1
|
Xie W, Ding B, Lou J, Wang X, Guo X, Zhu J. Metformin attenuates white matter injury in neonatal mice through activating NRF2/HO-1/NF-κB pathway. Int Immunopharmacol 2024; 141:112961. [PMID: 39163687 DOI: 10.1016/j.intimp.2024.112961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024]
Abstract
White matter injury (WMI) is a major form of brain injury that occurs in preterm infants and develops into lifelong disabilities, including cerebral palsy, impaired cognitive function, and psychiatric disorders. Metformin (MET) has been reported to have neuroprotective effects. However, whether MET is responsible for neuroprotection against WMI remains unclear. In this study, we established a WMI model in neonatal mice to explore the neuroprotective effects of MET and attempted to elucidate its potential mechanisms. Our results showed that MET increased the expression of myelin basic protein (MBP), oligodendrocyte transcription factor 2 (Olig2), and CC1, improved the thickness and density of the myelin sheath, and reduced oxidative stress and microglial infiltration after chronic hypoxia induction. Moreover, MET improved memory, learning, and motor abilities as well as relieved anxiety-like behaviors in mice with WMI. These protective effects of MET may involve the upregulation of the nuclear factor erythroid 2-related factor 2 (NRF2)/heme oxygenase-1(HO-1)/NF-κB pathway related protein expressions. In addition, the NRF2 inhibitor ML385 could significantly reverse the effects of MET. In conclusion, this study suggested that MET attenuated chronic hypoxia-induced WMI through activating the NRF2/HO-1/NF-κB pathway, indicating that MET might be a promising therapeutic option for WMI.
Collapse
Affiliation(s)
- Weiwei Xie
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Pediatrics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 150 Ximen Street, Linhai, Zhejiang, China
| | - Bingqing Ding
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia Lou
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyi Wang
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoling Guo
- Scientific Research Department, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China.
| | - Jianghu Zhu
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China.
| |
Collapse
|
2
|
Starke N, Challa NVD, Yuan H, Chen S, Duncan MR, Cabrera Ranaldi ED, de Rivero Vaccari JP, Schott A, Aguilar AC, Lee YS, Khan A, Duara J, Tan A, Benny M, Schmidt AF, Young K, Bancalari E, Claure N, Wu S. Extracellular Vesicle ASC: A Novel Mediator for Lung-Brain Axis in Preterm Brain Injury. Am J Respir Cell Mol Biol 2024; 71:464-480. [PMID: 38959416 PMCID: PMC11450310 DOI: 10.1165/rcmb.2023-0402oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 07/03/2024] [Indexed: 07/05/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) and neurodevelopmental impairment are among the most common morbidities affecting preterm infants. Although BPD is a predictor of poor neurodevelopmental outcomes, it is currently uncertain how BPD contributes to brain injury in preterm infants. Extracellular vesicles (EVs) are involved in interorgan communication in diverse pathological processes. ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain) is pivotal in inflammasome assembly and activation of inflammatory response. We assessed expression profiles of the alveolar macrophage (AM) markers CD11b, CD11c, and CD206 as well as ASC in EVs isolated from the plasma of preterm infants at risk for BPD at 1 week of age. We found that infants on higher fraction of inspired oxygen therapy (HO2⩾30%) had increased concentrations of AM-derived EV-ASC compared with infants on lower fraction of inspired oxygen (LO2<30%). To assess the function of these EVs, we performed adoptive transfer experiments by injecting them into the circulation of newborn mice. We discovered that mice that received EVs from infants on HO2 had increased lung inflammation, decreased alveolarization, and disrupted vascular development, the hallmarks of BPD. Importantly, these EVs crossed the blood-brain barrier, and the EVs from infants on HO2 caused inflammation, reduced cell survival, and increased cell death, with features of pyroptosis and necroptosis in the hippocampus. These results highlight a novel role for AM-derived EV-ASC in mediating the lung-to-brain cross-talk that is critical in the pathogenesis of BPD and brain injury and identify potential novel targets for preventing and treating BPD and brain injury in preterm infants.
Collapse
Affiliation(s)
- Natalie Starke
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Naga Venkata Divya Challa
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Huijun Yuan
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Shaoyi Chen
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Matthew R. Duncan
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | | | | | - Alini Schott
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Ana Cecilia Aguilar
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Yee-Shuan Lee
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, Florida
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, Florida
| | - Jo Duara
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - April Tan
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Merline Benny
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Augusto F. Schmidt
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Karen Young
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Eduardo Bancalari
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Nelson Claure
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| | - Shu Wu
- Division of Neonatology, Department of Pediatrics, Batchelor Children’s Research Institute, Holtz Children’s Hospital
| |
Collapse
|
3
|
Torres-González C, Ricardo-Garcell J, Alvarez-Núñez D, Galindo-Aldana G. Intellectual Development in Mexican Preterm Children at Risk of Perinatal Brain Damage: A Longitudinal Study. CHILDREN (BASEL, SWITZERLAND) 2024; 11:652. [PMID: 38929232 PMCID: PMC11201988 DOI: 10.3390/children11060652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024]
Abstract
Preterm birth accounts for about 10% of births worldwide. Studying risk factors for perinatal brain damage is essential, as findings suggest that almost 20% of disabilities are linked to risks in the early stages of development. This research aimed to study longitudinal changes in intelligence from 6 to 8 years of age in a sample of 39 preterm children with a history of risk of brain damage and a control group of 35 children born at term. The Wechsler Intelligence Scale (WISC-IV) was used to measure cognitive ability at six, seven, and eight years old. The results showed that the preterm group obtained significantly lower scores than the control group. The working memory indicator significantly affected the interaction between age and prematurity. We consider it crucial to expand the knowledge we have about the neurocognitive development of premature infants, both in specific cognitive domains and in age ranges, so that the information obtained can help predict the probability of presenting cognitive alterations from early stages. This, therefore, helps in implementing intervention strategies and programs based on scientific evidence, and their design is complemented by clinical experience and empirical and theoretical knowledge of the different professionals involved in infant cognitive intervention.
Collapse
Affiliation(s)
- Cynthia Torres-González
- Faculty of Administrative, Social, and Engineering Sciences, Universidad Autónoma de Baja California, State Hwy No. 3, Guadalupe Victoria, Mexicali 21720, Baja California, Mexico;
| | - Josefina Ricardo-Garcell
- Neurodevelopmental Research Unit “Augusto Fernandez Guardiola”, Institute of Neurobiology, Autonomous University of Mexico, Boulevard Juriquilla 3001, La Mesa, Juriquilla 76230, Querétaro, Mexico
| | - Daniel Alvarez-Núñez
- CETyS University, Calzada CETYS s/n. Col. Rivera, Mexicali 21259, Baja California, Mexico;
| | - Gilberto Galindo-Aldana
- Faculty of Administrative, Social, and Engineering Sciences, Universidad Autónoma de Baja California, State Hwy No. 3, Guadalupe Victoria, Mexicali 21720, Baja California, Mexico;
| |
Collapse
|
4
|
Shah DK, Pereira S, Lodygensky GA. Long-Term Neurologic Consequences following Fetal Growth Restriction: The Impact on Brain Reserve. Dev Neurosci 2024; 47:139-146. [PMID: 38740013 PMCID: PMC11965808 DOI: 10.1159/000539266] [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: 10/09/2023] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Fetal growth restriction (FGR) corresponds to the fetus's inability to achieve an adequate weight gain based on genetic potential and gestational age. It is an important cause of morbidity and mortality. SUMMARY In this review, we address the challenges of diagnosis and classification of FGR. We review how chronic fetal hypoxia impacts brain development. We describe recent advances on placental and fetal brain imaging using magnetic resonance imaging and how they offer new noninvasive means to study growth restriction in humans. We go on to review the impact of FGR on brain integrity in the neonatal period, later childhood, and adulthood and review available therapies. KEY MESSAGES FGR consequences are not limited to the perinatal period. We hypothesize that impaired brain reserve, as defined by structure and size, may predict some concerning epidemiological data of impaired cognitive outcomes and dementia with aging in this group of patients. BACKGROUND Fetal growth restriction (FGR) corresponds to the fetus's inability to achieve an adequate weight gain based on genetic potential and gestational age. It is an important cause of morbidity and mortality. SUMMARY In this review, we address the challenges of diagnosis and classification of FGR. We review how chronic fetal hypoxia impacts brain development. We describe recent advances on placental and fetal brain imaging using magnetic resonance imaging and how they offer new noninvasive means to study growth restriction in humans. We go on to review the impact of FGR on brain integrity in the neonatal period, later childhood, and adulthood and review available therapies. KEY MESSAGES FGR consequences are not limited to the perinatal period. We hypothesize that impaired brain reserve, as defined by structure and size, may predict some concerning epidemiological data of impaired cognitive outcomes and dementia with aging in this group of patients.
Collapse
Affiliation(s)
- Divyen K. Shah
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Neonatal Intensive Care, Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Susana Pereira
- Obstetrics and Maternity Care, Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Gregory A. Lodygensky
- Department of Pediatrics, University of Montréal, Montréal, QC, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
5
|
Drobyshevsky A, Synowiec S, Goussakov I, Fabres R, Lu J, Caplan M. Intestinal microbiota modulates neuroinflammatory response and brain injury after neonatal hypoxia-ischemia. Gut Microbes 2024; 16:2333808. [PMID: 38533575 PMCID: PMC10978030 DOI: 10.1080/19490976.2024.2333808] [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: 08/10/2023] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
Premature infants lack a normal intestinal microbial community and also at risk of perinatal hypoxic-ischemic (HI) brain injury, which is considered to be one of the major factors for motor, sensory, and cognitive deficits. We hypothesized that neonatal gut microbiota composition modulated the immune reaction and severity of neonatal H-I brain injury. Neonatal C57BL/6J mouse pups were exposed to H-I protocol consisting of permanent left carotid artery ligation, followed by 8% hypoxia for 60 min. Microbial manipulation groups included 1) antibiotic treatment, E18 (maternal) to P5; 2) antibiotic treatment E18 to P5 + E. coli gavage; 3) antibiotic treatment E18 to P5 + B. infantis gavage; and 4) saline to pups with dams getting fresh water. The extent of brain injury and recovery was measured on MRI. Edematous injury volume was significantly higher in E. coli group than that in B. infantis group and in fresh water group. Gene expression in brains of pro-inflammatory cytokines (IL1β, IL6, IL2, TNF-α and toll-like receptors 2-6) were elevated to a greater extent in the E. coli group at P10, no injury, and at P13, 72 hours after H-I relative to sham control and B. infantis groups. Significant effects of microbiome and brain injury and interaction of these factors were found in abundance of major phyla. The neuroinflammatory response and brain injury after neonatal hypoxia-ischemia are affected by intestinal microbiota, providing opportunities for therapeutic intervention through targeting the early colonization and development of the gut microbiota.
Collapse
Affiliation(s)
| | - Sylvia Synowiec
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Ivan Goussakov
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Rafael Fabres
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Jing Lu
- Department of Pediatrics, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Michael Caplan
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, USA
| |
Collapse
|
6
|
Farias-Jofre M, Romero R, Galaz J, Xu Y, Miller D, Garcia-Flores V, Arenas-Hernandez M, Winters AD, Berkowitz BA, Podolsky RH, Shen Y, Kanninen T, Panaitescu B, Glazier CR, Pique-Regi R, Theis KR, Gomez-Lopez N. Blockade of IL-6R prevents preterm birth and adverse neonatal outcomes. EBioMedicine 2023; 98:104865. [PMID: 37944273 PMCID: PMC10665693 DOI: 10.1016/j.ebiom.2023.104865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Preterm birth preceded by spontaneous preterm labour often occurs in the clinical setting of sterile intra-amniotic inflammation (SIAI), a condition that currently lacks treatment. METHODS Proteomic and scRNA-seq human data were analysed to evaluate the role of IL-6 and IL-1α in SIAI. A C57BL/6 murine model of SIAI-induced preterm birth was developed by the ultrasound-guided intra-amniotic injection of IL-1α. The blockade of IL-6R by using an aIL-6R was tested as prenatal treatment for preterm birth and adverse neonatal outcomes. QUEST-MRI evaluated brain oxidative stress in utero. Targeted transcriptomic profiling assessed maternal, foetal, and neonatal inflammation. Neonatal biometrics and neurodevelopment were tested. The neonatal gut immune-microbiome was evaluated using metagenomic sequencing and immunophenotyping. FINDINGS IL-6 plays a critical role in the human intra-amniotic inflammatory response, which is associated with elevated concentrations of the alarmin IL-1α. Intra-amniotic injection of IL-1α resembles SIAI, inducing preterm birth (7% vs. 50%, p = 0.03, Fisher's exact test) and neonatal mortality (18% vs. 56%, p = 0.02, Mann-Whitney U-test). QUEST-MRI revealed no foetal brain oxidative stress upon in utero IL-1α exposure (p > 0.05, mixed linear model). Prenatal treatment with aIL-6R abrogated IL-1α-induced preterm birth (50% vs. 7%, p = 0.03, Fisher's exact test) by dampening inflammatory processes associated with the common pathway of labour. Importantly, aIL-6R reduces neonatal mortality (56% vs. 22%, p = 0.03, Mann-Whitney U-test) by crossing from the mother to the amniotic cavity, dampening foetal organ inflammation and improving growth. Beneficial effects of prenatal IL-6R blockade carried over to neonatal life, improving survival, growth, neurodevelopment, and gut immune homeostasis. INTERPRETATION IL-6R blockade can serve as a strategy to treat SIAI, preventing preterm birth and adverse neonatal outcomes. FUNDING NICHD/NIH/DHHS, Contract HHSN275201300006C. WSU Perinatal Initiative in Maternal, Perinatal and Child Health.
Collapse
Affiliation(s)
- Marcelo Farias-Jofre
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, 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.
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Andrew D Winters
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MO, USA
| | - Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine; Detroit, MI, USA
| | - Robert H Podolsky
- Division of Biostatistics and Design Methodology, Center for Translational Research, Children's National Hospital, Silver Spring, MD, USA
| | - Yimin Shen
- Department of Radiology, School of Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tomi Kanninen
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Panaitescu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Catherine R Glazier
- UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Roger Pique-Regi
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kevin R Theis
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MO, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MO, USA; Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Rojas DB, Vizuete AFK, de Andrade VS, de Andrade RB, Gemelli T, Kim TDH, Gonçalves CA, Leipnitz G, Wannmacher CMD. Lipopolysaccharide impairs neurodevelopment and induces changes in astroglial reactivity, antioxidant defenses and bioenergetics in the cerebral cortex of neonatal rats. Int J Dev Neurosci 2023; 83:600-614. [PMID: 37477051 DOI: 10.1002/jdn.10288] [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: 03/31/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/22/2023] Open
Abstract
Neonates have an immature immune system, which increases their vulnerability to infectious agents and inflammatory insults. The administration of the immunostimulatory agent lipopolysaccharide (LPS) has been shown to induce the expression of pro-inflammatory cytokines and cause behavior alterations in rodents at different ages. However, the effects of LPS administration during the neonatal period and its consequences during immune system maturation remain to be elucidated. We showed here that a single intraperitoneal administration of LPS in rats on postnatal day (PND) 7 caused early and variable alterations in TNF-α, S100B and GFAP levels in the cerebral cortex, CSF and serum of the animals, indicating long-term induction of neuroinflammation and astroglial reactivity. However, on PND 21, only GFAP levels were increased by LPS. Additionally, LPS induced oxidative stress and altered energy metabolism enzymes in the cerebral cortex on PND 21, and caused neurodevelopment impairment over time. These data suggest that neuroinflammation induction during the neonatal period induces glial reactivity, oxidative stress and bioenergetic disruption that may lead to neurodevelopment impairment and cognitive deficit in adult life.
Collapse
Affiliation(s)
- Denise Bertin Rojas
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Adriana Fernanda K Vizuete
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Vivian Strassburger de Andrade
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Tanise Gemelli
- Universidade do Vale do Rio dos Sinos, São Leopoldo, Rio Grande do Sul, Brazil
| | - Tomas Duk Hwa Kim
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos Alberto Gonçalves
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
- Departmento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Departmento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Clovis Milton Duval Wannmacher
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Departmento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
8
|
Gervasi MT, Romero R, Cainelli E, Veronese P, Tran MR, Jung E, Suksai M, Bosco M, Gotsch F. Intra-amniotic inflammation in the mid-trimester of pregnancy is a risk factor for neuropsychological disorders in childhood. J Perinat Med 2023; 51:363-378. [PMID: 36173676 PMCID: PMC10010737 DOI: 10.1515/jpm-2022-0255] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/17/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Intra-amniotic inflammation is a subclinical condition frequently caused by either microbial invasion of the amniotic cavity or sterile inflammatory stimuli, e.g., alarmins. An accumulating body of evidence supports a role for maternal immune activation in the genesis of fetal neuroinflammation and the occurrence of neurodevelopmental disorders such as cerebral palsy, schizophrenia, and autism. The objective of this study was to determine whether fetal exposure to mid-trimester intra-amniotic inflammation is associated with neurodevelopmental disorders in children eight to 12 years of age. METHODS This is a retrospective case-control study comprising 20 children with evidence of prenatal exposure to intra-amniotic inflammation in the mid-trimester and 20 controls matched for gestational age at amniocentesis and at delivery. Amniotic fluid samples were tested for concentrations of interleukin-6 and C-X-C motif chemokine ligand 10, for bacteria by culture and molecular microbiologic methods as well as by polymerase chain reaction for eight viruses. Neuropsychological testing of children, performed by two experienced psychologists, assessed cognitive and behavioral domains. Neuropsychological dysfunction was defined as the presence of an abnormal score (<2 standard deviations) on at least two cognitive tasks. RESULTS Neuropsychological dysfunction was present in 45% (9/20) of children exposed to intra-amniotic inflammation but in only 10% (2/20) of those in the control group (p=0.03). The relative risk (RR) of neuropsychological dysfunction conferred by amniotic fluid inflammation remained significant after adjusting for gestational age at delivery [aRR=4.5 (1.07-16.7)]. Of the 11 children diagnosed with neuropsychological dysfunction, nine were delivered at term and eight of them had mothers with intra-amniotic inflammation. Children exposed to intra-amniotic inflammation were found to have abnormalities in neuropsychological tasks evaluating complex skills, e.g., auditory attention, executive functions, and social skills, whereas the domains of reasoning, language, and memory were not affected in the cases and controls. CONCLUSIONS Asymptomatic sterile intra-amniotic inflammation in the mid-trimester of pregnancy, followed by a term birth, can still confer to the offspring a substantial risk for neurodevelopmental disorders in childhood. Early recognition and treatment of maternal immune activation in pregnancy may be a strategy for the prevention of subsequent neurodevelopmental disorders in offspring.
Collapse
Affiliation(s)
- Maria Teresa Gervasi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Gynaecology and Obstetrics Unit, Department of Women’s and Children’s Health, University Hospital of Padua, Padua, Italy
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, 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
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, MI, USA
| | - Elisa Cainelli
- Department of General Psychology, University of Padova, Padova, Italy
| | - Paola Veronese
- Maternal-Fetal Medicine Unit, Department of Women’s and Children’s Health, AOPD, Padua, Italy
| | - Maria Rosa Tran
- Gynaecology and Obstetrics Unit, Department of Women’s and Children’s Health, University Hospital of Padua, Padua, Italy
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
9
|
Sex-specific inflammatory and white matter effects of prenatal opioid exposure: a pilot study. Pediatr Res 2023; 93:604-611. [PMID: 36280708 PMCID: PMC9998341 DOI: 10.1038/s41390-022-02357-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 09/01/2022] [Accepted: 10/11/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Preclinical data demonstrate that opioids modulate brain reward signaling through an inflammatory cascade, but this relationship has yet to be studied in opioid-exposed neonates. METHODS Saliva samples of 54 opioid-exposed and sex- and age-matched non-exposed neonates underwent transcriptomic analysis of inflammatory and reward genes. A subset of 22 neonates underwent brain magnetic resonance imaging (MRI) to evaluate white matter injury commonly associated with inflammatory response. Gene expression and brain MRI were compared between opioid- and non-exposed neonates and further stratified by sex and pharmacotherapy need. RESULTS Opioid-exposed females regardless of pharmacotherapy need had higher expression of inflammatory genes than their male counterparts, with notable differences in the expression of CCL2 and CXCL1 in females requiring pharmacotherapy (p = 0.01 and 0.06, respectively). Opioid-exposed males requiring pharmacotherapy had higher expression of DRD2 than exposed females (p = 0.07), validating our prior research. Higher expression of IL1β, IL6, TNFα, and IL10 was seen in opioid-exposed neonates with T1 white matter hyperintensity (WMH) compared to exposed neonates without WMH (p < 0.05). CONCLUSION Prenatal opioid exposure may promote inflammation resulting in changes in reward signaling and white matter injury in the developing brain, with unique sex-specific effects. The actions of opioids through non-neuronal pathways need further investigation. IMPACT Opioid-exposed neonates are at risk for punctate T1 white matter hyperintensity (WMH). Females carry a greater propensity for WMH. Salivary transcriptomic data showed significantly higher expression of inflammatory genes in opioid-exposed neonates with WMH than those without WMH, irrespective of pharmacotherapy need. Adding to prior studies, our findings suggest that prenatal opioid exposure may modulate white matter injury and reward signaling through a pro-inflammatory process that is sex specific. This novel study highlights the short-term molecular and structural effects of prenatal opioids and the need to elucidate the long-term impact of prenatal opioid exposure.
Collapse
|
10
|
Lu J, Martin CR, Claud EC. Neurodevelopmental outcome of infants who develop necrotizing enterocolitis: The gut-brain axis. Semin Perinatol 2023; 47:151694. [PMID: 36572620 PMCID: PMC9974904 DOI: 10.1016/j.semperi.2022.151694] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Necrotizing enterocolitis (NEC) poses a significant risk for neurodevelopmental impairment in extremely preterm infants. The gut microbiota shapes the development of the gut, immune system, and the brain; and dysbiosis drive neonatal morbidities including NEC. In this chapter, we delineate a gut-brain axis linking gut microbiota to the adverse neurological outcomes in NEC patients. We propose that in NEC, immaturity of the microbiome along with aberrant gut microbiota-driven immaturity of the gut barrier and immune system can lead to effects including systemic inflammation and circulating microbial mediators. This nexus of gut microbiota-driven systemic effects further interacts with a likewise underdeveloped blood-brain barrier to regulate neuroinflammation and neurodevelopment. Targeting deviant gut-brain axis signaling presents an opportunity to improve the neurodevelopmental outcomes of NEC patients.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pediatrics, Division of Biological Sciences, University of Chicago, Pritzker School of Medicine, Chicago, Illinois 60637, United States
| | - Camilia R Martin
- Department of Pediatrics, Division of Newborn Medicine, Weill Cornell Medicine, New York, New York 10021, United States
| | - Erika C Claud
- Department of Pediatrics, Division of Biological Sciences, University of Chicago, Pritzker School of Medicine, Chicago, Illinois 60637, United States.
| |
Collapse
|
11
|
Li Q, Shen J, Lv H, Liu Y, Chen Y, Zhou C, Shi J. Incidence, risk factors, and outcomes in electroencephalographic seizures after mechanical circulatory support: A systematic review and meta-analysis. Front Cardiovasc Med 2022; 9:872005. [PMID: 35990978 PMCID: PMC9381842 DOI: 10.3389/fcvm.2022.872005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeTo estimate the overall incidence, risk factors, and clinical outcomes of electroencephalographic (EEG) seizures for adults and children after mechanical circulatory support (MCS).Method and measurementsThis systematic review and meta-analysis were carried out in accordance with the PRISMA (Preferred Reporting Items for Systematic Review and Meta-Analysis) guidance document. MEDLINE EMBASE and CENTRAL were investigated for relevant studies. The related information was retrieved by two independent reviewers and all analyses were conducted by STATA (version 16.0; Stata Corporation, College Station, TX, United States).ResultSixty studies including 36,191 adult and 55,475 pediatric patients with MCS were enrolled for evaluation. The study showed that the overall incidence of EEG seizures in adults was 2% (95%CI: 1–3%), in which 1% (95%CI: 1–2%) after cardiopulmonary bypass (CPB), and 3% (95%CI: 1–6%) after extracorporeal membrane oxygenation (ECMO). For pediatrics patients, the incidence of EEG seizures was 12% (95%CI: 11–14%), among which 12% (9–15%) after CPB and 13% (11–15%) after ECMO. The major risk factors of EEG seizures after MCS in adults were redo surgery (coefficient = 0.0436, p = 0.044), and COPD (coefficient = 0.0749, p = 0.069). In addition, the gestational week of CPB (coefficient = 0.0544, p = 0.080) and respiratory failure of ECMO (coefficient = –0.262, p = 0.019) were also indicated to be associated with EEG seizures in pediatrics.ConclusionEEG seizures after MCS were more common in pediatrics than in adults. In addition, the incidence of EEG seizure after ECMO was higher than CPB both in adults and children. It is expected that appropriate measures should be taken to control modifiable risk factors, thus improving the prognosis and increasing the long-term survival rate of MCS patients.Systematic Review Registration[https://www.crd.york.ac.uk/prospero], identifier [CRD42021287288].
Collapse
|
12
|
Pierre WC, Zhang E, Londono I, De Leener B, Lesage F, Lodygensky GA. Non-invasive in vivo MRI detects long-term microstructural brain alterations related to learning and memory impairments in a model of inflammation-induced white matter injury. Behav Brain Res 2022; 428:113884. [DOI: 10.1016/j.bbr.2022.113884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/18/2022] [Accepted: 04/03/2022] [Indexed: 11/28/2022]
|
13
|
Pierre WC, Londono I, Quiniou C, Chemtob S, Lodygensky GA. Modulatory effect of IL‐1 inhibition following lipopolysaccharide‐induced neuroinflammation in neonatal microglia and astrocytes. Int J Dev Neurosci 2022; 82:243-260. [DOI: 10.1002/jdn.10179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 02/23/2022] [Accepted: 03/17/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Wyston C. Pierre
- Sainte‐Justine Hospital and Research Center, Department of Pediatrics Université de Montréal Montréal, Québec Canada
- Department of Pharmacology and Physiology Université de Montréal Montréal Canada
| | - Irène Londono
- Sainte‐Justine Hospital and Research Center, Department of Pediatrics Université de Montréal Montréal, Québec Canada
| | - Christiane Quiniou
- Sainte‐Justine Hospital and Research Center, Department of Pediatrics Université de Montréal Montréal, Québec Canada
| | - Sylvain Chemtob
- Sainte‐Justine Hospital and Research Center, Department of Pediatrics Université de Montréal Montréal, Québec Canada
- Department of Pharmacology and Physiology Université de Montréal Montréal Canada
- Department of Pharmacology and Therapeutics McGill University Montréal Canada
| | - Gregory A. Lodygensky
- Sainte‐Justine Hospital and Research Center, Department of Pediatrics Université de Montréal Montréal, Québec Canada
- Department of Pharmacology and Physiology Université de Montréal Montréal Canada
| |
Collapse
|
14
|
He L, Wang C, Simujide H, Aricha H, Zhang J, Liu B, Zhang C, Cui Y, Aorigele C. Effect of Early Pathogenic Escherichia coli Infection on the Intestinal Barrier and Immune Function in Newborn Calves. Front Cell Infect Microbiol 2022; 12:818276. [PMID: 35265533 PMCID: PMC8900010 DOI: 10.3389/fcimb.2022.818276] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/04/2022] [Indexed: 11/30/2022] Open
Abstract
We studied the effect of early pathogenic Escherichia coli infection on newborn calves’ intestinal barrier and immune function. A total of 64 newborn Holstein male calves (40–43 kg) were divided into two groups: normal (NG) and test (TG), each with 32 heads. At the beginning of the experiment, the TG calves were orally administered pathogenic E. coli O1 (2.5 × 1011 CFU/mL, 100 mL) to establish a calf diarrhea model. In contrast, the NG calves were given the same amount of normal saline. During the 30 d trial period, the feeding and management of the two groups remained constant. Enzyme-linked immunosorbent assay, quantification PCR, and high-throughput 16S rRNA sequencing technology were used to detect indicators related to the intestinal barrier and immune function in the calf serum and tissues. Pathogenic E. coli O1 had a significant effect on calf diarrhea in the TG; it increased the bovine diamine oxidase (P < 0.05) and endotoxin levels in the serum and decreased (P < 0.05) the intestinal trefoil factor (P < 0.05), Occludin, Claudin-1, and Zonula Occludens 1 (ZO-1) levels in the colon tissue, as well as downregulated the mRNA expression of Occludin, Claudin-1,and ZO-1 in the colon mucosa, leading to increased intestinal permeability and impaired intestinal barrier function. Additionally, pathogenic E. coli had a significant impact on the diversity of colonic microbial flora, increasing the relative abundance of Proteobacteria at the phylum level and decreasing the levels of Firmicutes and Bacteroides. At the genus level, the relative abundance of Escherichia and Shigella in the TG increased significantly (P < 0.05), whereas that of Bacteroides, Butyricicoccus, Rikenellaceae_RC9_gut_group, Blautia, and Lactobacillus was significantly decreased (P < 0.05). In addition, the level of IL-6 in the serum of the TG calves was significantly increased (P < 0.05), whereas the IL-4 and IL-10 levels were significantly decreased (P < 0.05), compared to those in the NG calves. Thus, pathogenic E. coli induced diarrhea early in life disrupts intestinal barrier and impairs immune function in calves.
Collapse
Affiliation(s)
- Lina He
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Chunjie Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Huasai Simujide
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Han Aricha
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Jian Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Bo Liu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Chen Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yinxue Cui
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Chen Aorigele
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
- *Correspondence: Chen Aorigele,
| |
Collapse
|
15
|
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, SWITZERLAND) 2021; 8:917. [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
| | - 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
|
16
|
Neonates Have a Lot More at Stake When It Comes to Infections. Clin Perinatol 2021; 48:xv-xvii. [PMID: 34030824 PMCID: PMC9754888 DOI: 10.1016/j.clp.2021.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Ali A, Zambrano R, Duncan MR, Chen S, Luo S, Yuan H, Chen P, Benny M, Schmidt A, Young K, Kerr N, de Rivero Vaccari JP, Keane RW, Dietrich WD, Wu S. Hyperoxia-activated circulating extracellular vesicles induce lung and brain injury in neonatal rats. Sci Rep 2021; 11:8791. [PMID: 33888735 PMCID: PMC8062626 DOI: 10.1038/s41598-021-87706-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/16/2021] [Indexed: 01/02/2023] Open
Abstract
Hyperoxia-induced lung injury plays a key role in the development of bronchopulmonary dysplasia (BPD), characterized by inflammatory injury and impaired lung development in preterm infants. Although BPD is a predictor of poor neurodevelopmental outcomes, currently it is uncertain how lung injury contributes to brain injury in preterm infants. Extracellular vesicles (EVs) are a heterogeneous group of cell-derived membranous structures that regulate intercellular and inter-organ communications. Gasdermin D (GSDMD) has emerged as a key executor of inflammasome-mediated cell death and inflammation. In this study, we utilized a neonatal rat model of BPD to assess if hyperoxia stimulates lung release of circulating EVs and if these EVs induce lung and brain injury. We found that hyperoxia-exposed rats had elevated numbers of plasma-derived EVs compared to rats maintained in room air. These EVs also had increased cargos of surfactant protein C, a marker of type II alveolar epithelial cells (AEC), and the active (p30) form of GSDMD. When these EVs were adoptively transferred into normal newborn rats via intravenous injection, they were taken up both by lung and brain tissues. Moreover, EVs from hyperoxic animals induced not only the pathological hallmarks of BPD, but also brain inflammatory injury in recipient rats, as well as inducing cell death in cultured pulmonary vascular endothelial cells and neural stem cells (NSC). Similarly, hyperoxia-exposed cultured AEC-like cells released EVs that also contained increased GSDMD-p30 and these EVs induced pyroptotic cell death in NSC. Overall, these data indicate that hyperoxia-activated circulating EVs mediate a lung to brain crosstalk resulting in brain injury and suggest a mechanism that links lung injury and neurodevelopmental impairment in BPD infants.
Collapse
Affiliation(s)
- Anum Ali
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Ronald Zambrano
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Matthew R Duncan
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Shaoyi Chen
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Shihua Luo
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Huijun Yuan
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Pingping Chen
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Merline Benny
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Augusto Schmidt
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Karen Young
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Nadine Kerr
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert W Keane
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - Shu Wu
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA.
| |
Collapse
|
18
|
Ophelders DR, Gussenhoven R, Klein L, Jellema RK, Westerlaken RJ, Hütten MC, Vermeulen J, Wassink G, Gunn AJ, Wolfs TG. Preterm Brain Injury, Antenatal Triggers, and Therapeutics: Timing Is Key. Cells 2020; 9:E1871. [PMID: 32785181 PMCID: PMC7464163 DOI: 10.3390/cells9081871] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 02/08/2023] Open
Abstract
With a worldwide incidence of 15 million cases, preterm birth is a major contributor to neonatal mortality and morbidity, and concomitant social and economic burden Preterm infants are predisposed to life-long neurological disorders due to the immaturity of the brain. The risks are inversely proportional to maturity at birth. In the majority of extremely preterm infants (<28 weeks' gestation), perinatal brain injury is associated with exposure to multiple inflammatory perinatal triggers that include antenatal infection (i.e., chorioamnionitis), hypoxia-ischemia, and various postnatal injurious triggers (i.e., oxidative stress, sepsis, mechanical ventilation, hemodynamic instability). These perinatal insults cause a self-perpetuating cascade of peripheral and cerebral inflammation that plays a critical role in the etiology of diffuse white and grey matter injuries that underlies a spectrum of connectivity deficits in survivors from extremely preterm birth. This review focuses on chorioamnionitis and hypoxia-ischemia, which are two important antenatal risk factors for preterm brain injury, and highlights the latest insights on its pathophysiology, potential treatment, and future perspectives to narrow the translational gap between preclinical research and clinical applications.
Collapse
Affiliation(s)
- Daan R.M.G. Ophelders
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ruth Gussenhoven
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
| | - Luise Klein
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Mental Health and Neuroscience (MHeNS), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Reint K. Jellema
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
| | - Rob J.J. Westerlaken
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Matthias C. Hütten
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Jeroen Vermeulen
- Department of Pediatric Neurology, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands;
| | - Guido Wassink
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland 1023, New Zealand; (G.W.); (A.J.G.)
| | - Alistair J. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Private bag 92019, Auckland 1023, New Zealand; (G.W.); (A.J.G.)
| | - Tim G.A.M. Wolfs
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands; (D.R.M.G.O.); (R.G.); (L.K.); (R.K.J.); (R.J.J.W.); (M.C.H.)
- School for Oncology and Developmental Biology (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
19
|
Abstract
Cerebral palsy (CP), defined as a group of nonprogressive disorders of movement and posture, is the most common cause of severe neurodisability in children. The prevalence of CP is the same across the globe, affecting approximately 17 million people worldwide. Cerebral Palsy is an umbrella term used to describe the disease due to its inherent heterogeneity. For instance, CP has multiple (1) causes; (2) clinical types; (3) patterns of neuropathology on brain imaging and (4) it's associated with several developmental pathologies such as intellectual disability, autism, epilepsy, and visual impairment. Understanding its physiopathology is crucial to developing protective strategies. Despite its importance, there is still insufficient progress in the areas of CP prediction, early diagnosis, treatment, and prevention. Herein we describe the current risk factors and biomarkers used for the diagnosis and prediction of CP. With the advancement in biomarker discovery, we predict that our understanding of the etiopathophysiology of CP will also increase, lending to more opportunities for developing novel treatments and prognosis.
Collapse
Affiliation(s)
- Zeynep Alpay Savasan
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Division, Beaumont Health System, Royal Oak, MI, United States; Oakland University-William Beaumont School of Medicine, Beaumont Health, Royal Oak, MI, United States.
| | - Sun Kwon Kim
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Division, Beaumont Health System, Royal Oak, MI, United States; Oakland University-William Beaumont School of Medicine, Beaumont Health, Royal Oak, MI, United States
| | - Kyung Joon Oh
- Beaumont Research Institute, Beaumont Health, Royal Oak, MI, United States; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, South Korea
| | - Stewart F Graham
- Oakland University-William Beaumont School of Medicine, Beaumont Health, Royal Oak, MI, United States; Beaumont Research Institute, Beaumont Health, Royal Oak, MI, United States
| |
Collapse
|
20
|
Pierre WC, Akakpo L, Londono I, Pouliot P, Chemtob S, Lesage F, Lodygensky GA. Assessing therapeutic response non-invasively in a neonatal rat model of acute inflammatory white matter injury using high-field MRI. Brain Behav Immun 2019; 81:348-360. [PMID: 31247289 DOI: 10.1016/j.bbi.2019.06.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 05/20/2019] [Accepted: 06/22/2019] [Indexed: 12/19/2022] Open
Abstract
Perinatal infection and inflammatory episodes in preterm infants are associated with diffuse white matter injury (WMI) and adverse neurological outcomes. Inflammation-induced WMI was previously shown to be linked with later hippocampal atrophy as well as learning and memory impairments in preterm infants. Early evaluation of injury load and therapeutic response with non-invasive tools such as multimodal magnetic resonance imaging (MRI) would greatly improve the search of new therapeutic approaches in preterm infants. Our aim was to evaluate the potential of multimodal MRI to detect the response of interleukin-1 receptor antagonist (IL-1Ra) treatment, known for its neuroprotective properties, during the acute phase of injury on a model of neonatal WMI. Rat pups at postnatal day 3 (P3) received intracerebral injection of lipopolysaccharide with systemic IL-1Ra therapy. 24 h later (P4), rats were imaged with multimodal MRI to assess microstructure by diffusion tensor imaging (DTI) and neurochemical profile of the hippocampus with 1H-magnetic resonance spectroscopy. Astrocyte and microglial activation, apoptosis and the mRNA expression of pro-inflammatory and necroptotic markers were assessed. During the acute phase of injury, neonatal LPS exposure altered the concentration of hippocampus metabolites related to neuronal integrity, neurotransmission and membrane integrity and induced diffusivity restriction. Just 24 h after initiation of therapy, early indication of IL-1Ra neuroprotective effect could be detected in vivo by non-invasive spectroscopy and DTI, and confirmed with immunohistochemical evaluation and mRNA expression of inflammatory markers and cell death. In conclusion, multimodal MRI, particularly DTI, can detect not only injury but also the acute therapeutic effect of IL-1Ra suggesting that MRI could be a useful non-invasive tool to follow, at early time points, the therapeutic response in preterm infants.
Collapse
Affiliation(s)
- Wyston C Pierre
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada; Department of Pharmacology, Université de Montréal, Montréal, Canada
| | - Luis Akakpo
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada; École Polytechnique de Montréal, Montreal, QC, Canada
| | - Irène Londono
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Philippe Pouliot
- École Polytechnique de Montréal, Montreal, QC, Canada; Montreal Heart Institute, Montreal, QC, Canada
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada; Department of Pharmacology, Université de Montréal, Montréal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Frédéric Lesage
- École Polytechnique de Montréal, Montreal, QC, Canada; Montreal Heart Institute, Montreal, QC, Canada
| | - Gregory A Lodygensky
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada; Department of Pharmacology, Université de Montréal, Montréal, Canada; Montreal Heart Institute, Montreal, QC, Canada.
| |
Collapse
|
21
|
Zhang L, Wang H. Long Non-coding RNA in CNS Injuries: A New Target for Therapeutic Intervention. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 17:754-766. [PMID: 31437654 PMCID: PMC6709344 DOI: 10.1016/j.omtn.2019.07.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 07/17/2019] [Accepted: 07/17/2019] [Indexed: 12/15/2022]
Abstract
CNS injuries, such as traumatic brain injury (TBI), subarachnoid hemorrhage (SAH), intracerebral hemorrhage (ICH), and cerebral ischemic stroke, are important causes of death and long-term disability worldwide. As an important class of pervasive genes involved in many pathophysiological processes, long non-coding RNAs (lncRNAs) have received attention in the past decades. Multiple studies indicate that lncRNAs are abundant in the CNS and have a key role in brain function as well as many neurological disorders, especially in CNS injuries. Several investigations have deciphered that regulation of lncRNAs exert pro-angiogenesis, anti-apoptosis, and anti-inflammation effects in CNS injury via different molecules and pathways, including microRNA (miRNA), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphatidylinositol-4,5-bisphosphate 3-kinase/protein kinase B (PI3K/AKT), Notch, and p53. Thus, lncRNAs show great promise as molecular targets in CNS injuries. In this article, we provide an updated review of the current state of our knowledge about the relationship between lncRNAs and CNS injuries, highlighting the specific roles of lncRNAs in CNS injuries.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
22
|
Vaes JEG, Vink MA, de Theije CGM, Hoebeek FE, Benders MJNL, Nijboer CHA. The Potential of Stem Cell Therapy to Repair White Matter Injury in Preterm Infants: Lessons Learned From Experimental Models. Front Physiol 2019; 10:540. [PMID: 31143126 PMCID: PMC6521595 DOI: 10.3389/fphys.2019.00540] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Diffuse white matter injury (dWMI) is a major cause of morbidity in the extremely preterm born infant leading to life-long neurological impairments, including deficits in cognitive, motor, sensory, psychological, and behavioral functioning. At present, no treatment options are clinically available to combat dWMI and therefore exploration of novel strategies is urgently needed. In recent years, the pathophysiology underlying dWMI has slowly started to be unraveled, pointing towards the disturbed maturation of oligodendrocytes (OLs) as a key mechanism. Immature OL precursor cells in the developing brain are believed to be highly sensitive to perinatal inflammation and cerebral oxygen fluctuations, leading to impaired OL differentiation and eventually myelination failure. OL lineage development under normal and pathological circumstances and the process of (re)myelination have been studied extensively over the years, often in the context of other adult and pediatric white matter pathologies such as stroke and multiple sclerosis (MS). Various studies have proposed stem cell-based therapeutic strategies to boost white matter regeneration as a potential strategy against a wide range of neurological diseases. In this review we will discuss experimental studies focusing on mesenchymal stem cell (MSC) therapy to reduce white matter injury (WMI) in multiple adult and neonatal neurological diseases. What lessons have been learned from these previous studies and how can we translate this knowledge to application of MSCs for the injured white matter in the preterm infant? A perspective on the current state of stem cell therapy will be given and we will discuss different important considerations of MSCs including cellular sources, timing of treatment and administration routes. Furthermore, we reflect on optimization strategies that could potentially reinforce stem cell therapy, including preconditioning and genetic engineering of stem cells or using cell-free stem cell products, to optimize cell-based strategy for vulnerable preterm infants in the near future.
Collapse
Affiliation(s)
- Josine E G Vaes
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Marit A Vink
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Caroline G M de Theije
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Freek E Hoebeek
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Cora H A Nijboer
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
23
|
Alpay Savasan Z, Yilmaz A, Ugur Z, Aydas B, Bahado-Singh RO, Graham SF. Metabolomic Profiling of Cerebral Palsy Brain Tissue Reveals Novel Central Biomarkers and Biochemical Pathways Associated with the Disease: A Pilot Study. Metabolites 2019; 9:metabo9020027. [PMID: 30717353 PMCID: PMC6409919 DOI: 10.3390/metabo9020027] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 12/17/2022] Open
Abstract
Cerebral palsy (CP) is one of the most common causes of motor disability in childhood, with complex and heterogeneous etiopathophysiology and clinical presentation. Understanding the metabolic processes associated with the disease may aid in the discovery of preventive measures and therapy. Tissue samples (caudate nucleus) were obtained from post-mortem CP cases (n = 9) and age- and gender-matched control subjects (n = 11). We employed a targeted metabolomics approach using both 1H NMR and direct injection liquid chromatography-tandem mass spectrometry (DI/LC-MS/MS). We accurately identified and quantified 55 metabolites using 1H NMR and 186 using DI/LC-MS/MS. Among the 222 detected metabolites, 27 showed significant concentration changes between CP cases and controls. Glycerophospholipids and urea were the most commonly selected metabolites used to develop predictive models capable of discriminating between CP and controls. Metabolomics enrichment analysis identified folate, propanoate, and androgen/estrogen metabolism as the top three significantly perturbed pathways. We report for the first time the metabolomic profiling of post-mortem brain tissue from patients who died from cerebral palsy. These findings could help to further investigate the complex etiopathophysiology of CP while identifying predictive, central biomarkers of CP.
Collapse
Affiliation(s)
- Zeynep Alpay Savasan
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Division, Beaumont Health System, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
- Oakland University-William Beaumont School of Medicine, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
| | - Ali Yilmaz
- Beaumont Research Institute, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
| | - Zafer Ugur
- Beaumont Research Institute, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
| | - Buket Aydas
- Departments of Mathematics and Computer Sciences, Albion College, 611 E. Porter St., Albion, MI 49224, USA.
| | - Ray O Bahado-Singh
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Division, Beaumont Health System, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
- Oakland University-William Beaumont School of Medicine, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
| | - Stewart F Graham
- Oakland University-William Beaumont School of Medicine, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
- Beaumont Research Institute, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
| |
Collapse
|
24
|
Finch-Edmondson M, Morgan C, Hunt RW, Novak I. Emergent Prophylactic, Reparative and Restorative Brain Interventions for Infants Born Preterm With Cerebral Palsy. Front Physiol 2019; 10:15. [PMID: 30745876 PMCID: PMC6360173 DOI: 10.3389/fphys.2019.00015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/08/2019] [Indexed: 12/13/2022] Open
Abstract
Worldwide, an estimated 15 million babies are born preterm (<37 weeks' gestation) every year. Despite significant improvements in survival rates, preterm infants often face a lifetime of neurodevelopmental disability including cognitive, behavioral, and motor impairments. Indeed, prematurity remains the largest risk factor for the development of cerebral palsy. The developing brain of the preterm infant is particularly fragile; preterm babies exhibit varying severities of cerebral palsy arising from reductions in both cerebral white and gray matter volumes, as well as altered brain microstructure and connectivity. Current intensive care therapies aim to optimize cardiovascular and respiratory function to protect the brain from injury by preserving oxygenation and blood flow. If a brain injury does occur, definitive diagnosis of cerebral palsy in the first few hours and weeks of life is difficult, especially when the lesions are subtle and not apparent on cranial ultrasound. However, early diagnosis of mildly affected infants is critical, because these are the patients most likely to respond to emergent treatments inducing neuroplasticity via high-intensity motor training programs and regenerative therapies involving stem cells. A current controversy is whether to test universal treatment in all infants at risk of brain injury, accepting that some patients never required treatment, because the perceived potential benefits outweigh the risk of harm. Versus, waiting for a diagnosis before commencing targeted treatment for infants with a brain injury, and potentially missing the therapeutic window. In this review, we discuss the emerging prophylactic, reparative, and restorative brain interventions for infants born preterm, who are at high risk of developing cerebral palsy. We examine the current evidence, considering the timing of the intervention with relation to the proposed mechanism/s of action. Finally, we consider the development of novel markers of preterm brain injury, which will undoubtedly lead to improved diagnostic and prognostic capability, and more accurate instruments to assess the efficacy of emerging interventions for this most vulnerable group of infants.
Collapse
Affiliation(s)
- Megan Finch-Edmondson
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Catherine Morgan
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Rod W. Hunt
- Department of Neonatal Medicine, The Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
- Neonatal Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Iona Novak
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
25
|
Agrawal S, Rao S, Nathan EA, Patole S. Effect of probiotics on C-reactive protein levels in preterm infants: Secondary analysis of a randomized controlled trial. J Neonatal Perinatal Med 2018; 11:165-171. [PMID: 29843270 DOI: 10.3233/npm-181763] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Excessive inflammation is associated with adverse outcomes in preterm infants. C- reactive protein (CRP) is a marker of inflammation/infection. Probiotics have anti-inflammatory properties. Randomized controlled trials (RCTs) in preterm infants have not reported effect of probiotics on CRP. AIM To evaluate effect of probiotics on CRP in preterm infants who had participated in a RCT of Bifidobacterium breve (B. breve) m-16v. METHODS Data on all infants (GA <33 weeks, n = 159) enrolled in the RCT was analyzed. For study purpose, CRP <15 mg/L and ≤10 mg/L was considered normal for the first week, and thereafter respectively. Mixed logistic regression modelling was used to assess probiotic effect on CRP levels. RESULTS There were 1579 CRP measurements (Probiotic: 851 vs. Placebo: 728). Baseline characteristics and number [Median (IQR)] of CRP estimations per infant [l0 (5, 20) vs. 10 (6, 17), p = 0.861] were comparable between probiotic vs. placebo group. There was no significant difference in the proportion of infants with high CRP over time (treatment by weekly time points interaction, p = 0.187), and across all time points between probiotic and placebo group (adjusted OR: 1.62, 95% CI: 0.91-2.88, p = 0.102)CONCLUSION:B. breve m-16v did not decrease CRP levels in preterm infants born <33 weeks.
Collapse
Affiliation(s)
- S Agrawal
- Department of Neonatal Paediatrics, KEM Hospital, Perth, WA, Australia.,Department of Neonatal Paediatrics, Princess Margaret Hospital for Children, Perth, WA, Australia.,Centre for Neonatal Research and Education, School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia
| | - S Rao
- Department of Neonatal Paediatrics, KEM Hospital, Perth, WA, Australia.,Department of Neonatal Paediatrics, Princess Margaret Hospital for Children, Perth, WA, Australia.,Centre for Neonatal Research and Education, School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia
| | - E A Nathan
- Women and Infants Research Foundation, King Edward Memorial Hospital for Women, Perth, WA, Australia.,School of Women's and Infants' Health, University of Western Australia, Perth, WA, Australia
| | - S Patole
- Department of Neonatal Paediatrics, KEM Hospital, Perth, WA, Australia.,Centre for Neonatal Research and Education, School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
26
|
Zhu T, Yuan T, Yu H, Gu W, Chen X, Jiang P. The phosphodiesterase-4 inhibitor Rolipram promotes cognitive function recovery in prenatal Escherichia coli infected offspring. J Matern Fetal Neonatal Med 2018; 33:2166-2175. [PMID: 30373424 DOI: 10.1080/14767058.2018.1542682] [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: 01/22/2023]
Abstract
Objective: Preterm infants are especially vulnerable to intrauterine infection-induced brain injury, which is closely relevant with cognitive deficits and cerebral palsy. Rolipram, a phosphodiesterase-4 inhibitor, can improve cognition in rodents. However, the underlying roles and mechanisms are not well investigated.Methods: In the present study, we used intrauterine Escherichia coli (E. coli) infected model. Escherichia coli was inoculated into pregnant rats' uterine cervix at embryonic day 15 (E15) while the control group was given normal saline. Rolipram was administered by intraperitoneal (i.p.) injection once daily from postnatal day (P) 1-7. Morris water maze test was used for cognitive behavior test. Hippocampal neural stem/precursor cells (NSPCs) proliferation and neuronal differentiation were studied by immunofluorescent staining. The expressions of p-CREB, p-Akt, TrkB and BDNF were estimated by western-blot analysis.Results: The data showed that Rolipram could ameliorate cognitive deficits and enhance NSPCs proliferation and neuronal differentiation in intrauterine infected offspring. Additionally, Rolipram could significantly increase p-CREB/CREB, p-Akt/Akt, TrkB and BDNF levels.Conclusions: These results suggested that Rolipram might play a neuroprotective role to promote cognitive function recovery after intrauterine infection. And hippocampal NSPCs proliferation and neuronal differentiation might be enhanced via CREB/Akt/BDNF signal transduction.
Collapse
Affiliation(s)
- Tao Zhu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huimin Yu
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weizhong Gu
- Central Laboratory, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Chen
- Central Laboratory, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peifang Jiang
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Departments of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
Cakan P, Yildiz S, Ozgocer T, Yildiz A, Vardi N. Maternal viral mimetic administration at the beginning of fetal hypothalamic nuclei development accelerates puberty in female rat offspring. Can J Physiol Pharmacol 2018; 96:506-514. [DOI: 10.1139/cjpp-2016-0535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This study aimed to investigate the effects of maternal viral infection during a critical time window of fetal hypothalamic development on timing of puberty in the female offspring. For that purpose, a viral mimetic (i.e., synthetic double-strand RNA, namely, polyinosinic–polycytidylic acid, poly (I:C)) or saline was injected (i.p.) to the pregnant rats during the beginning (day 12 of pregnancy, n = 5 for each group) or at the end of this time window (day 14 of pregnancy, n = 5 for each group). Four study groups were formed from the female pups (n = 9–10 pups/group). Following weaning of pups, vaginal opening and vaginal smearing was studied daily until 2 sequential estrous cycles were observed. During the second diestrus phase, blood samples were taken for progesterone, leptin, corticosterone, follicle-stimulating hormone, and luteinizing hormone. Maternal poly (I:C) injection on day 12 of pregnancy increased body mass and reduced the time to puberty in the female offspring. Neither poly (I:C) nor timing of injection affected other parameters studied (p > 0.05). It has been shown for the first time that maternal viral infection during the beginning of fetal hypothalamic development might hasten puberty by increasing body mass in rat offspring.
Collapse
Affiliation(s)
- Pinar Cakan
- Department of Physiology, Faculty of Medicine, University of Inonu, 44280 Malatya, Turkey
| | - Sedat Yildiz
- Department of Physiology, Faculty of Medicine, University of Inonu, 44280 Malatya, Turkey
| | - Tuba Ozgocer
- Department of Physiology, Faculty of Medicine, University of Inonu, 44280 Malatya, Turkey
| | - Azibe Yildiz
- Department of Histology, Faculty of Medicine, University of Inonu, 44280 Malatya, Turkey
| | - Nigar Vardi
- Department of Histology, Faculty of Medicine, University of Inonu, 44280 Malatya, Turkey
| |
Collapse
|
28
|
Gussenhoven R, Westerlaken RJJ, Ophelders DRMG, Jobe AH, Kemp MW, Kallapur SG, Zimmermann LJ, Sangild PT, Pankratova S, Gressens P, Kramer BW, Fleiss B, Wolfs TGAM. Chorioamnionitis, neuroinflammation, and injury: timing is key in the preterm ovine fetus. J Neuroinflammation 2018; 15:113. [PMID: 29673373 PMCID: PMC5907370 DOI: 10.1186/s12974-018-1149-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/04/2018] [Indexed: 01/11/2023] Open
Abstract
Background Antenatal infection (i.e., chorioamnionitis) is an important risk factor for adverse neurodevelopmental outcomes after preterm birth. Destructive and developmental disturbances of the white matter are hallmarks of preterm brain injury. Understanding the temporal effects of antenatal infection in relation to the onset of neurological injury is crucial for the development of neurotherapeutics for preterm infants. However, these dynamics remain unstudied. Methods Time-mated ewes were intra-amniotically injected with lipopolysaccharide at 5, 12, or 24 h or 2, 4, 8, or 15 days before preterm delivery at 125 days gestational age (term ~ 150 days). Post mortem analyses for peripheral immune activation, neuroinflammation, and white matter/neuronal injury were performed. Moreover, considering the neuroprotective potential of erythropoietin (EPO) for perinatal brain injury, we evaluated (phosphorylated) EPO receptor (pEPOR) expression in the fetal brain following LPS exposure. Results Intra-amniotic exposure to this single bolus of LPS resulted in a biphasic systemic IL-6 and IL-8 response. In the developing brain, intra-amniotic LPS exposure induces a persistent microgliosis (IBA-1 immunoreactivity) but a shorter-lived increase in the pro-inflammatory marker COX-2. Cell death (caspase-3 immunoreactivity) was only observed when LPS exposure was greater than 8 days in the white matter, and there was a reduction in the number of (pre) oligodendrocytes (Olig2- and PDGFRα-positive cells) within the white matter at 15 days post LPS exposure only. pEPOR expression displayed a striking biphasic regulation following LPS exposure which may help explain contradicting results among clinical trials that tested EPO for the prevention of preterm brain injury. Conclusion We provide increased understanding of the spatiotemporal pathophysiological changes in the preterm brain following intra-amniotic inflammation which may aid development of new interventions or implement interventions more effectively to prevent perinatal brain damage.
Collapse
Affiliation(s)
- Ruth Gussenhoven
- Department of Pediatrics, Maastricht University Medical Center, 6202, AZ, Maastricht, The Netherlands.,School for Mental Health and Neuroscience (MHeNs), Maastricht University Medical Center, 6229, ER, Maastricht, The Netherlands
| | - Rob J J Westerlaken
- Department of Pediatrics, Maastricht University Medical Center, 6202, AZ, Maastricht, The Netherlands
| | - Daan R M G Ophelders
- Department of Pediatrics, Maastricht University Medical Center, 6202, AZ, Maastricht, The Netherlands.,School of Oncology and Developmental Biology (GROW), Maastricht University Medical Center, 6229, ER, Maastricht, the Netherlands
| | - Alan H Jobe
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45208, USA
| | - Matthew W Kemp
- School of Women's and Infants' Health, The University of Western Australia (M550), Crawley, WA, 6009, Australia
| | - Suhas G Kallapur
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45208, USA
| | - Luc J Zimmermann
- Department of Pediatrics, Maastricht University Medical Center, 6202, AZ, Maastricht, The Netherlands.,School of Oncology and Developmental Biology (GROW), Maastricht University Medical Center, 6229, ER, Maastricht, the Netherlands
| | - Per T Sangild
- Department of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg DK 1870 C, Copenhagen, Denmark.,Departments of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, 2100, Denmark
| | - Stanislava Pankratova
- Department of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg DK 1870 C, Copenhagen, Denmark.,Departments of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, 2100, Denmark
| | - Pierre Gressens
- Department of Perinatal Imaging and Health, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas Hospital, London, SE1 7EH, UK.,PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,PremUP, Université Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center, 6202, AZ, Maastricht, The Netherlands.,School for Mental Health and Neuroscience (MHeNs), Maastricht University Medical Center, 6229, ER, Maastricht, The Netherlands.,School of Oncology and Developmental Biology (GROW), Maastricht University Medical Center, 6229, ER, Maastricht, the Netherlands
| | - Bobbi Fleiss
- Department of Perinatal Imaging and Health, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas Hospital, London, SE1 7EH, UK.,PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,PremUP, Université Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Tim G A M Wolfs
- Department of Pediatrics, Maastricht University Medical Center, 6202, AZ, Maastricht, The Netherlands. .,School of Oncology and Developmental Biology (GROW), Maastricht University Medical Center, 6229, ER, Maastricht, the Netherlands. .,Department of BioMedical Engineering, Maastricht University Medical Center, 6229, ER, Maastricht, The Netherlands.
| |
Collapse
|
29
|
Lu J, Lu L, Yu Y, Cluette-Brown J, Martin CR, Claud EC. Effects of Intestinal Microbiota on Brain Development in Humanized Gnotobiotic Mice. Sci Rep 2018; 8:5443. [PMID: 29615691 PMCID: PMC5882882 DOI: 10.1038/s41598-018-23692-w] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/19/2018] [Indexed: 12/23/2022] Open
Abstract
Poor growth in the Neonatal Intensive Care Unit is associated with an increased risk for poor neurodevelopmental outcomes for preterm infants, however the mechanism is unclear. The microbiome has increasingly been recognized as a modifiable environmental factor to influence host development. Here we explore the hypothesis that the microbiome influences both growth phenotype and brain development. A germ free mouse transfaunation model was used to examine the effects of preterm infant microbiotas known to induce either high growth or low growth phenotypes on postnatal brain development. The microbiome which induced the low growth phenotype was associated with decreases in the neuronal markers NeuN and neurofilament-L as well as the myelination marker MBP when compared to the microbiome associated with the high growth phenotype. Additionally, poor growth phenotype-associated microbiota was associated with increased neuroinflammation marked by increased Nos1, as well as alteration in IGF-1 pathway including decreased circulating and brain IGF-1, decreased circulating IGFBP3, and increased Igfbp3 brain mRNA expression. This study suggests that growth-associated microbiota can influence early neuron and oligodendrocyte development and that this effect may be mediated by effects on neuroinflammation and circulating IGF-1.
Collapse
Affiliation(s)
- Jing Lu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Lei Lu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Yueyue Yu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Joanne Cluette-Brown
- Beth Israel Deaconess Medical Center, Division of Gastroenterology, Boston, MA, 02215, USA
| | - Camilia R Martin
- Beth Israel Deaconess Medical Center, Harvard Medical School, Department of Neonatology and Division of Translational Research, Boston, MA, 02215, USA
| | - Erika C Claud
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA.
| |
Collapse
|
30
|
Noureldein M, Mardare R, Pickard J, Shing HL, Eisenhut M. Cerebrospinal fluid protein and glucose levels in neonates with a systemic inflammatory response without meningitis. Fluids Barriers CNS 2018. [PMID: 29540199 PMCID: PMC5853144 DOI: 10.1186/s12987-018-0095-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND It has been estimated that paediatric meningitis without elevated CSF white cell count (pleocytosis) accounts for 0.5-12% of all cases of bacterial meningitis. CSF protein and glucose measurements are therefore essential in management but may be neglected in clinical practice. In order to improve recognition of bacterial meningitis in neonates and to enable adequate management and audit, we investigated whether a systemic inflammatory response in the absence of meningitis is associated with elevated CSF protein and reduced CSF glucose levels. A further aim was to determine whether abnormal levels of these parameters were associated with increased incidence of neurological damage. METHODS As part of an audit into management of abnormal CSF findings in neonates, we conducted a retrospective analysis of neonates without meningitis as evident from normal CSF white blood cell counts and negative CSF culture. We compared data from neonates with fever (temperature > 38.0 °C) and/or elevated C-reactive protein (CRP) levels (> 5 mg/l) (possible sepsis) with data from neonates without fever or CRP elevation. RESULTS We analysed results from a total of 244 neonates. CSF protein levels were 0.89 g/l (SD 0.37) in neonates without fever or elevated CRP (n = 26) and not significantly different from neonates with possible sepsis (n = 218) with 0.92 g/l (SD 0.40). CSF glucose levels in infants with possible sepsis were 2.71 (SD 0.83) mmol/l and not significantly different from infants without sepsis with 2.55 mmol/l (SD 0.34). CONCLUSIONS CSF protein and glucose levels are not affected by a systemic inflammatory response syndrome if there is no meningitis.
Collapse
Affiliation(s)
- Mona Noureldein
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton, LU4ODZ, UK
| | - Roxana Mardare
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton, LU4ODZ, UK
| | - Jack Pickard
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton, LU4ODZ, UK
| | - Hoi Lun Shing
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton, LU4ODZ, UK
| | - Michael Eisenhut
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton, LU4ODZ, UK.
| |
Collapse
|
31
|
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: 21] [Impact Index Per Article: 3.0] [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
|
32
|
Gussenhoven R, Ophelders DRMG, Kemp MW, Payne MS, Spiller OB, Beeton ML, Stock SJ, Cillero-Pastor B, Barré FPY, Heeren RMA, Kessels L, Stevens B, Rutten BP, Kallapur SG, Jobe AH, Kramer BW, Wolfs TGAM. The Paradoxical Effects of Chronic Intra-Amniotic Ureaplasma parvum Exposure on Ovine Fetal Brain Development. Dev Neurosci 2017; 39:472-486. [PMID: 28848098 DOI: 10.1159/000479021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/24/2017] [Indexed: 01/23/2023] Open
Abstract
Chorioamnionitis is associated with adverse neurodevelopmental outcomes in preterm infants. Ureaplasma spp. are the microorganisms most frequently isolated from the amniotic fluid of women diagnosed with chorioamnionitis. However, controversy remains concerning the role of Ureaplasma spp. in the pathogenesis of neonatal brain injury. We hypothesize that reexposure to an inflammatory trigger during the perinatal period might be responsible for the variation in brain outcomes of preterms following Ureaplasma-driven chorioamnionitis. To investigate these clinical scenarios, we performed a detailed multimodal study in which ovine neurodevelopmental outcomes were assessed following chronic intra-amniotic Ureaplasma parvum (UP) infection either alone or combined with subsequent lipopolysaccharide (LPS) exposure. We show that chronic intra-amniotic UP exposure during the second trimester provoked a decrease in astrocytes, increased oligodendrocyte numbers, and elevated 5-methylcytosine levels. In contrast, short-term LPS exposure before preterm birth induced increased microglial activation, myelin loss, elevation of 5-hydroxymethylcytosine levels, and lipid profile changes. These LPS-induced changes were prevented by chronic preexposure to UP (preconditioning). These data indicate that chronic UP exposure has dual effects on preterm brain development in utero. On the one hand, prolonged UP exposure causes detrimental cerebral changes that may predispose to adverse postnatal clinical outcomes. On the other, chronic intra-amniotic UP exposure preconditions the brain against a second inflammatory hit. This study demonstrates that microbial interactions and the timing and duration of the inflammatory insults determine the effects on the fetal brain. Therefore, this study helps to understand the complex and diverse postnatal neurological outcomes following UP driven chorioamnionitis.
Collapse
Affiliation(s)
- Ruth Gussenhoven
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
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
|
Magalhães RC, Moreira JM, Vieira ÉLM, Rocha NP, Miranda DM, Simões e Silva AC. Urinary Levels of IL-1 β and GDNF in Preterm Neonates as Potential Biomarkers of Motor Development: A Prospective Study. Mediators Inflamm 2017; 2017:8201423. [PMID: 28553016 PMCID: PMC5434239 DOI: 10.1155/2017/8201423] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/22/2017] [Indexed: 01/07/2023] Open
Abstract
Objectives. To evaluate the association between inflammatory biomarkers, neurotrophic factors, birth conditions, and the presence of motor development abnormalities in preterm neonates. Methods. Plasma and urinary levels of cytokines (IL-1β, IL-6, IL-10, TNF, and IL-12p70), chemokines (CXCL8/IL-8, CCL2/MCP-1, CCL5/RANTES, CXCL10/IP-10, and CXCL9/MIG), and neurotrophic factors (BDNF and GDNF) were evaluated in 40 preterm neonates born between 28 and 32 incomplete weeks of gestation, at four distinct time points: at birth (umbilical cord blood) (T0), at 48 (T1), at 72 hours (T2), and at 3 weeks after birth (T3). Biomarkers levels were compared between different time points and then associated with Test of Infant Motor Performance (TIMP) percentiles. Results. Maternal age, plasma, and urinary concentrations of inflammatory molecules and neurotrophic factors were significantly different between groups with normal versus lower than expected motor development. Higher levels of GDNF were found in the group with lower than expected motor development, while IL-1β and CXCL8/IL-8 values were higher in the group with typical motor development. Conclusion. Measurements of cytokines and neurotrophic factors in spot urine may be useful in the follow-up of motor development in preterm neonates.
Collapse
Affiliation(s)
- Rafael Coelho Magalhães
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Janaina Matos Moreira
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Érica Leandro Marciano Vieira
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Natália Pessoa Rocha
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Débora Marques Miranda
- Instituto Nacional de Ciência e Tecnologia de Medicina Molecular, Faculdade de Medicina, UFMG, Belo Horizonte, MG, Brazil
| | - Ana Cristina Simões e Silva
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia de Medicina Molecular, Faculdade de Medicina, UFMG, Belo Horizonte, MG, Brazil
| |
Collapse
|
35
|
Langham A. MRI for premature neonatal brain injury: a case report. J Med Radiat Sci 2017; 64:152-155. [PMID: 28263044 PMCID: PMC5454326 DOI: 10.1002/jmrs.226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 12/05/2016] [Accepted: 01/27/2017] [Indexed: 01/12/2023] Open
Abstract
This case report aims to extend analytical thinking and clinical reasoning of clinicians and radiographers when presented with diagnosing premature neonatal brain injuries (PNBI). The report considers the uses and merit of magnetic resonance imaging (MRI) in the primary assessment of PNBI. The traditional technique of cranial ultrasound as the first modality of choice can have several limitations, which includes a lower temporal resolution in its ability to differentiate grey-white matter distribution patterns, lower spatial resolution in its ability to accurately map white matter fibre tracts and distribution patterns which are critical in white matter injury pathological events. In this specific case report, MRI was useful for the assessment of haemorrhagic brain injury post partum.Therefore, should MRI be considered, the primary imaging modality in these cases when the concerns about PNBI is presented? This case study explores the current trends in MRI neonatal brain imaging and advancements being made in this field.
Collapse
|
36
|
Ten VS. Mitochondrial dysfunction in alveolar and white matter developmental failure in premature infants. Pediatr Res 2017; 81:286-292. [PMID: 27901512 PMCID: PMC5671686 DOI: 10.1038/pr.2016.216] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/19/2016] [Indexed: 01/11/2023]
Abstract
At birth, some organs in premature infants are not developed enough to meet challenges of the extra-uterine life. Although growth and maturation continues after premature birth, postnatal organ development may become sluggish or even arrested, leading to organ dysfunction. There is no clear mechanistic concept of this postnatal organ developmental failure in premature neonates. This review introduces a concept-forming hypothesis: Mitochondrial bioenergetic dysfunction is a fundamental mechanism of organs maturation failure in premature infants. Data collected in support of this hypothesis are relevant to two major diseases of prematurity: white matter injury and broncho-pulmonary dysplasia. In these diseases, totally different clinical manifestations are defined by the same biological process, developmental failure of the main functional units-alveoli in the lungs and axonal myelination in the brain. Although molecular pathways regulating alveolar and white matter maturation differ, proper bioenergetic support of growth and maturation remains critical biological requirement for any actively developing organ. Literature analysis suggests that successful postnatal pulmonary and white matter development highly depends on mitochondrial function which can be inhibited by sublethal postnatal stress. In premature infants, sublethal stress results mostly in organ maturation failure without excessive cellular demise.
Collapse
Affiliation(s)
- Vadim S. Ten
- Department of Pediatrics, Division of Neonatology, Columbia University, New York, New York
| |
Collapse
|
37
|
Graham EM, Burd I, Everett AD, Northington FJ. Blood Biomarkers for Evaluation of Perinatal Encephalopathy. Front Pharmacol 2016; 7:196. [PMID: 27468268 PMCID: PMC4942457 DOI: 10.3389/fphar.2016.00196] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/20/2016] [Indexed: 12/17/2022] Open
Abstract
Recent research in identification of brain injury after trauma shows many possible blood biomarkers that may help identify the fetus and neonate with encephalopathy. Traumatic brain injury shares many common features with perinatal hypoxic-ischemic encephalopathy. Trauma has a hypoxic component, and one of the 1st physiologic consequences of moderate-severe traumatic brain injury is apnea. Trauma and hypoxia-ischemia initiate an excitotoxic cascade and free radical injury followed by the inflammatory cascade, producing injury in neurons, glial cells and white matter. Increased excitatory amino acids, lipid peroxidation products, and alteration in microRNAs and inflammatory markers are common to both traumatic brain injury and perinatal encephalopathy. The blood-brain barrier is disrupted in both leading to egress of substances normally only found in the central nervous system. Brain exosomes may represent ideal biomarker containers, as RNA and protein transported within the vesicles are protected from enzymatic degradation. Evaluation of fetal or neonatal brain derived exosomes that cross the blood-brain barrier and circulate peripherally has been referred to as the "liquid brain biopsy." A multiplex of serum biomarkers could improve upon the current imprecise methods of identifying fetal and neonatal brain injury such as fetal heart rate abnormalities, meconium, cord gases at delivery, and Apgar scores. Quantitative biomarker measurements of perinatal brain injury and recovery could lead to operative delivery only in the presence of significant fetal risk, triage to appropriate therapy after birth and measure the effectiveness of treatment.
Collapse
Affiliation(s)
- Ernest M. Graham
- Division of Maternal-Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Irina Burd
- Division of Maternal-Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Integrated Research Center for Fetal Medicine, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Allen D. Everett
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Division of Cardiology, Department of Pediatrics, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Frances J. Northington
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of MedicineBaltimore, MD, USA
| |
Collapse
|