1
|
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
|
2
|
Hernández-Rodríguez J, Pérez-Hernández J, Flores-Espinosa P, Olmos-Ortiz A, Velazquez P, Zamora-Escudero R, Islas-López M, Helguera-Repetto AC, Hernández-Bones K, Rodríguez-Flores S, Jiménez-Escutia R, Fortanel-Fonseca A, Flores-Pliego A, Lopez-Vancell R, Zaga-Clavellina V. Galectin-1 Elicits a Tissue-Specific Anti-Inflammatory and Anti-Degradative Effect Upon LPS-Induced Response in an Ex Vivo Model of Human Fetal Membranes Modeling an Intraamniotic Inflammation. Am J Reprod Immunol 2024; 92:e70016. [PMID: 39575516 PMCID: PMC11582940 DOI: 10.1111/aji.70016] [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: 08/22/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/24/2024] Open
Abstract
PROBLEM Intrauterine infection is one of the most jeopardizing conditions associated with adverse outcomes, including preterm birth; however, multiple tolerance mechanisms operate at the maternal-fetal interface to avoid the rejection of the fetus. Among the factors that maintain the uterus as an immunoprivileged site, Galectin-1 (Gal-1), an immunomodulatory glycan-binding protein secreted by the maternal-fetal unit, is pivotal in promoting immune cell homeostasis. This work aimed to evaluate the role of Gal-1 during a lipopolysaccharide (LPS)-induced-inflammatory milieu. METHOD OF STUDY Using an ex vivo culture with two independent compartments, human fetal membranes at term were pretreated with 40 and 80 ng/mL of Gal-1, then to reproduce an intraamniotic inflammation, the fetal side of membranes was stimulated with 500 ng/mL of LPS for 24 h. The concentrations of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, monocyte chemoattractant protein (MCP1), macrophage inflammatory protein (MIP1) α, regulated upon activation normal T cell expressed and secreted (RANTES), and matrix metalloproteinase (MMP)-9 were measured in both amnion and choriodecidua compartments. RESULTS In a tissue-specific fashion profile, pretreatment with the physiologic concentration of Gal-1 significantly diminished the LPS-dependent secretion of TNF-α, IL-1β, Il-6, MCP1, MIP1α, RANTES, and MMP-9. CONCLUSION Gal-1 elicits an anti-inflammatory effect on the human fetal membranes stimulated with LPS, which supports the hypothesis that Gal-1 is part of the immunomodulatory mechanisms intended to stop the harmful effect of inflammation of the maternal-fetal interface.
Collapse
Affiliation(s)
- Jazmin Hernández-Rodríguez
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
- Laboratorio de Patología Experimental UME, Unidad de Medicina Experimental, Facultad de Medicina UNAM, Ciudad Universitaria, Ciudad de México, Mexico
| | - Jesús Pérez-Hernández
- Laboratorio de Patología Experimental UME, Unidad de Medicina Experimental, Facultad de Medicina UNAM, Ciudad Universitaria, Ciudad de México, Mexico
| | - Pilar Flores-Espinosa
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Andrea Olmos-Ortiz
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Pilar Velazquez
- Departamento de Ginecología y Obstetricia, Hospital Ángeles México, Ciudad de México, Mexico
| | | | - Marcela Islas-López
- Ginecología y Obstetricia, Hospital Ángeles Lomas-UNAM, Huixquilucan, Mexico
| | | | - Karla Hernández-Bones
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
- Posgrado en Ciencias Médicas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Samara Rodríguez-Flores
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
- Posgrado en Ciencias Médicas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Rodrigo Jiménez-Escutia
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | | | - Arturo Flores-Pliego
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Rosario Lopez-Vancell
- Laboratorio de Patología Experimental UME, Unidad de Medicina Experimental, Facultad de Medicina UNAM, Ciudad Universitaria, Ciudad de México, Mexico
| | | |
Collapse
|
3
|
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:NPM230224. [PMID: 39365327 DOI: 10.3233/npm-230224] [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: 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)
- J Saito
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Kanagawa, Japan
| | - J Shibasaki
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Kanagawa, Japan
| | - K Yamamoto
- Department of Biostatistics, School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
| | - M Fujita
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Kanagawa, Japan
| | - K Toyoshima
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Kanagawa, Japan
| |
Collapse
|
4
|
Branagan A, Molloy EJ, Badawi N, Nelson KB. Causes and Terminology in Neonatal Encephalopathy: What is in a Name? Neonatal Encephalopathy, Hypoxic-ischemic Encephalopathy or Perinatal Asphyxia. Clin Perinatol 2024; 51:521-534. [PMID: 39095093 DOI: 10.1016/j.clp.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Neurologic depression in term/near-term neonates (neonatal encephalopathy, NE) is uncommon with modern obstetric care. Asphyxial birth, with or without co-factors, accounts for a minority of NE, while maldevelopment (congenital malformations, growth aberrations, genetic, metabolic and placental abnormalities) plays an enlarging role in identifying etiologic subgroups of NE. The terms NE and hypoxic-ischemic encephalopathy (HIE) have not been employed uniformly, hampering research and clinical care. The authors propose the term NE as an early working-diagnosis, to be supplemented by a diagnosis of NE due to HIE or to other factors, as a final diagnosis once workup is complete.
Collapse
Affiliation(s)
- Aoife Branagan
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Trinity Translational Medicine Institute (TTMI), St James Hospital & Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland; Department of Paediatrics, The Coombe Hospital, 32 Kickham Road, Inchicore, Dublin 8, Dublin D08W2T0, Ireland; Health Research Board Neonatal Encephalopathy PhD Training Network (NEPTuNE), Ireland
| | - Eleanor J Molloy
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Trinity Translational Medicine Institute (TTMI), St James Hospital & Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland; Department of Paediatrics, The Coombe Hospital, 32 Kickham Road, Inchicore, Dublin 8, Dublin D08W2T0, Ireland; Health Research Board Neonatal Encephalopathy PhD Training Network (NEPTuNE), Ireland; Department of Neonatology, Children's Health Ireland, Dublin, Ireland; Neurodisability, Children's Health Ireland (CHI) at Tallaght, Dublin, Ireland; Department of Paediatrics, Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.
| | - Nadia Badawi
- Cerebral Palsy Alliance Research Institute, Specialty of Child & Adolescent Health, Sydney Medical School; Faculty of Medicine & Health, Department of Paediatrics, The University of Sydney, PO Box 171, Allambie Heights, Sydney, New South Wales 2100, Australia; Grace Centre for Newborn Intensive Care, Sydney Children's Hospital Network, The University of Sydney, Westmead, New South Wales, Australia
| | - Karin B Nelson
- National Institutes of Health, National Institute of Neurological Diseases and Stroke, 050 Military Road NEW, Apt 815, Washington, DC 20015, USA
| |
Collapse
|
5
|
Tekgul H, Koroğlu OA, Tanrıverdi M, Yalaz M, Terek D, Aktan G, Akisu M, Kültürsay N. The net impact of clinical seizures on outcome characteristics in infants with neonatal encephalopathies at 12 months of age. Seizure 2024; 116:133-139. [PMID: 36588060 DOI: 10.1016/j.seizure.2022.12.007] [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: 11/08/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022] Open
Abstract
PURPOSE To assess the impact of clinical neonatal seizures on outcome characteristics of preterm and term newborns with neonatal encephalopathy (NE). METHODS We designed a prospective comparative study with 53 babies (preterm neonates: 26 and term neonates: 27) with NE: group 1 (preterm neonates with seizures, n = 13), group 2 (preterm neonates without seizures, n = 13), group 3 (term neonates with seizures, n = 13) and group 4 (term neonates without seizures, n = 14). The functional outcome characteristics of the survivors were assessed by the Ankara Developmental Screening Inventory (ADSI) and the Guide for Monitoring Child Development (GMCD) at 12 months of age. RESULTS Clinically defined acute symptomatic seizures were diagnosed with prompt conventional EEG / amplitude-integrated EEG in preterm (92.3%) and term neonates (81.4%) with etiology-specific diagnoses of NE. There were no differences between the study groups regarding seizure semiology and EEG characteristics. A primary adverse outcome was defined in 22 (41.5%) of the cohort. However, only 15.3% of infants had an unfavorable functional outcome with ADSI at 12 months. Among the survivors, there was no significant difference between the study groups regarding ADSI scores. The GMDC test revealed normal development in 50% of survivors with seizures in the preterm group and 83% in the term group. CONCLUSION There was no significant difference between the characteristics of functional outcomes at 12 months in preterm and term neonates with NE for clinical seizures.
Collapse
Affiliation(s)
- Hasan Tekgul
- Department of Pediatrics, Division of Child Neurology, Ege University Medical Faculty, Turkey.
| | - Ozge A Koroğlu
- Department of Pediatrics, Division of Neonatology, Ege University Medical Faculty, Turkey
| | - Mahir Tanrıverdi
- Department of Pediatrics, Ege University Medical Faculty, Turkey
| | - Mehmet Yalaz
- Department of Pediatrics, Division of Neonatology, Ege University Medical Faculty, Turkey
| | - Demet Terek
- Department of Pediatrics, Division of Neonatology, Ege University Medical Faculty, Turkey
| | - Gül Aktan
- Department of Pediatrics, Division of Child Neurology, Ege University Medical Faculty, Turkey
| | - Mete Akisu
- Department of Pediatrics, Division of Neonatology, Ege University Medical Faculty, Turkey
| | - Nilgün Kültürsay
- Department of Pediatrics, Division of Neonatology, Ege University Medical Faculty, Turkey
| |
Collapse
|
6
|
Gonzalez FF, Voldal E, Comstock BA, Mayock DE, Goodman AM, Cornet MC, Wu TW, Redline RW, Heagerty P, Juul SE, Wu YW. Placental Histologic Abnormalities and 2-Year Outcomes in Neonatal Hypoxic-Ischemic Encephalopathy. Neonatology 2023; 120:760-767. [PMID: 37742617 PMCID: PMC10711751 DOI: 10.1159/000533652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/12/2023] [Indexed: 09/26/2023]
Abstract
OBJECTIVE We aimed to examine the association between placental abnormalities and neurodevelopmental outcomes in a multicenter cohort of newborn infants with hypoxic-ischemic encephalopathy (HIE) that underwent therapeutic hypothermia. We hypothesized that subjects with acute placental abnormalities would have reduced risk of death or neurodevelopmental impairment (NDI) at 2 years of age after undergoing therapeutic hypothermia compared to subjects without acute placental changes. STUDY DESIGN Among 500 subjects born at ≥36 weeks gestation with moderate or severe HIE enrolled in the High-dose Erythropoietin for Asphyxia and Encephalopathy (HEAL) Trial, a placental pathologist blinded to clinical information reviewed clinical pathology reports to determine the presence of acute only, chronic only, or both acute and chronic histologic abnormalities. We calculated adjusted relative risks (aRRs) for associations between placental pathologic abnormalities and death or NDI at age 2 years, adjusting for HIE severity, treatment assignment, and site. RESULT 321/500 subjects (64%) had available placental pathology reports. Placental abnormalities were characterized as acute only (20%), chronic only (21%), both acute and chronic (43%), and none (15%). The risk of death or NDI was not statistically different between subjects with and without an acute placental abnormality (46 vs. 53%, aRR 1.1, 95% confidence interval (CI): 0.9, 1.4). Subjects with two or more chronic lesions were more likely to have an adverse outcome than subjects with no chronic abnormalities, though this did not reach statistical significance (55 vs. 45%, aRR 1.24, 95% CI: 0.99, 1.56). CONCLUSION Placental pathologic findings were not independently associated with risk of death or NDI in subjects with HIE. The relationship between multiple chronic placental lesions and HIE outcomes deserves further study.
Collapse
Affiliation(s)
- Fernando F. Gonzalez
- Department of Pediatrics, UCSF Benioff Children’s Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Emily Voldal
- Department Biostatistics, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bryan A. Comstock
- Department Biostatistics, University of Washington, Seattle, WA, USA
| | - Dennis E. Mayock
- Division of Neonatology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Amy M. Goodman
- Department of Neurology and Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA
| | - Marie-Coralie Cornet
- Department of Pediatrics, UCSF Benioff Children’s Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Tai-Wei Wu
- Division of Neonatology, Fetal and Neonatal Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Raymond W. Redline
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Patrick Heagerty
- Department Biostatistics, University of Washington, Seattle, WA, USA
| | - Sandra E. Juul
- Division of Neonatology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Yvonne W. Wu
- Department of Pediatrics, UCSF Benioff Children’s Hospital, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology and Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
7
|
Lenahan A, Mietzsch U, Wood TR, Callahan KP, Weiss EM, Miller DE, German K, Natarajan N, Puia-Dumitrescu M, Esposito V, Kolnik S, Law JB. Characteristics, Genetic Testing, and Diagnoses of Infants with Neonatal Encephalopathy Not Due to Hypoxic Ischemic Encephalopathy: A Cohort Study. J Pediatr 2023; 260:113533. [PMID: 37269901 DOI: 10.1016/j.jpeds.2023.113533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/29/2023] [Accepted: 05/30/2023] [Indexed: 06/05/2023]
Abstract
OBJECTIVE To characterize the presentation and evaluation of infants with neonatal encephalopathy (NE) not due to hypoxic-ischemic encephalopathy (non-HIE NE) and to describe the genetic abnormalities identified. STUDY DESIGN Retrospective cohort study of 193 non-HIE NE neonates admitted to a level IV NICU from 2015 through 2019. For changes in testing over time, Cochrane-Armitage test for trend was used with a Bonferroni-corrected P-value, and comparison between groups was performed using Fisher exact test. RESULT The most common symptom of non-HIE NE was abnormal tone in 47% (90/193). Ten percent (19/193) died prior to discharge, and 48% of survivors (83/174) required medical equipment at discharge. Forty percent (77/193) underwent genetic testing as an inpatient. Of 52 chromosomal studies, 54 targeted tests, and 16 exome sequences, 10%, 41%, and 69% were diagnostic, respectively, with no difference in diagnostic rates between infants with and without an associated congenital anomaly and/or dysmorphic feature. Twenty-eight genetic diagnoses were identified. CONCLUSIONS Neonates with non-HIE NE have high rates of morbidity and mortality and may benefit from early genetic testing, even in the absence of other exam findings. This study broadens our knowledge of genetic conditions underlying non-HIE NE, which may enable families and care teams to anticipate the needs of the individual, allow early initiation of targeted therapies, and facilitate decisions surrounding goals of care.
Collapse
Affiliation(s)
- Arthur Lenahan
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Ulrike Mietzsch
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Thomas R Wood
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Katharine Press Callahan
- Department of Pediatrics, Children's Hospital of Philadelphia, PA; Department of Medical Ethics and Health Policy, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Elliott M Weiss
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Danny E Miller
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Kendell German
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Niranjana Natarajan
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA; Division of Pediatric Neurology, Department of Neurology, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Mihai Puia-Dumitrescu
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Valentine Esposito
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Sarah Kolnik
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Janessa B Law
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA.
| |
Collapse
|
8
|
Zamstein O, Wainstock T, Sheiner E. Intrapartum Maternal Fever and Long-Term Infectious Morbidity of the Offspring. J Clin Med 2023; 12:jcm12093329. [PMID: 37176769 PMCID: PMC10179301 DOI: 10.3390/jcm12093329] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/29/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Maternal intrapartum fever can lead to various maternal and neonatal complications and is attributed to various etiologies including infectious and non-infectious processes. In this study, we evaluated whether intrapartum fever affects the offspring's tendency to long-term infectious morbidity. A population-based cohort analysis including deliveries between 1991 and 2021 was conducted. The incidence of hospitalizations of the offspring up to the age of 18 years, due to various infectious conditions, was compared between pregnancies complicated by intrapartum fever and those that were not. A Kaplan-Meier survival curve was used to assess cumulative hospitalization incidence. A Cox proportional hazards model was used to control for confounders. Overall, 538 of the 356,356 included pregnancies were complicated with fever. A higher rate of pediatric hospitalizations due to various infectious conditions was found among the exposed group, which was significant for viral, fungal and ENT infections (p < 0.05 for all). The total number of infectious-related hospitalizations was significantly higher (30.1% vs. 24.1%; OR = 1.36; p = 0.001), as was the cumulative incidence of hospitalizations. This association remained significant after controlling for confounders using a Cox proportional hazards model (adjusted HR = 1.21; 95% CI 1.04-1.41, p = 0.016). To conclude, fever diagnosed close to delivery may influence offspring susceptibility to pediatric infections.
Collapse
Affiliation(s)
- Omri Zamstein
- The Obstetrics and Gynecology Division, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva POB 151, Israel
| | - Tamar Wainstock
- The Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva POB 653, Israel
| | - Eyal Sheiner
- The Obstetrics and Gynecology Division, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva POB 151, Israel
| |
Collapse
|
9
|
Mir IN, Uddin N, Liao J, Brown LS, Leon R, Chalak LF, Savani RC, Rosenfeld CR. Placental clearance not synthesis tempers exaggerated pro-inflammatory cytokine response in neonates exposed to chorioamnionitis. Pediatr Res 2023; 93:675-681. [PMID: 35690685 DOI: 10.1038/s41390-022-02147-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/03/2022] [Accepted: 05/17/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND The source and clearance of cytokines in the fetal circulation in term pregnancies complicated by chorioamnionitis remains unclear as are the contributions of placental transport, synthesis, and clearance. The objectives of the study were to determine (1) fetal and/or placental contributions to synthesis and/or clearance of inflammatory and anti-inflammatory cytokines in term pregnancies complicated by chorioamnionitis and (2) whether this differs in pregnancies further complicated by fetal hypoxia. METHODS Prospective cohort study of pregnancies >37 weeks gestational age that included: Group 1, uncomplicated cesarean delivery without labor (n = 20); Group 2, uncomplicated vaginal delivery (n = 30); Group 3, pregnancies complicated by chorioamnionitis (n = 10); Group 4, complicated by chorioamnionitis + fetal hypoxia (n = 10). Umbilical arterial (UmA) and venous (UmV) blood were assayed for IL-1β, IL-2, IL-6, IL-8, TNFα, and IL-10. RESULTS IL-6 and IL-8 were below assay detection in UmA and UmV blood in Group 1 and increased in Group 2 (P < 0.01), UmA»UmV (P < 0.01). Their concentrations increased further in Groups 3 and 4 (P = 0.003), UmA»UmV. Placental clearance was concentration dependent that approaches saturation in the presence of chorioamnionitis. CONCLUSIONS Marked increases in fetal synthesis of IL-6 and IL-8 occur in chorioamnionitis. Synthesis increase further when complicated by fetal hypoxia. Cytokine removal occurs via placental concentration-dependent mechanisms, potentially contributing to adverse fetal effects. IMPACT The source and role of the placenta in synthesis and/or clearance of inflammatory mediators in term pregnancies complicated by clinical chorioamnionitis are unclear; however, conventional wisdom suggests the placenta is their source. This is the first study demonstrating that circulating concentrations of fetal IL-6 and IL-8 in clinical chorioamnionitis ± birth asphyxia in term pregnancies are of fetal origin. Circulating fetal inflammatory cytokines are cleared by concentration-dependent placental mechanisms that are nearly saturated in chorioamnionitis ± fetal hypoxia. These observations provide additional insight into understanding the fetal immune response in term pregnancies complicated by clinical chorioamnionitis.
Collapse
Affiliation(s)
- Imran N Mir
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Naseem Uddin
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jie Liao
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Center for Pulmonary & Vascular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Larry S Brown
- Parkland Health and Hospital Systems, Dallas, TX, USA
| | - Rachel Leon
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lina F Chalak
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rashmin C Savani
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,The Center for Pulmonary & Vascular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Charles R Rosenfeld
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
10
|
Rasineni GK, Panigrahy N, Rath SN, Chinnaboina M, Konanki R, Chirla DK, Madduri S. Diagnostic and Therapeutic Roles of the "Omics" in Hypoxic-Ischemic Encephalopathy in Neonates. Bioengineering (Basel) 2022; 9:498. [PMID: 36290466 PMCID: PMC9598631 DOI: 10.3390/bioengineering9100498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Perinatal asphyxia and neonatal encephalopathy remain major causes of neonatal mortality, despite the improved availability of diagnostic and therapeutic tools, contributing to neurological and intellectual disabilities worldwide. An approach using a combination of clinical data, neuroimaging, and biochemical parameters is the current strategy towards the improved diagnosis and prognosis of the outcome in neonatal hypoxic-ischemic encephalopathy (HIE) using bioengineering methods. Traditional biomarkers are of little use in this multifactorial and variable phenotype-presenting clinical condition. Novel systems of biology-based "omics" approaches (genomics, transcriptome proteomics, and metabolomics) may help to identify biomarkers associated with brain and other tissue injuries, predicting the disease severity in HIE. Biomarker studies using omics technologies will likely be a key feature of future neuroprotective treatment methods and will help to assess the successful treatment and long-term efficacy of the intervention. This article reviews the roles of different omics as biomarkers of HIE and outlines the existing knowledge of our current understanding of the clinical use of different omics molecules as novel neonatal brain injury biomarkers, which may lead to improved interventions related to the diagnostic and therapeutic aspects of HIE.
Collapse
Affiliation(s)
- Girish Kumar Rasineni
- LCMS Division, Tenet Medcorp Pvt. Ltd., 54 Kineta Towers Road No 3, Banjara Hills, Hyderabad 500034, India
| | | | - Subha Narayan Rath
- Regenerative Medicine and Stem Cell Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana 502284, India
| | - Madhurarekha Chinnaboina
- LCMS Division, Tenet Medcorp Pvt. Ltd., 54 Kineta Towers Road No 3, Banjara Hills, Hyderabad 500034, India
| | - Ramesh Konanki
- Department of Pediatric Neurology, Rainbow Children’s Hospital, Hyderabad 500034, India
| | - Dinesh Kumar Chirla
- Department of Neonatology, Rainbow Children’s Hospital, Hyderabad 500034, India
| | - Srinivas Madduri
- Bioengineering and Regenerative Medicine, Department of Biomedical Engineering, University of Basel, University Hospital Basel, 4001 Basel, Switzerland
- Department of Surgery, Bioengineering and Neuroregeneration, University of Geneva, University Hospital Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
11
|
Al Amrani F, Sébire G, Chen MF, Wintermark P, Saint-Martin C. Distinctive Neuroimaging Pattern in Term Newborns With Neonatal Placental Encephalopathy: A Case Series. Pediatr Neurol 2022; 126:74-79. [PMID: 34740136 DOI: 10.1016/j.pediatrneurol.2021.09.020] [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: 05/19/2021] [Revised: 09/11/2021] [Accepted: 09/27/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Identifying antepartum versus intrapartum timing and the cause of neonatal encephalopathy (NE) often remains elusive owing to our limited understanding of the underlying pathophysiological processes and lack of appropriate biomarkers. OBJECTIVES This retrospective observational study describes a case series of term newborns with NE who displayed a recognizable magnetic resonance imaging pattern of immediately postnatal brain abnormalities that rapidly evolved toward cavitation. Our aim is to (1) report this neuroimaging pattern, (2) look for placental determinants, and (3) depict the outcome. DESIGN/METHODS This is a unicentric retrospective case series reporting the clinical, radiological, and laboratory findings of NE associated with a distinctive neuroimaging pattern, that is, immediately postnatal extensive corticosubcortical T2 hyperintensities, followed by rapid corticosubcortical cavitation that does not match the neuroimaging picture of intrapartum hypoxic-ischemic encephalopathy (HIE). RESULTS Seven term newborns presented bilateral corticosubcortical hyperintensities that were detected on T2 between day of life (DOL) 1-4, which rapidly evolved toward cystic encephalomalacia, that is, between DOL9 and DOL12. All these newborns presented with moderate/severe NE. The outcome was either neonatal death or quadriplegic cerebral palsy and epilepsy. None of the reported patients fulfilled the criteria of a high likelihood of acute intrapartum hypoxic-ischemic or quadriplegic cerebral palsy. All these newborns were exposed to chronic and/or acute placental inflammation and/or hypoxic-ischemic. CONCLUSIONS To further define the antepartum causes of NE, early neuroimaging and a placental examination are recommended. Brain T2 hyperintense injuries before DOL4 followed by rapid cavitation before DOL12 might be biomarkers of NE from an antepartum/placental origin.
Collapse
Affiliation(s)
- Fatema Al Amrani
- Division of Pediatric Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University, Montreal, Quebec, Canada; Pediatric Neurology Unit, Child Health Department, Sultan Qaboos University Hospital, Al Khod, Muscat, Sultanate of Oman
| | - Guillaume Sébire
- Division of Pediatric Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University, Montreal, Quebec, Canada; Child Health and Human Development Program, Research Institute of McGill University Health Center, Montreal, Quebec, Canada.
| | - Moy Fong Chen
- Department of Pathology, Research Institute of McGill University Health Center, Montreal, Quebec, Canada
| | - Pia Wintermark
- Child Health and Human Development Program, Research Institute of McGill University Health Center, Montreal, Quebec, Canada; Division of Neonatology, Department of Pediatrics, Montreal Children's Hospital, Montreal, Quebec, Canada
| | - Christine Saint-Martin
- Division of Pediatric Medical Imaging, Department of Radiology, Montreal Children's Hospital, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Vasilakos LK, Steinbrekera B, Santillan DA, Santillan MK, Brandt DS, Dagle D, Roghair RD. Umbilical Cord Blood Leptin and IL-6 in the Presence of Maternal Diabetes or Chorioamnionitis. Front Endocrinol (Lausanne) 2022; 13:836541. [PMID: 35197933 PMCID: PMC8859102 DOI: 10.3389/fendo.2022.836541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/17/2022] [Indexed: 12/05/2022] Open
Abstract
Diabetes during pregnancy is associated with elevated maternal insulin, leptin and IL-6. Within the placenta, IL-6 can further stimulate leptin production. Despite structural similarities and shared roles in inflammation, leptin and IL-6 have contrasting effects on neurodevelopment, and the relative importance of maternal diabetes or chorioamnionitis on fetal hormone exposure has not been defined. We hypothesized that there would be a positive correlation between IL-6 and leptin with progressively increased levels in pregnancies complicated by maternal diabetes and chorioamnionitis. To test this hypothesis, cord blood samples were obtained from 104 term infants, including 47 exposed to maternal diabetes. Leptin, insulin, and IL-6 were quantified by multiplex assay. Factors independently associated with hormone levels were identified by univariate and multivariate linear regression. Unlike IL-6, leptin and insulin were significantly increased by maternal diabetes. Maternal BMI and birth weight were independent predictors of leptin and insulin with birth weight the strongest predictor of leptin. Clinically diagnosed chorioamnionitis and neonatal sepsis were associated with increased IL-6 but not leptin. Among appropriate for gestational age infants without sepsis, IL-6 and leptin were strongly correlated (R=0.6, P<0.001). In summary, maternal diabetes and birth weight are associated with leptin while chorioamnionitis is associated with IL-6. The constraint of the positive association between leptin and IL-6 to infants without sepsis suggests that the term infant and placenta may have a limited capacity to increase cord blood levels of the neuroprotective hormone leptin in the presence of increased cord blood levels of the potential neurotoxin IL-6.
Collapse
Affiliation(s)
- Lauren K. Vasilakos
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Baiba Steinbrekera
- Department of Pediatrics, University of South Dakota, Sioux Falls, SD, United States
| | - Donna A. Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Mark K. Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Debra S. Brandt
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Daniel Dagle
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Robert D. Roghair
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- *Correspondence: Robert D. Roghair,
| |
Collapse
|
13
|
Vitaliti G, Falsaperla R. Chorioamnionitis, Inflammation and Neonatal Apnea: Effects on Preterm Neonatal Brainstem and on Peripheral Airways: Chorioamnionitis and Neonatal Respiratory Functions. CHILDREN-BASEL 2021; 8:children8100917. [PMID: 34682182 PMCID: PMC8534519 DOI: 10.3390/children8100917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/01/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022]
Abstract
Background: The present manuscript aims to be a narrative review evaluating the association between inflammation in chorioamnionitis and damage on respiratory centers, peripheral airways, and lungs, explaining the pathways responsible for apnea in preterm babies born by delivery after chorioamnionitis. Methods: A combination of keywords and MESH words was used, including: "inflammation", "chorioamnionitis", "brainstem", "cytokines storm", "preterm birth", "neonatal apnea", and "apnea physiopathology". All identified papers were screened for title and abstracts by the two authors to verify whether they met the proper criteria to write the topic. Results: Chorioamnionitis is usually associated with Fetal Inflammatory Response Syndrome (FIRS), resulting in injury of brain and lungs. Literature data have shown that infections causing chorioamnionitis are mostly associated with inflammation and consequent hypoxia-mediated brain injury. Moreover, inflammation and infection induce apneic episodes in neonates, as well as in animal samples. Chorioamnionitis-induced inflammation favors the systemic secretion of pro-inflammatory cytokines that are involved in abnormal development of the respiratory centers in the brainstem and in alterations of peripheral airways and lungs. Conclusions: Preterm birth shows a suboptimal development of the brainstem and abnormalities and altered development of peripheral airways and lungs. These alterations are responsible for reduced respiratory control and apnea. To date, mostly animal studies have been published. Therefore, more clinical studies on the role of chorioamninitis-induced inflammation on prematurity and neonatal apnea are necessary.
Collapse
Affiliation(s)
- Giovanna Vitaliti
- Unit of Pediatrics, Department of Medical Sciences, Section of Pediatrics, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: ; Tel.: +39-34-0471-0614
| | - Raffaele Falsaperla
- Pediatrics and Pediatric Emergency Operative Unit, Azienda Ospedaliero Universitaria Policlinico G.Rodolico-San Marco, San Marco Hospital, University of Catania, 95124 Catania, Italy;
- Neonatal Intensive Care Unit, Azienda Ospedaliero Universitaria Policlinico G.Rodolico-San Marco, San Marco Hospital, San Marco Hospital, University of Catania, 95124 Catania, Italy
| |
Collapse
|
14
|
Chalak L, Redline RW, Goodman AM, Juul SE, Chang T, Yanowitz TD, Maitre N, Mayock DE, Lampland AL, Bendel-Stenzel E, Riley D, Mathur AM, Rao R, Van Meurs KP, Wu TW, Gonzalez FF, Flibotte J, Mietzsch U, Sokol GM, Ahmad KA, Baserga M, Weitkamp JH, Poindexter BB, Comstock BA, Wu YW. Acute and Chronic Placental Abnormalities in a Multicenter Cohort of Newborn Infants with Hypoxic-Ischemic Encephalopathy. J Pediatr 2021; 237:190-196. [PMID: 34144032 DOI: 10.1016/j.jpeds.2021.06.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/28/2021] [Accepted: 06/14/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To examine the frequency of placental abnormalities in a multicenter cohort of newborn infants with hypoxic-ischemic encephalopathy (HIE) and to determine the association between acuity of placental abnormalities and clinical characteristics of HIE. STUDY DESIGN Infants born at ≥36 weeks of gestation (n = 500) with moderate or severe HIE were enrolled in the High-dose Erythropoietin for Asphyxia and Encephalopathy Trial. A placental pathologist blinded to clinical information reviewed clinical pathology reports to determine the presence of acute and chronic placental abnormalities using a standard classification system. RESULTS Complete placental pathologic examination was available for 321 of 500 (64%) trial participants. Placental abnormalities were identified in 273 of 321 (85%) and were more common in infants ≥40 weeks of gestation (93% vs 81%, P = .01). A combination of acute and chronic placental abnormalities (43%) was more common than either acute (20%) or chronic (21%) abnormalities alone. Acute abnormalities included meconium staining of the placenta (41%) and histologic chorioamnionitis (39%). Chronic abnormalities included maternal vascular malperfusion (25%), villitis of unknown etiology (8%), and fetal vascular malperfusion (6%). Infants with chronic placental abnormalities exhibited a greater mean base deficit at birth (-15.9 vs -14.3, P = .049) than those without such abnormalities. Patients with HIE and acute placental lesions had older mean gestational ages (39.1 vs 38.0, P < .001) and greater rates of clinically diagnosed chorioamnionitis (25% vs 2%, P < .001) than those without acute abnormalities. CONCLUSIONS Combined acute and chronic placental abnormalities were common in this cohort of infants with HIE, underscoring the complex causal pathways of HIE. TRIAL REGISTRATION ClinicalTrials.gov: NCT02811263.
Collapse
Affiliation(s)
- Lina Chalak
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX.
| | - Raymond W Redline
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Amy M Goodman
- Department of Neurology, University of California San Francisco, San Francisco, CA
| | - Sandra E Juul
- Division of Neonatology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Taeun Chang
- Department of Neurology, Children's National Hospital, George Washington School of Medicine, Washington, DC
| | - Toby D Yanowitz
- Division of Newborn Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC and Magee Womens Hospital of UPMC, Pittsburgh, PA
| | - Nathalie Maitre
- Department of Pediatrics and Research Institute, Nationwide Children's Hospital, Columbus, OH
| | - Dennis E Mayock
- Division of Neonatology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | | | - Ellen Bendel-Stenzel
- Division of Neonatal Medicine, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN
| | - David Riley
- Department of Pediatrics, Cook Children's Medical Center, Texas Christian University and University of North Texas Health Science Center School of Medicine, Fort Worth, TX
| | - Amit M Mathur
- Department of Pediatrics/Neonatal-Perinatal Medicine, Saint Louis University School of Medicine, St Louis, MO
| | - Rakesh Rao
- Division of Newborn-Medicine, Department of Pediatrics, Washington University School of Medicine, St Louis, MO
| | - Krisa P Van Meurs
- Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - Tai-Wei Wu
- Division of Neonatology, Fetal and Neonatal Institute, Children's Hospital Los Angeles, Los Angeles, CA; Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Fernando F Gonzalez
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - John Flibotte
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Ulrike Mietzsch
- Division of Neonatology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Gregory M Sokol
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | | | - Mariana Baserga
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT
| | | | - Brenda B Poindexter
- Division of Neonatology, Department of Pediatrics, Children's Healthcare of Atlanta and Emory University, Atlanta, GA
| | - Bryan A Comstock
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Yvonne W Wu
- Department of Neurology, University of California San Francisco, San Francisco, CA
| |
Collapse
|
15
|
Penn AA, Wintermark P, Chalak LF, Armstrong J, Redline R, Scher MS, Nelson KB. Placental contribution to neonatal encephalopathy. Semin Fetal Neonatal Med 2021; 26:101276. [PMID: 34420894 DOI: 10.1016/j.siny.2021.101276] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Placental assessment, although currently underused, can inform our understanding of the etiology and timing of Neonatal Encephalopathy (NE). We review our current understanding of the links between placental dysfunction and NE and how this information may inform clinical decisions, now and in the future, emphasizing the four major placental lesions associated with NE. In addition, we discuss maternal and fetal factors that are hypothesized to contribute to specific placental pathologies, especially innate or acquired thrombophilias. We outline the importance of assessing placenta across trimesters and after delivery. As this field continues to evolve, currently available placental histopathological examination methods may need to be combined with advanced prenatal molecular and imaging assessments of placenta and be applied in well-designed studies in large representative populations to better define the links between placental dysfunction and NE.
Collapse
Affiliation(s)
- A A Penn
- Division of Neonatology, Department of Pediatrics, Columbia University, New York, NY, USA.
| | - P Wintermark
- Division of Newborn Medicine, Montreal Children's Hospital, Montreal, Canada
| | - L F Chalak
- Neonatal-Perinatal Medicine, Department of Pediatrics, UT Southwestern Medical Center, Dallas, USA
| | - J Armstrong
- Department of Pediatrics (Section of Child Neurology, Neurology, and OB/GYN), University of Colorado Anschutz Medical Campus, Hemophilia and Thrombosis Center, Aurora, CO, USA
| | - R Redline
- Department of Pathology, UH Cleveland Medical Center, Cleveland, OH, USA
| | - M S Scher
- Case Western Reserve University School of Medicine, Department of Pediatrics, Department of Neurology, Rainbow Babies and Children's Hospital/MacDonald Hospital for Women, UH Cleveland Medical Center, Cleveland, OH, USA
| | - K B Nelson
- National Institutes of Health, National Institute of Neurological Diseases and Stroke, Bethesda, MD, USA
| | | |
Collapse
|
16
|
Danladi J, Sabir H. Perinatal Infection: A Major Contributor to Efficacy of Cooling in Newborns Following Birth Asphyxia. Int J Mol Sci 2021; 22:ijms22020707. [PMID: 33445791 PMCID: PMC7828225 DOI: 10.3390/ijms22020707] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/05/2021] [Accepted: 01/09/2021] [Indexed: 12/19/2022] Open
Abstract
Neonatal encephalopathy (NE) is a global burden, as more than 90% of NE occurs in low- and middle-income countries (LMICs). Perinatal infection seems to limit the neuroprotective efficacy of therapeutic hypothermia. Efforts made to use therapeutic hypothermia in LMICs treating NE has led to increased neonatal mortality rates. The heat shock and cold shock protein responses are essential for survival against a wide range of stressors during which organisms raise their core body temperature and temporarily subject themselves to thermal and cold stress in the face of infection. The characteristic increase and decrease in core body temperature activates and utilizes elements of the heat shock and cold shock response pathways to modify cytokine and chemokine gene expression, cellular signaling, and immune cell mobilization to sites of inflammation, infection, and injury. Hypothermia stimulates microglia to secret cold-inducible RNA-binding protein (CIRP), which triggers NF-κB, controlling multiple inflammatory pathways, including nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasomes and cyclooxygenase-2 (COX-2) signaling. Brain responses through changes in heat shock protein and cold shock protein transcription and gene-expression following fever range and hyperthermia may be new promising potential therapeutic targets.
Collapse
Affiliation(s)
- Jibrin Danladi
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital University of Bonn, 53127 Bonn, Germany;
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Correspondence:
| | - Hemmen Sabir
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital University of Bonn, 53127 Bonn, Germany;
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| |
Collapse
|
17
|
Zarate MA, Wesolowski SR, Nguyen LM, De Dios RK, Wilkening RB, Rozance PJ, Wright CJ. In utero inflammatory challenge induces an early activation of the hepatic innate immune response in late gestation fetal sheep. Innate Immun 2020; 26:549-564. [PMID: 32538259 PMCID: PMC7556190 DOI: 10.1177/1753425920928388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 12/15/2022] Open
Abstract
Chorioamnionitis is associated with inflammatory end-organ damage in the fetus. Tissues in direct contact with amniotic fluid drive a pro-inflammatory response and contribute to this injury. However, due to a lack of direct contact with the amniotic fluid, the liver contribution to this response has not been fully characterized. Given its role as an immunologic organ, we hypothesized that the fetal liver would demonstrate an early innate immune response to an in utero inflammatory challenge. Fetal sheep (131 ± 1 d gestation) demonstrated metabolic acidosis and high cortisol and norepinephrine values within 5 h of exposure to intra-amniotic LPS. Likewise, expression of pro-inflammatory cytokines increased significantly at 1 and 5 h of exposure. This was associated with NF-κB activation, by inhibitory protein IκBα degradation, and nuclear translocation of NF-κB subunits (p65/p50). Corroborating these findings, LPS exposure significantly increased pro-inflammatory innate immune gene expression in fetal sheep hepatic macrophages in vitro. Thus, an in utero inflammatory challenge induces an early hepatic innate immune response with systemic metabolic and stress responses. Within the fetal liver, hepatic macrophages respond robustly to LPS exposure. Our results demonstrate that the fetal hepatic innate immune response must be considered when developing therapeutic approaches to attenuate end-organ injury associated with in utero inflammation.
Collapse
Affiliation(s)
- Miguel A Zarate
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Stephanie R Wesolowski
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Leanna M Nguyen
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Robyn K De Dios
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Randall B Wilkening
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Paul J Rozance
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
18
|
Pazandak C, Mir IN, Brown LS, Chalak LF. Placental Pathology, Cerebral Blood Flow, and Intraventricular Hemorrhage in Preterm Infants: Is There a Link? Pediatr Neurol 2020; 108:65-69. [PMID: 32451157 DOI: 10.1016/j.pediatrneurol.2020.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 01/02/2020] [Accepted: 01/05/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND There is growing evidence to support an association between placental inflammation and neurological sequelae of preterm infants. The goal of this study is to evaluate the relationship between placental pathology, post-natal Doppler cerebral resistive indices (RI's), and intraventricular hemorrhage (IVH) in premature infants. METHODS In a retrospective cohort study, preterm infants born between 23 0/7 and 32 6/7 weeks' gestation at Parkland Hospital were examined with placental pathology and serial ultrasound Doppler to evaluate for the primary outcome of IVH and death. RESULTS A total of 255 infants were included, and 166 (65%) had at least one significant placental pathology, most commonly chorioamnionitis. Infants with placental pathologies were significantly more likely to have mothers with clinical chorioamnionitis and to have lower gestational ages. There was no observed association between placental pathology and IVH or death. Secondary analysis demonstrated that resistive indices obtained from the first and second head ultrasounds were not different in infants with IVH. CONCLUSION In this study, we observed a high rate of placental pathologies but no alterations in cerebral indices on ultrasound, or differences in rates of IVH or death. Additional studies are necessary to delineate the relationship between placental pathology, white matter brain injury, and outcomes.
Collapse
Affiliation(s)
- Christine Pazandak
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, UT Southwestern Medical School, Dallas, Texas
| | - Imran N Mir
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, UT Southwestern Medical School, Dallas, Texas
| | - L Steven Brown
- Parkland Health and Hospital Systems, Department of Pediatrics, UT Southwestern Medical School, Dallas, Texas
| | - Lina F Chalak
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, UT Southwestern Medical School, Dallas, Texas.
| |
Collapse
|
19
|
Benmakhlouf Y, Zian Z, Nourouti NG, Barakat A, Mechita MB. Potential Cytokine Biomarkers in Intellectual Disability. Endocr Metab Immune Disord Drug Targets 2020; 21:569-576. [PMID: 32600239 DOI: 10.2174/1871530320666200628024944] [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: 01/31/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 11/22/2022]
Abstract
Intellectual disability (ID), previously called mental retardation, is the most common neurodevelopmental disorder characterized by life-long intellectual and adaptive functioning impairments that have an impact on individuals, families, and society. Its prevalence is estimated to 3% of the general population and its etiology is still insufficiently understood. Besides the involvement of genetic and environmental factors, immunological dysfunctions have been also suggested to contribute to the pathophysiology of ID. Over the years, immune biomarkers related to ID have gained significant attention and researchers have begun to look at possible cytokine profiles in individuals suffered from this disorder. In fact, in addition to playing crucial physiological roles in the majority of normal neurodevelopmental processes, cytokines exert an important role in neuroinflammation under pathological conditions, and interactions between the immune system and central nervous system have long been under investigation. Cytokine levels imbalance has been reported associated with some behavioral characteristics and the onset of some syndromic forms of ID. In this review, we will focus on immunological biomarkers, especially the cytokine profiles that have been identified in people with ID. Thus, data reported and discussed in the present paper may provide additional information to start further studies and to plan strategies for early identification and managing of ID.
Collapse
Affiliation(s)
- Yousra Benmakhlouf
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Zeineb Zian
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Naima G Nourouti
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Amina Barakat
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Mohcine B Mechita
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| |
Collapse
|
20
|
Chalak L, Ferriero DM, Gressens P, Molloy E, Bearer C. A 20 years conundrum of neonatal encephalopathy and hypoxic ischemic encephalopathy: are we closer to a consensus guideline? Pediatr Res 2019; 86:548-549. [PMID: 31450231 DOI: 10.1038/s41390-019-0547-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/26/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Lina Chalak
- UT Southwestern Medical Center of Dallas, Dallas, TX, USA.
| | | | | | | | - Cynthia Bearer
- Department of Pediatrics, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
21
|
Sutton JA, Rogers LM, Dixon B, Kirk L, Doster R, Algood HM, Gaddy JA, Flaherty R, Manning SD, Aronoff DM. Protein kinase D mediates inflammatory responses of human placental macrophages to Group B Streptococcus. Am J Reprod Immunol 2019; 81:e13075. [PMID: 30582878 PMCID: PMC6459189 DOI: 10.1111/aji.13075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/11/2018] [Accepted: 12/18/2018] [Indexed: 01/22/2023] Open
Abstract
PROBLEM During pregnancy, Group B Streptococcus (GBS) can infect fetal membranes to cause chorioamnionitis, resulting in adverse pregnancy outcomes. Macrophages are the primary resident phagocyte in extraplacental membranes. Protein kinase D (PKD) was recently implicated in mediating pro-inflammatory macrophage responses to GBS outside of the reproductive system. This work aimed to characterize the human placental macrophage inflammatory response to GBS and address the extent to which PKD mediates such effects. METHOD Primary human placental macrophages were infected with GBS in the presence or absence of a specific, small molecule PKD inhibitor, CRT 0066101. Macrophage phenotypes were characterized by evaluating gene expression, cytokine release, assembly of the NLRP3 inflammasome, and NFκB activation. RESULTS GBS evoked a strong inflammatory phenotype characterized by the release of inflammatory cytokines (TNFα, IL-1β, IL-6 (P ≤ 0.05), NLRP3 inflammasome assembly (P ≤ 0.0005), and NFκB activation (P ≤ 0.05). Pharmacological inhibition of PKD suppressed these responses, newly implicating a role for PKD in mediating immune responses of primary human placental macrophages to GBS. CONCLUSION PKD plays a critical role in mediating placental macrophage inflammatory activation in response to GBS infection.
Collapse
Affiliation(s)
- Jessica A. Sutton
- Department of Microbiology and Immunology, Meharry Medical College School of Medicine, Nashville, TN, 37208, USA
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lisa M. Rogers
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Beverly Dixon
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Leslie Kirk
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ryan Doster
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Holly M. Algood
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee, U.S.A
| | - Jennifer A. Gaddy
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee, U.S.A
| | - Rebecca Flaherty
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Shannon D. Manning
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - David M. Aronoff
- Department of Microbiology and Immunology, Meharry Medical College School of Medicine, Nashville, TN, 37208, USA
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
22
|
Scher MS. Fetal neurology: Principles and practice with a life-course perspective. HANDBOOK OF CLINICAL NEUROLOGY 2019; 162:1-29. [PMID: 31324306 DOI: 10.1016/b978-0-444-64029-1.00001-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Clinical service, educational, and research components of a fetal/neonatal neurology program are anchored by the disciplines of developmental origins of health and disease and life-course science as programmatic principles. Prenatal participation provides perspectives on maternal, fetal, and placental contributions to health or disease for fetal and subsequent neonatal neurology consultations. This program also provides an early-life diagnostic perspective for neurologic specialties concerned with brain health and disease throughout childhood and adulthood. Animal models and birth cohort studies have demonstrated how the science of epigenetics helps to understand gene-environment interactions to better predict brain health or disease. Fetal neurology consultations provide important diagnostic contributions during critical or sensitive periods of brain development when future neurotherapeutic interventions will maximize adaptive neuroplasticity. Age-specific normative neuroinformatics databases that employ computer-based strategies to integrate clinical/demographic, neuroimaging, neurophysiologic, and genetic datasets will more accurately identify either symptomatic patients or those at risk for brain disorders who would benefit from preventive, rescue, or reparative treatment choices throughout the life span.
Collapse
Affiliation(s)
- Mark S Scher
- Division of Pediatric Neurology, Case Western Reserve University, Cleveland, OH, United States.
| |
Collapse
|
23
|
Aslam S, Strickland T, Molloy EJ. Neonatal Encephalopathy: Need for Recognition of Multiple Etiologies for Optimal Management. Front Pediatr 2019; 7:142. [PMID: 31058120 PMCID: PMC6477286 DOI: 10.3389/fped.2019.00142] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/26/2019] [Indexed: 12/18/2022] Open
Abstract
Neonatal encephalopathy (NE) is associated with high mortality and morbidity. Factors predisposing to NE can be antenatal, perinatal, or a combination of both. Antenatal maternal factors, familial factors, genetic predisposition, hypoxic ischemic encephalopathy, infections, placental abnormalities, thrombophilia, coagulation defects, and metabolic disorders all have been implicated in the pathogenesis of NE. At present, therapeutic hypothermia is the only treatment available, regardless of etiology. Recognizing the etiology of NE involved can also guide investigations such as metabolic and sepsis workups to ensure optimal management. Understanding the etiology of NE may allow the development of targeted adjunctive therapies related to the underlying mechanism and develop preventative strategies.
Collapse
Affiliation(s)
- Saima Aslam
- Paediatrics, National Maternity Hospital, Dublin, Ireland.,UCD School of Medicine & Medical Sciences, University College Dublin, Dublin, Ireland
| | - Tammy Strickland
- Paediatrics, National Maternity Hospital, Dublin, Ireland.,Trinity College Translational Medicine Institute, Academic Paediatrics, Trinity College Dublin, National Children's Hospital, Dublin, Ireland.,Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Eleanor J Molloy
- Paediatrics, National Maternity Hospital, Dublin, Ireland.,UCD School of Medicine & Medical Sciences, University College Dublin, Dublin, Ireland.,Trinity College Translational Medicine Institute, Academic Paediatrics, Trinity College Dublin, National Children's Hospital, Dublin, Ireland.,Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland.,Neonatology, Our Lady's Children's Hospital, Drimnagh, Ireland
| |
Collapse
|
24
|
Murray DM. Biomarkers in neonatal hypoxic-ischemic encephalopathy-Review of the literature to date and future directions for research. HANDBOOK OF CLINICAL NEUROLOGY 2019; 162:281-293. [PMID: 31324315 DOI: 10.1016/b978-0-444-64029-1.00013-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The widespread introduction of therapeutic hypothermia as a standard of care in hypoxic-ischemic encephalopathy (HIE) has brought increasing pressure on clinicians to make an early and accurate assessment of the degree of hypoxic injury (HI) that has occurred and the severity of the encephalopathy that will ensue. No single blood-based marker is currently robust enough to detect significant HI or predict outcome. However, research in the field has been active in the last 10 years and we know that HIE is associated with predictable alterations in the expression of a number of inflammatory proteins, neuron-specific proteins, metabolite pathways, and microRNA. These alterations evolve quickly over the first hours and days of life. Predictive power varies depending on the timing of measurement of the biomarker, the sample type, and the case mix of the cohort examined. Combining clinical data with biochemical measurements is currently the most likely path toward improved detection and prediction of outcome in neonatal HIE.
Collapse
Affiliation(s)
- Deirdre M Murray
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.
| |
Collapse
|
25
|
δ-Opioid Receptor-Nrf-2-Mediated Inhibition of Inflammatory Cytokines in Neonatal Hypoxic-Ischemic Encephalopathy. Mol Neurobiol 2018; 56:5229-5240. [PMID: 30560518 DOI: 10.1007/s12035-018-1452-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/07/2018] [Indexed: 12/11/2022]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) causes serious neurological disability; there are, however, currently few promising therapies for it. We have recently shown that δ-opioid receptor (DOR) is neuroprotective by downregulating TNF-α. Since hypoxia-ischemia (HI) triggers a robust inflammatory response, which exacerbates HI brain damage, we investigated, in this study, whether DOR activation could regulate inflammatory cytokine expression, thereby playing a protective effect on the neonatal brain under HI. Twenty-five neonatal rats were randomly divided into five groups: (1) control (control); (2) HI; (3) HI with saline (HI + NS); (4) DOR activation with UFP-512 (a potent and specific DOR agonist) under HI conditions (HI + U); and (5) DOR inhibition using NT treatment under HI conditions (HI + NT). The rats were sacrificed by decapitation at 24 h after HI, and their brains were rapidly removed for measurements. The protein expression of TNF-α, IL-6, ICAM-1, IL-10, IL-18, NQO-1, Nrf-2, and HO-1 was measured using Western blot. In the hemispheres exposed to HI, DOR activation significantly decreased the expressions of TNF-α, IL-6, and ICAM-1 in the cortex, while it significantly increased IL-10 and had no effect on IL-18 in the same region. In contrast, DOR had no appreciable effect on inflammatory cytokine expression in non-cortical tissues including hippocampal, subcortical, and cerebellar tissues. Moreover, HI stress triggered an upregulation of Nrf-2 nuclear protein as well as some of its downstream anti-inflammatory genes such as HO-1 and NQO-1 in the cortex, while DOR activation further augmented such a protective reaction against HI injury. DOR plays an important role in protecting against HI by regulating the expression of inflammatory and anti-inflammatory cytokines in the cortex, which is likely mediated by the Nrf-2/HO-1/NQO-1 signaling.
Collapse
|
26
|
Mir IN, Chalak LF, Liao J, Johnson-Welch S, Brown LS, Longoria C, Savani RC, Rosenfeld CR. Fetal-placental crosstalk occurs through fetal cytokine synthesis and placental clearance. Placenta 2018; 69:1-8. [PMID: 30213477 DOI: 10.1016/j.placenta.2018.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/13/2018] [Accepted: 07/09/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cytokines modulate fetal well-being and contribute to parturition. Their origin in fetal blood, whether maternal, placental or fetal, at the time of parturition remains unclear. OBJECTIVE To determine fetal and placental contributions to circulating fetal cytokines by measuring umbilical arterial (UmA) and venous (UmV) concentration differences in uncomplicated term pregnancies in the absence and presence of labor. METHODS Term uncomplicated pregnancies were assessed: Group 1 were not in labor and delivered by elective cesarean section (n = 20); Group 2 delivered vaginally following uncomplicated pregnancy and labor (n = 30). UmA and UmV blood was collected before delivery of the placenta to measure circulating cytokines. Placental tissue was collected for histology and to determine cytokine contents and localization. RESULTS Group 1 UmA and UmV IL-10 concentrations were similar (504 ± 15 and 468 ± 16 pg/ml, respectively; P ≥ 0.1); other cytokines were below level of detection. During labor, IL-10 concentrations increased 15-34%, but placental contents decreased. Group 2 UmA IL-6 and IL-8 concentrations increased (P < 0.001) to 16.7 ± 1.6 and 18.4 ± 4.3 pg/ml, respectively, but were less (P < 0.001) in UmV, 0.29 ± 0.2 and 0.74 ± 0.3 pg/ml, respectively, demonstrating placental clearances ≥97%. This was associated with >6-fold increases in placental IL-6/IL-8 contents (P < 0.001) and chorioamniotic infiltration of activated maternal neutrophils. IL-6 and IL-10 were localized to villous syncytiotrophoblasts. CONCLUSIONS In uncomplicated term pregnancies fetal circulating IL-10 is likely of placental origin, whereas IL-6/IL-8 are derived from the fetus, increase during parturition, and circulating levels are modulated by non-saturable placental clearance, revealing a novel pathway for fetal-placental crosstalk and signaling.
Collapse
Affiliation(s)
- Imran N Mir
- Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center, USA.
| | - Lina F Chalak
- Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center, USA
| | - Jie Liao
- Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center, USA
| | - Sarah Johnson-Welch
- Department of Pediatrics, Department of Pathology, University of Texas Southwestern Medical Center, USA
| | | | - Christopher Longoria
- Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center, USA
| | - Rashmin C Savani
- Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center, USA; The Center for Pulmonary & Vascular Biology, University of Texas Southwestern Medical Center, USA
| | - Charles R Rosenfeld
- Division of Neonatal-Perinatal Medicine, University of Texas Southwestern Medical Center, USA
| |
Collapse
|
27
|
Boskabadi H, Moradi A, Zakerihamidi M. Interleukins in diagnosis of perinatal asphyxia: A systematic review. Int J Reprod Biomed 2018; 17. [PMID: 31435616 PMCID: PMC6653496 DOI: 10.18502/ijrm.v17i5.4598] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/25/2018] [Accepted: 09/12/2018] [Indexed: 11/24/2022] Open
Abstract
Background Biochemical markers including interleukins (ILs) has been proposed for early diagnosis of asphyxia. Objective This study has aimed to systematically review the significance of IL measurements in the diagnosis of perinatal asphyxia. Materials and Methods PubMed, Cochrane Library, Web of Science, Embase, and Scopus databases before 2017 were searched for the following keywords: asphyxia, neonatal, interleukin, and diagnosis. A total of 13 out of 300 searched papers were finally selected for evaluation. Interleukins under study were IL6 and interleukin 1 β (IL-1 β ). Interleukins had been measured in 10 studies by serum samples, 2 studies by samples of Cerebro Spinal Fluid (CSF), and 1 study by sample of umbilical cord blood. The inclusion criteria were: studies on neonates, with adequate information from the test results and studies using markers other than ILs to detect asphyxia; however, studies with only abstracts available were excluded. Results Research on the issue suggests that IL6 > 41 Pg/dl has the sensitivity of 84.88% and the specificity of 85.43%, whereas IL-1 β > 4.7 Pg/dl has the sensitivity of 78% and specificity of 83% in the diagnosis of neonatal asphyxia. Among diagnostic ILs for neonatal asphyxia, combination of IL6 and IL-1 β had the highest sensitivity, that is, 92.9%. Conclusion IL6 and IL-1 β of serum samples were used in the early diagnosis of perinatal asphyxia and are useful predictors for the outcomes of perinatal asphyxia and its intensity. In addition, simultaneous evaluation of IL-1 β and IL6 can improve the sensitivity of the early diagnosis of perinatal asphyxia.
Collapse
Affiliation(s)
- Hassan Boskabadi
- Department of Pediatrics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Moradi
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Zakerihamidi
- Department of Midwifery, Faculty of Medical Sciences, Islamic Azad University, Tonekabon Branch, Tonekabon, Iran
| |
Collapse
|
28
|
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
|
29
|
Abstract
Abstract
Background: Perinatal asphyxia is a major cause of neurologic morbidity and mortality in infants. Objective: Determine the serum level of interleukin-6 (IL-6) in neonates with perinatal asphyxia and its relation to the severity of hypoxic-ischemic encephalopathy and short term neurological outcome. Methods: Serum IL-6 levels were measured at birth, and at 24 and 48 hour post-partum in 37 consecutive uninfected neonates with peri-natal asphyxia and 45 randomly selected healthy newborns. Results: Serum IL-6 concentrations in the infants who developed hypoxic-ischemic encephalopathy was 43 folds higher compared to values in the normal infants (p < 0.001) and 1.9 folds higher as compared to infants with asphyxia who did not subsequently develop hypoxic-ischemic encephalopathy (p <0.001). Serum IL-6 concentrations were also related to the degree of hypoxic-ischemic encephalopathy and neurologicaldevelopmental outcomes at the time of discharge. Conclusion: Serum levels of IL-6 increased in neonates with asphyxia, and this was most pronounced in neonates with adverse outcomes.
Collapse
|
30
|
Stojanovska V, Miller SL, Hooper SB, Polglase GR. The Consequences of Preterm Birth and Chorioamnionitis on Brainstem Respiratory Centers: Implications for Neurochemical Development and Altered Functions by Inflammation and Prostaglandins. Front Cell Neurosci 2018; 12:26. [PMID: 29449803 PMCID: PMC5799271 DOI: 10.3389/fncel.2018.00026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/17/2018] [Indexed: 11/16/2022] Open
Abstract
Preterm birth is a major cause for neonatal morbidity and mortality, and is frequently associated with adverse neurological outcomes. The transition from intrauterine to extrauterine life at birth is particularly challenging for preterm infants. The main physiological driver for extrauterine transition is the establishment of spontaneous breathing. However, preterm infants have difficulty clearing lung liquid, have insufficient surfactant levels, and underdeveloped lungs. Further, preterm infants have an underdeveloped brainstem, resulting in reduced respiratory drive. These factors facilitate the increased requirement for respiratory support. A principal cause of preterm birth is intrauterine infection/inflammation (chorioamnionitis), and infants with chorioamnionitis have an increased risk and severity of neurological damage, but also demonstrate impaired autoresuscitation capacity and prevalent apnoeic episodes. The brainstem contains vital respiratory centers which provide the neural drive for breathing, but the impact of preterm birth and/or chorioamnionitis on this brain region is not well understood. The aim of this review is to provide an overview of the role and function of the brainstem respiratory centers, and to highlight the proposed mechanisms of how preterm birth and chorioamnionitis may affect central respiratory functions.
Collapse
Affiliation(s)
- Vanesa Stojanovska
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University and Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University and Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University and Hudson Institute of Medical Research, Melbourne, VIC, Australia
| |
Collapse
|
31
|
Li B, Concepcion K, Meng X, Zhang L. Brain-immune interactions in perinatal hypoxic-ischemic brain injury. Prog Neurobiol 2017; 159:50-68. [PMID: 29111451 PMCID: PMC5831511 DOI: 10.1016/j.pneurobio.2017.10.006] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/26/2017] [Indexed: 01/07/2023]
Abstract
Perinatal hypoxia-ischemia remains the primary cause of acute neonatal brain injury, leading to a high mortality rate and long-term neurological deficits, such as behavioral, social, attentional, cognitive and functional motor deficits. An ever-increasing body of evidence shows that the immune response to acute cerebral hypoxia-ischemia is a major contributor to the pathophysiology of neonatal brain injury. Hypoxia-ischemia provokes an intravascular inflammatory cascade that is further augmented by the activation of resident immune cells and the cerebral infiltration of peripheral immune cells response to cellular damages in the brain parenchyma. This prolonged and/or inappropriate neuroinflammation leads to secondary brain tissue injury. Yet, the long-term effects of immune activation, especially the adaptive immune response, on the hypoxic-ischemic brain still remain unclear. The focus of this review is to summarize recent advances in the understanding of post-hypoxic-ischemic neuroinflammation triggered by the innate and adaptive immune responses and to discuss how these mechanisms modulate the brain vulnerability to injury. A greater understanding of the reciprocal interactions between the hypoxic-ischemic brain and the immune system will open new avenues for potential immunomodulatory therapy in the treatment of neonatal brain injury.
Collapse
Affiliation(s)
- Bo Li
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Katherine Concepcion
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Xianmei Meng
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
32
|
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
|
33
|
Ziemka-Nalecz M, Jaworska J, Zalewska T. Insights Into the Neuroinflammatory Responses After Neonatal Hypoxia-Ischemia. J Neuropathol Exp Neurol 2017; 76:644-654. [DOI: 10.1093/jnen/nlx046] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
34
|
Kumar N, Nandula P, Menden H, Jarzembowski J, Sampath V. Placental TLR/NLR expression signatures are altered with gestational age and inflammation. J Matern Fetal Neonatal Med 2017; 30:1588-1595. [PMID: 27440318 DOI: 10.1080/14767058.2016.1214705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To quantify changes in placental expression of Toll-like receptors (TLRs) and nuclear oligomerization domain (NOD)-like receptors (NLRs) gene with (1) advancing gestational age (GA) and (2) exposure to chorioamnionitis (CA) and preterm premature rupture of membrane (PPROM). METHODS Placental tissue was collected at the time of birth from 83 subjects with live birth pregnancies from 24- to 40-week gestation between 2009 and 2013. Real-time RT-PCR analysis of 13 TLR/NLR genes involved in bacterial sensing was performed using specific probes. RESULTS Of 83 patients enrolled, 61 were preterm (<37 weeks). 23 (27%) had evidence of CA; and 33 (39.8%) had PPROM. 15 (18%) had both CA and PPROM (CP). 42 (50%) had neither CA nor PPROM (C/P). Only RIPK2 (p = 0.0025) and TLR4 (p = 0.0005) were found to increase progressively with GA. We found significant changes in TLR5 (p = 0.01) with CA, NFKBIA (p = 0.016) with PPROM, NKKBIA (p = 0.003), and NFKB1 (p = 0.009) with CA and PPROM. CONCLUSION RIPK2 (mediator of NOD-dependent NF-kB signaling) and TLR4 progressively increased with GA. We speculate this upregulation may be involved in initiating labor and delivery at term. Increase in NFKBIA seen in PPROM and CA might represent a counter regulatory mechanism to decrease inflammation in these conditions. This study provides new information on relationships between GA, CA/PPROM, and TLR/NLR signaling in the placenta.
Collapse
Affiliation(s)
- Navin Kumar
- a Division of Neonatology, Hurley Medical Center , Flint, MI , USA
| | - Padma Nandula
- b Division of Neonatology, Clinical Center, University of Florida Health at Jacksonville , Jacksonville, FL , USA
| | - Heather Menden
- c Division of Neonatology, Children's Mercy Hospital , Kansas City, MO , USA
| | - Jason Jarzembowski
- d Department of Pathology, Medical College of Wisconsin and Children's Hospital of Wisconsin , Milwaukee, WI , USA
| | - Venkatesh Sampath
- c Division of Neonatology, Children's Mercy Hospital , Kansas City, MO , USA
| |
Collapse
|
35
|
Huang L, Zhao F, Qu Y, Zhang L, Wang Y, Mu D. Animal models of hypoxic-ischemic encephalopathy: optimal choices for the best outcomes. Rev Neurosci 2017; 28:31-43. [PMID: 27559689 DOI: 10.1515/revneuro-2016-0022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/15/2016] [Indexed: 12/21/2022]
Abstract
AbstractHypoxic-ischemic encephalopathy (HIE), a serious disease leading to neonatal death, is becoming a key area of pediatric neurological research. Despite remarkable advances in the understanding of HIE, the explicit pathogenesis of HIE is unclear, and well-established treatments are absent. Animal models are usually considered as the first step in the exploration of the underlying disease and in evaluating promising therapeutic interventions. Various animal models of HIE have been developed with distinct characteristics, and it is important to choose an appropriate animal model according to the experimental objectives. Generally, small animal models may be more suitable for exploring the mechanisms of HIE, whereas large animal models are better for translational studies. This review focuses on the features of commonly used HIE animal models with respect to their modeling strategies, merits, and shortcomings, and associated neuropathological changes, providing a comprehensive reference for improving existing animal models and developing new animal models.
Collapse
Affiliation(s)
- Lan Huang
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Fengyan Zhao
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Yi Qu
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Li Zhang
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Yan Wang
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Dezhi Mu
- 1Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 2Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, Sichuan University, Chengdu 610041, China
- 3Department of Pediatrics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
36
|
Harding B, Conception K, Li Y, Zhang L. Glucocorticoids Protect Neonatal Rat Brain in Model of Hypoxic-Ischemic Encephalopathy (HIE). Int J Mol Sci 2016; 18:ijms18010017. [PMID: 28025500 PMCID: PMC5297652 DOI: 10.3390/ijms18010017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/10/2016] [Accepted: 12/19/2016] [Indexed: 11/27/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) resulting from asphyxia in the peripartum period is the most common cause of neonatal brain damage and can result in significant neurologic sequelae, including cerebral palsy. Currently therapeutic hypothermia is the only accepted treatment in addition to supportive care for infants with HIE, however, many additional neuroprotective therapies have been investigated. Of these, glucocorticoids have previously been shown to have neuroprotective effects. HIE is also frequently compounded by infectious inflammatory processes (sepsis) and as such, the infants may be more amenable to treatment with an anti-inflammatory agent. Thus, the present study investigated dexamethasone and hydrocortisone treatment given after hypoxic-ischemic (HI) insult in neonatal rats via intracerebroventricular (ICV) injection and intranasal administration. In addition, we examined the effects of hydrocortisone treatment in HIE after lipopolysaccharide (LPS) sensitization in a model of HIE and sepsis. We found that dexamethasone significantly reduced rat brain infarction size when given after HI treatment via ICV injection; however it did not demonstrate any neuroprotective effects when given intranasally. Hydrocortisone after HI insult also significantly reduced brain infarction size when given via ICV injection; and the intranasal administration showed to be protective of brain injury in male rats at a dose of 300 µg. LPS sensitization did significantly increase the brain infarction size compared to controls, and hydrocortisone treatment after LPS sensitization showed a significant decrease in brain infarction size when given via ICV injection, as well as intranasal administration in both genders at a dose of 300 µg. To conclude, these results show that glucocorticoids have significant neuroprotective effects when given after HI injury and that these effects may be even more pronounced when given in circumstances of additional inflammatory injury, such as neonatal sepsis.
Collapse
Affiliation(s)
- Benjamin Harding
- Division of Neonatology, Department of Pediatrics, Loma Linda University Children's Hospital, Loma Linda, CA 92354, USA.
| | - Katherine Conception
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Yong Li
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Lubo Zhang
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
37
|
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
|
38
|
Orrock JE, Panchapakesan K, Vezina G, Chang T, Harris K, Wang Y, Knoblach S, Massaro AN. Association of brain injury and neonatal cytokine response during therapeutic hypothermia in newborns with hypoxic-ischemic encephalopathy. Pediatr Res 2016; 79:742-7. [PMID: 26717001 PMCID: PMC4853239 DOI: 10.1038/pr.2015.280] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cytokines have been proposed as mediators of neonatal brain injury via neuroinflammatory pathways triggered by hypoxia-ischemia. Limited data are available on cytokine profiles in larger cohorts of newborns with hypoxic-ischemic encephalopathy (HIE) undergoing therapeutic hypothermia (TH). METHODS Serum cytokines interleukin (IL)-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-13, tumor necrosis factor-α, and interferon-γ were measured in newborns with HIE at 24 and 72 h of TH. Differences between infants with favorable (survivors with mild/no magnetic resonance imaging (MRI) injury) vs. adverse outcome (death or moderate/severe MRI injury) were compared using mixed models to adjust for covariates. RESULTS Data from 36 term newborns with HIE (favorable outcome: n = 20, adverse outcome: n = 16) were evaluated. Cytokines IL-1β, IL-2, IL-6, IL-8, IL-10, and IL-13 were elevated in the adverse relative to favorable outcome group at 24 h. IL-6 remained significantly elevated in the adverse outcome group at 72 h. IL-6 and IL-10 remained significantly associated with outcome group after controlling for covariates. CONCLUSION Inflammatory cytokines are elevated in HIE newborns with brain injury by MRI. In particular, IL-6 and IL-10 were associated with adverse outcomes after controlling for baseline characteristics and severity of presentation. These data suggest that cytokine response may identify infants in need of additional neuroprotective interventions.
Collapse
Affiliation(s)
- Janet E. Orrock
- Pediatric Residency Program, Children's National Health System, Washington, DC, USA
| | - Karuna Panchapakesan
- Center for Genetic Medicine Research, Children's Research Institute, Washington, DC, USA
| | - Gilbert Vezina
- Division of Diagnostic Imaging and Radiology, Children's National Health System, Washington, DC, USA,The George Washington University School of Medicine, Washington DC, USA
| | - Taeun Chang
- Division of Neurology, Children's National Health System, Washington, DC, USA,The George Washington University School of Medicine, Washington DC, USA
| | - Kari Harris
- Division of Neurology, Children's National Health System, Washington, DC, USA
| | - Yunfei Wang
- Division of Biostatistics and Study Methodology, Children's National Health System, Washington, DC, USA,The George Washington University School of Medicine, Washington DC, USA
| | - Susan Knoblach
- Center for Genetic Medicine Research, Children's Research Institute, Washington, DC, USA,The George Washington University School of Medicine, Washington DC, USA
| | - An N. Massaro
- Division of Neonatology, Children's National Health System, Washington, DC, USA,The George Washington University School of Medicine, Washington DC, USA
| |
Collapse
|
39
|
Gold JR, Chaffin K, Burgess BA, Morley PS. Factors Associated With Nonsurvival in Foals Diagnosed With Perinatal Asphyxia Syndrome. J Equine Vet Sci 2016. [DOI: 10.1016/j.jevs.2015.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
40
|
Risk of autistic disorder after exposure to general anaesthesia and surgery: a nationwide, retrospective matched cohort study. Eur J Anaesthesiol 2016; 32:303-10. [PMID: 25101714 DOI: 10.1097/eja.0000000000000130] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Deficits of learning, memory and cognition have been observed in newborn animals exposed to general anaesthetics. However, conclusions from clinical studies conducted in humans to investigate the relationship between anaesthesia and neurodevelopmental disorders have been inconsistent. Autistic disorder is typically recognised earlier than other neurobehavioural disorders. Although certain genes apparently contribute to autistic disorder susceptibility, other factors such as perinatal insults and exposure to neurotoxic agents may play a crucial role in gene-environmental interaction. OBJECTIVE This study was designed to investigate the association of exposure to general anaesthesia/surgery with autistic disorder. We hypothesised that exposure to general anaesthesia and surgery before 2 years of age is associated with an increased risk of developing autistic disorder. DESIGN A retrospective matched-cohort study. SETTING A medical university. Data from the National Health Insurance Research Database of Taiwan from 2001 to 2010 were analysed. PATIENTS The birth cohort included 114,435 children, among whom 5197 were exposed to general anaesthesia and surgery before the age of 2 years. The 1 : 4 matched controls comprised 20,788 children. MAIN OUTCOME MEASURES The primary endpoint was the diagnosis of autistic disorder after the first exposure to general anaesthesia and surgery. RESULTS No differences were found in the incidence of autistic disorder between the exposed group (0.96%) and the unexposed controls (0.89%) (P = 0.62). Cox proportional regression showed that the hazard ratio of exposure to general anaesthesia and surgery was 0.93 [95% confidence interval (95% CI) 0.57 to 1.53] after adjusting for potential confounders. Age at first exposure did not influence the risk of autistic disorder. No relationship was found between the total number of exposures and the risk of autistic disorder. CONCLUSION Exposure to general anaesthesia and surgery before the age of 2 years age at first exposure and number of exposures were not associated with the development of autistic disorder.
Collapse
|
41
|
Romero R, Chaemsaithong P, Docheva N, Korzeniewski SJ, Tarca AL, Bhatti G, Xu Z, Kusanovic JP, Dong Z, Yoon BH, Hassan SS, Chaiworapongsa T, Yeo L, Kim YM, Kim YM. Clinical chorioamnionitis at term V: umbilical cord plasma cytokine profile in the context of a systemic maternal inflammatory response. J Perinat Med 2016; 44:53-76. [PMID: 26360486 PMCID: PMC5625297 DOI: 10.1515/jpm-2015-0121] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 07/02/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Microbial invasion of the fetus due to intra-amniotic infection can lead to a systemic inflammatory response characterized by elevated concentrations of cytokines in the umbilical cord plasma/serum. Clinical chorioamnionitis represents the maternal syndrome often associated with intra-amniotic infection, although other causes of this syndrome have been recently described. The objective of this study was to characterize the umbilical cord plasma cytokine profile in neonates born to mothers with clinical chorioamnionitis at term, according to the presence or absence of bacteria and/or intra-amniotic inflammation. MATERIALS AND METHODS A cross-sectional study was conducted, including patients with clinical chorioamnionitis at term (n=38; cases) and those with spontaneous term labor without clinical chorioamnionitis (n=77; controls). Women with clinical chorioamnionitis were classified according to the results of amniotic fluid culture, broad-range polymerase chain reaction coupled with electrospray ionization mass spectrometry (PCR/ESI-MS) and amniotic fluid interleukin (IL)-6 concentration into three groups: 1) no intra-amniotic inflammation; 2) intra-amniotic inflammation without detectable microorganisms; or 3) microbial-associated intra-amniotic inflammation. A fetal inflammatory response syndrome (FIRS) was defined as an umbilical cord plasma IL-6 concentration >11 pg/mL. The umbilical cord plasma concentrations of 29 cytokines were determined with sensitive and specific V-PLEX immunoassays. Nonparametric statistical methods were used for analysis, adjusting for a false discovery rate of 5%. RESULTS 1) Neonates born to mothers with clinical chorioamnionitis at term (considered in toto) had significantly higher median umbilical cord plasma concentrations of IL-6, IL-12p70, IL-16, IL-13, IL-4, IL-10 and IL-8, but significantly lower interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF)-α concentrations than neonates born to mothers with spontaneous term labor without clinical chorioamnionitis; 2) neonates born to mothers with clinical chorioamnionitis at term but without intra-amniotic inflammation had higher concentrations of IL-6, IL-12p70, IL-13, IL-4, IL-5, and IL-8, but lower IFN-γ, than neonates not exposed to clinical chorioamnionitis, suggesting that maternal fever in the absence of intra-amniotic inflammation leads to a change in the fetal cytokine network; 3) there were significant, positive correlations between maternal and umbilical cord plasma IL-6 and IL-8 concentrations (IL-6: Spearman correlation=0.53; P<0.001; IL-8: Spearman correlation=0.42; P<0.001), consistent with placental transfer of cytokines; 4) an elevated fetal plasma IL-6 (>11 pg/mL), the diagnostic criterion for FIRS, was present in 21% of cases (8/38), and all these neonates were born to mothers with proven intra-amniotic infection; and 5) FIRS was associated with a high concentration of umbilical cord plasma IL-8, IL-10 and monocyte chemoattractant protein (MCP)-1. CONCLUSIONS Neonates born to mothers with clinical chorioamnionitis at term had higher concentrations of umbilical cord plasma cytokines than those born to mothers without clinical chorioamnionitis. Even neonates exposed to clinical chorioamnionitis but not to intra-amniotic inflammation had elevated concentrations of multiple cytokines, suggesting that intrapartum fever alters the fetal immune response.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA,Department of Molecular Obstetrics and Genetics, Wayne State University, Detroit, MI, USA
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nikolina Docheva
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Steven J. Korzeniewski
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Zhonghui Xu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Juan P. Kusanovic
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Center for Research and Innovation in Maternal-Fetal Medicine (CIMAF). Department of Obstetrics and Gynecology, Sótero del Río Hospital, Santiago, Chile,Department of Obstetrics and Gynecology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Zhong Dong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bo Hyun Yoon
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Sonia S. Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Lami Yeo
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yeon Mee Kim
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA,Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan Korea
| | | |
Collapse
|
42
|
Jenkins DD, Wiest DB, Mulvihill DM, Hlavacek AM, Majstoravich SJ, Brown TR, Taylor JJ, Buckley JR, Turner RP, Rollins LG, Bentzley JP, Hope KE, Barbour AB, Lowe DW, Martin RH, Chang EY. Fetal and Neonatal Effects of N-Acetylcysteine When Used for Neuroprotection in Maternal Chorioamnionitis. J Pediatr 2016; 168:67-76.e6. [PMID: 26545726 PMCID: PMC4698030 DOI: 10.1016/j.jpeds.2015.09.076] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 08/25/2015] [Accepted: 09/29/2015] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To evaluate the clinical safety of antenatal and postnatal N-acetylcysteine (NAC) as a neuroprotective agent in maternal chorioamnionitis in a randomized, controlled, double-blinded trial. STUDY DESIGN Twenty-two mothers >24 weeks gestation presenting within 4 hours of diagnosis of clinical chorioamnionitis were randomized with their 24 infants to NAC or saline treatment. Antenatal NAC (100 mg/kg/dose) or saline was given intravenously every 6 hours until delivery. Postnatally, NAC (12.5-25 mg/kg/dose, n = 12) or saline (n = 12) was given every 12 hours for 5 doses. Doppler studies of fetal umbilical and fetal and infant cerebral blood flow, cranial ultrasounds, echocardiograms, cerebral oxygenation, electroencephalograms, and serum cytokines were evaluated before and after treatment, and 12, 24, and 48 hours after birth. Magnetic resonance spectroscopy and diffusion imaging were performed at term age equivalent. Development was followed for cerebral palsy or autism to 4 years of age. RESULTS Cardiovascular measures, cerebral blood flow velocity and vascular resistance, and cerebral oxygenation did not differ between treatment groups. Cerebrovascular coupling was disrupted in infants with chorioamnionitis treated with saline but preserved in infants treated with NAC, suggesting improved vascular regulation in the presence of neuroinflammation. Infants treated with NAC had higher serum anti-inflammatory interleukin-1 receptor antagonist and lower proinflammatory vascular endothelial growth factor over time vs controls. No adverse events related to NAC administration were noted. CONCLUSIONS In this cohort of newborns exposed to chorioamnionitis, antenatal and postnatal NAC was safe, preserved cerebrovascular regulation, and increased an anti-inflammatory neuroprotective protein. TRIAL REGISTRATION ClinicalTrials.gov: NCT00724594.
Collapse
Affiliation(s)
- Dorothea D. Jenkins
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Donald B. Wiest
- Department of Clinical Pharmacy and Outcome Science, Medical University of South Carolina, Charleston, SC
| | - Denise M. Mulvihill
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Anthony M. Hlavacek
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | | | - Truman R. Brown
- Department of Neuroscience’s Center for Advanced Imaging Research, Medical University of South Carolina, Charleston, SC
| | - Joseph J. Taylor
- Department of Neuroscience’s Center for Advanced Imaging Research, Medical University of South Carolina, Charleston, SC,Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC
| | - Jason R. Buckley
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Robert P. Turner
- Department of Clinical Pediatrics and Neurology, University of South Carolina School of Medicine and Palmetto Health Richland Children’s Hospital, Columbia, SC
| | | | - Jessica P. Bentzley
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Kathryn E. Hope
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Andrew B. Barbour
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Danielle W. Lowe
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Renee H. Martin
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC
| | - Eugene Y. Chang
- Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
43
|
Mir IN, Johnson-Welch SF, Nelson DB, Brown LS, Rosenfeld CR, Chalak LF. Placental pathology is associated with severity of neonatal encephalopathy and adverse developmental outcomes following hypothermia. Am J Obstet Gynecol 2015; 213:849.e1-7. [PMID: 26408082 DOI: 10.1016/j.ajog.2015.09.072] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 08/19/2015] [Accepted: 09/15/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Although neonatal encephalopathy (NE) due to perinatal asphyxia accounts for a notable proportion of brain injury, the causal pathway remains largely unexplained. We sought to determine the association of placental pathology with: (1) severity of NE in the first 6 hours postnatal, and (2) abnormal neurodevelopmental outcomes (NDO) in neonates requiring hypothermia therapy. STUDY DESIGN This is a retrospective cohort study of neonates ≥36 weeks' gestation born at Parkland Hospital, Dallas, TX, from January 2006 through November 2011 with NE. Placental histology was reviewed and validated by a pediatric pathologist blinded to outcomes. Abnormal NDO was defined as death or Bayley-III score of <85 at 18-24 months of age. RESULTS Of 86,274 neonates ≥36 weeks' gestation, 120 had evidence of a combination of perinatal acidosis and NE. In all, 47 had mild NE and received no treatment, while 73 had moderate (n = 70) or severe (n = 3) NE and received systemic hypothermia. Nine neonates died and all survivors receiving hypothermia had a Bayley-III assessment at 22 ± 7 (SD) months of age. Chorioamnionitis with or without fetal response and patchy/diffuse chronic villitis were found to be independently associated with severity of NE (P < .001). Univariate logistic regression revealed an association with a diagnosis of major placental pathology (odds ratio, 3.5; 95% confidence interval, 1.1-11.4) and abnormal outcomes following cooling. Specifically, diffuse chronic villitis (odds ratio, 9.29; 95% confidence interval, 1.11-77.73) was the only individual predictor of abnormal NDO following hypothermia therapy. CONCLUSION Placental inflammatory villitis appears to be a harbinger of abnormal outcomes in neonates with NE, spanning to the 18-24 month NDO.
Collapse
|
44
|
Affiliation(s)
- J. R. Gold
- Department of Clinical Sciences; Washington State University; Pullman USA
| |
Collapse
|
45
|
|
46
|
Adenosine A1 receptors contribute to immune regulation after neonatal hypoxic ischemic brain injury. Purinergic Signal 2015; 12:89-101. [PMID: 26608888 DOI: 10.1007/s11302-015-9482-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/28/2015] [Indexed: 12/20/2022] Open
Abstract
Neonatal brain hypoxic ischemia (HI) often results in long-term motor and cognitive impairments. Post-ischemic inflammation greatly effects outcome and adenosine receptor signaling modulates both HI and immune cell function. Here, we investigated the influence of adenosine A1 receptor deficiency (A1R(-/-)) on key immune cell populations in a neonatal brain HI model. Ten-day-old mice were subjected to HI. Functional outcome was assessed by open locomotion and beam walking test and infarction size evaluated. Flow cytometry was performed on brain-infiltrating cells, and semi-automated analysis of flow cytometric data was applied. A1R(-/-) mice displayed larger infarctions (+33%, p < 0.05) and performed worse in beam walking tests (44% more mistakes, p < 0.05) than wild-type (WT) mice. Myeloid cell activation after injury was enhanced in A1R(-/-) versus WT brains. Activated B lymphocytes expressing IL-10 infiltrated the brain after HI in WT, but were less activated and did not increase in relative frequency in A1R(-/-). Also, A1R(-/-) B lymphocytes expressed less IL-10 than their WT counterparts, the A1R antagonist DPCPX decreased IL-10 expression whereas the A1R agonist CPA increased it. CD4(+) T lymphocytes including FoxP3(+) T regulatory cells, were unaffected by genotype, whereas CD8(+) T lymphocyte responses were smaller in A1R(-/-) mice. Using PCA to characterize the immune profile, we could discriminate the A1R(-/-) and WT genotypes as well as sham operated from HI-subjected animals. We conclude that A1R signaling modulates IL-10 expression by immune cells, influences the activation of these cells in vivo, and affects outcome after HI.
Collapse
|
47
|
Masaoka N, Nakajima Y, Morooka M, Tashiro H, Wada M, Maruta K, Iwane E, Yamashiro M. The impact of intrauterine infection on fetal brain damage assessed by S100B protein concentrations in umbilical cord arteries. J Matern Fetal Neonatal Med 2015; 29:2464-9. [PMID: 26421445 DOI: 10.3109/14767058.2015.1087501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To assess the impact of intrauterine infection on fetal brain damage by measuring S100B protein concentration in umbilical cord arteries. METHODS In the intrauterine infection cases determined by pathology of 25 deliveries (Group I) and non-infection cases of 35 deliveries as control (Group C), we compared gestational age at delivery, birth weight, fetal heart rate monitoring during labor, Apgar score, umbilical cord artery pH and S100B protein concentrations in umbilical arteries measured by two-site immunoradiometric assay kit. RESULTS (1) There was no significant correlation between pH and concentration of S100B protein. (2) Gestational age at delivery was found to be earlier in Group I, resulting in lower birth weights, when compared with Group C. (3) There was no significant difference between two groups concerning Apgar scores, pH. (4) S100B protein concentrations in Group I was significantly higher than those of Group C (3.9 7 ± 0.66 versus 1.8 9 ± 0.56 μg/L, p < 0.05). (5) The concentration of S100B protein in severe chorioamnionitis (CAM) cases were significantly higher than those of mild CAM and control cases. CONCLUSION Higher concentration of S100B protein in Group I suggests that intrauterine infection itself has a serious risk factor on fetal brain damage.
Collapse
Affiliation(s)
- Naoki Masaoka
- a Department of Obstetrics and Gynecology , Tokyo Women's Medical University Yachiyo Medical Center , Chiba , Japan
| | - Yoshiyuki Nakajima
- a Department of Obstetrics and Gynecology , Tokyo Women's Medical University Yachiyo Medical Center , Chiba , Japan
| | - Masako Morooka
- a Department of Obstetrics and Gynecology , Tokyo Women's Medical University Yachiyo Medical Center , Chiba , Japan
| | - Hidefumi Tashiro
- a Department of Obstetrics and Gynecology , Tokyo Women's Medical University Yachiyo Medical Center , Chiba , Japan
| | - Masami Wada
- a Department of Obstetrics and Gynecology , Tokyo Women's Medical University Yachiyo Medical Center , Chiba , Japan
| | - Kana Maruta
- a Department of Obstetrics and Gynecology , Tokyo Women's Medical University Yachiyo Medical Center , Chiba , Japan
| | - Eriko Iwane
- a Department of Obstetrics and Gynecology , Tokyo Women's Medical University Yachiyo Medical Center , Chiba , Japan
| | - Miwako Yamashiro
- a Department of Obstetrics and Gynecology , Tokyo Women's Medical University Yachiyo Medical Center , Chiba , Japan
| |
Collapse
|
48
|
Keane M, Fallon R, Riordan A, Shaw B. Markedly raised levels of C-reactive protein are associated with culture-proven sepsis or necrotising enterocolitis in extremely preterm neonates. Acta Paediatr 2015; 104:e289-93. [PMID: 25703293 DOI: 10.1111/apa.12978] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 12/22/2014] [Accepted: 02/12/2015] [Indexed: 11/28/2022]
Abstract
AIM A serious inflammatory process is suspected when C-reactive protein (CRP) is very high, and we established the causes and outcomes when CRP was >100 mg/L in neonates. METHODS We retrospectively reviewed all 277 episodes where CRP exceeded 100 mg/L between January 2007 and December 2011 at a tertiary neonatal unit. RESULTS Of the 6025 neonates admitted during the study period, 258 had CRP >100 mg/L at least once. The overall mortality rate was 44/258 (17%); 36 died within 7 days of CRP >100 mg/L, and 34 were extremely preterm infants. CRP exceeded 100 mg/L in 106 infants within the first 3 days of life - 74 term, 25 preterm and seven extremely preterm - with no infection identified in 81%. In contrast, infections were found in 87% of the 171 episodes from day four of life - 129 extremely preterm, 23 preterm and 19 term - predominantly coagulase-negative staphylococcus sepsis and necrotising enterocolitis. CONCLUSION Markedly elevated CRP in the first 3 days of life was most likely to affect term neonates (74/106) with no infectious cause (81%). However, CRP >100 mg/L from the fourth day of life was most likely to affect extremely preterm neonates (129/171) and have an infectious cause (87%).
Collapse
Affiliation(s)
| | - Rachel Fallon
- Neonatal Unit; Liverpool Women's Hospital; Liverpool UK
| | - Andrew Riordan
- Neonatal Unit; Liverpool Women's Hospital; Liverpool UK
- Department of Paediatric Infectious diseases; Alder Hey Children's NHS Foundation Trust; Liverpool UK
| | - Ben Shaw
- Neonatal Unit; Liverpool Women's Hospital; Liverpool UK
| |
Collapse
|
49
|
Lachapelle J, Chen M, Oskoui M, Ali N, Brown R, Wintermark P. Placental pathology in asphyxiated newborns treated with therapeutic hypothermia. J Neonatal Perinatal Med 2015; 8:33-40. [PMID: 25766201 DOI: 10.3233/npm-15814068] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE To describe the placental findings in asphyxiated newborns treated with hypothermia and to determine their association with the presence and severity of later brain injury. METHODS Prospective cohort study of the placentas of asphyxiated newborns treated with hypothermia, in whom later brain injury was defined by brain imaging and/or autopsy results. RESULTS Of the 142 asphyxiated newborns meeting the criteria for hypothermia, 73% had placenta and brain MRI/autopsy available for analysis. Fifty-one percent of these newborns developed brain injury. Sixty-five percent had microscopic placental findings involving the fetal vascular supply, which were comparable in asphyxiated newborns developing or not developing brain injury. Among the asphyxiated newborns with normal placental growth, the placental microscopic findings tended to be more common in those developing brain injury compared to those who did not: chorionic plate meconium in 50% compared to 36%, chorioamnionitis in 75% compared to 44%, and villitis of unknown etiology in 67% compared to 33%, but this did not reach statistical significance. CONCLUSIONS Antenatal placental processes are common in term asphyxiated newborns treated with hypothermia. The placenta of each asphyxiated term newborn treated with hypothermia should be carefully examined to better understand its role in the progression from perinatal depression to brain injury.
Collapse
Affiliation(s)
- J Lachapelle
- Department of Pathology, McGill University, Montreal, Canada
| | - M Chen
- Department of Pathology, McGill University, Montreal, Canada
- Department of Gynecology and Obstetrics, McGill University, Montreal, Canada
| | - M Oskoui
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Department of Pediatrics, McGill University, Montreal, Canada
| | - N Ali
- Department of Pediatrics, McGill University, Montreal, Canada
| | - R Brown
- Department of Gynecology and Obstetrics, McGill University, Montreal, Canada
| | - P Wintermark
- Department of Pediatrics, McGill University, Montreal, Canada
| |
Collapse
|
50
|
Ek CJ, D'Angelo B, Baburamani AA, Lehner C, Leverin AL, Smith PLP, Nilsson H, Svedin P, Hagberg H, Mallard C. Brain barrier properties and cerebral blood flow in neonatal mice exposed to cerebral hypoxia-ischemia. J Cereb Blood Flow Metab 2015; 35:818-27. [PMID: 25627141 PMCID: PMC4420855 DOI: 10.1038/jcbfm.2014.255] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 12/05/2014] [Accepted: 12/08/2014] [Indexed: 11/09/2022]
Abstract
Insults to the developing brain often result in irreparable damage resulting in long-term deficits in motor and cognitive functions. The only treatment today for hypoxic-ischemic encephalopathy (HIE) in newborns is hypothermia, which has limited clinical benefit. We have studied changes to the blood-brain barriers (BBB) as well as regional cerebral blood flow (rCBF) in a neonatal model of HIE to further understand the underlying pathologic mechanisms. Nine-day old mice pups, brain roughly equivalent to the near-term human fetus, were subjected to hypoxia-ischemia. Hypoxia-ischemia increased BBB permeability to small and large molecules within hours after the insult, which normalized in the following days. The opening of the BBB was associated with changes to BBB protein expression whereas gene transcript levels were increased showing direct molecular damage to the BBB but also suggesting compensatory mechanisms. Brain pathology was closely related to reductions in rCBF during the hypoxia as well as the areas with compromised BBB showing that these are intimately linked. The transient opening of the BBB after the insult is likely to contribute to the pathology but at the same time provides an opportunity for therapeutics to better reach the infarcted areas in the brain.
Collapse
Affiliation(s)
- C Joakim Ek
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Barbara D'Angelo
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ana A Baburamani
- 1] Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden [2] Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Christine Lehner
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Department of Traumatology and Sport Injuries, Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria; Austrian Cluster for Tissue Regeneration
| | - Anna-Lena Leverin
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter L P Smith
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Holger Nilsson
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Svedin
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- 1] Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK [2] Departments of Obstetrics and Gynecology, Institute for Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Department of Physiology, Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|