modifications, including protein misfolding and clearance. Among proteins affected by prenatal hypoxia are a key enzyme of the cholinergic system-acetylcholinesterase, and the amyloid precursor protein (APP), both of which have important roles in brain function. Disruption of their expression and metabolism caused by prenatal hypoxia can also result, apart from early cognitive dysfunctions, in development of neurodegeneration in later life. Another group of enzymes affected by prenatal hypoxia are peptidases involved in catabolism of neuropeptides, including amyloid-β peptide (Aβ). The decrease in the activity of neprilysin and other amyloid-degrading enzymes observed after prenatal hypoxia could result over the years in an Aβ clearance deficit and accumulation of its toxic species which cause neuronal cell death and development of neurodegeneration. Applying various approaches to restore expression of neuronal genes disrupted by prenatal hypoxia during postnatal development opens an avenue for therapeutic compensation of cognitive dysfunctions and prevention of Aβ accumulation in the aging brain and the model of prenatal hypoxia in rodents can be used as a reliable tool for assessment of their efficacy.
Collapse
Affiliation(s)
- Natalia N. Nalivaeva
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Anthony J. Turner
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Igor A. Zhuravin
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Research Centre, Saint-Petersburg State Pediatric Medical University, St. Petersburg, Russia
| |
Collapse
|
33
|
Boardman JP, Ireland G, Sullivan G, Pataky R, Fleiss B, Gressens P, Miron V. The Cerebrospinal Fluid Inflammatory Response to Preterm Birth. Front Physiol 2018; 9:1299. [PMID: 30258368 PMCID: PMC6144928 DOI: 10.3389/fphys.2018.01299] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022] Open
Abstract
Background: Preterm birth is the leading risk factor for perinatal white matter injury, which can lead to motor and neuropsychiatric impairment across the life course. There is an unmet clinical need for therapeutics. White matter injury is associated with an altered inflammatory response in the brain, primarily led by microglia, and subsequent hypomyelination. However, microglia can release both damaging and trophic factors in response to injury, and a comprehensive assessment of these factors in the preterm central nervous system (CNS) has not been carried out. Method: A custom antibody array was used to assess relative levels of 50 inflammation- and myelination-associated proteins in the cerebrospinal fluid (CSF) of preterm infants in comparison to term controls. Results: Fifteen proteins differed between the groups: BDNF, BTC, C5a, FasL, Follistatin, IL-1β, IL-2, IL-4, IL-9, IL-17A, MIP-1α, MMP8, SPP1, TGFβ, and TNFβ (p < 0.05). To investigate the temporal regulation of these proteins after injury, we mined a gene expression dataset of microglia isolated from a mouse model of developmental white matter injury. Microglia in the experimental model showed dynamic temporal expression of genes encoding these proteins, with an initial and sustained pro-inflammatory response followed by a delayed anti-inflammatory response, and a continuous expression of genes predicted to inhibit healthy myelination. Conclusion: Preterm CSF shows a distinct neuroinflammatory profile compared to term controls, suggestive of a complex neural environment with concurrent damaging and reparative signals. We propose that limitation of pro-inflammatory responses, which occur early after perinatal insult, may prevent expression of myelination-suppressive genes and support healthy white matter development.
Collapse
Affiliation(s)
- James P Boardman
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, United Kingdom
| | - Graeme Ireland
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Gemma Sullivan
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Rozalia Pataky
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Bobbi Fleiss
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, United Kingdom.,PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,PremUP, Paris, France
| | - Pierre Gressens
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, United Kingdom.,PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,PremUP, Paris, France
| | - Veronique Miron
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
34
|
Sun J, Pan X, Christiansen LI, Yuan XL, Skovgaard K, Chatterton DEW, Kaalund SS, Gao F, Sangild PT, Pankratova S. Necrotizing enterocolitis is associated with acute brain responses in preterm pigs. J Neuroinflammation 2018; 15:180. [PMID: 29885660 PMCID: PMC5994241 DOI: 10.1186/s12974-018-1201-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/14/2018] [Indexed: 12/24/2022] Open
Abstract
Background Necrotizing enterocolitis (NEC) is an acute gut inflammatory disorder that occurs in preterm infants in the first weeks after birth. Infants surviving NEC often show impaired neurodevelopment. The mechanisms linking NEC lesions with later neurodevelopment are poorly understood but may include proinflammatory signaling in the immature brain. Using preterm pigs as a model for preterm infants, we hypothesized that severe intestinal NEC lesions are associated with acute effects on the developing hippocampus. Methods Cesarean-delivered preterm pigs (n = 117) were reared for 8 days and spontaneously developed variable severity of NEC lesions. Neonatal arousal, physical activity, and in vitro neuritogenic effects of cerebrospinal fluid (CSF) were investigated in pigs showing NEC lesions in the colon (Co-NEC) or in the small intestine (Si-NEC). Hippocampal transcriptome analysis and qPCR were used to assess gene expressions and their relation to biological processes, including neuroinflammation, and neural plasticity. Microglia activation was quantified by stereology. The neuritogenic response to selected proteins was investigated in primary cultures of hippocampal neurons. Results NEC development rapidly reduced the physical activity of pigs, especially when lesions occurred in the small intestine. Si-NEC and Co-NEC were associated with 27 and 12 hippocampal differentially expressed genes (DEGs), respectively. These included genes related to neuroinflammation (i.e., S100A8, S100A9, IL8, IL6, MMP8, SAA, TAGLN2) and hypoxia (i.e., PDK4, IER3, TXNIP, AGER), and they were all upregulated in Si-NEC pigs. Genes related to protection against oxidative stress (HBB, ALAS2) and oligodendrocytes (OPALIN) were downregulated in Si-NEC pigs. CSF collected from NEC pigs promoted neurite outgrowth in vitro, and the S100A9 and S100A8/S100A9 proteins may mediate the neuritogenic effects of NEC-related CSF on hippocampal neurons. NEC lesions did not affect total microglial cell number but markedly increased the proportion of Iba1-positive amoeboid microglial cells. Conclusions NEC lesions, especially when present in the small intestine, are associated with changes to hippocampal gene expression that potentially mediate neuroinflammation and disturbed neural circuit formation via enhanced neuronal differentiation. Early brain-protective interventions may be critical for preterm infants affected by intestinal NEC lesions to reduce their later neurological dysfunctions. Electronic supplementary material The online version of this article (10.1186/s12974-018-1201-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing Sun
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark
| | - Xiaoyu Pan
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark
| | - Line I Christiansen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark
| | - Xiao-Long Yuan
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800, Kgs. Lyngby, Denmark
| | - Dereck E W Chatterton
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark.,Department of Food Science, University of Copenhagen, DK-1958, Frederiksberg C, Denmark
| | - Sanne S Kaalund
- Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospitals, DK-2400, Copenhagen, Denmark
| | - Fei Gao
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, 518000, Shenzhen, China
| | - Per T Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark. .,Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen, Denmark.
| | - Stanislava Pankratova
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Blegdamsvej 9, DK-2100, Copenhagen, Denmark. .,Laboratory of Neural Plasticity, Department of Neuroscience, University of Copenhagen, DK-2200, Copenhagen, Denmark.
| |
Collapse
|
35
|
Chalak L, Latremouille S, Mir I, Sánchez PJ, Sant'Anna G. A review of the conundrum of mild hypoxic-ischemic encephalopathy: Current challenges and moving forward. Early Hum Dev 2018; 120:88-94. [PMID: 29506900 DOI: 10.1016/j.earlhumdev.2018.02.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A review of the conundrum called mild hypoxic-ischemic encephalopathy (HIE) is provided. During the past decades, the definition of HIE has evolved to accommodate the short window of time required for the initiation of therapeutic hypothermia. Also, neurological evaluations have changed with the use of simpler staging systems that can be applied within the first 6 h of life. In this review, we discuss the challenges in the identification of newborns with "mild HIE" within 6 h after birth, the limitations in the existing early biomarkers of brain injury, and the current knowledge gaps in the long term neurodevelopmental outcomes of infants diagnosed with mild HIE. Progress in the understanding of mild HIE and its sequelae continues to be hindered by the lack of a standardized definition for mild HIE that will reliably identify at-risk infants who may benefit from neuroprotective strategies.
Collapse
Affiliation(s)
- Lina Chalak
- University of Texas Southwestern Medical Center, TX, Dallas, USA.
| | | | - Imran Mir
- University of Texas Southwestern Medical Center, TX, Dallas, USA
| | - Pablo J Sánchez
- Nationwide Children's Hospital - The Ohio State University College of Medicine, Columbus, OH, USA
| | | |
Collapse
|
36
|
Multiple Postnatal Infections in Newborns Born Preterm Predict Delayed Maturation of Motor Pathways at Term-Equivalent Age with Poorer Motor Outcomes at 3 Years. J Pediatr 2018; 196:91-97.e1. [PMID: 29398063 DOI: 10.1016/j.jpeds.2017.12.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/21/2017] [Accepted: 12/15/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To evaluate whether the number of postnatal infections is associated with abnormal white matter maturation and poorer motor neurodevelopmental outcomes at 36 months of corrected age. STUDY DESIGN A prospective longitudinal cohort study was undertaken of 219 newborns born preterm at 24-32 weeks of gestational age recruited between 2006 and 2013 with magnetic resonance imaging of the brain both early in life and at term-equivalent age. Postnatal infection was defined as any clinical infection or positive culture ≥72 hours after birth. White matter maturation was assessed by magnetic resonance spectroscopic imaging, magnetic resonance diffusion tensor imaging, and tract-based spatial statistics. Neurodevelopmental outcomes were assessed in 175 (82% of survivors) infants with Bayley Scales of Infant and Toddler Development-III composite scores and Peabody Developmental Motor Scales at 35 months of corrected age (IQR 34-37 months). Infection groups were compared via the Fisher exact test, Kruskal-Wallis test, and generalized estimating equations. RESULTS Of 219 neonates born preterm (median gestational age 27.9 weeks), 109 (50%) had no postnatal infection, 83 (38%) had 1 or 2 infections, and 27 (12%) had ≥3 infections. Infants with postnatal infections had more cerebellar hemorrhage. Infants with ≥3 infections had lower N-acetylaspartate/choline in the white matter and basal ganglia regions, lower fractional anisotropy in the posterior limb of the internal capsule, and poorer maturation of the corpus callosum, optic radiations, and posterior limb of the internal capsule on tract-based spatial statistics analysis as well as poorer Bayley Scales of Infant and Toddler Development-III (P = .02) and Peabody Developmental Motor Scales, Second Edition, motor scores (P < .01). CONCLUSIONS In newborns born preterm, ≥3 postnatal infections predict impaired development of the motor pathways and poorer motor outcomes in early childhood.
Collapse
|
37
|
TRPM2 Channel Aggravates CNS Inflammation and Cognitive Impairment via Activation of Microglia in Chronic Cerebral Hypoperfusion. J Neurosci 2018; 38:3520-3533. [PMID: 29507145 DOI: 10.1523/jneurosci.2451-17.2018] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 01/25/2018] [Accepted: 02/12/2018] [Indexed: 01/05/2023] Open
Abstract
Chronic cerebral hypoperfusion is a characteristic seen in widespread CNS diseases, including neurodegenerative and mental disorders, and is commonly accompanied by cognitive impairment. Recently, several studies demonstrated that chronic cerebral hypoperfusion can induce the excessive inflammatory responses that precede neuronal dysfunction; however, the precise mechanism of cognitive impairment due to chronic cerebral hypoperfusion remains unknown. Transient receptor potential melastatin 2 (TRPM2) is a Ca2+-permeable channel that is abundantly expressed in immune cells and is involved in aggravation of inflammatory responses. Therefore, we investigated the pathophysiological role of TRPM2 in a mouse chronic cerebral hypoperfusion model with bilateral common carotid artery stenosis (BCAS). When male mice were subjected to BCAS, cognitive dysfunction and white matter injury at day 28 were significantly improved in TRPM2 knock-out (TRPM2-KO) mice compared with wild-type (WT) mice, whereas hippocampal damage was not observed. There were no differences in blood-brain barrier breakdown and H2O2 production between the two genotypes at 14 and 28 d after BCAS. Cytokine production was significantly suppressed in BCAS-operated TRPM2-KO mice compared with WT mice at day 28. In addition, the number of Iba1-positive cells gradually decreased from day 14. Moreover, daily treatment with minocycline significantly improved cognitive perturbation. Surgical techniques using bone marrow chimeric mice revealed that activated Iba1-positive cells in white matter could be brain-resident microglia, not peripheral macrophages. Together, these findings suggest that microglia contribute to the aggravation of cognitive impairment by chronic cerebral hypoperfusion, and that TRPM2 may be a potential target for chronic cerebral hypoperfusion-related disorders.SIGNIFICANCE STATEMENT Chronic cerebral hypoperfusion is manifested in a wide variety of CNS diseases, including neurodegenerative and mental disorders that are accompanied by cognitive impairment; however, the underlying mechanisms require clarification. Here, we used a chronic cerebral hypoperfusion mouse model to investigate whether TRPM2, a Ca2+-permeable cation channel highly expressed in immune cells, plays a destructive role in the development of chronic cerebral hypoperfusion-induced cognitive impairment, and propose a new hypothesis in which TRPM2-mediated activation of microglia, not macrophages, specifically contributes to the pathology through the aggravation of inflammatory responses. These findings shed light on the understanding of the mechanisms of chronic cerebral hypoperfusion-related inflammation, and are expected to provide a novel therapeutic molecule for cognitive impairment in CNS diseases.
Collapse
|
38
|
Bennet L, Dhillon S, Lear CA, van den Heuij L, King V, Dean JM, Wassink G, Davidson JO, Gunn AJ. Chronic inflammation and impaired development of the preterm brain. J Reprod Immunol 2018; 125:45-55. [DOI: 10.1016/j.jri.2017.11.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 11/13/2017] [Accepted: 11/24/2017] [Indexed: 12/17/2022]
|
39
|
Šumanović-Glamuzina D, Čulo F, Čulo MI, Konjevoda P, Jerković-Raguž M. A comparison of blood and cerebrospinal fluid cytokines (IL-1β, IL-6, IL-18, TNF-α) in neonates with perinatal hypoxia. Bosn J Basic Med Sci 2017; 17:203-210. [PMID: 28418828 PMCID: PMC5581968 DOI: 10.17305/bjbms.2017.1381] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 11/15/2016] [Indexed: 11/16/2022] Open
Abstract
Perinatal hypoxia-ischemia is a specific and important pathological event in neonatal care practice. The data on relationship between the concentrations of cytokines in blood and cerebrospinal fluid (CSF) and perinatal brain injury are scarce. The aim of this study is to evaluate changes in interleukin (IL-1β, IL-6, and IL-18) and tumor necrosis factor alpha (TNF-α) levels in newborns with perinatal hypoxia (PNH). CSF and serum samples of 35 term and near-term (35-40 weeks) newborns with PNH, at the age of 3-96 hours, were analyzed using enzyme-linked immunosorbent assay. Control group consisted of 25 non-asphyxic/non-hypoxic infants of the same age sampled for clinically suspected perinatal meningitis, but proven negative and healthy otherwise. The cytokine values in CSF and serum samples were determined in relation to initial hypoxic-ischemic encephalopathy (HIE) staged according the Sarnat/Sarnat method, and compared with neurological outcome at 12 months of age estimated using Amiel-Tison procedure. The concentrations of IL-6 and TNF-α in serum of PNH patients were significantly higher compared to control group (p = 0.0407 and p = 0.023, respectively). No significant difference between average values of cytokines in relation to the stage of HIE was observed. Significantly higher levels of IL-6 and IL-18 corresponded to a mildly abnormal neurological outcome, while higher levels of IL-6 and TNF-α corresponded to a severely abnormal neurological outcome, at 12 months of age. Elevated serum levels of IL-6 and TNF-α better corresponded with hypoxia/ischemia compared to CSF values, within 96 hours of birth. Also, higher serum levels of IL-6, TNF-α, and IL-18 corresponded better with abnormal neurological outcome at 12 months of age, compared to CSF values.
Collapse
Affiliation(s)
- Darinka Šumanović-Glamuzina
- Department of Neonatology and Intensive Care, Clinic for Child Diseases, University Hospital Mostar, Mostar, Bosnia and Herzegovina.
| | | | | | | | | |
Collapse
|
40
|
Eisenhut M, Choudhury S. In Premature Newborns Intraventricular Hemorrhage Causes Cerebral Vasospasm and Associated Neurodisability via Heme-Induced Inflammasome-Mediated Interleukin-1 Production and Nitric Oxide Depletion. Front Neurol 2017; 8:423. [PMID: 28868047 PMCID: PMC5563367 DOI: 10.3389/fneur.2017.00423] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 08/04/2017] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Intraventricular hemorrhage (IVH) occurs in 60-70% of neonates weighing 500-750 g and 10-20% of those weighing 1,000-1,500 g. All forms of IVH have been associated with neurocognitive deficits. Both subarachnoid and IVHs have been associated with delayed vasospasm leading to neurological deficits. Pathways linking hemoglobin release from blood clots to vasospasm include heme-induced activation of inflammasomes releasing interleukin-1 (IL-1) that can cause calcium dependent and independent vasospasm. Free hemoglobin is a potent scavenger of nitric oxide (NO). Depletion of NO, a potent endogenous vasodilator, has been associated with features of vasospasm. HYPOTHESIS In premature newborns, IVH causes cerebral vasospasm and associated neurodisability via heme-induced increased inflammasome-mediated IL-1 production and NO depletion. CONFIRMATION OF HYPOTHESIS AND IMPLICATIONS This hypothesis could be confirmed in the IVH animal model with visualization of any associated vasospasm by angiography and in newborns with IVH by transcranial Doppler ultrasonography and correlation with cerebrospinal fluid IL-1 and NO metabolite levels. Confirmation of the role of heme in activation of inflammasomes causing IL-1 production and NO binding could be achieved by measuring the effect of heme scavenging interventions on IL-1 levels and levels of NO metabolites. In addition to removal of the accumulated blood of an IVH by drainage, irrigation, and fibrinolytic therapy intrathecal application of vasodilators and heme scavenging agents like haptoglobin and haemopexin and systemic treatment with inhibitors of inflammasomes like telmisartan could be used to prevent and treat cerebral vasospasm, and thus reduce the risk of associated brain injury in premature neonates.
Collapse
Affiliation(s)
- Michael Eisenhut
- Pediatric Department, Luton and Dunstable University Hospital NHS Foundation Trust, Luton, Bedfordshire, United Kingdom
| | - Samyami Choudhury
- Pediatric Department, Luton and Dunstable University Hospital NHS Foundation Trust, Luton, Bedfordshire, United Kingdom
| |
Collapse
|
41
|
Patra A, Huang H, Bauer JA, Giannone PJ. Neurological consequences of systemic inflammation in the premature neonate. Neural Regen Res 2017; 12:890-896. [PMID: 28761416 PMCID: PMC5514858 DOI: 10.4103/1673-5374.208547] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Despite substantial progress in neonatal care over the past two decades leading to improved survival of extremely premature infants, extreme prematurity continues to be associated with long term neurodevelopmental impairments. Cerebral white matter injury is the predominant form of insult in preterm brain leading to adverse neurological consequences. Such brain injury pattern and unfavorable neurologic sequelae is commonly encountered in premature infants exposed to systemic inflammatory states such as clinical or culture proven sepsis with or without evidence of meningitis, prolonged mechanical ventilation, bronchopulmonary dysplasia, necrotizing enterocolitis and chorioamnionitis. Underlying mechanisms may include cytokine mediated processes without direct entry of pathogens into the brain, developmental differences in immune response and complex neurovascular barrier system that play a critical role in regulating the cerebral response to various systemic inflammatory insults in premature infants. Understanding of these pathologic mechanisms and clinical correlates of such injury based on serum biomarkers or brain imaging findings on magnetic resonance imaging will pave way for future research and translational therapeutic opportunities for the developing brain.
Collapse
Affiliation(s)
- Aparna Patra
- OMNI Academic Service Line and Division of Neonatology, Department of Pediatrics, Kentucky Children's Hospital, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Hong Huang
- OMNI Academic Service Line and Division of Neonatology, Department of Pediatrics, Kentucky Children's Hospital, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - John A Bauer
- OMNI Academic Service Line and Division of Neonatology, Department of Pediatrics, Kentucky Children's Hospital, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Peter J Giannone
- OMNI Academic Service Line and Division of Neonatology, Department of Pediatrics, Kentucky Children's Hospital, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
42
|
|
43
|
Pataky R, Howie FA, Girardi G, Boardman JP. Complement C5a is present in CSF of human newborns and is elevated in association with preterm birth. J Matern Fetal Neonatal Med 2016; 30:2413-2416. [PMID: 27806664 PMCID: PMC5556752 DOI: 10.1080/14767058.2016.1251896] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neuroinflammation contributes to developmental brain injury associated with preterm birth, but the mediators that drive it are incompletely understood. Previous studies have shown that complement C5a is present and injurious in the brains of foetal mice exposed to preterm labour. Here, we demonstrate that C5a is present in the cerebrospinal fluid of newborn human infants and that levels are elevated in those born preterm. The difference is not explained by systemic infection. Complement activation in the neonatal brain and its role as a potential therapeutic target in preterm brain injury warrant further study. Activation in the neonatal brain and its role as a potential therapeutic target for preterm brain injury warrants further study.
Collapse
Affiliation(s)
- Rozalia Pataky
- a MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute , Edinburgh , UK and
| | - Forbes A Howie
- a MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute , Edinburgh , UK and
| | - Guillermina Girardi
- b Department of Women's Health , King's College London, St Thomas' Hospital , London , UK
| | - James P Boardman
- a MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute , Edinburgh , UK and
| |
Collapse
|
44
|
Schmidt AF, Kannan PS, Chougnet CA, Danzer SC, Miller LA, Jobe AH, Kallapur SG. Intra-amniotic LPS causes acute neuroinflammation in preterm rhesus macaques. J Neuroinflammation 2016; 13:238. [PMID: 27596440 PMCID: PMC5011884 DOI: 10.1186/s12974-016-0706-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/25/2016] [Indexed: 01/06/2023] Open
Abstract
Background Chorioamnionitis is associated with an increased risk of brain injury in preterm neonates. Inflammatory changes in brain could underlie this injury. Here, we evaluated whether neuroinflammation is induced by chorioamnionitis in a clinically relevant model. Methods Rhesus macaque fetuses were exposed to either intra-amniotic (IA) saline, or IA lipopolysaccharide (LPS) (1 mg) 16 or 48 h prior to delivery at 130 days (85 % of gestation) (n = 4–5 animals/group). We measured cytokines in the cerebrospinal fluid (CSF), froze samples from the left brain for molecular analysis, and immersion fixed the right brain hemisphere for immunohistology. We analyzed the messenger RNA (mRNA) levels of the pro-inflammatory cytokines IL-1β, CCL2, TNF-α, IL-6, IL-8, IL-10, and COX-2 in the periventricular white matter (PVWM), cortex, thalamus, hippocampus, and cerebellum by RT-qPCR. Brain injury was assessed by immunohistology for myelin basic protein (MBP), IBA1 (microglial marker), GFAP (astrocyte marker), OLIG2 (oligodendrocyte marker), NeuN (neuronal marker), CD3 (T cells), and CD14 (monocytes). Microglial proliferation was assessed by co-immunostaining for IBA1 and Ki67. Data were analyzed by ANOVA with Tukey’s post-test. Results IA LPS increased mRNA expression of pro-inflammatory cytokines in the PVWM, thalamus, and cerebellum, increased IL-6 concentration in the CSF, and increased apoptosis in the periventricular area after 16 h. Microglial proliferation in the white matter was increased 48 h after IA LPS. Conclusions LPS-induced chorioamnionitis caused neuroinflammation, microglial proliferation, and periventricular apoptosis in a clinically relevant model of chorioamnionitis in fetal rhesus macaques. These findings identify specific responses in the fetal brain and support the hypothesis that neuroinflammatory changes may mediate the adverse neurodevelopmental outcomes associated with chorioamnionitis. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0706-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Augusto F Schmidt
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Paranthaman S Kannan
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lisa A Miller
- California National Primate Research Center and Department of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| | - Alan H Jobe
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Suhas G Kallapur
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
| |
Collapse
|
45
|
Abstract
Although therapies in addition to whole-body cooling are being developed to treat the neonate at risk for hypoxic-ischemic encephalopathy, we have no quickly measured serum inflammatory or neuronal biomarkers to acutely and accurately identify brain injury or to follow the efficacy of therapy. This review covers inflammatory serum biomarkers in the setting of birth asphyxia that can help assess the degree or severity of encephalopathy at birth and neurodevelopmental outcomes. These biomarkers still need to be independently validated in large cohorts before they are ready for clinical implementation in practice.
Collapse
Affiliation(s)
- Lina F Chalak
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Room F3.312B, Dallas, TX 75390-9063, USA.
| |
Collapse
|
46
|
The Developing Microbiome of the Preterm Infant. Clin Ther 2016; 38:733-9. [PMID: 26947798 DOI: 10.1016/j.clinthera.2016.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/03/2016] [Accepted: 02/03/2016] [Indexed: 12/26/2022]
Abstract
PURPOSE To determine the importance of the neonatal microbiome in intestinal and overall health. METHOD A review of existing literature. FINDINGS AND IMPLICATIONS The microbiome is increasingly understood to have a significant role in health and disease. However, the microbiome of the preterm infant is unique, with simple microbial communities exposed to a consistent diet in a regulated environment, and development from naive to stable under the influence of the neonatal intensive care unit. This early microbiome encounters a still developing host and thus has the potential to program fundamental pathways with implications for neonatal and later outcomes.
Collapse
|
47
|
Pre- and Perinatal Ischemia-Hypoxia, the Ischemia-Hypoxia Response Pathway, and ADHD Risk. Behav Genet 2016; 46:467-77. [DOI: 10.1007/s10519-016-9784-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 02/10/2016] [Indexed: 02/06/2023]
|
48
|
Krishnadas R, Nicol A, Sassarini J, Puri N, Burden AD, Leman J, Combet E, Pimlott S, Hadley D, McInnes IB, Cavanagh J. Circulating tumour necrosis factor is highly correlated with brainstem serotonin transporter availability in humans. Brain Behav Immun 2016; 51:29-38. [PMID: 26255693 DOI: 10.1016/j.bbi.2015.08.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/04/2015] [Accepted: 08/04/2015] [Indexed: 11/24/2022] Open
Abstract
Preclinical studies demonstrate that pro-inflammatory cytokines increase serotonin transporter availability and function, leading to depressive symptoms in rodent models. Herein we investigate associations between circulating inflammatory markers and brainstem serotonin transporter (5-HTT) availability in humans. We hypothesised that higher circulating inflammatory cytokine concentrations, particularly of tumour necrosis factor (TNF-α), would be associated with greater 5-HTT availability, and that TNF-α inhibition with etanercept (sTNFR:Fc) would in turn reduce 5-HTT availability. In 13 neurologically healthy adult women, plasma TNF-α correlated significantly with 5-HTT availability (rho=0.6; p=0.03) determined by [(123)I]-beta-CIT SPECT scanning. This association was replicated in an independent sample of 12 patients with psoriasis/psoriatic arthritis (rho=0.76; p=0.003). Indirect effects analysis, showed that there was a significant overlap in the variance explained by 5-HTT availability and TNF-α concentrations on BDI scores. Treatment with etanercept for 6-8weeks was associated with a significant reduction in 5-HTT availability (Z=2.09; p=0.03; r=0.6) consistent with a functional link. Our findings confirm an association between TNF-α and 5-HTT in both the basal physiological and pathological condition. Modulation of both TNF-α and 5-HTT by etanercept indicate the presence of a mechanistic pathway whereby circulating inflammatory cytokines are related to central nervous system substrates underlying major depression.
Collapse
Affiliation(s)
- Rajeev Krishnadas
- Sackler Institute of Psychobiological Research, Institute of Health and Wellbeing, University of Glasgow, United Kingdom.
| | - Alice Nicol
- Institute of Neurological Sciences, Southern General Hospital, Glasgow, United Kingdom
| | - Jen Sassarini
- School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Navesh Puri
- Sackler Institute of Psychobiological Research, Institute of Health and Wellbeing, University of Glasgow, United Kingdom
| | - A David Burden
- Department of Dermatology, Western Infirmary, Glasgow, United Kingdom
| | - Joyce Leman
- Department of Dermatology, Western Infirmary, Glasgow, United Kingdom
| | - Emilie Combet
- School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Sally Pimlott
- West of Scotland Radionuclide Dispensary, Greater Glasgow and Clyde NHS Trust, United Kingdom
| | - Donald Hadley
- Institute of Neurological Sciences, Southern General Hospital, Glasgow, United Kingdom
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, United Kingdom
| | - Jonathan Cavanagh
- Sackler Institute of Psychobiological Research, Institute of Health and Wellbeing, University of Glasgow, United Kingdom.
| |
Collapse
|
49
|
Basu S, Agarwal P, Anupurba S, Shukla R, Kumar A. Elevated plasma and cerebrospinal fluid interleukin-1 beta and tumor necrosis factor-alpha concentration and combined outcome of death or abnormal neuroimaging in preterm neonates with early-onset clinical sepsis. J Perinatol 2015; 35:855-61. [PMID: 26226245 DOI: 10.1038/jp.2015.86] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/30/2015] [Accepted: 06/16/2015] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Prematurity and sepsis are the major contributors of neonatal mortality and neurodevelopmental sequelae. The present study was conducted to measure the plasma and cerebrospinal fluid (CSF) concentration of interleukin (IL)-1β and tumor necrotic factor (TNF)-α in preterm neonates with early-onset clinical sepsis (EOCS), and to find out their association with combined outcome of death or abnormal neuroimaging. STUDY DESIGN Thirty-two preterm (⩽34 weeks) neonates with EOCS and 32 gestational age-matched, healthy neonates served as cases and controls, respectively. Samples were collected soon after birth. Neonates were followed up clinically and by serial cranial ultrasonography (CUS) until discharge and subsequently by magnetic resonance imaging (MRI) of brain until 1 year. Developmental screening was done by Denver Developmental Screening test-II. RESULT In EOCS group, no neonate had any clinical/microbiological evidence of meningitis. Blood culture was positive in 17 (53%). CUS was abnormal in 12 (37%) (intracranial hemorrhage-11, periventricular leukomalacia-1). Ten (31%) neonates expired. Significant elevation of plasma and CSF IL-1β and TNF-α was observed in the EOCS group. On follow-up, seven (22%) neonates showed evidence of white matter damage in MRI, two of them had developmental delay and microcephaly. Plasma and CSF IL-1β and TNF-α concentration were significantly elevated in deceased neonates and those with abnormal neuroimaging. Both biomarkers demonstrated high predictive accuracy for poor outcome in receiver-operating curve analysis. CONCLUSION Elevation of plasma and CSF IL-1β and TNF-α is associated with an increase in the combined outcome of death or abnormal neuroimaging in preterm neonates with EOCS in the absence of clinical/microbiological evidence of meningitis with high predictive accuracy.
Collapse
Affiliation(s)
- S Basu
- Division of Neonatology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - P Agarwal
- Division of Neonatology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - S Anupurba
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - R Shukla
- Department of Radiodiagnosis, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - A Kumar
- Division of Neonatology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
50
|
Jin C, Londono I, Mallard C, Lodygensky GA. New means to assess neonatal inflammatory brain injury. J Neuroinflammation 2015; 12:180. [PMID: 26407958 PMCID: PMC4583178 DOI: 10.1186/s12974-015-0397-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 09/10/2015] [Indexed: 01/23/2023] Open
Abstract
Preterm infants are especially vulnerable to infection-induced white matter injury, associated with cerebral palsy, cognitive and psychomotor impairment, and other adverse neurological outcomes. The etiology of such lesions is complex and multifactorial. Furthermore, timing and length of exposure to infection also influence neurodevelopmental outcomes. Different mechanisms have been posited to mediate the observed brain injury including microglial activation followed by subsequent release of pro-inflammatory species, glutamate-induced excitotoxicity, and vulnerability of developing oligodendrocytes to cerebral insults. The prevalence of such neurological impairments requires an urgent need for early detection and effective neuroprotective strategies. Accordingly, noninvasive methods of monitoring disease progression and therapy effectiveness are essential. While diagnostic tools using biomarkers from bodily fluids may provide useful information regarding potential risks of developing neurological diseases, the use of magnetic resonance imaging/spectroscopy has emerged as a promising candidate for such purpose. Various pharmacological agents have demonstrated protective effects in the immature brain in animal models; however, few studies have progressed to clinical trials with promising results.
Collapse
Affiliation(s)
- Chen Jin
- Department of Pediatrics, Sainte-Justine Hospital and Research Center, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada.
| | - Irene Londono
- Department of Pediatrics, Sainte-Justine Hospital and Research Center, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada.
| | - Carina Mallard
- Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden.
| | - Gregory A Lodygensky
- Department of Pediatrics, Sainte-Justine Hospital and Research Center, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada. .,Montreal Heart Institute, 5000 Rue Bélanger, Montréal, Québec, Canada. .,Department of Neuroscience and Pharmacology, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|