1
|
Bitar L, Stonestreet BS, Lim YP, Qiu J, Chen X, Mir IN, Chalak LF. Association between decreased cord blood inter-alpha inhibitor levels and neonatal encephalopathy at birth. Early Hum Dev 2024; 193:106036. [PMID: 38733833 DOI: 10.1016/j.earlhumdev.2024.106036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Inter-alpha inhibitor proteins (IAIPs) are structurally related proteins found in the systemic circulation with immunomodulatory anti-inflammatory properties. Reduced levels are found in inflammatory related conditions including sepsis and necrotizing enterocolitis, and in neonatal rodents after exposure to hypoxia ischemia. In the current study, cord blood IAIP levels were measured in neonates with and without exposure to hypoxic-ischemic encephalopathy (HIE). METHODS This is a prospective cohort study including infants born ≥36 weeks over a one-year period. Term pregnancies were divided into two groups: a "reference control" (uncomplicated term deliveries), and "moderate to severe HIE" (qualifying for therapeutic hypothermia). IAIPs were quantified using a sensitive ELISA on the cord blood samples. RESULTS The study included 57 newborns: Reference control group (n = 13) and moderate/severe HIE group (n = 44). Measurement of IAIP cord blood concentrations in moderate to severe HIE group [278.2 (138.0, 366.0) μg/ml] revealed significantly lower IAIP concentrations compared with the control group [418.6 (384.5, 445.0) μg/ml] (p = 0.002). CONCLUSIONS These findings suggest a potential role for IAIPs as indicators of neonates at risk for HIE. IAIP levels could have diagnostic implications in the management of HIE. Future research is required to explore the relationship between HIE and IAIPs as biomarkers for disease severity. CATEGORY OF STUDY Translational.
Collapse
Affiliation(s)
- Lynn Bitar
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Barbara S Stonestreet
- The Alpert Medical School of Brown University, Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI, United States of America
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI, United States of America; The Alpert Medical School of Brown University, Department of Pathology and Laboratory Medicine, Providence, RI, United States of America
| | - Joseph Qiu
- ProThera Biologics, Inc., Providence, RI, United States of America
| | - Xiaodi Chen
- The Alpert Medical School of Brown University, Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI, United States of America
| | - Imran N Mir
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Lina F Chalak
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States of America.
| |
Collapse
|
2
|
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
|
3
|
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: 6.0] [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
|
4
|
Kushwah S, Kumar A, Verma A, Basu S, Kumar A. Comparison of fractional anisotropy and apparent diffusion coefficient among hypoxic ischemic encephalopathy stages 1, 2, and 3 and with nonasphyxiated newborns in 18 areas of brain. Indian J Radiol Imaging 2021; 27:447-456. [PMID: 29379241 PMCID: PMC5761173 DOI: 10.4103/ijri.ijri_384_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Purpose To determine the area and extent of injury in hypoxic encephalopathy stages by diffusion tensor imaging (DTI) using parameters apparent diffusion coefficient (ADC) and fractional anisotropy (FA) values and their comparison with controls without any evidence of asphyxia. To correlate the outcome of hypoxia severity clinically and significant changes on DTI parameter. Materials and Methods DTI was done in 50 cases at median age of 12 and 20 controls at median age of 7 days. FA and apparent diffusion coefficient (ADC) were measured in several regions of interest (ROI). Continuous variables were analyzed using Student's t-test. Categorical variables were compared by Fisher's exact test. Comparison among multiple groups was done using analysis of variance (ANOVA) and post hoc Bonferroni test. Results Abnormalities were more easily and accurately determined in ROI with the help of FA and ADC values. When compared with controls FA values were significantly decreased and ADC values were significantly increased in cases, in ROI including both right and left side of thalamus, basal ganglia, posterior limb of internal capsule, cerebral peduncle, corticospinal tracts, frontal, parietal, temporal, occipital with P value < 0.05. The extent of injury was maximum in stage-III. There was no significant difference among males and females. Conclusion Compared to conventional magnetic resonance imaging (MRI), the evaluation of FA and ADC values using DTI can determine the extent and severity of injury in hypoxic encephalopathy. It can be used for early determination of brain injury in these patients.
Collapse
Affiliation(s)
- Supriya Kushwah
- Department of Paediatrics, Yenepoya Medical College, Mangalore, Karnataka, India
| | - Ashok Kumar
- Department of Paediatrics, Institute of Medical Sciences, Varanasi, Uttar Pradesh, India
| | - Ashish Verma
- Department of Paediatrics, Institute of Medical Sciences, Varanasi, Uttar Pradesh, India
| | - Sriparna Basu
- Department of Paediatrics, Institute of Medical Sciences, Varanasi, Uttar Pradesh, India
| | - Ashutosh Kumar
- Department of Anaesthesia, KMC, Mangalore, Karnataka, India
| |
Collapse
|
5
|
Parfenova H, Pourcyrous M, Fedinec AL, Liu J, Basuroy S, Leffler CW. Astrocyte-produced carbon monoxide and the carbon monoxide donor CORM-A1 protect against cerebrovascular dysfunction caused by prolonged neonatal asphyxia. Am J Physiol Heart Circ Physiol 2018; 315:H978-H988. [PMID: 30028198 DOI: 10.1152/ajpheart.00140.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neonatal asphyxia leads to cerebrovascular disease and neurological complications via a mechanism that may involve oxidative stress. Carbon monoxide (CO) is an antioxidant messenger produced via a heme oxygenase (HO)-catalyzed reaction. Cortical astrocytes are the major cells in the brain that express constitutive HO-2 isoform. We tested the hypothesis that CO, produced by astrocytes, has cerebroprotective properties during neonatal asphyxia. We developed a survival model of prolonged asphyxia in newborn pigs that combines insults of severe hypoxia, hypercapnia, and acidosis while avoiding extreme hypotension and cerebral blood flow reduction. During the 60-min asphyxia, CO production by brain and astrocytes was continuously elevated. Excessive formation of reactive oxygen species during asphyxia/reventilation was potentiated by the HO inhibitor tin protoporphyrin, suggesting that endogenous CO has antioxidant effects. Cerebral vascular outcomes tested 24 and 48 h after asphyxia demonstrated the sustained impairment of cerebral vascular responses to astrocyte- and endothelium-specific vasodilators. Postasphyxia cerebral vascular dysfunction was aggravated in newborn pigs pretreated with tin protoporphyrin to inhibit brain HO/CO. The CO donor CO-releasing molecule-A1 (CORM-A1) reduced brain oxidative stress during asphyxia/reventilation and prevented postasphyxia cerebrovascular dysfunction. The antioxidant and antiapoptotic effects of HO/CO and CORM-A1 were confirmed in primary cultures of astrocytes from the neonatal pig brain exposed to glutamate excitotoxicity. Overall, prolonged neonatal asphyxia leads to neurovascular injury via an oxidative stress-mediated mechanism that is counteracted by an astrocyte-based constitutive antioxidant HO/CO system. We propose that gaseous CO or CO donors can be used as novel approaches for prevention of neonatal brain injury caused by prolonged asphyxia. NEW & NOTEWORTHY Asphyxia in newborn infants may lead to lifelong neurological disabilities. Using the model of prolonged asphyxia in newborn piglets, we propose novel antioxidant therapy based on systemic administration of low doses of a carbon monoxide donor that prevent loss of cerebral blood flow regulation and may improve the neurological outcome of asphyxia.
Collapse
Affiliation(s)
- Helena Parfenova
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Massroor Pourcyrous
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Alex L Fedinec
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Jianxiong Liu
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Shyamali Basuroy
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Charles W Leffler
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
| |
Collapse
|
6
|
Luan Y, Zhang X, Zhang Y, Dong Y. MicroRNA-210 Protects PC-12 Cells Against Hypoxia-Induced Injury by Targeting BNIP3. Front Cell Neurosci 2017; 11:285. [PMID: 29018330 PMCID: PMC5614931 DOI: 10.3389/fncel.2017.00285] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 09/01/2017] [Indexed: 11/30/2022] Open
Abstract
MicroRNA (miR)-210 is the most consistently and predominantly up-regulated miR in response to hypoxia in multiple cancer cells. The roles of miR-210 in rat adrenal gland pheochromocytoma (PC-12) cells remain unknown. We aimed to explore the possible effect of miR-210 in neonatal brain injury. We explored the potential molecular mechanism by using PC-12 cells under hypoxia. Scramble miRs, miR-210 mimic, miR-210 inhibitor or its negative control were respectively transfected into PC-12 cells. Cell viability, migration, invasion and apoptosis were also assessed to evaluate hypoxia-induced cell injury. The expression level of miR-210 was identified by quantitative real-time polymerase chain reaction (qRT-PCR) analysis. Apoptosis-related protein expression as well as key kinases in the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signal pathway was studied by Western blot analysis. Hypoxia suppressed cell viability, migration and invasion, but promoted apoptosis through activation of mitochondrial- and caspase-dependent pathways. Hypoxia markedly induced up-regulation of miR-210 in PC-12 cells. Overexpression of miR-210 protected PC-12 cells against hypoxia-induced injury. Bcl-2 adenovirus E1B 19 kDa-interacting protein 3 (BNIP3) was proven to be a target gene of miR-210 in PC-12 cells. miR-210 overexpression ameliorated the hypoxia-induced injury in PC-12 cells by down-regulating BNIP3. Hypoxia-induced alterations of key kinases in the PI3K/AKT/mTOR signal pathway were affected by aberrant expression of BNIP3. These findings suggested that miR-210 protected PC-12 cells against hypoxia-induced injury by targeting BNIP3, involving the PI3K/AKT/mTOR signal pathway.
Collapse
Affiliation(s)
- Yonggang Luan
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| | - Xiaoli Zhang
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| | - Yongli Zhang
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| | - Yubin Dong
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| |
Collapse
|
7
|
Abstract
"Golden Hour" of neonatal life is defined as the first hour of post-natal life in both preterm and term neonates. This concept in neonatology has been adopted from adult trauma where the initial first hour of trauma management is considered as golden hour. The "Golden hour" concept includes practicing all the evidence based intervention for term and preterm neonates, in the initial sixty minutes of postnatal life for better long-term outcome. Although the current evidence supports the concept of golden hour in preterm and still there is no evidence seeking the benefit of golden hour approach in term neonates, but neonatologist around the globe feel the importance of golden hour concept equally in both preterm and term neonates. Initial first hour of neonatal life includes neonatal resuscitation, post-resuscitation care, transportation of sick newborn to neonatal intensive care unit, respiratory and cardiovascular support and initial course in nursery. The studies that evaluated the concept of golden hour in preterm neonates showed marked reduction in hypothermia, hypoglycemia, intraventricular hemorrhage (IVH), bronchopulmonary dysplasia (BPD), and retinopathy of prematurity (ROP). In this review article, we will discuss various components of neonatal care that are included in "Golden hour" of preterm and term neonatal care.
Collapse
Affiliation(s)
- Deepak Sharma
- National Institute of Medical Science, Jaipur, Rajasthan India
| |
Collapse
|
8
|
Anderson KB, Poloyac SM, Kochanek PM, Empey PE. Effect of Hypothermia and Targeted Temperature Management on Drug Disposition and Response Following Cardiac Arrest: A Comprehensive Review of Preclinical and Clinical Investigations. Ther Hypothermia Temp Manag 2016; 6:169-179. [PMID: 27622966 DOI: 10.1089/ther.2016.0003] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Targeted temperature management (TTM) has been shown to reduce mortality and improve neurological outcomes in out-of-hospital cardiac arrest (CA) patients and in neonates with hypoxic-ischemic encephalopathy (HIE). TTM has also been associated with adverse drug events in the critically ill patient due to its effect on drug pharmacokinetics (PKs) and pharmacodynamics (PDs). We aim to evaluate the current literature on the effect of TTM on drug PKs and PDs following CA. MEDLINE/PubMed databases were searched for publications, which include the MeSH terms hypothermia, drug metabolism, drug transport, P450, critical care, cardiac arrest, hypoxic-ischemic encephalopathy, pharmacokinetics, and pharmacodynamics between July 2006 and October 2015. Twenty-three studies were included in this review. The studies demonstrate that hypothermia impacts PK parameters and increases concentrations of cytochrome-P450-metabolized drugs in the cooling and rewarming phase. Furthermore, the current data demonstrate a combined effect of CA and hypothermia on drug PK. Importantly, these effects can last greater than 4-5 days post-treatment. Limited evidence suggests hypothermia-mediated changes in the Phase II metabolism and the Phase III transport of drugs. Hypothermia also has been shown to potentially decrease the effect of specific drugs at the receptor level. Therapeutic hypothermia, as commonly deployed/applied during TTM, alters PK, and elevates concentrations of several commonly used medications. Hypothermia-mediated effects are an important factor when dosing and monitoring patients undergoing TTM treatment.
Collapse
Affiliation(s)
- Kacey B Anderson
- 1 Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Samuel M Poloyac
- 1 Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Patrick M Kochanek
- 2 Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Philip E Empey
- 3 Department of Pharmacy and Therapeutics, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
9
|
Lan KM, Tien LT, Cai Z, Lin S, Pang Y, Tanaka S, Rhodes PG, Bhatt AJ, Savich RD, Fan LW. Erythropoietin Ameliorates Neonatal Hypoxia-Ischemia-Induced Neurobehavioral Deficits, Neuroinflammation, and Hippocampal Injury in the Juvenile Rat. Int J Mol Sci 2016; 17:289. [PMID: 26927081 PMCID: PMC4813153 DOI: 10.3390/ijms17030289] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 01/13/2016] [Accepted: 02/05/2016] [Indexed: 12/11/2022] Open
Abstract
The hematopoietic growth factor erythropoietin (EPO) has been shown to be neuroprotective against hypoxia-ischemia (HI) in Postnatal Day 7 (P7)–P10 or adult animal models. The current study was aimed to determine whether EPO also provides long-lasting neuroprotection against HI in P5 rats, which is relevant to immature human infants. Sprague-Dawley rats at P5 were subjected to right common carotid artery ligation followed by an exposure to 6% oxygen with balanced nitrogen for 1.5 h. Human recombinant EPO (rEPO, at a dose of 5 units/g) was administered intraperitoneally one hour before or immediately after insult, followed by additional injections at 24 and 48 h post-insult. The control rats were injected with normal saline following HI. Neurobehavioral tests were performed on P8 and P20, and brain injury was examined on P21. HI insult significantly impaired neurobehavioral performance including sensorimotor, locomotor activity and cognitive ability on the P8 and P20 rats. HI insult also resulted in brain inflammation (as indicated by microglia activation) and neuronal death (as indicated by Jade B positive staining) in the white matter, striatum, cortex, and hippocampal areas of the P21 rat. Both pre- and post-treatment with rEPO significantly improved neurobehavioral performance and protected against the HI-induced neuronal death, microglia activation (OX42+) as well as loss of mature oligodendrocytes (APC-CC1+) and hippocampal neurons (Nissl+). The long-lasting protective effects of rEPO in the neonatal rat HI model suggest that to exert neurotrophic activity in the brain might be an effective approach for therapeutic treatment of neonatal brain injury induced by hypoxia-ischemia.
Collapse
Affiliation(s)
- Kuo-Mao Lan
- Department of Anesthesiology, Chi-Mei General Hospital, Tainan 71004, Taiwan.
- School of Medicine, Fu Jen Catholic University, Xinzhuang District, New Taipei City 24205, Taiwan.
| | - Lu-Tai Tien
- School of Medicine, Fu Jen Catholic University, Xinzhuang District, New Taipei City 24205, Taiwan.
| | - Zhengwei Cai
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | - Shuying Lin
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | - Yi Pang
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | - Sachiko Tanaka
- Department of Pharmacology, Toxicology & Therapeutics, Division of Toxicology, School of Pharmacy, Showa University, Shingawa-ku, Tokyo 142-8555, Japan.
| | - Philip G Rhodes
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | - Abhay J Bhatt
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | - Renate D Savich
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | - Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
10
|
Nnorom CC, Davis C, Fedinec AL, Howell K, Jaggar JH, Parfenova H, Pourcyrous M, Leffler CW. Contributions of KATP and KCa channels to cerebral arteriolar dilation to hypercapnia in neonatal brain. Physiol Rep 2014; 2:2/8/e12127. [PMID: 25168876 PMCID: PMC4246596 DOI: 10.14814/phy2.12127] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mechanisms by which Pco2 controls cerebral vascular tone remain uncertain. We hypothesize that potassium channel activation contributes to the neonatal cerebrovascular dilation in response to increases in Paco2. To test this hypothesis, experiments were performed on newborn pigs with surgically implanted, closed cranial windows. Hypercapnia was induced by ventilation with elevated Pco2 gas in the absence and presence of the KATP channel inhibitor, glibenclamide and/or the KCa channel inhibitor, paxillin. Dilations to pinacidil, a selective KATP channel activator, without and with glibenclamide, were used to evaluate the efficacy of KATP channel inhibition. Dilations to NS1619, a selective KCa channel activator, without and with paxillin, were used to evaluate the efficacy of KCa channel inhibition. Cerebrovascular responses to the KATP and KCa channel activators, pinacidil and NS1619, respectively, cAMP‐dependent dilator, isoproterenol, and cGMP‐dependent dilator, sodium nitroprusside (SNP), were used to evaluate the selectivity of glibenclamide and paxillin. Glibenclamide blocked dilation to pinacidil, but did not inhibit dilations to NS1619, isoproterenol, or SNP. Glibenclamide prior to hypercapnia decreased mean pial arteriole dilation ~60%. Glibenclamide treatment during hypercapnia constricted arterioles ~35%. The level of hypercapnia, Paco2 between 50 and 75 mmHg, did not appear to be involved in efficacy of glibenclamide in blocking dilation to Paco2. Similarly to glibenclamide and KATP channel inhibition, paxillin blocked dilation to the KCa channel agonist, NS1619, and attenuated, but did not block, arteriolar dilation to hypercapnia. Treatment with both glibenclamide and paxillin abolished dilation to hypercapnia. Therefore, either glibenclamide or paxillin that block dilation to their channel agonists, pinacidil or NS1619, respectively, only partially inhibit dilation to hypercapnia. Block of both KATP and KCa channels completely prevent dilation hypercapnia. These data suggest hypercapnia activates both KATP and KCa channels leading to cerebral arteriolar dilation in newborn pigs. Mechanisms by which Pco2 controls vascular tone remain uncertain. We hypothesize KATP and KCa channel activation contributes to the neonatal cerebrovascular dilation in response to increases in Paco2. Presented data support this hypothesis.
Collapse
Affiliation(s)
- Chukwuma C Nnorom
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Corinne Davis
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Alexander L Fedinec
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Khadesia Howell
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Jonathan H Jaggar
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Helena Parfenova
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Massroor Pourcyrous
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Charles W Leffler
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
11
|
Behavioral and histological outcomes following neonatal HI injury in a preterm (P3) and term (P7) rodent model. Behav Brain Res 2013; 259:85-96. [PMID: 24185032 DOI: 10.1016/j.bbr.2013.10.038] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/15/2013] [Accepted: 10/24/2013] [Indexed: 11/22/2022]
Abstract
Hypoxia-ischemia (HI) occurs when blood and/or oxygen delivery to the brain is compromised. HI injuries can occur in infants born prematurely (<37 weeks gestational age) or at very low birth weight (<1500 g), as well as in term infants with birth complications. In both preterm and term HI populations, brain injury is associated with subsequent behavioral deficits. Neonatal HI injury can be modeled in rodents (e.g., the Rice-Vannucci method, via cautery of right carotid followed by hypoxia). When this injury is induced early in life (between postnatal day (P)1-5), neuropathologies typical of human preterm HI are modeled. When injury is induced later (P7-12), neuropathologies typical of those seen in HI term infants are modeled. The current study sought to characterize the similarities/differences between outcomes following early (P3) and late (P7) HI injury in rats. Male rats with HI injury on P3 or P7, as well as sham controls, were tested on a variety of behavioral tasks in both juvenile and adult periods. Results showed that P7 HI rats displayed deficits on motor learning, rapid auditory processing (RAP), and other learning/memory tasks, as well as a reduction in volume in various neuroanatomical structures. P3 HI animals showed only transient deficits on RAP tasks in the juvenile period (but not in adulthood), yet robust deficits on a visual attention task in adulthood. P3 HI animals did not show any significant reductions in brain volume that we could detect. These data suggest that: (1) behavioral deficits following neonatal HI are task-specific depending on timing of injury; (2) P3 HI rats showed transient deficits on RAP tasks; (3) the more pervasive behavioral deficits seen following P7 HI injury were associated with substantial global tissue loss; and (4) persistent deficits in attention in P3 HI subjects might be linked to neural connectivity disturbances rather than a global loss of brain volume, given that no such pathology was found. These combined findings can be applied to our understanding of differing long-term outcomes following neonatal HI injury in premature versus term infants.
Collapse
|
12
|
Heringhaus A, Blom MD, Wigert H. Becoming a parent to a child with birth asphyxia-From a traumatic delivery to living with the experience at home. Int J Qual Stud Health Well-being 2013; 8:1-13. [PMID: 23639330 PMCID: PMC3643077 DOI: 10.3402/qhw.v8i0.20539] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/03/2013] [Indexed: 11/14/2022] Open
Abstract
The aim of this study is to describe the experiences of becoming a parent to a child with birth asphyxia treated with hypothermia in the neonatal intensive care unit (NICU). In line with the medical advances, the survival of critically ill infants with increased risk of morbidity is increasing. Children who survive birth asphyxia are at a higher risk of functional impairments, cerebral palsy (CP), or impaired vision and hearing. Since 2006, hypothermia treatment following birth asphyxia is used in many of the Swedish neonatal units to reduce the risk of brain injury. To date, research on the experience of parenthood of the child with birth asphyxia is sparse. To improve today's neonatal care delivery, health-care providers need to better understand the experiences of becoming a parent to a child with birth asphyxia. A total of 26 parents of 16 children with birth asphyxia treated with hypothermia in a Swedish NICU were interviewed. The transcribed interview texts were analysed according to a qualitative latent content analysis. We found that the experience of becoming a parent to a child with birth asphyxia treated with hypothermia at the NICU was a strenuous journey of overriding an emotional rollercoaster, that is, from being thrown into a chaotic situation which started with a traumatic delivery to later processing the difficult situation of believing the child might not survive or was to be seriously affected by the asphyxia. The prolonged parent-infant separation due to the hypothermia treatment and parents' fear of touching the infant because of the high-tech equipment seemed to hamper the parent-infant bonding. The adaption of the everyday life at home seemed to be facilitated by the follow-up information of the doctor after discharge. The results of this study underline the importance of family-centered support during and also after the NICU discharge.
Collapse
Affiliation(s)
- Alina Heringhaus
- Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | | | | |
Collapse
|
13
|
QIU JIE, ZHOU XIAOYU, ZHOU XIAOGUANG, CHENG RUI, LIU HAIYING, LI YONG. Neuroprotective effects of microRNA-210 against oxygen-glucose deprivation through inhibition of apoptosis in PC12 cells. Mol Med Rep 2013; 7:1955-9. [DOI: 10.3892/mmr.2013.1431] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 04/10/2013] [Indexed: 11/06/2022] Open
|
14
|
Merrill L. Therapeutic hypothermia to treat hypoxic ischemic encephalopathy in newborns: implications for nurses. Nurs Womens Health 2012; 16:126-134. [PMID: 22900770 DOI: 10.1111/j.1751-486x.2012.01718.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Hypoxic ischemic encephalopathy (HIE) in newborns is caused by an injury to the brain following a hypoxic or an ischemic event during the peripartum, intrapartum or postpartum period. HIE may result in death or cause serious impairment in survivors, and remains a significant cause of morbidity and mortality among neonates. Mild hypothermia as a treatment for HIE is commonly used to treat moderate to severe HIE, with promising results. Nurses play an integral role in identifying newborns at risk of developing HIE.
Collapse
|
15
|
Reynolds R, Talmage S. "Caution! Contents should be cold": developing a whole-body hypothermia program. Neonatal Netw 2011; 30:225-30. [PMID: 21729853 DOI: 10.1891/0730-0832.30.4.225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is characterized as brain injury that results from lack of oxygen or blood flow to the brain in the perinatal period. Neonatal whole-body hypothermia and selective head cooling are becoming increasingly common care practices across the U.S. and Canada for infants with moderate-to-severe HIE because of the demonstrated ability of these approaches to reduce reperfusion injury to the brain. Health care professionals must develop a clinical care path for these fragile infants. For best results, induced hypothermia should be initiated within six hours of birth; therefore, care must be organized and provided without delay. This article provides bedside clinicians with care recommendations for infants being treated with these new interventions.
Collapse
|
16
|
Beckstrom AC, Humston EM, Snyder LR, Synovec RE, Juul SE. Application of comprehensive two-dimensional gas chromatography with time-of-flight mass spectrometry method to identify potential biomarkers of perinatal asphyxia in a non-human primate model. J Chromatogr A 2011; 1218:1899-906. [PMID: 21353677 PMCID: PMC3064854 DOI: 10.1016/j.chroma.2011.01.086] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 01/09/2011] [Accepted: 01/31/2011] [Indexed: 12/19/2022]
Abstract
Perinatal asphyxia is a leading cause of brain injury in infants, occurring in 2-4 per 1000 live births. The clinical response to asphyxia is variable and difficult to predict with current diagnostic tests. Reliable biomarkers are needed to help predict the timing and severity of asphyxia, as well as response to treatment. Two-dimensional gas chromatography-time-of-flight-mass spectrometry (GC×GC-TOFMS) was used herein, in conjunction with chemometric data analysis approaches for metabolomic analysis in order to identify significant metabolites affected by birth asphyxia. Blood was drawn before and after 15 or 18 min of cord occlusion in a Macaca nemestrina model of perinatal asphyxia. Postnatal samples were drawn at 5 min of age (n=20 subjects). Metabolomic profiles of asphyxiated animals were compared to four controls delivered at comparable gestational age. Fifty metabolites with the greatest change pre- to post-asphyxia were identified and quantified. The metabolic profile of post-asphyxia samples showed marked variability compared to the pre-asphyxia samples. Fifteen of the 50 metabolites showed significant elevation in response to asphyxia, ten of which remained significant upon comparison to the control animals. This metabolomic analysis confirmed lactate and creatinine as markers of asphyxia and discovered new metabolites including succinic acid and malate (intermediates in the Krebs cycle) and arachidonic acid (a brain fatty acid and inflammatory marker) as potential biomarkers. GC×GC-TOFMS coupled with chemometric data analysis are useful tools to identify acute biomarkers of brain injury. Further study is needed to correlate these metabolites with severity of disease, and response to treatment.
Collapse
Affiliation(s)
| | | | - Laura R. Snyder
- Department of Pathology, University of Washington, Seattle, WA 98195
| | - Robert E. Synovec
- Department of Chemistry, University of Washington, Seattle, WA 98195
| | - Sandra E. Juul
- Department of Pediatrics, University of Washington, Seattle, WA 98195
| |
Collapse
|
17
|
Abstract
Hypoxia-ischemia in the perinatal period is an important cause of cerebral palsy and associated disabilities in children. There has been significant research progress in hypoxic-ischemic encephalopathy over the last 2 decades, and many new molecular mechanisms have been identified. Despite all these advances, therapeutic interventions are still limited. In this article the authors discuss several molecular pathways involved in hypoxia-ischemia, and potential therapeutic targets.
Collapse
Affiliation(s)
- Ali Fatemi
- Assistant Professor of Neurology and Pediatrics, Kennedy Krieger Institute, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Mary Ann Wilson
- Associate Professor of Neurology and Neuroscience, Kennedy Krieger Institute, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael V. Johnston
- Blum-Moser Chair for Pediatric Neurology at the Kennedy Krieger Institute, Professor of Neurology, Pediatrics, Physical Medicine and Rehabilitation, Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
18
|
Abstract
Neonatal resuscitation is an attempt to facilitate the dynamic transition from fetal to neonatal physiology. This article outlines the current practices in delivery room management of the neonate. Developments in cardiopulmonary resuscitation techniques for term and preterm infants and advances in the areas of cerebral resuscitation and thermoregulation are reviewed. Resuscitation in special circumstances (such as the presence of congenital anomalies) are also covered. The importance of communication with other members of the health care team and the family is discussed. Finally, future trends in neonatal resuscitation are explored.
Collapse
Affiliation(s)
- Anand K Rajani
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA.
| | | | | |
Collapse
|
19
|
Abstract
UNLABELLED Hypothermia is the first treatment for newborns with hypoxic-ischaemic encephalopathy (HIE) with consistent evidence of a reduction in the risk of death or severe disability. This paper addresses a number of ethical and practical issues faced by clinicians as cooling moves from an experimental treatment into practice. These issues are not unique to therapeutic hypothermia. They include the extrapolation of evidence from trials to clinical care, as well as the impact of hypothermia on prognosis and withdrawal of life-sustaining treatment. CONCLUSION Hypothermia is a promising new therapy, but further research will be necessary to help resolve some of the ethical concerns associated with its use in newborns with HIE .
Collapse
|
20
|
Mathur AM, Smith JR, Donze A. Hypothermia and hypoxic-ischemic encephalopathy: guideline development using the best evidence. Neonatal Netw 2008; 27:271-86. [PMID: 18697657 DOI: 10.1891/0730-0832.27.4.271] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BABY AVA WAS DELIVERED AT 39 weeks gestation by emergency cesarean section following a prolapsed cord. Her mother was 23 years old, and this was her first pregnancy, which had been uneventful. She was Group B Streptococcus negative. The mother’s membranes ruptured one hour prior to arrival at the hospital, and she presented in labor. She was afebrile with stable vital signs. When initially examined, the cord was found prolapsed in the vaginal canal. She was immediately placed in a knee-chest posture and rushed to the operating room.
Collapse
Affiliation(s)
- Amit Mohan Mathur
- Washington University School of Medicine, St. Louis Children's Hospital, USA
| | | | | |
Collapse
|