1
|
Garg PM, Shenberger JS, Ostrander M, Inder TE, Garg PP. Gut-Brain Axis in Preterm Infants with Surgical Necrotizing Enterocolitis. Am J Perinatol 2025. [PMID: 40112874 DOI: 10.1055/a-2563-0878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Necrotizing enterocolitis (NEC) affects 5 to 10% of very low-birth-weight infants and remains a leading cause of mortality and long-term morbidity. Preterm infants with NEC, especially those requiring surgery, have higher inflammatory markers in the blood, severe white matter abnormalities on brain imaging, and adverse neurodevelopmental outcomes. This review presents current evidence regarding the clinical factors associated with brain injury in preterm infants with NEC needing surgical intervention. Studies that evaluate neuroprotective strategies to prevent brain injury are greatly needed to improve neurodevelopmental outcomes in high-risk preterm infants with NEC. · NEC is associated with white matter, grey matter, and cerebellar injury in neonates.. · Clinical and histopathological factors are associated with gut-associated brain injury in NEC.. · Neuroprotective strategies and intervention are greatly needed in infants with surgical NEC..
Collapse
Affiliation(s)
- Parvesh M Garg
- Department of Pediatrics/Neonatology, Wake Forest University, Winston Salem, North Carolina
| | - Jeffrey S Shenberger
- Department of Pediatrics/Neonatology, Connecticut Children's, Hartford, Connecticut
| | - Mckenzie Ostrander
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Terrie E Inder
- Children Hospital of Orange County, University of California, Irvine, Orange, California
| | - Padma P Garg
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
2
|
Younge N, Patel RM. Probiotics and the Risk of Infection. Clin Perinatol 2025; 52:87-100. [PMID: 39892956 PMCID: PMC11789005 DOI: 10.1016/j.clp.2024.10.006] [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] [Indexed: 02/04/2025]
Abstract
Probiotic use has increased in preterm infants and may reduce the risk of necrotizing enterocolitis. Probiotic-associated infection is a concern for infants receiving probiotic supplementation in the neonatal intensive care unit, as highlighted by a recent case and subsequent action by the United States Food and Drug Administration. Based on reports to date, invasive infection is an infrequent but known risk of probiotic supplementation. In this article, we discuss the epidemiology and pathophysiology of invasive infection in preterm infants, review the benefits and risks of probiotic as regulations and available products continue to evolve.
Collapse
Affiliation(s)
- Noelle Younge
- Department of Pediatrics, Duke University, 2400 Pratt Street, DUMC Box 2739, Durham, NC 27705, USA
| | - Ravi M Patel
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Arthur M. Blank Hospital, 2220 North Druid Hills Road NE, CL.06323, Atlanta, GA 30329, USA.
| |
Collapse
|
3
|
Irzan FI, Retinasamy T, Wen WR, Sheng ETM, Shaikh MF, Arulsamy A. The Role of HMGB1 in Infection-Related Cognitive Deficits. FRONT BIOSCI-LANDMRK 2025; 30:25544. [PMID: 40018921 DOI: 10.31083/fbl25544] [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: 07/04/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 03/01/2025]
Abstract
Infectious diseases caused by fungi, viruses, or bacteria can have a profound impact on human cognition. This can be due to either direct spread to the central nervous system (CNS) or indirect neuroinflammation. Ultimately causing neuronal damage and even neurodegeneration. Deteriorations in cognition, such as poor encoding and attention deficits, have been reported secondary to infectious diseases. Preclinical studies have identified the underlying mechanisms of these infection-related cognitive effects, such as through blood-brain barrier (BBB) disruption and M1 microglial polarization. These mechanisms are spearheaded by inflammatory markers that are released/initiated by the pathogens over the course of the infection. Among them, the high mobility group box 1 (HMGB1) protein is a common biomarker implicated across several infection-related cognitive deficits. Understanding these effects and mechanisms is crucial for the development of strategies to prevent and treat infection-related cognitive impairment. This review will thus consolidate and elucidate the current knowledge on the potential role of HMGB1 as a therapeutic target for infection-related cognitive impairments. This review will not only advance scientific understanding but also have significant clinical and public health implications, especially considering recent global health challenges. Based on the selected articles, extracellular HMGB1, as opposed to intracellular HMGB1, acts as damage-associated molecular patterns (DAMPs) or alarmins when released in the peripheries secondary to inflammasome activation. Due to their low molecular weight, they then enter the CNS through routes such as retrograde transport along the afferent nerves, or simple diffusion across the impaired BBB. This results in further disruption of the brain microenvironment due to the dysregulation of other regulatory pathways. The outcome is structural neuronal changes and cognitive impairment. Given its key role in neuroinflammation, HMGB1 holds promise as both a biomarker for diagnostic detection and a potential therapeutic target candidate for preventing infection-related cognitive impairment.
Collapse
Affiliation(s)
- Fathima Ijaza Irzan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Wong Ruo Wen
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Edward Ting Ming Sheng
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, NSW 2800, Australia
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| |
Collapse
|
4
|
Thompson DK, Cai S, Kelly CE, Alexander B, Matthews LG, Mainzer R, Doyle LW, Cheong JLY, Inder TE, Yang JYM, Anderson PJ. Brain volume and neurodevelopment at 13 years following sepsis in very preterm infants. Pediatr Res 2025; 97:744-750. [PMID: 39003332 DOI: 10.1038/s41390-024-03407-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/05/2024] [Accepted: 06/17/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Associations of neonatal infection with brain growth and later neurodevelopmental outcomes in very preterm (VP) infants are unclear. This study aimed to assess associations of neonatal sepsis in VP infants with (1) brain growth from term-equivalent age to 13 years; and (2) 13-year brain volume and neurodevelopmental outcomes. METHODS 224 infants born VP ( < 30 weeks' gestation/<1250 g birthweight) were recruited. Longitudinal brain volumes for 68 cortical and 14 subcortical regions were derived from MRI at term-equivalent, 7 and/or 13 years of age for 216 children (79 with neonatal sepsis and 137 without). 177 children (79%) had neurodevelopmental assessments at age 13. Of these, 63 with neonatal sepsis were compared with 114 without. Brain volumetric growth trajectories across time points were compared between sepsis and no-sepsis groups using mixed effects models. Linear regressions compared brain volume and neurodevelopmental outcome measures at 13 years between sepsis and no sepsis groups. RESULTS Growth trajectories were similar and there was little evidence for differences in brain volumes or neurodevelopmental domains at age 13 years between those with or without sepsis. CONCLUSIONS Neonatal sepsis in children born VP does not appear to disrupt subsequent brain development, or to have functional consequences in early adolescence. IMPACT STATEMENT Neonatal sepsis has been associated with poorer short-term neurodevelopmental outcomes and reduced brain volumes in very preterm infants. This manuscript provides new insights into the long-term brain development and neurodevelopmental outcomes of very preterm-born children who did or did not have neonatal sepsis. We found that regional brain volumes up to 13 years, and neurodevelopmental outcomes at age 13, were similar between those with and without neonatal sepsis. The links between neonatal sepsis and long-term neurodevelopment remain unclear.
Collapse
Affiliation(s)
- Deanne K Thompson
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia.
- Developmental Imaging, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia.
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia.
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Faculty of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, 3800, Australia.
| | - Shirley Cai
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Developmental Imaging, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Melbourne Medicine School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Claire E Kelly
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Developmental Imaging, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Faculty of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Bonnie Alexander
- Developmental Imaging, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Neuroscience Advanced Clinical Imaging Service (NACIS), Department of Neurosurgery, Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Lillian G Matthews
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Developmental Imaging, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Rheanna Mainzer
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Lex W Doyle
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
- Neonatal Services, The Royal Women's Hospital, Parkville, VIC, 3052, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Jeanie L Y Cheong
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
- Neonatal Services, The Royal Women's Hospital, Parkville, VIC, 3052, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Terrie E Inder
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Center for Neonatal Research, Children's Hospital of Orange County, Orange, CA, 92866, USA
- Department of Pediatrics, University of California, Irvine, CA, 92697, USA
| | - Joseph Y M Yang
- Developmental Imaging, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
- Neuroscience Advanced Clinical Imaging Service (NACIS), Department of Neurosurgery, Royal Children's Hospital, Parkville, VIC, 3052, Australia
- Neuroscience Research, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Peter J Anderson
- Victorian Infant Brain Studies, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Faculty of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
5
|
Miselli F, Costantini RC, Maugeri M, Deonette E, Mazzotti S, Bedetti L, Lugli L, Rossi K, Roversi M, Berardi A. Outcome prediction for late-onset sepsis after premature birth. Pediatr Res 2025:10.1038/s41390-025-03814-7. [PMID: 39824936 DOI: 10.1038/s41390-025-03814-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 01/20/2025]
Abstract
BACKGROUND Our aim was to develop a quantitative model for immediately estimating the risk of death and/or brain injury in late-onset sepsis (LOS) in preterm infants, based on objective and measurable data available at the time sepsis is first suspected (i.e., time of blood culture collection). METHODS Retrospective study on neonates ≤36 weeks' gestation with a positive blood and/or cerebrospinal fluid culture after 72 hours from birth. RESULTS Among 3217 preterm live births, 94 cases were included (median gestational age 26.5 weeks' IQR 25.0;28.0), of whom 26 (27.7%) had poor outcomes (17 death; 9 brain injuries). Infants with poor outcomes showed lower postnatal age (11.5 vs 12.5 days, p < 0.001), lower mean blood pressure (30.5 vs 43 mmHg, p < 0.001) and higher lactate levels (4.4 vs 1.5 mmol/l, p < 0.001). Our multivariable model showed good discrimination and calibration (c statistic=0.8618, Hosmer-Lemeshow p = 0.8532), stratifying the population into 3 groups: low-risk (sensitivity 97%, specificity 52%), middle-risk, and high-risk (sensitivity 77%, specificity 80%). CONCLUSION This predictive model performs well as a practical and easy-to-use tool to help clinicians early identify the sickest neonates who may benefit from timely and aggressive support (e.g., central line, haemodynamic assessment) and close monitoring (e.g., 1:1 nursing assignment, frequent reassessments). IMPACT We lack data to early identify the severity of neonatal late-onset sepsis in preterm infants. Delay in treatment contributes to poor prognosis. We developed a model for early prediction of poor outcomes (mortality and brain injuries). The model utilizes immediately available and measurable data at the time sepsis is first suspected. This can help clinicians in tailoring management based on individual risks.
Collapse
Affiliation(s)
- Francesca Miselli
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Via Università, 41121, Modena, Italy.
- Neonatal Intensive Care Unit, University Hospital of Modena, Via del Pozzo, 41124, Modena, Italy.
| | - Riccardo Cuoghi Costantini
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Via del Pozzo, 41124, Modena, Italy
| | - Melissa Maugeri
- Medicine and Surgery School, University of Modena and Reggio Emilia, Via Università, 41121, Modena, Italy
| | - Elisa Deonette
- Pediatric Post-Graduate School, University Hospital of Modena and Reggio Emilia, Via del Pozzo, 41224, Modena, Italy
| | - Sofia Mazzotti
- Pediatric Post-Graduate School, University Hospital of Modena and Reggio Emilia, Via del Pozzo, 41224, Modena, Italy
| | - Luca Bedetti
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Via Università, 41121, Modena, Italy
- Neonatal Intensive Care Unit, University Hospital of Modena, Via del Pozzo, 41124, Modena, Italy
| | - Licia Lugli
- Neonatal Intensive Care Unit, University Hospital of Modena, Via del Pozzo, 41124, Modena, Italy
| | - Katia Rossi
- Neonatal Intensive Care Unit, University Hospital of Modena, Via del Pozzo, 41124, Modena, Italy
| | - MariaFederica Roversi
- Neonatal Intensive Care Unit, University Hospital of Modena, Via del Pozzo, 41124, Modena, Italy
| | - Alberto Berardi
- Neonatal Intensive Care Unit, University Hospital of Modena, Via del Pozzo, 41124, Modena, Italy
| |
Collapse
|
6
|
Liong S. Detecting the early warning signs of neonatal brain injury. Brain Behav Immun 2025; 123:948-949. [PMID: 39505050 DOI: 10.1016/j.bbi.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024] Open
Affiliation(s)
- Stella Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
| |
Collapse
|
7
|
Lawrence SM, Wynn JL, Gordon SM. Neonatal bacteremia and sepsis. REMINGTON AND KLEIN'S INFECTIOUS DISEASES OF THE FETUS AND NEWBORN INFANT 2025:183-232.e25. [DOI: 10.1016/b978-0-323-79525-8.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
8
|
Garg PM, South AM, Inder T, Shenberger JS. Hypotension: a modifiable risk factor impacting surgical NEC-associated AKI and brain injury in preterm infants. Pediatr Res 2024; 96:1125-1126. [PMID: 38714861 PMCID: PMC11524779 DOI: 10.1038/s41390-024-03244-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/13/2024] [Indexed: 11/01/2024]
Affiliation(s)
- Parvesh Mohan Garg
- Department of Pediatrics/Neonatology, Brenner Children's at Atrium Health Wake Forest Baptist and Wake Forest University School of Medicine, Winston Salem, NC, USA.
| | - Andrew M South
- Department of Pediatrics/Nephrology, Brenner Children's at Atrium Health Wake Forest Baptist and Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Terrie Inder
- Children Hospital of Orange County, University of California, Irvine, Orange, CA, USA
| | - Jeffrey S Shenberger
- Department of Pediatrics, Connecticut Children's and The University of Connecticut School of Medicine, Hartford, CT, USA
| |
Collapse
|
9
|
Meyer S, Hess S, Poryo M, Papan C, Simon A, Welcker S, Ehrlich A, Ruckes C. Study draft: "UVC-You Will See" study: longer vs. shorter umbilical venous catheter (UVC) dwell time (6-10 vs. 1-5 days) in very premature infants with birth weight < 1250 g and/or gestational age < 30 weeks. Wien Med Wochenschr 2024; 174:217-224. [PMID: 38869762 PMCID: PMC11347460 DOI: 10.1007/s10354-024-01047-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Umbilical venous catheters (UVCs) are often used in preterm infants. Their use is associated with complications (infections, clot formation, organ injury). Very preterm infants with acquired bloodstream infection are at a higher risk for death and important morbidities (e.g., adverse neurodevelopmental outcomes). It is standard clinical practice to remove UVCs in the first days of life. Replacement of intravenous access is often performed using percutaneously inserted central catheters (PICCs). It is unclear whether serial central line use affects the rates of catheter-related complications. METHODS A multicenter randomized controlled trial (random group assignment) was performed in 562 very premature (gestational age < 30 weeks) and/or very low birth weight infants (< 1250 g) requiring an UVC for administration of parenteral nutrition and/or drugs. Group allocation was random. HYPOTHESIS A UVC dwell time of 6-10 days (281 infants) is not associated with an increased rate of central venous catheter (UVC, PICC)-related complications compared to 1-5 days (281 infants), and a longer UVC dwell time will significantly reduce the number of painful, invasive procedures associated with the need for vascular access as well as radiation exposure, use of antibiotics, and medical costs. PRIMARY OUTCOME PARAMETER The number of catheter-related bloodstream infections and/or catheter-related thromboses and/or catheter-associated organ injuries related to the use of UVC/PICC was the primary outcome. CONCLUSION Extending the UVC dwell time may significantly reduce the number of painful invasive procedures, with the potential to positively impact not only long-term pain perception but also important social competencies (attention, learning, and behavior). Thus, the "UVC-You Will See" study has the potential to substantially change current neonatal intensive care practice.
Collapse
Affiliation(s)
- Sascha Meyer
- Department of Pediatrics and Neonatology, University Hospital Saarland, Homburg, Germany.
| | - Steffi Hess
- Kinder- und Jugendmedizin, Klinikum Saarbrücken Winterberg, Saarbrücken, Germany
| | - Martin Poryo
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg, Germany
| | - Cihan Papan
- Institute for Hygiene and Public Health, University Hospital Bonn, Bonn, Germany
| | - Arne Simon
- Department of Pediatric Hematology and Oncology, and Infectious Diseases, Saarland University Medical Center, Homburg, Germany
| | - Silvia Welcker
- Franz-Lust Klinik für Kinder und Jugendliche, Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76135, Karlsruhe, Germany
| | - Anne Ehrlich
- Interdisziplinäres Zentrum für Klinische Studien (IZKS), Mainz, Germany
| | - Christian Ruckes
- Interdisziplinäres Zentrum für Klinische Studien (IZKS), Mainz, Germany
| |
Collapse
|
10
|
Epstein AA, Janos SN, Menozzi L, Pegram K, Jain V, Bisset LC, Davis JT, Morrison S, Shailaja A, Guo Y, Chao AS, Abdi K, Rikard B, Yao J, Gregory SG, Fisher K, Pittman R, Erkanli A, Gustafson KE, Carrico CWT, Malcolm WF, Inder TE, Cotten CM, Burt TD, Shinohara ML, Maxfield CM, Benner EJ. Subventricular zone stem cell niche injury is associated with intestinal perforation in preterm infants and predicts future motor impairment. Cell Stem Cell 2024; 31:467-483.e6. [PMID: 38537631 PMCID: PMC11129818 DOI: 10.1016/j.stem.2024.03.001] [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: 09/24/2023] [Revised: 02/11/2024] [Accepted: 03/01/2024] [Indexed: 04/07/2024]
Abstract
Brain injury is highly associated with preterm birth. Complications of prematurity, including spontaneous or necrotizing enterocolitis (NEC)-associated intestinal perforations, are linked to lifelong neurologic impairment, yet the mechanisms are poorly understood. Early diagnosis of preterm brain injuries remains a significant challenge. Here, we identified subventricular zone echogenicity (SVE) on cranial ultrasound in preterm infants following intestinal perforations. The development of SVE was significantly associated with motor impairment at 2 years. SVE was replicated in a neonatal mouse model of intestinal perforation. Examination of the murine echogenic subventricular zone (SVZ) revealed NLRP3-inflammasome assembly in multiciliated FoxJ1+ ependymal cells and a loss of the ependymal border in this postnatal stem cell niche. These data suggest a mechanism of preterm brain injury localized to the SVZ that has not been adequately considered. Ultrasound detection of SVE may serve as an early biomarker for neurodevelopmental impairment after inflammatory disease in preterm infants.
Collapse
Affiliation(s)
- Adrian A Epstein
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Sara N Janos
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Luca Menozzi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kelly Pegram
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Logan C Bisset
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Joseph T Davis
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Samantha Morrison
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Aswathy Shailaja
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Yingqiu Guo
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Agnes S Chao
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Khadar Abdi
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Blaire Rikard
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA; Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Kimberley Fisher
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Rick Pittman
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Al Erkanli
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Kathryn E Gustafson
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | | | - William F Malcolm
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Terrie E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - C Michael Cotten
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Trevor D Burt
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA; Children's Health and Discovery Initiative, Duke University School of Medicine, Durham, NC, USA
| | - Mari L Shinohara
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Charles M Maxfield
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA.
| | - Eric J Benner
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
11
|
Dubner SE, Rickerich L, Bruckert L, Poblaciones RV, Sproul D, Scala M, Feldman HM, Travis KE. Early, low-dose hydrocortisone and near-term brain connectivity in extremely preterm infants. Pediatr Res 2024; 95:1028-1034. [PMID: 38030826 DOI: 10.1038/s41390-023-02903-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 10/17/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Postnatal steroids are used to prevent bronchopulmonary dysplasia in extremely preterm infants but may have adverse effects on brain development. We assessed connectivity metrics of major cerebral and cerebellar white matter pathways at near-term gestational age among infants who did or did not receive a standardized regimen of hydrocortisone during the first 10 days of life. METHODS Retrospective cohort study. PARTICIPANTS Infants born <28 weeks: Protocol group (n = 33) received at least 50% and not more than 150% of an intended standard dose of 0.5 mg/kg hydrocortisone twice daily for 7 days, then 0.5 mg/kg per day for 3 days; Non-Protocol group (n = 22), did not receive protocol hydrocortisone or completed <50% of the protocol dose. We assessed group differences in near-term diffusion MRI mean fractional anisotropy (FA) and mean diffusivity (MD) across the corticospinal tract, inferior longitudinal fasciculus, corpus callosum and superior cerebellar peduncle. RESULTS Groups were comparable in gestational age, post-menstrual age at scan, medical complications, bronchopulmonary dysplasia, and necrotizing enterocolitis. No significant large effect group differences were identified in mean FA or MD in any cerebral or cerebellar tract. CONCLUSION(S) Low dose, early, postnatal hydrocortisone was not associated with significant differences in white matter tract microstructure at near-term gestational age. IMPACT This study compared brain microstructural connectivity as a primary outcome among extremely preterm infants who did or did not receive early postnatal hydrocortisone. Low dose hydrocortisone in the first 10 days of life was not associated with significant differences in white matter microstructure in major cerebral and cerebellar pathways. Hydrocortisone did not have a significant effect on early brain white matter circuits.
Collapse
Affiliation(s)
- Sarah E Dubner
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Lucy Rickerich
- Program in Human Biology, Stanford University, Stanford, CA, USA
| | - Lisa Bruckert
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Rocío Velasco Poblaciones
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Dawson Sproul
- Program in Human Biology, Stanford University, Stanford, CA, USA
| | - Melissa Scala
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Heidi M Feldman
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Katherine E Travis
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
12
|
Gold A, Wray J, Kosmach-Park B, Bannister L, Wichart J, Graham A, Piotrowski C, Mayersohn G, Shellmer DA, Patterson C. Allied health and nursing practices in pediatric solid organ transplantation: An international survey. Pediatr Transplant 2024; 28:e14541. [PMID: 37550265 DOI: 10.1111/petr.14541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 02/09/2023] [Accepted: 03/22/2023] [Indexed: 08/09/2023]
Abstract
INTRODUCTION With improved survival in pediatric solid organ transplantation (SOT) care has focused on optimizing functional, developmental, and psychosocial outcomes, roles often supported by Allied Health and Nursing professionals (AHNP). However, there is a scarcity of research examining frameworks of clinical practice. METHODS The International Pediatric Transplant Association AHNP Committee developed and disseminated an online survey to transplant centers as a quality improvement project to explore AHNP practice issues. Participant responses were characterized using descriptive statistics, and free-text comments were thematically analyzed. Responses were compared across professional groups; Group 1: Advanced Practice Providers, Group 2: Nursing, Group 3: Allied Health. RESULTS The survey was completed by 119 AHNP from across the globe, with responses predominantly (78%) from North America. Half of respondents had been working in pediatric transplant for 11+ years. Two-thirds of respondents were formally funded to provide transplant care; however, of these not funded, over half (57%) were allied health, compared to just 6% of advance practice providers. Advanced practice/nursing groups typically provided care to one organ program, with allied health providing care for multiple organ programs. Resource constraints were barriers to practice across all groups and countries. CONCLUSION In this preliminary survey exploring AHNP roles, professionals provided a range of specialized clinical care. Challenges to practice were funding and breadth of care, highlighting the need for additional resources, alongside the development of clinical practice guidelines for defining, and supporting the role of AHNP within pediatric SOT. Professional organizations, such as IPTA, can offer professional advocacy.
Collapse
Affiliation(s)
- Anna Gold
- The Department of Psychology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Transplant and Regenerative Medicine Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jo Wray
- Heart and Lung Directorate, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Beverly Kosmach-Park
- Department of Transplant Surgery, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Philadelphia, USA
| | - Louise Bannister
- Transplant and Regenerative Medicine Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jenny Wichart
- Department of Pharmacy, Alberta Children's Hospital, Alberta Health Services, Calgary, Alberta, Canada
| | - Ashley Graham
- Transplant and Regenerative Medicine Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Occupational Science & Occupational Therapy, The University of Toronto, Toronto, Ontario, Canada
| | - Caroline Piotrowski
- Department of Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gillian Mayersohn
- St. Louis Children's Hospital, St. Louis, Missouri, USA
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Diana A Shellmer
- Department of Surgery, University of Pittsburgh, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Catherine Patterson
- Transplant and Regenerative Medicine Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Department of Rehabilitation Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physical Therapy, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Pardy C, Berkes S, D'Souza R, Fox G, Davidson JR, Yardley IE. Complete Resection of Necrotic Bowel Improves Survival in NEC Without Compromising Enteral Autonomy. J Pediatr Surg 2024; 59:206-210. [PMID: 37957101 DOI: 10.1016/j.jpedsurg.2023.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/09/2023] [Indexed: 11/15/2023]
Abstract
AIMS Controversy persists regarding operative strategy for necrotising enterocolitis (NEC). Some surgeons advocate resecting all necrotic bowel, whilst others defunction with a stoma, leaving diseased bowel in situ to preserve bowel length. We reviewed our institutional experience of both approaches. METHODS Neonates undergoing laparotomy for NEC May 2015-2019 were identified. Data extracted from electronic records included: demographics, neonatal Sequential Organ Failure Assessment (nSOFA) score at surgery, operative findings, and procedure performed. Neonates were assigned to two groups according to operative strategy: complete resection of necrotic bowel (CR) or necrotic bowel left in situ (LIS). Primary outcome was survival, and secondary outcome was enteral autonomy. Outcomes were compared between groups. RESULTS Fifty neonates were identified. Six were excluded: 4 with NEC totalis and 2 with no visible necrosis or histological confirmation of NEC. Of the 44 remaining neonates, 27 were in the CR group and 17 in the LIS group. 32 neonates survived to discharge (73%). On univariate analysis, survival was associated with lower nSOFA score (P = 0.003), complete resection of necrotic bowel (OR 9.0, 95% CI [1.94-41.65]), and being born outside the surgical centre (OR 5.11 [1.23-21.28]). On Cox regression multivariate analysis, complete resection was still strongly associated with survival (OR 4.87 [1.51-15.70]). 28 of the 32 survivors (88%) achieved enteral autonomy. There was no association between operative approach and enteral autonomy (P = 0.373), or time to achieve this. CONCLUSION Complete resection of necrotic bowel during surgery for NEC significantly improves likelihood of surviving without negatively impacting remaining bowel function. LEVEL OF EVIDENCE III.
Collapse
Affiliation(s)
- Caroline Pardy
- Evelina London Children's Hospital, London, United Kingdom.
| | | | - Rashmi D'Souza
- Evelina London Children's Hospital, London, United Kingdom
| | - Grenville Fox
- Evelina London Children's Hospital, London, United Kingdom
| | - Joseph R Davidson
- Evelina London Children's Hospital, London, United Kingdom; GOS-UCL Institute of Child Health, London, United Kingdom
| | - Iain E Yardley
- Evelina London Children's Hospital, London, United Kingdom; Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
14
|
Thomas R, Bijlsma MW, Gonçalves BP, Nakwa FL, Velaphi S, Heath PT. Long-term impact of serious neonatal bacterial infections on neurodevelopment. Clin Microbiol Infect 2024; 30:28-37. [PMID: 37084940 DOI: 10.1016/j.cmi.2023.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/27/2023] [Accepted: 04/15/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND Neonatal bacterial infections have long been recognized as an important cause of acute morbidity and mortality, but long-term neurodevelopmental consequences have not been comprehensively described and discussed. OBJECTIVES We aimed to summarize evidence on the pathogenesis, diagnosis, and epidemiology of long-term sequelae after neonatal bacterial sepsis and meningitis. We also discuss approaches for future studies to quantify the public health impact of neonatal infection-associated neurodevelopmental impairment. SOURCES We identified studies, both research articles and reviews, which provide mechanistic information on the long-term disease, as well as epidemiological studies that describe the frequency of neurodevelopmental impairment in children with and, for comparison, without a history of neonatal bacterial infection. Tools currently used in clinical practice and research settings to assess neurodevelopmental impairment were also reviewed. CONTENT We first enumerate potential direct and indirect mechanisms that can lead to brain injury following neonatal infections. We then discuss summary data, either frequencies or measures of association, from epidemiological studies. Risk factors that predict long-term outcomes are also described. Finally, we describe clinical approaches for identifying children with neurodevelopmental impairment and provide an overview of common diagnostic tools. IMPLICATIONS The limited number of studies that describe the long-term consequences of neonatal infections, often undertaken in high-income settings and using variable designs and diagnostic tools, are not sufficient to inform clinical practice and policy prioritization. Multi-country studies with follow-up into adolescence, standardized diagnostic approaches, and local comparator groups are needed, especially in low and middle-income countries where the incidence of neonatal sepsis is high.
Collapse
Affiliation(s)
- Reenu Thomas
- Department of Pediatrics and Child Health, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Merijn W Bijlsma
- Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands; Department of Pediatrics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | | | - Firdose L Nakwa
- Department of Pediatrics and Child Health, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sithembiso Velaphi
- Department of Pediatrics and Child Health, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Paul T Heath
- Centre for Neonatal and Paediatric Infection, St George's, University of London, London, UK
| |
Collapse
|
15
|
Garg PM, Pittman I, Yi J, Shetty A, Taylor C, Reddy K, Inder TE, Varshney N, Hillegass WB, Garg PP. Clinical correlates of cerebellar injury in preterm infants with surgical necrotizing enterocolitis. J Neonatal Perinatal Med 2024; 17:705-716. [PMID: 39269856 PMCID: PMC11489020 DOI: 10.3233/npm-240022] [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] [Indexed: 09/15/2024]
Abstract
BACKGROUND The preterm infants are at risk of cerebellar injury and the risk factors for necrotizing enterocolitis (NEC) associated cerebellar injury are not fully understood. AIM Determine the risk factors of cerebellar injury in infants with surgical necrotizing enterocolitis (NEC). METHODS Retrospective study compared clinical/pathological information between surgical NEC infants with and those without cerebellar injury detected on brain MRI obtained at term equivalent age. Cerebellar Injury patterns that we identified on MRI brain were cerebellar hemorrhage, siderosis and/or cerebellar volume loss. RESULTS Cerebellar injury (21/65, 32.3%) in preterm infants with NEC was associated with patent ductus arteriosus (PDA) (18/21(85.7%) vs. 25/44(56.8%); p = 0.021), blood culture positive sepsis (13/21 (61.9%) vs. 11/44 (25%); p = 0.004) following NEC, predominantly grew gram positive bacteria (9/21(42.9%) vs. 4/44(9.1%); p = 0.001), greater red cell transfusion, higher rates of cholestasis following NEC and differences in intestinal histopathology (more hemorrhagic and reparative lesions) on univariate analysis. Those with cerebellar injury had higher grade white matter injury (14/21 (66.7%) vs. 4/44(9.1%) p = 0.0005) and higher-grade ROP (70.6% vs. 38.5%; p = 0.027) than those without cerebellar injury.On multilogistic regression, the positive blood culture sepsis (OR 3.9, CI 1.1-13.7, p = 0.03), PDA (OR 4.5, CI 1.0-19.9, p = 0.04) and severe intestinal pathological hemorrhage (grade 3-4) (OR 16.9, CI 2.1-135.5, p = 0.007) were independently associated with higher risk of cerebellar injury. CONCLUSION Preterm infants with surgical NEC with positive blood culture sepsis, PDA, and severe intestinal hemorrhagic lesions (grade 3-4) appear at greater risk for cerebellar injury.
Collapse
Affiliation(s)
- P M Garg
- Department of Pediatrics/Neonatology, Wake Forest University, Winston Salem, NC, USA
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - I Pittman
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - J Yi
- Frank Porter Graham Child Development Institute, University of North Carolina at Chapel Hill, NC, USA
| | - A Shetty
- Department of Pediatrics/Infectious Disease, Wake Forest University, Winston Salem, NC, USA
| | - C Taylor
- Department of Radiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - K Reddy
- Department of Radiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - T E Inder
- Children Hospital of Orange County, University of California, Irvine, Orange, CA, USA
| | - N Varshney
- Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - W B Hillegass
- Department of Data Science, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - P P Garg
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
16
|
Garg PM, Pittman I, Taylor C, Reddy K, Varshney N, Hillegass WB, Shetty A, Yi J, Inder T, Garg P. Clinical Correlates of Cerebellar Injury in Preterm Infants with Surgical Necrotizing Enterocolitis. RESEARCH SQUARE 2023:rs.3.rs-3720723. [PMID: 38168331 PMCID: PMC10760219 DOI: 10.21203/rs.3.rs-3720723/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Objective Determine the risk factors of cerebellar injury in infants with surgical necrotizing enterocolitis (NEC). Methods Retrospective study compared clinical/pathological information between surgical NEC infants with and those without cerebellar injury. Results Infants with cerebellar injury (21/65, 32.3%) had significantly more hemorrhagic and the reparative lesions on the intestinal histopathology, had patent ductus arteriosus (PDA) more often, received red cell transfusion frequently, had blood culture positive sepsis and grew gram positive organisms more often and had cholestasis frequently following NEC than those without cerebellar injury. On multilogistic regression, the positive blood culture sepsis (OR 3.9, CI 1.1-13.7, p = 0.03), PDA (OR 4.5, CI 1.0-19.9, p = 0.04) and severe hemorrhage (grade 3-4)(OR 16.9, CI 2.1-135.5, p = 0.007) were independently associated with higher risk of cerebellar injury. Conclusion The cerebellar injury was most likely associated with positive blood culture sepsis following NEC, PDA, and severe hemorrhage lesions (grade 3-4) in infants with surgical NEC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Joe Yi
- 3. Frank Porter Graham Child Development Institute, University of North Carolina
| | | | | |
Collapse
|
17
|
Martinez M, Yu W, Menden HL, Lei T, Monaghan-Nichols P, Sampath V. Butyrate suppresses experimental necrotizing enterocolitis-induced brain injury in mice. Front Pediatr 2023; 11:1284085. [PMID: 38130941 PMCID: PMC10733464 DOI: 10.3389/fped.2023.1284085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
Background Necrotizing enterocolitis (NEC) is a devastating disease in premature infants, and 50% of infants with surgical NEC develop neurodevelopmental defects. The mechanisms by which NEC-induced cytokine release and activation of inflammatory cells in the brain mediate neuronal injury, and whether enteral immunotherapy attenuates NEC-associated brain injury remain understudied. Based on our prior work, which demonstrated that experimental NEC-like intestinal injury is attenuated by the short-chain fatty acid, butyrate, in this study, we hypothesize that NEC-induced brain injury would be suppressed by enteral butyrate supplementation. Methods A standardized NEC mouse model [enteral formula feeding, lipopolysaccharide (LPS), and hypoxia] was used. Mice were randomized into the following groups: control, NEC, butyrate pretreated NEC, and butyrate control. NEC scoring (1-4 with 4 representing severe injury) was performed on ileal sections using a validated scoring system. Intestinal and brain lysates were used to assess inflammation, proinflammatory signaling, and apoptosis. Results NEC-induced intestinal injury was attenuated by butyrate supplementation. NEC-induced microglial activation in the cerebral cortex and hippocampus was suppressed with butyrate. NEC increased the number of activated microglial cells but decreased the number of oligodendrocytes. Butyrate pretreatment attenuated these changes. Increased activation of proinflammatory Toll-like receptor signaling, cytokine expression, and induction of GFAP and IBA1 in the cerebral cortex observed with NEC was suppressed with butyrate. Conclusion Experimental NEC induced inflammation and activation of microglia in several regions of the brain, most prominently in the cortex. NEC-induced neuroinflammation was suppressed with butyrate pretreatment. The addition of short-chain fatty acids to diet may be used to attenuate NEC-induced intestinal injury and neuroinflammation in preterm infants.
Collapse
Affiliation(s)
- Maribel Martinez
- Division of Neonatology, Department of Pediatrics, Children’s Mercy Kansas City, Kansas, MO, United States
- Neonatal Diseases Research Program, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas, MO, United States
| | - Wei Yu
- Division of Neonatology, Department of Pediatrics, Children’s Mercy Kansas City, Kansas, MO, United States
- Neonatal Diseases Research Program, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas, MO, United States
| | - Heather L. Menden
- Division of Neonatology, Department of Pediatrics, Children’s Mercy Kansas City, Kansas, MO, United States
- Neonatal Diseases Research Program, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas, MO, United States
| | - Tianhua Lei
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas, MO, United States
| | - Paula Monaghan-Nichols
- Department of Biomedical Sciences, University of Missouri Kansas City School of Medicine, Kansas, MO, United States
| | - Venkatesh Sampath
- Division of Neonatology, Department of Pediatrics, Children’s Mercy Kansas City, Kansas, MO, United States
- Neonatal Diseases Research Program, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas, MO, United States
| |
Collapse
|
18
|
Kurul Ş, Beckers FLM, Vermeulen MJ, Suurland J, Hasbek JE, Ramakers CRB, Simons SHP, Reiss IKM, Taal HR. Inflammation, sepsis severity and neurodevelopmental outcomes of late-onset sepsis in preterm neonates. Pediatr Res 2023; 94:2026-2032. [PMID: 37468719 DOI: 10.1038/s41390-023-02742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/15/2023] [Accepted: 07/03/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND The aim of this study was to investigate the association between inflammatory biomarkers (C-reactive protein (CRP), procalcitonin (PCT) and interleukin-6 (IL-6)) and sepsis severity (neonatal-Sequential-Organ-Failure-Assessment (nSOFA)) and neurodevelopmental outcomes at 2 years, among very preterm neonates. METHODS Data on preterm neonates (gestational age <30 weeks) from 2016 until 2020 were reviewed. Outcomes of interest were NDI (no, mild, severe) and the motor and cognitive score on the Dutch-Bayley-Scales-of-Infant-and-Toddler-Development (Bayley-III-NL) assessed at the corrected age of 2 years. Logistic and linear regression analysis were used for categorical and continuous outcomes, respectively. All analyses were adjusted for gestational age, sex and birthweight-for-gestational-age SD-score. RESULTS In total 410 patients were eligible for analysis. Maximum CRP concentrations were associated with lower motor and cognitive scores (effect estimate -0.03 points,(95% CI -0.07; -0.00) and -0.03 points,(95% CI -0.06; -0.004), respectively) and increased risk of severe NDI (odds ratio (OR) 1.01, (95% CI 1.00; 1.01)). High nSOFA scores (≥4) during sepsis episodes were associated with an increased risk of mild NDI (OR 2.01, (95% CI 1.34; 3.03)). There were no consistent associations between IL-6, PCT and the outcomes of interest. CONCLUSION High CRP concentrations and sepsis severity in preterm neonates seem to be associated with neurodevelopmental outcomes in survivors at the age of 2 years. IMPACT STATEMENT The level of inflammation and sepsis severity are associated with neurodevelopmental outcome in preterm neonates at 2 years of corrected age. Sepsis is a major health issue in preterm neonates and can lead to brain damage and impaired neurodevelopment. Biomarkers can be determined to assess the level of inflammation. However, the relation of inflammatory biomarkers with neurodevelopmental outcome is not known. The level of inflammation and sepsis severity are related to neurodevelopmental outcome in preterm neonates. Maximum CRP concentration and high nSOFA scores are associated with an increased risk of neurodevelopmental impairment in survivors at the corrected age of 2 years.
Collapse
Affiliation(s)
- Şerife Kurul
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Famke L M Beckers
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Marijn J Vermeulen
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Jill Suurland
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jasmin E Hasbek
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | | | - Sinno H P Simons
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Irwin K M Reiss
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - H Rob Taal
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands.
| |
Collapse
|
19
|
Russ JB, Ostrem BEL. Acquired Brain Injuries Across the Perinatal Spectrum: Pathophysiology and Emerging Therapies. Pediatr Neurol 2023; 148:206-214. [PMID: 37625929 DOI: 10.1016/j.pediatrneurol.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/29/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
The development of the central nervous system can be directly disrupted by a variety of acquired factors, including infectious, inflammatory, hypoxic-ischemic, and toxic insults. Influences external to the fetus also impact neurodevelopment, including placental health, maternal comorbidities, adverse experiences, environmental exposures, and social determinants of health. Acquired perinatal brain insults tend to affect the developing brain in a stage-specific manner that reflects the susceptible cell types, developmental processes, and risk factors present at the time of the insult. In this review, we discuss the pathophysiology, neurodevelopmental outcomes, and management of common acquired perinatal brain conditions. In the fetal brain, we divide insults based on trimester, and in the postnatal brain, we focus on common pathologies that have a presentation dependent on gestational age at birth: white matter injury and germinal matrix hemorrhage/intraventricular hemorrhage in preterm infants and hypoxic-ischemic encephalopathy in term infants. Although specific treatments for fetal and newborn brain disorders are currently limited, we emphasize therapies in preclinical or early clinical phases of the development pipeline. The growing number of novel cell type- and stage-specific emerging therapies suggests that in the near future we may have a dramatically improved ability to treat acquired perinatal brain disorders and to mitigate the associated neurodevelopmental consequences.
Collapse
Affiliation(s)
- Jeffrey B Russ
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina
| | - Bridget E L Ostrem
- Department of Neurology, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
20
|
Mangalesh S, Toth CA. Preterm infant retinal OCT markers of perinatal health and retinopathy of prematurity. Front Pediatr 2023; 11:1238193. [PMID: 37808559 PMCID: PMC10551634 DOI: 10.3389/fped.2023.1238193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
The increasing survival of preterm infants has led to the importance of improving long-term outcomes associated with preterm birth. Antenatal and perinatal insults not only impact mortality, but also long-term disability. While in the intensive care nursery, preterm infants are also exposed to various stressors that lead to long-term cognitive deficits. It is therefore critical to identify early, low-stress, non-invasive biomarkers for preterm infant health. Optical coherence tomography (OCT) is a powerful imaging modality that has recently been adapted to the infant population and provides noninvasive, high-resolution, cross-sectional imaging of the infant eye at the bedside with low stress relative to conventional examination. In this review we delve into discussing the associations between preterm systemic health factors and OCT-based retinal findings and their potential contribution to the development of non-invasive biomarkers for infant health and for retinopathy of prematurity (ROP).
Collapse
Affiliation(s)
| | - Cynthia A. Toth
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
21
|
Chao AS, Matak P, Pegram K, Powers J, Hutson C, Jo R, Dubois L, Thompson JW, Smith PB, Jain V, Liu C, Younge NE, Rikard B, Reyes EY, Shinohara ML, Gregory SG, Goldberg RN, Benner EJ. 20-αHydroxycholesterol, an oxysterol in human breast milk, reverses mouse neonatal white matter injury through Gli-dependent oligodendrogenesis. Cell Stem Cell 2023; 30:1054-1071.e8. [PMID: 37541211 PMCID: PMC10625465 DOI: 10.1016/j.stem.2023.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 05/21/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023]
Abstract
White matter injuries (WMIs) are the leading cause of neurologic impairment in infants born premature. There are no treatment options available. The most common forms of WMIs in infants occur prior to the onset of normal myelination, making its pathophysiology distinctive, thus requiring a tailored approach to treatment. Neonates present a unique opportunity to repair WMIs due to a transient abundance of neural stem/progenitor cells (NSPCs) present in the germinal matrix with oligodendrogenic potential. We identified an endogenous oxysterol, 20-αHydroxycholesterol (20HC), in human maternal breast milk that induces oligodendrogenesis through a sonic hedgehog (shh), Gli-dependent mechanism. Following WMI in neonatal mice, injection of 20HC induced subventricular zone-derived oligodendrogenesis and improved myelination in the periventricular white matter, resulting in improved motor outcomes. Targeting the oligodendrogenic potential of postnatal NSPCs in neonates with WMIs may be further developed into a novel approach to mitigate this devastating complication of preterm birth.
Collapse
Affiliation(s)
- Agnes S Chao
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Pavle Matak
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Kelly Pegram
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - James Powers
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Collin Hutson
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Rebecca Jo
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Laura Dubois
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomics and Computational Biology, School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - J Will Thompson
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomics and Computational Biology, School of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - P Brian Smith
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Vaibhav Jain
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Noelle E Younge
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Blaire Rikard
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Estefany Y Reyes
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Simon G Gregory
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Ronald N Goldberg
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Eric J Benner
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
22
|
Gravina G, Ardalan M, Chumak T, Nilsson AK, Ek JC, Danielsson H, Svedin P, Pekny M, Pekna M, Sävman K, Hellström A, Mallard C. Proteomics identifies lipocalin-2 in neonatal inflammation associated with cerebrovascular alteration in mice and preterm infants. iScience 2023; 26:107217. [PMID: 37496672 PMCID: PMC10366453 DOI: 10.1016/j.isci.2023.107217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/07/2023] [Accepted: 06/22/2023] [Indexed: 07/28/2023] Open
Abstract
Staphylococcus (S.) epidermidis is the most common nosocomial coagulase-negative staphylococci infection in preterm infants. Clinical signs of infection are often unspecific and novel markers to complement diagnosis are needed. We investigated proteomic alterations in mouse brain after S. epidermidis infection and in preterm infant blood. We identified lipocalin-2 (LCN2) as a crucial protein associated with cerebrovascular changes and astrocyte reactivity in mice. We further proved that LCN2 protein expression was associated with endothelial cells but not astrocyte reactivity. By combining network analysis and differential expression approaches, we identified LCN2 linked to blood C-reactive protein levels in preterm infants born <28 weeks of gestation. Blood LCN2 levels were associated with similar alterations of cytokines and chemokines in both infected mice and human preterm infants with increased levels of C-reactive protein. This experimental and clinical study suggests that LCN2 may be a marker of preterm infection/inflammation associated with cerebrovascular changes and neuroinflammation.
Collapse
Affiliation(s)
- Giacomo Gravina
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryam Ardalan
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Translational Neuropsychiatric Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tetyana Chumak
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders K. Nilsson
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joakim C. Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hanna Danielsson
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Sach’s Children’s and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Pernilla Svedin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- University of Newcastle, Newcastle, NSW, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Marcela Pekna
- University of Newcastle, Newcastle, NSW, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Laboratory of Regenerative Neurobiology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Karin Sävman
- Department of Pediatrics, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska Academy, Gothenburg, Sweden
- Region Västra Götaland, Department of Neonatology, The Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ann Hellström
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
23
|
Fontana C, Schiavolin P, Ardemani G, Amerotti DA, Pesenti N, Bonfanti C, Boggini T, Gangi S, Porro M, Squarza C, Giannì ML, Persico N, Mosca F, Fumagalli M. To be born twin: effects on long-term neurodevelopment of very preterm infants-a cohort study. Front Pediatr 2023; 11:1217650. [PMID: 37528875 PMCID: PMC10389041 DOI: 10.3389/fped.2023.1217650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/26/2023] [Indexed: 08/03/2023] Open
Abstract
Objective To examine the effect of twin birth on long-term neurodevelopmental outcomes in a cohort of Italian preterm infants with very low birth weight. Study design We performed a retrospective cohort study on children born in a tertiary care centre. We included children born between 1 January 2007 and 31 December 2013 with a gestational age (GA) of ≤32 weeks and birth weight of <1,500 g. The infants born from twin pregnancies complicated by twin-to-twin transfusion syndrome and from higher-order multiple pregnancies were excluded. The children were evaluated both at 2 years corrected age and 5 years chronological age with Griffiths mental development scales revised (GMDS-R). The linear mixed effects models were used to study the effect of being a twin vs. being a singleton on GMDS-R scores, adjusting for GA, being born small for gestational age, sex, length of NICU stay, socio-economic status, and comorbidity score (CS) calculated as the sum of the weights associated with each of the major morbidities of the infants. Results A total of 301 children were included in the study, of which 189 (62.8%) were singletons and 112 (37.2%) were twins; 23 out of 112 twins were monochorionic (MC). No statistically significant differences were observed between twins and singletons in terms of mean general quotient and subscales at both 2 and 5 years. No effect of chorionicity was found when comparing scores of MC and dichorionic twins vs. singletons; however, after adjusting for the CS, the MC twins showed lower scores in the hearing and language and performance subscales at 5 years. Conclusion Overall, in our cohort of children born very preterm, twin infants were not at higher risk of neurodevelopmental impairment compared with singletons at pre-school age.
Collapse
Affiliation(s)
- Camilla Fontana
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Paola Schiavolin
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Giulia Ardemani
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | - Nicola Pesenti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
- Department of Statistics and Quantitative Methods, Division of Biostatistics, Epidemiology and Public Health, University of Milano-Bicocca, Milan, Italy
| | - Chiara Bonfanti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Tiziana Boggini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Silvana Gangi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Matteo Porro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pediatric Physical Medicine and Rehabilitation Service, Milan, Italy
| | - Chiara Squarza
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Maria Lorella Giannì
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Nicola Persico
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Prenatal Diagnosis and Fetal Surgery Unit, Milan, Italy
| | - Fabio Mosca
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Monica Fumagalli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
24
|
Soltani Kouhbanani S, Arabi SM, Zarenezhad S. Does the Frontal Brain Electrical Activity Mediate the Effect of Home Executive Function Environment and Screen Time on Children's Executive Function? J Genet Psychol 2023; 184:430-445. [PMID: 37335540 DOI: 10.1080/00221325.2023.2223653] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Executive functions play an important role in various developmental aspects of children; however, environmental factors influencing individual differences in children's executive function and their neural substructures, particularly in middle childhood, are rarely investigated. Therefore, the current study aimed to investigate the relationship between the home executive function environment (HEFE) and screen time with the executive function of children aged 8-12 years by employing the mediating variables of alpha, beta, and theta waves. The parents of 133 normal children completed Barkley Deficits in Executive Functioning, HEFE, and Screen Time Scales. Alpha, beta, and theta brain waves were also measured. Data were examined using correlational and path analysis. The results suggested a positive and significant relationship between home executive functions and the executive functions of children. Furthermore, the results indicated an inverse and significant relationship between screen time and executive function. The results also proved the mediating role of alpha, beta, and theta brain waves in the relationship between screen time and the children's executive function. Environmental factors (such as home environment and screen time) affect the function of brain waves and, thus, the daily executive function of children.
Collapse
Affiliation(s)
- Sakineh Soltani Kouhbanani
- Department of Educational Sciences, Educational Sciences and Psychology Faculty, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyedeh Manizheh Arabi
- Department of Motor Behavior, Faculty of Sports Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Somayeh Zarenezhad
- Department of Educational Sciences, Educational Sciences and Psychology Faculty, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
25
|
Mesfin FM, Manohar K, Shelley WC, Brokaw JP, Liu J, Ma M, Markel TA. Stem cells as a therapeutic avenue for active and long-term complications of Necrotizing Enterocolitis. Semin Pediatr Surg 2023; 32:151311. [PMID: 37276782 PMCID: PMC10330659 DOI: 10.1016/j.sempedsurg.2023.151311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating neonatal intestinal disease associated with significant morbidity and mortality. Although decades of research have been dedicated to understanding the pathogenesis of NEC and developing therapies, it remains the leading cause of death among neonatal gastrointestinal diseases. Mesenchymal stem cells (MSCs) have garnered significant interest recently as potential therapeutic agents for the treatment of NEC. They have been shown to rescue intestinal injury and reduce the incidence and severity of NEC in various preclinical animal studies. MSCs and MSC-derived organoids and tissue engineered small intestine (TESI) have shown potential for the treatment of long-term sequela of NEC such as short bowel syndrome, neurodevelopmental delay, and chronic lung disease. Although the advances made in the use of MSCs are promising, further research is needed prior to the widespread use of these cells for the treatment of NEC.
Collapse
Affiliation(s)
- Fikir M Mesfin
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Krishna Manohar
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - W Christopher Shelley
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John P Brokaw
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jianyun Liu
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Troy A Markel
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Riley Hospital for Children at Indiana University Health, Indianapolis, IN, USA.
| |
Collapse
|
26
|
Efstathiou N, Soubasi V, Koliakos G, Kantziou K, Kyriazis G, Slavakis A, Dermentzoglou V, Michalettou I, Drosou-Agakidou V. Beyond brain injury biomarkers: chemoattractants and circulating progenitor cells as biomarkers of endogenous rehabilitation effort in preterm neonates with encephalopathy. Front Pediatr 2023; 11:1151787. [PMID: 37292373 PMCID: PMC10244884 DOI: 10.3389/fped.2023.1151787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/26/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction Preclinical work and studies in adults have shown that endogenous regeneration efforts that involve mobilization of progenitor cells take place after brain injury. However, kinetics of endogenous circulating progenitor cells (CPCs) in preterm neonates is not well described, particularly their possible role regarding brain injury and regeneration. We aimed to assess the kinetics of CPCs in neonates with encephalopathy of prematurity in relation to brain injury biomarkers, chemoattractants and relevant antenatal and postanal clinical factors, in an effort to outline the related pathophysiology. Materials and methods 47 preterm neonates (of 28-33 weeks GA) were enrolled: 31 newborns with no or minimal brain injury (grade I IVH) and 16 prematures with encephalopathy (grade III or IV IVH, PVL or infarct). Peripheral blood samples obtained on days 1, 3, 9, 18 and 45 after birth were analyzed using flow cytometry, focusing on EPCs (early and late Endothelial Progenitor Cells), HSCs (Hematopoietic Stem Cells) and VSELs (Very Small Embryonic-Like Stem Cells). At the same time-points serum levels of S100B, Neuron-specific Enolase (NSE), Erythropoietin (EPO), Insulin-like growth factor-1 (IGF-1) and SDF-1 were also measured. Neonates were assessed postnatally with brain MRI, and with Bayley III developmental test at 2 years of corrected age. Results Preterms with brain injury proved to have significant increase of S100B and NSE, followed by increase of EPO and enhanced mobilization mainly of HSCs, eEPCs and lEPCs. IGF-1 was rather decreased in this group of neonates. IGF-1 and most CPCs were intense decreased in cases of antenatal or postnatal inflammation. S100B and NSE correlated with neuroimaging and language scale in Bayley III test, providing good prognostic ability. Conclusion The observed pattern of CPCs' mobilization and its association with neurotrophic factors following preterm brain injury indicate the existence of an endogenous brain regeneration process. Kinetics of different biomarkers and associations with clinical factors contribute to the understanding of the related pathophysiology and might help to early discriminate neonates with adverse outcome. Timely appropriate enhancement of the endogenous regeneration effort, when it is suppressed and insufficient, using neurotrophic factors and exogenous progenitor cells might be a powerful therapeutic strategy in the future to restore brain damage and improve the neurodevelopmental outcome in premature infants with brain injury.
Collapse
Affiliation(s)
- N. Efstathiou
- 1st Neonatal Department and NICU, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - V. Soubasi
- 2nd Neonatal Department and NICU, Papageorgiou General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - G. Koliakos
- Biochemistry Department, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - K. Kantziou
- 1st Neonatal Department and NICU, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - G. Kyriazis
- Immunology Laboratory, Pulmonology Department, Papanikolaou General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - A. Slavakis
- Biochemistry Department, Hippokration General Hospital, Thessaloniki, Greece
| | - V. Dermentzoglou
- Child Radiologist, Radiology Department, Agia Sofia Pediatric Hospital, Athens, Greece
| | - I. Michalettou
- Child Occupational Τherapist, Hippokration General Hospital, Thessaloniki, Greece
| | - V. Drosou-Agakidou
- 1st Neonatal Department and NICU, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
27
|
Hess S, Poryo M, Böttger R, Franz A, Klotz D, Linnemann K, Ott T, Pöschl J, Schroth M, Stein A, Ralser E, Reutter H, Thome UH, Wieg C, Ehrlich A, Ruckes C, Wagenpfeil S, Zemlin M, Papan C, Simon A, Bay J, Meyer S. Umbilical venous catheter- and peripherally inserted central catheter-associated complications in preterm infants with birth weight < 1250 g : Results from a survey in Austria and Germany. Wien Med Wochenschr 2023; 173:161-167. [PMID: 35939216 PMCID: PMC10147741 DOI: 10.1007/s10354-022-00952-z] [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: 05/12/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND OBJECTIVE Umbilical venous catheters (UVC) and peripherally inserted central catheters (PICC) are commonly used in preterm infants but have been associated with a number of serious complications. We performed a survey in Austria and Germany to assess the use of UVCs and PICCs in preterm infants with a birth weight < 1250 g and associated rates of catheter-related adverse events. METHODS Electronic survey of participating centers of the NeoVitaA trial. Main outcome parameter was the reported rates of UVC- and PICC-associated complications (infection, thrombosis, emboli, organ injury, arrhythmia, dislocation, miscellaneous). RESULTS In total, 20 neonatal intensive care units (NICU) providing maximal intensive care in Austria and Germany (level I) were contacted, with a senior neonatologist response rate of 12/20 (60%). The reported rates for UVC with a dwell time of 1-10 days were bacterial infection: 4.2 ± 3.4% (range 0-10%); thrombosis: 7.3 ± 7.1% (0-20%); emboli: 0.9 ± 2.0% (0-5%); organ injury: 1.1 ± 1.9% (0-5%); cardiac arrhythmia: 2.2 ± 2.5% (0-5%); and dislocation: 5.4 ± 8.7% (0-30%); and for PICCs with a dwell time of 1-14 days bacterial infection: 15.0 ± 3.4% (range 2.5-30%); thrombosis; 4.3 ± 3.5% (0-10%); emboli: 0.8 ± 1.6% (0-5%); organ injury: 1.5 ± 2.3% (0-5%); cardiac arrhythmia: 1.5 ± 2.3% (0-5%), and dislocation: 8.5 ± 4.6% (0-30%). CONCLUSION The catheter-related complication rates reported in this survey differed between UVCs and PICCs and were higher than those reported in the literature. To generate more reliable data on this clinically important issue, we plan to perform a large prospective multicenter randomized controlled trial investigating the non-inferiority of a prolonged UVC dwell time (up to 10 days) against the early change (up to 5 days) to a PICC.
Collapse
Affiliation(s)
- Steffi Hess
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, Kirrberger Str., Building 9, 66421, Homburg, Germany
| | - Martin Poryo
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg, Germany
| | - Ralf Böttger
- University Children's Hospital, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Axel Franz
- University Children's Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Daniel Klotz
- Department of Neonatology, Center for Pediatrics, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Knud Linnemann
- University Children's Hospital, Greifswald University Hospital, Greifswald, Germany
| | - Torsten Ott
- University Children's Hospital, University Hospital Muenster, Münster, Germany
| | - Johannes Pöschl
- University Children's Hospital, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Anja Stein
- University Children's Hospital, Essen University Hospital, Essen, Germany
| | - Elisabeth Ralser
- University Children's Hospital, Medical University of Innsbruck, Innsbruck, Austria
| | - Heiko Reutter
- Division of Neonatology and Pediatric Intensive Care Medicine, Department of Pediatric and Adolescent Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Ulrich H Thome
- University Children's Hospital, University of Leipzig Medical Center, Leipzig, Germany
| | - Christian Wieg
- Children's Hospital Aschaffenburg, Aschaffenburg, Germany
| | - Anne Ehrlich
- Interdisziplinäres Zentrum für Klinische Studien (IZKS), Johannes Gutenberg-Universität, Mainz, Germany
| | - Christian Ruckes
- Interdisziplinäres Zentrum für Klinische Studien (IZKS), Johannes Gutenberg-Universität, Mainz, Germany
| | - Stefan Wagenpfeil
- Institute for Medical Biometry, Epidemiology and Medical Informatics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, Kirrberger Str., Building 9, 66421, Homburg, Germany
| | - Cihan Papan
- Department of Medical Microbiology and Hospital Hygiene, Saarland University Medical Center, Homburg, Germany
| | - Arne Simon
- Department of Pediatric Hematology and Oncology, Infectious Diseases, Saarland University Medical Center, Homburg, Germany
| | - Johannes Bay
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, Kirrberger Str., Building 9, 66421, Homburg, Germany
| | - Sascha Meyer
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, Kirrberger Str., Building 9, 66421, Homburg, Germany.
| |
Collapse
|
28
|
Conole ELS, Vaher K, Cabez MB, Sullivan G, Stevenson AJ, Hall J, Murphy L, Thrippleton MJ, Quigley AJ, Bastin ME, Miron VE, Whalley HC, Marioni RE, Boardman JP, Cox SR. Immuno-epigenetic signature derived in saliva associates with the encephalopathy of prematurity and perinatal inflammatory disorders. Brain Behav Immun 2023; 110:322-338. [PMID: 36948324 DOI: 10.1016/j.bbi.2023.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/12/2023] [Accepted: 03/16/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND Preterm birth is closely associated with a phenotype that includes brain dysmaturation and neurocognitive impairment, commonly termed Encephalopathy of Prematurity (EoP), of which systemic inflammation is considered a key driver. DNA methylation (DNAm) signatures of inflammation from peripheral blood associate with poor brain imaging outcomes in adult cohorts. However, the robustness of DNAm inflammatory scores in infancy, their relation to comorbidities of preterm birth characterised by inflammation, neonatal neuroimaging metrics of EoP, and saliva cross-tissue applicability are unknown. METHODS Using salivary DNAm from 258 neonates (n = 155 preterm, gestational age at birth 23.28 - 34.84 weeks, n = 103 term, gestational age at birth 37.00 - 42.14 weeks), we investigated the impact of a DNAm surrogate for C-reactive protein (DNAm CRP) on brain structure and other clinically defined inflammatory exposures. We assessed i) if DNAm CRP estimates varied between preterm infants at term equivalent age and term infants, ii) how DNAm CRP related to different types of inflammatory exposure (maternal, fetal and postnatal) and iii) whether elevated DNAm CRP associated with poorer measures of neonatal brain volume and white matter connectivity. RESULTS Higher DNAm CRP was linked to preterm status (-0.0107 ± 0.0008, compared with -0.0118 ± 0.0006 among term infants; p < 0.001), as well as perinatal inflammatory diseases, including histologic chorioamnionitis, sepsis, bronchopulmonary dysplasia, and necrotising enterocolitis (OR range |2.00 | to |4.71|, p < 0.01). Preterm infants with higher DNAm CRP scores had lower brain volume in deep grey matter, white matter, and hippocampi and amygdalae (β range |0.185| to |0.218|). No such associations were observed for term infants. Association magnitudes were largest for measures of white matter microstructure among preterms, where elevated epigenetic inflammation associated with poorer global measures of white matter integrity (β range |0.206| to |0.371|), independent of other confounding exposures. CONCLUSIONS Inflammatory-related DNAm captures the allostatic load of inflammatory burden in preterm infants. Such DNAm measures complement biological and clinical metrics when investigating the determinants of neurodevelopmental differences.
Collapse
Affiliation(s)
- Eleanor L S Conole
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK; Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK.
| | - Kadi Vaher
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Manuel Blesa Cabez
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Gemma Sullivan
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Anna J Stevenson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Jill Hall
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Lee Murphy
- Edinburgh Clinical Research Facility, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Michael J Thrippleton
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Edinburgh Clinical Research Facility, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Alan J Quigley
- Imaging Department, Royal Hospital for Children and Young People, Edinburgh, EH16 4TJ, UK
| | - Mark E Bastin
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Veronique E Miron
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Heather C Whalley
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - James P Boardman
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; MRC Centre for Reproductive Health, Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Simon R Cox
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK.
| |
Collapse
|
29
|
Hess S, Poryo M, Ruckes C, Papan C, Ehrlich A, Ebrahimi-Fakhari D, Bay JS, Wagenpfeil S, Simon A, Meyer S. Assessment of an umbilical venous catheter dwell-time of 8-14 days versus 1-7 days in very low birth weight infacts (UVC - You Will See): a pilot single-center, randomized controlled trial. Early Hum Dev 2023; 179:105752. [PMID: 36958105 DOI: 10.1016/j.earlhumdev.2023.105752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND Umbilical venous catheters (UVCs) are used for central vascular access in preterm infants, but controversy exits with regard to the optimum dwell-time. PATIENTS AND METHODS Prospective, randomized controlled trial at a level III University neonatal intensive care unit (NICU), comparing a UVC dwell-time of 1-7 days (control group) to 8-14 days (intervention group) in very low birth weight (VLBW) infants. PRIMARY OUTCOME PARAMETER Number of infants requiring additional peripherally inserted central catheters (PICC) after removal of UVC. SECONDARY OUTCOME PARAMETERS Total number of central lines (CL = UVC and PICCs) until time point of full enteral feeds (130-160 mL/kg/d), total number of intravenous vascular catheters, number of CL-associated complications (infection, thrombosis/emboli, organ injury, secondary CL dislocation), number of X-rays for assessment of CL positioning, and days of therapy (DOT) (teicoplanin) for CL-associated blood stream infections (CLABSI). RESULTS Of 116 patients screened for eligibility, 63 patients were enrolled - control group: 31 infants, mean gestational age (GA) 280 weeks (standard deviation (SD) 2.6 weeks), mean birth weight (BW) 988.9 g (SD 322.0 g); intervention group: 32 infants, mean GA 285 weeks (SD 3.0 weeks), mean BW 1078.9 g (SD 324.6 g). In the control group, 28 infants required additional PICCs versus 16 in the intervention group (p < 0.001); total number of CLs: control group n = 58 versus intervention group n = 28; p < 0.001, and the total number of venous vascular devices was also significantly higher in the control group (109 versus 61; p = 0.04). No significant differences were seen with regard to CL-associated complications (p = 0.09). The number of X-rays for assessment of correct CL-position significantly lower in the intervention group (144 versus 96; p = 0.03). In the intervention group, length of hospital stay was significantly shorter (88.1 (SD: 35.3 days) versus 68.1 (SD: 32.6 days); p = 0.03) and GA significantly lower at discharge from the hospital (404: SD: 33 weeks) versus 385: SD: 25 weeks; p = 0.02. No differences existed with regard to neonatal morbidities and mortality at 36 weeks gestational age. CONCLUSIONS A longer UVC dwell-time of up to 14 days significantly decreased the number of painful invasive vascular procedures and radiation exposure, and shortened the length of the hospital stay. The findings of our pilot study should be confirmed in a larger, multi-center RCT with the primary focus on catheter-associated complications.
Collapse
Affiliation(s)
- Steffi Hess
- Saarland University Medical Center, Department of General Pediatrics and Neonatology, Homburg, Germany
| | - Martin Poryo
- Saarland University Medical Center, Department of Pediatric Cardiology, Homburg, Germany
| | - Christian Ruckes
- Interdisziplinäres Zentrum für Klinische Studien (IZKS), Iohannes Gutenberg-University, Mainz, Germany
| | - Cihan Papan
- Center for Infectious Diseases, Institute of Medical Microbiology and Hygiene, Saarland University, Homburg, Germany; Institute for Hygiene and Public Health, University Hospital Bonn, Bonn, Germany
| | - Anne Ehrlich
- Interdisziplinäres Zentrum für Klinische Studien (IZKS), Iohannes Gutenberg-University, Mainz, Germany
| | | | - Johannes Saaradonna Bay
- Saarland University Medical Center, Department of General Pediatrics and Neonatology, Homburg, Germany
| | - Stefan Wagenpfeil
- Saarland University Medical Center, Institute for Medical Biometry, Epidemiology, and Medical Informatics (IMBEI), Homburg, Germany
| | - Arne Simon
- Saarland University Medical Center, Department of Pediatric Hematology and Oncology, Infectious Diseases, Homburg, Germany
| | - Sascha Meyer
- Saarland University Medical Center, Department of General Pediatrics and Neonatology, Homburg, Germany; Franz-Lust Klinik für Kinder- und Jgendmedizin, Karlsruhe, Germany.
| |
Collapse
|
30
|
França A. The Role of Coagulase-Negative Staphylococci Biofilms on Late-Onset Sepsis: Current Challenges and Emerging Diagnostics and Therapies. Antibiotics (Basel) 2023; 12:antibiotics12030554. [PMID: 36978421 PMCID: PMC10044083 DOI: 10.3390/antibiotics12030554] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Infections are one of the most significant complications of neonates, especially those born preterm, with sepsis as one of the principal causes of mortality. Coagulase-negative staphylococci (CoNS), a group of staphylococcal species that naturally inhabit healthy human skin and mucosa, are the most common cause of late-onset sepsis, especially in preterms. One of the risk factors for the development of CoNS infections is the presence of implanted biomedical devices, which are frequently used for medications and/or nutrient delivery, as they serve as a scaffold for biofilm formation. The major concerns related to CoNS infections have to do with the increasing resistance to multiple antibiotics observed among this bacterial group and biofilm cells’ increased tolerance to antibiotics. As such, the treatment of CoNS biofilm-associated infections with antibiotics is increasingly challenging and considering that antibiotics remain the primary form of treatment, this issue will likely persist in upcoming years. For that reason, the development of innovative and efficient therapeutic measures is of utmost importance. This narrative review assesses the current challenges and emerging diagnostic tools and therapies for the treatment of CoNS biofilm-associated infections, with a special focus on late-onset sepsis.
Collapse
Affiliation(s)
- Angela França
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- LABBELS—Associate Laboratory in Biotechnology and Bioengineering and Microelectromechanical Systems, Braga and Guimarães, Portugal
| |
Collapse
|
31
|
Sugita K, Muto M, Murakami M, Yano K, Harumatsu T, Onishi S, Yamada K, Yamada W, Matsukubo M, Kawano T, Machigashira S, Torikai M, Ishihara C, Tokuhisa T, Ibara S, Ieiri S. Does protocol miconazole administration improve mortality and morbidity on surgical necrotizing enterocolitis? Pediatr Surg Int 2023; 39:102. [PMID: 36738350 DOI: 10.1007/s00383-023-05390-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/26/2023] [Indexed: 02/05/2023]
Abstract
PURPOSE Our previous clinical pilot study reported that miconazole (MCZ) prevented morbidity from surgical necrotizing enterocolitis (NEC). The present study re-investigated this effect in a long-term cohort over 20 years. METHODS We conducted a retrospective cohort study from April 1998 to March 2020. A total of 1169 extremely low-birth-weight infants (ELBWIs) admitted to our neonatal intensive care unit, including 45 with NEC (3.8%), underwent surgery. Since 2002, protocol MCZ administration for 3 weeks has been applied for neonates born before 26 weeks' gestation or weighing under 1000 g. We compared the background characteristics and clinical outcomes between patients with and without MCZ administration. RESULTS The morbidity rate decreased after applying the MCZ protocol, but no improvement in mortality was seen. A propensity score-matched analysis indicated that treated patients by MCZ showed a delay in developing surgical NEC by 12 days. The MCZ protocol also helped increase body weight at surgery. Prophylactic MCZ administration did not improve the neurological development of the language-social and postural-motor domains in the surgical NEC patients. But cognitive-adaptive domain caught up by a chronological age of 3 years old. CONCLUSIONS Revising the protocol to extend the dosing period may improve the outcomes of surgical NEC after the onset.
Collapse
Affiliation(s)
- Koshiro Sugita
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Mitsuru Muto
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Masakazu Murakami
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Keisuke Yano
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Toshio Harumatsu
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Shun Onishi
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Koji Yamada
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Waka Yamada
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Makoto Matsukubo
- Department of Pediatric Surgery, Kagoshima City Hospital, Kagoshima, Japan
| | - Takafumi Kawano
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Seiro Machigashira
- Department of Pediatric Surgery, Kagoshima City Hospital, Kagoshima, Japan
| | - Motofumi Torikai
- Department of Pediatric Surgery, Kagoshima City Hospital, Kagoshima, Japan
| | - Chie Ishihara
- Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Takuya Tokuhisa
- Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Satoshi Ibara
- Department of Neonatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Satoshi Ieiri
- Department of Pediatric Surgery, Research Field in Medical and Health Sciences, Medical and Dental Area, Research and Education Assembly, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan.
| |
Collapse
|
32
|
Shen LL, Mangalesh S, Michalak SM, McGeehan B, Sarin N, Finkle J, Winter KP, Tran-Viet D, Benner EJ, Vajzovic L, Freedman SF, Younge N, Cotten CM, El-Dairi M, Ying GS, Toth C. Associations between systemic health and retinal nerve fibre layer thickness in preterm infants at 36 weeks postmenstrual age. Br J Ophthalmol 2023; 107:242-247. [PMID: 34389548 PMCID: PMC8858642 DOI: 10.1136/bjophthalmol-2021-319254] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/30/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND/AIMS Neonatal insults from systemic diseases have been implicated in the pathway of impaired neurodevelopment in preterm infants. We aimed to investigate the associations between systemic health factors and retinal nerve fibre layer (RNFL) thickness in preterm infants. METHODS We prospectively enrolled infants and imaged both eyes at 36±1 weeks postmenstrual age (PMA) using a hand-held optical coherence tomography system at the bedside in the Duke intensive care nurseries. We evaluated associations between RNFL thickness and 29 systemic health factors using univariable and multivariable regression models. RESULTS 83 infants with RNFL thickness measures were included in this study. Based on the multivariable model, RNFL thickness was positively associated with infant weight at imaging and was negatively associated with sepsis/necrotising enterocolitis (NEC). RNFL thickness was 10.4 µm (95% CI -15.9 to -4.9) lower in infants with than without sepsis/NEC in the univariable analysis (p<0.001). This difference remained statistically significant after adjustment for confounding variables in various combinations (birth weight, birthweight percentile, gestational age, infant weight at imaging and growth velocity). A 250 g increase in infant weight at imaging was associated with a 3.1 µm (95% CI 2.1 to 4.2) increase in RNFL thickness in the univariable analysis (p<0.001). CONCLUSIONS Low infant weight and sepsis/NEC were independently associated with thinner RNFL in preterm infants at 36 weeks PMA. To our knowledge, this study is the first to suggest that sepsis/NEC may affect retinal neurodevelopment. Future longitudinal studies are needed to investigate this relationship further.
Collapse
Affiliation(s)
- Liangbo L Shen
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA
| | - Shwetha Mangalesh
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Suzanne M Michalak
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Brendan McGeehan
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Neeru Sarin
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Joanne Finkle
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Katrina P Winter
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Du Tran-Viet
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Eric J Benner
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Lejla Vajzovic
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sharon F Freedman
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Noelle Younge
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - C Michael Cotten
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Mays El-Dairi
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Gui-Shuang Ying
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cynthia Toth
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| |
Collapse
|
33
|
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
|
34
|
Wang Y, Hang C, Hu J, Li C, Zhan C, Pan J, Yuan T. Role of gut-brain axis in neurodevelopmental impairment of necrotizing enterocolitis. Front Neurosci 2023; 17:1059552. [PMID: 36743802 PMCID: PMC9894661 DOI: 10.3389/fnins.2023.1059552] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a common gastrointestinal disease of preterm infants with high morbidity and mortality. In survivors of NEC, one of the leading causes of long-term morbidity is the development of severe neurocognitive injury. The exact pathogenesis of neurodevelopmental delay in NEC remains unknown, but microbiota is considered to have dramatic effects on the development and function of the host brain via the gut-brain axis. In this review, we discuss the characteristics of microbiota of NEC, the impaired neurological outcomes, and the role of the complex interplay between the intestinal microbiota and brain to influence neurodevelopment in NEC. The increasing knowledge of microbial-host interactions has the potential to generate novel therapies for manipulating brain development in the future.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China
| | - Chengcheng Hang
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China
| | - Jun Hu
- Department of Surgical Intensive Care Unit, Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Chen Li
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China
| | - Canyang Zhan
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China
| | - Jiarong Pan
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China
| | - Tianming Yuan
- Department of Neonatology, Children’s Hospital of Zhejiang University, Hangzhou, China,*Correspondence: Tianming Yuan,
| |
Collapse
|
35
|
Manohar K, Mesfin FM, Liu J, Shelley WC, Brokaw JP, Markel TA. Gut-Brain cross talk: The pathogenesis of neurodevelopmental impairment in necrotizing enterocolitis. Front Pediatr 2023; 11:1104682. [PMID: 36873645 PMCID: PMC9975605 DOI: 10.3389/fped.2023.1104682] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating condition of multi-factorial origin that affects the intestine of premature infants and results in high morbidity and mortality. Infants that survive contend with several long-term sequelae including neurodevelopmental impairment (NDI)-which encompasses cognitive and psychosocial deficits as well as motor, vision, and hearing impairment. Alterations in the gut-brain axis (GBA) homeostasis have been implicated in the pathogenesis of NEC and the development of NDI. The crosstalk along the GBA suggests that microbial dysbiosis and subsequent bowel injury can initiate systemic inflammation which is followed by pathogenic signaling cascades with multiple pathways that ultimately lead to the brain. These signals reach the brain and activate an inflammatory cascade in the brain resulting in white matter injury, impaired myelination, delayed head growth, and eventual downstream NDI. The purpose of this review is to summarize the NDI seen in NEC, discuss what is known about the GBA, explore the relationship between the GBA and perinatal brain injury in the setting of NEC, and finally, highlight the existing research into possible therapies to help prevent these deleterious outcomes.
Collapse
Affiliation(s)
- Krishna Manohar
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Fikir M Mesfin
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Jianyun Liu
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - W Christopher Shelley
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - John P Brokaw
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Troy A Markel
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States.,Riley Hospital for Children, Indiana University Health, Indianapolis, IN, United States
| |
Collapse
|
36
|
Cho H, Lee EH, Lee KS, Heo JS. Machine learning-based risk factor analysis of necrotizing enterocolitis in very low birth weight infants. Sci Rep 2022; 12:21407. [PMID: 36496465 PMCID: PMC9741654 DOI: 10.1038/s41598-022-25746-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
This study used machine learning and a national prospective cohort registry database to analyze the major risk factors of necrotizing enterocolitis (NEC) in very low birth weight (VLBW) infants, including environmental factors. The data consisted of 10,353 VLBW infants from the Korean Neonatal Network database from January 2013 to December 2017. The dependent variable was NEC. Seventy-four predictors, including ambient temperature and particulate matter, were included. An artificial neural network, decision tree, logistic regression, naïve Bayes, random forest, and support vector machine were used to evaluate the major predictors of NEC. Among the six prediction models, logistic regression and random forest had the best performance (accuracy: 0.93 and 0.93, area under the receiver-operating-characteristic curve: 0.73 and 0.72, respectively). According to random forest variable importance, major predictors of NEC were birth weight, birth weight Z-score, maternal age, gestational age, average birth year temperature, birth year, minimum birth year temperature, maximum birth year temperature, sepsis, and male sex. To the best of our knowledge, the performance of random forest in this study was among the highest in this line of research. NEC is strongly associated with ambient birth year temperature, as well as maternal and neonatal predictors.
Collapse
Affiliation(s)
- Hannah Cho
- Department of Pediatrics, Korea University College of Medicine, Anam Hospital, 73 Goryeodae-Ro, Seongbuk-Gu, Seoul, 02841, Korea
- Department of Pediatrics, Korea University Anam Hospital, Seoul, Korea
| | - Eun Hee Lee
- Department of Pediatrics, Korea University College of Medicine, Anam Hospital, 73 Goryeodae-Ro, Seongbuk-Gu, Seoul, 02841, Korea
| | - Kwang-Sig Lee
- AI Center, Korea University College of Medicine, Anam Hospital, 73 Goryeodae-Ro, Seongbuk-Gu, Seoul, 02841, Korea.
| | - Ju Sun Heo
- Department of Pediatrics, Korea University College of Medicine, Anam Hospital, 73 Goryeodae-Ro, Seongbuk-Gu, Seoul, 02841, Korea.
- Department of Pediatrics, Korea University Anam Hospital, Seoul, Korea.
| |
Collapse
|
37
|
Garg PM, Paschal JL, Ansari MAY, Block D, Inagaki K, Weitkamp JH. Clinical impact of NEC-associated sepsis on outcomes in preterm infants. Pediatr Res 2022; 92:1705-1715. [PMID: 35352003 DOI: 10.1038/s41390-022-02034-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To determine risk factors and outcomes of necrotizing enterocolitis (NEC)-associated sepsis in infants with NEC. METHODS A retrospective review comparing demographic and clinical information in infants with and without NEC-associated sepsis (defined as positive blood culture at the time of NEC onset). RESULTS A total of 209 infants with medical (n = 98) and surgical NEC (n = 111) had a median gestational age of 27 weeks (IQR 25; 30.5) and a median birth weight of 910 g [IQR 655; 1138]. Fifty of 209 (23.9%) infants had NEC-associated sepsis. Infants with NEC-associated sepsis had lower median GA (26.4 vs. 27.4 weeks; p = 0.01), lower birth weight (745 vs. 930 g; p = 0.009), were more likely mechanically ventilated [p < 0.001], received dopamine [p < 0.001], had more evidence of acute kidney injury [60% vs. 38.4%, p = 0.01], longer postoperative ileus (16 [13.0; 22.0] vs. 12 [8; 16] days; p = 0.006), higher levels of C-reactive protein, lower platelet counts, longer hospitalization compared to infants without NEC-associated sepsis. On multivariate regression, cholestasis was an independent risk factor for NEC-associated sepsis (OR 2.94; 95% CI 1.1-8.8, p = 0.038). CONCLUSION NEC-associated sepsis was associated with greater hemodynamic support, acute kidney injury, longer postoperative ileus, and hospitalization on bivariate analysis, and cholestasis was associated with higher odds of sepsis on multi regression analysis. IMPACT NEC-associated sepsis was present in 24% of infants with NEC. Gram-positive bacteria, Gram-negative bacteria, and Candida were found in 15.3%, 10.5%, and 2.8% of cases, respectively. Infants with NEC-associated sepsis had a greater inflammatory response (CRP levels), received more blood transfusion before NEC onset, frequently needed assisted ventilation ionotropic support, and had acute kidney injury after NEC onset. NEC infants with Gram-negative sepsis had higher portal venous gas, received more platelet transfusions before NEC onset, and had higher CRP levels and lower median lymphocyte counts at 24 h after NEC onset than those with Gram-positive sepsis.
Collapse
Affiliation(s)
- Parvesh Mohan Garg
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Jaslyn L Paschal
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Md Abu Yusuf Ansari
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Danielle Block
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kengo Inagaki
- Department of Pediatrics/Infectious Disease, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jörn-Hendrik Weitkamp
- Department of Pediatrics/Neonatology, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, TN, USA
| |
Collapse
|
38
|
Hoberg K, Häusler M, Orlikowsky T, Lidzba K. Enhancing the Follow-up Assessment of Very Preterm Children with Regard to 5-Year IQ Considering Socioeconomic Status. Z Geburtshilfe Neonatol 2022; 226:405-415. [PMID: 35981549 DOI: 10.1055/a-1864-9895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Specifying peri- and postnatal factors in children born very preterm (VPT) that affect later outcome helps to improve long-term treatment. AIM To enhance the predictability of 5-year cognitive outcome by perinatal, 2-year developmental and socio-economic data. SUBJECTS AND OUTCOME MEASURES 92 VPT infants, born 2007-2009, gestational age<32 weeks and/or birthweight of 1500 g, were assessed longitudinally including basic neonatal, socio-economic (SES), 2-year Mental Developmental Index (MDI, Bayley Scales II), 5-year Mental Processing Composite (MPC, Kaufman-Assessment Battery for Children), and Language Screening for Preschoolers data. 5-year infants born VPT were compared to 34 term controls. RESULTS The IQ of 5-year infants born VPT was 10 points lower than that of term controls and influenced independently by preterm birth and SES. MDI, SES, birth weight and birth complications explained 48% of the variance of the MPC. The MDI proved highly predictive (r=0.6, R2=36%) for MPC but tended to underestimate the cognitive outcome. A total of 61% of the 2-year infants born VPT were already correctly classified (specificity of .93, sensitivity of .54). CHAID decision tree technique identified SES as decisive for the outcome for infants born VPT with medium MDI results (76-91): They benefit from effects associated to a higher SES, while those with a poor MDI outcome and a birth weight≤890 g do not. CONCLUSION Developmental follow-up of preterm children enhances the quality of prognosis and later outcome when differentially considering perinatal risks and SES.
Collapse
Affiliation(s)
- Kathrin Hoberg
- Department of Paediatrics, Social Paediatric Centre, Division of Neuropaediatrics and Social Paediatrics; University Hospital RWTH Aachen, RWTH Aachen University, Aachen, Germany
| | - Martin Häusler
- Department of Paediatrics, Division of Neuropediatrics and Social Pediatrics; University Hospital RWTH Aachen, RWTH Aachen University, Aachen, Germany
| | - Thorsten Orlikowsky
- Department of Paediatrics, Division of Neonatology; University Hospital RWTH Aachen, RWTH Aachen University, Aachen, Germany
| | - Karen Lidzba
- Division of Child Neurology, Department of Pediatrics, Pediatric Neurology, Inselspital University Hospital Bern, Bern, Switzerland
| |
Collapse
|
39
|
Iskusnykh IY, Chizhikov VV. Cerebellar development after preterm birth. Front Cell Dev Biol 2022; 10:1068288. [PMID: 36523506 PMCID: PMC9744950 DOI: 10.3389/fcell.2022.1068288] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/09/2022] [Indexed: 11/30/2022] Open
Abstract
Preterm birth and its complications and the associated adverse factors, including brain hemorrhage, inflammation, and the side effects of medical treatments, are the leading causes of neurodevelopmental disability. Growing evidence suggests that preterm birth affects the cerebellum, which is the brain region involved in motor coordination, cognition, learning, memory, and social communication. The cerebellum is particularly vulnerable to the adverse effects of preterm birth because key cerebellar developmental processes, including the proliferation of neural progenitors, and differentiation and migration of neurons, occur in the third trimester of a human pregnancy. This review discusses the negative impacts of preterm birth and its associated factors on cerebellar development, focusing on the cellular and molecular mechanisms that mediate cerebellar pathology. A better understanding of the cerebellar developmental mechanisms affected by preterm birth is necessary for developing novel treatment and neuroprotective strategies to ameliorate the cognitive, behavioral, and motor deficits experienced by preterm subjects.
Collapse
|
40
|
Mohammadi A, Higazy R, Gauda EB. PGC-1α activity and mitochondrial dysfunction in preterm infants. Front Physiol 2022; 13:997619. [PMID: 36225305 PMCID: PMC9548560 DOI: 10.3389/fphys.2022.997619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/09/2022] [Indexed: 11/26/2022] Open
Abstract
Extremely low gestational age neonates (ELGANs) are born in a relatively hyperoxic environment with weak antioxidant defenses, placing them at high risk for mitochondrial dysfunction affecting multiple organ systems including the nervous, respiratory, ocular, and gastrointestinal systems. The brain and lungs are highly affected by mitochondrial dysfunction and dysregulation in the neonate, causing white matter injury (WMI) and bronchopulmonary dysplasia (BPD), respectively. Adequate mitochondrial function is important in providing sufficient energy for organ development as it relates to alveolarization and axonal myelination and decreasing oxidative stress via reactive oxygen species (ROS) and reactive nitrogen species (RNS) detoxification. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) is a master regulator of mitochondrial biogenesis and function. Since mitochondrial dysfunction is at the root of WMI and BPD pathobiology, exploring therapies that can regulate PGC-1α activity may be beneficial. This review article describes several promising therapeutic agents that can mitigate mitochondrial dysfunction through direct and indirect activation and upregulation of the PGC-1α pathway. Metformin, resveratrol, omega 3 fatty acids, montelukast, L-citrulline, and adiponectin are promising candidates that require further pre-clinical and clinical studies to understand their efficacy in decreasing the burden of disease from WMI and BPD in preterm infants.
Collapse
Affiliation(s)
- Atefeh Mohammadi
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Randa Higazy
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
| | - Estelle B. Gauda
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- *Correspondence: Estelle B. Gauda,
| |
Collapse
|
41
|
Favrais G, Bokobza C, Saliba E, Chalon S, Gressens P. Alteration of the Oligodendrocyte Lineage Varies According to the Systemic Inflammatory Stimulus in Animal Models That Mimic the Encephalopathy of Prematurity. Front Physiol 2022; 13:881674. [PMID: 35928559 PMCID: PMC9343871 DOI: 10.3389/fphys.2022.881674] [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: 02/22/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
Preterm birth before the gestational age of 32 weeks is associated with the occurrence of specific white matter damage (WMD) that can compromise the neurological outcome. These white matter abnormalities are embedded in more global brain damage defining the encephalopathy of prematurity (EoP). A global reduction in white matter volume that corresponds to chronic diffuse WMD is the most frequent form in contemporary cohorts of very preterm infants. This WMD partly results from alterations of the oligodendrocyte (OL) lineage during the vulnerability window preceding the beginning of brain myelination. The occurrence of prenatal, perinatal and postnatal events in addition to preterm birth is related to the intensity of WMD. Systemic inflammation is widely recognised as a risk factor of WMD in humans and in animal models. This review reports the OL lineage alterations associated with the WMD observed in infants suffering from EoP and emphasizes the role of systemic inflammation in inducing these alterations. This issue is addressed through data on human tissue and imaging, and through neonatal animal models that use systemic inflammation to induce WMD. Interestingly, the OL lineage damage varies according to the inflammatory stimulus, i.e., the liposaccharide portion of the E.Coli membrane (LPS) or the proinflammatory cytokine Interleukin-1β (IL-1β). This discrepancy reveals multiple cellular pathways inducible by inflammation that result in EoP. Variable long-term consequences on the white matter morphology and functioning may be speculated upon according to the intensity of the inflammatory challenge. This hypothesis emerges from this review and requires further exploration.
Collapse
Affiliation(s)
- Geraldine Favrais
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
- Neonatology Unit, CHRU de Tours, Tours, France
- *Correspondence: Geraldine Favrais,
| | - Cindy Bokobza
- Inserm, NeuroDiderot, Université Paris Cité, Paris, France
| | - Elie Saliba
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | - Sylvie Chalon
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France
| | | |
Collapse
|
42
|
MRI based radiomics enhances prediction of neurodevelopmental outcome in very preterm neonates. Sci Rep 2022; 12:11872. [PMID: 35831452 PMCID: PMC9279296 DOI: 10.1038/s41598-022-16066-w] [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: 04/21/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
To predict adverse neurodevelopmental outcome of very preterm neonates. A total of 166 preterm neonates born between 24–32 weeks’ gestation underwent brain MRI early in life. Radiomics features were extracted from T1- and T2- weighted images. Motor, cognitive, and language outcomes were assessed at a corrected age of 18 and 33 months and 4.5 years. Elastic Net was implemented to select the clinical and radiomic features that best predicted outcome. The area under the receiver operating characteristic (AUROC) curve was used to determine the predictive ability of each feature set. Clinical variables predicted cognitive outcome at 18 months with AUROC 0.76 and motor outcome at 4.5 years with AUROC 0.78. T1-radiomics features showed better prediction than T2-radiomics on the total motor outcome at 18 months and gross motor outcome at 33 months (AUROC: 0.81 vs 0.66 and 0.77 vs 0.7). T2-radiomics features were superior in two 4.5-year motor outcomes (AUROC: 0.78 vs 0.64 and 0.8 vs 0.57). Combining clinical parameters and radiomics features improved model performance in motor outcome at 4.5 years (AUROC: 0.84 vs 0.8). Radiomic features outperformed clinical variables for the prediction of adverse motor outcomes. Adding clinical variables to the radiomics model enhanced predictive performance.
Collapse
|
43
|
Berken JA, Chang J. Neurologic consequences of neonatal necrotizing enterocolitis. Dev Neurosci 2022; 44:295-308. [PMID: 35697005 DOI: 10.1159/000525378] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 05/24/2022] [Indexed: 11/19/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease of the premature infant with high mortality and morbidity. Children who survive NEC have been shown to demonstrate neurodevelopmental delay, with significantly worse outcomes than from prematurity alone. The pathways leading to NEC-associated neurological impairments remain unclear, limiting the development of preventative and protective strategies. This review aims to summarize the existing clinical and experimental studies related to NEC-associated brain injury. We describe the current epidemiology of NEC, reported long-term neurodevelopmental outcomes among survivors, and proposed pathogenesis of brain injury in NEC. Highlighted are the potential connections between hypoxia-ischemia, nutrition, infection, gut inflammation, and the developing brain in NEC.
Collapse
Affiliation(s)
- Jonathan A Berken
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Jill Chang
- Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA,
- Division of Neonatal-Perinatal Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA,
| |
Collapse
|
44
|
Abstract
Advances in perinatal care have seen substantial improvements in survival without disability for extremely preterm infants. Protecting the developing brain and reducing neurodevelopmental sequelae of extremely preterm birth are strategic priorities for both research and clinical care. A number of evidence-based interventions exist for neuroprotection in micropreemies, inclusive of prevention of preterm birth and multiple births with implantation of only one embryo during in vitro fertilisation, as well as antenatal care to optimize fetal wellbeing, strategies for supporting neonatal transition, and neuroprotective developmental care. Avoidance of complications that trigger ischemia and inflammation is vital for minimizing brain dysmaturation and injury, particularly of the white matter. Neurodevelopmental surveillance, early diagnosis of cerebral palsy and early intervention are essential for optimizing long-term outcomes and quality of life. Research priorities include further evaluation of putative neuroprotective agents, and investigation of common neonatal interventions in trials adequately powered to assess neurodevelopmental outcome.
Collapse
|
45
|
He Y, Zhang Y, Li F, Shi Y. White Matter Injury in Preterm Infants: Pathogenesis and Potential Therapy From the Aspect of the Gut–Brain Axis. Front Neurosci 2022; 16:849372. [PMID: 35573292 PMCID: PMC9099073 DOI: 10.3389/fnins.2022.849372] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/04/2022] [Indexed: 12/12/2022] Open
Abstract
Very preterm infants who survive are at high risk of white matter injury (WMI). With a greater understanding of the pathogenesis of WMI, the gut microbiota has recently drawn increasing attention in this field. This review tries to clarify the possible mechanisms behind the communication of the gut bacteria and the immature brain via the gut–brain axis. The gut microbiota releases signals, such as microbial metabolites. These metabolites regulate inflammatory and immune responses characterized by microglial activation, which ultimately impact the differentiation of pre-myelinating oligodendrocytes (pre-OLs) and lead to WMI. Moreover, probiotics and prebiotics emerge as a promising therapy to improve the neurodevelopmental outcome. However, future studies are required to clarify the function of these above products and the optimal time for their administration within a larger population. Based on the existing evidence, it is still too early to recommend probiotics and prebiotics as effective treatments for WMI.
Collapse
Affiliation(s)
- Yu He
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Yuni Zhang
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Fang Li
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- *Correspondence: Fang Li,
| | - Yuan Shi
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Yuan Shi,
| |
Collapse
|
46
|
Brain injury in preterm infants with surgical necrotizing enterocolitis: clinical and bowel pathological correlates. Pediatr Res 2022; 91:1182-1195. [PMID: 34103675 DOI: 10.1038/s41390-021-01614-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND The objective of this study was to determine the risk factors and outcomes of white matter brain injury (WMBI) on magnetic resonance imaging (MRI) at term-equivalent age in infants with surgical necrotizing enterocolitis (NEC). METHODS This retrospective study compared clinical/pathological information between infants with and those without WMBI. RESULTS Out of 69 infants with surgical NEC, 17 (24.6%) had mild WMBI, 13 (18.8%) had moderate WMBI, and six (8.7%) had severe WMBI on the brain MRI. Several clinical factors (gestational age, more red blood cell (RBC) transfusions before NEC onset, pneumoperitoneum, earlier NEC onset age, postoperative ileus, acute kidney injury (AKI) by serum creatinine, postnatal steroids, hospital stay) and histopathological findings (necrosis, hemorrhage) had univariate associations with WMBI. Associations with RBC transfusion (odds ratio (OR) 23.6 [95% confidence interval (CI): 4.73-117.97]; p = 0.0001), age at NEC onset (OR 0.30 [95%CI: 0.11-0.84]; p = 0.021), necrosis (OR 0.10 [95%CI: 0.01-0.90]; p = 0.040), and bowel hemorrhage (OR 7.79 [95%CI: 2.19-27.72]; p = 0.002) persisted in multivariable association with grade 3-4 WMBI. The infants with WMBI had lower mean motor, cognitive, language scores, and higher ophthalmic morbidity at 2 years of age. CONCLUSIONS The WMBI was most likely associated with earlier NEC onset, higher RBC transfusions, and less necrosis and greater hemorrhage lesions on intestinal pathology in preterm infants with surgical NEC. IMPACT In preterm infants with surgical NEC, brain MRI showed injury in the white matter in 52%, gray matter in 10%, and cerebellar region in 30%. Preterm infants with severe WMBI (grade 3-4) had less necrosis and greater hemorrhagic lesions on histopathology of the bowel. Preterm infants with WMBI were more likely to have a more severe postoperative course, AKI, and longer length of hospitalization. Neuroprotective strategies to prevent brain injury in preterm infants with surgical NEC are needed with the goal of improving the neurodevelopmental outcomes.
Collapse
|
47
|
Li Y, Wang D, Li Z, Ouyang Z. PSB0788 ameliorates maternal inflammation-induced periventricular leukomalacia-like injury. Bioengineered 2022; 13:10224-10234. [PMID: 35436416 PMCID: PMC9161964 DOI: 10.1080/21655979.2022.2061296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Studies have shown that periventricular leukomalacia (PVL) is a distinctive form of cerebral white matter injury that pertains to myelination disturbances. Maternal inflammation is a main cause of white matter injury. Intrauterine inflammation cellular will be propagated to the developing brain by the entire maternal-placental-fetal axis, and triggers neural immune injury. As a low-affinity receptor, adenosine A2B receptor (A2BAR) requires high concentrations of adenosine to be significantly activated in pathological conditions. We hypothesized that in the maternal inflammation-induced PVL model, a selective A2BAR antagonist PSB0788 had the potential to prevent the injury. In this work, a total of 18 SD pregnant rats were divided into three groups, and treated with intraperitoneal injection of phosphate buffered saline (PBS), lipopolysaccharide (LPS), or LPS+PSB0788. Placental infection was determined by H&E staining and the inflammatory condition was determined by ELISA. Change of MBP, NG2 and CC-1 in the brain of the rats' offspring were detected by western blot and immunohistochemistry. Furthermore, LPS-induced maternal inflammation reduced the expression of MBP, which related to the decrease in the numbers of OPCs and mature oligodendrocytes in neonate rats. After treatment with PSB0788, the levels of MBP proteins increased in the rats' offspring, improved the remyelination. In conclusion, our study shows that the selective A2BAR antagonist PSB0788 plays an important role in promoting the normal development of OPCs in vivo by the maternal inflammation-induced PVL model. Future studies will focus on the mechanism of PSB0788 in this model.
Collapse
Affiliation(s)
- Yilu Li
- School of Chemistry and Chemical Engineering, South China University of Technology, scDFG Guangzhou, Guangdong, China
| | - Dan Wang
- Department of clinical medicine, Bengbu Medical College, Bengbu, Anhui, China,Department of clinical medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zhuoyang Li
- School of Chemistry and Chemical Engineering, South China University of Technology, scDFG Guangzhou, Guangdong, China,South China University of Technology-Zhuhai Institute of Modern Industrial Innovation, Zhuhai, Guangdong, China
| | - Zhi Ouyang
- South China University of Technology Hospital, South China University of Technology, Guangzhou, Guangdong, China,CONTACT Zhi Ouyang South China University of Technology Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
48
|
Neurodevelopmental outcomes at 6, 12, and 24 months of age in preterm infants with very low birth weights in Taiwan. J Formos Med Assoc 2022; 121:1804-1812. [DOI: 10.1016/j.jfma.2022.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 12/25/2022] Open
|
49
|
Zhao Q, Shi Q, Zhu Q, Hu Y, Zhang X. A mini-review of advances in intestinal flora and necrotizing enterocolitis. Lett Appl Microbiol 2022; 75:2-9. [PMID: 35138661 DOI: 10.1111/lam.13670] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/04/2022] [Accepted: 02/04/2022] [Indexed: 12/01/2022]
Abstract
Necrotizing enterocolitis (NEC) is a digestive disease that frequently occurs in premature infants with low gestational age and low birth weight, and seriously threatens the life of NEC patients. NEC pathogenesis is recognized to be affected by multiple factors, such as preterm birth, formula feeding, and low birth weight. As a popular object for the past decades, intestinal flora is commonly used in NEC-related studies, and intestinal disorder is considered as a critical risk factor for the occurrence and development of NEC. The colonization of abnormal microbiota into gastrointestinal micro-ecosystem can easily lead to the damage of intestinal mucosal barrier, destruction of immune function, inflammatory reaction, and further the occurrence of NEC. Although it is a low-cost and safe way to prevent and treat the NEC by early intervention of oral probiotics to regulate the intestinal homeostasis, more studies in the future are still encouraged to narrow the gap between theoretical guidance and practical application.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Occupational and Environment Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - Qiwei Shi
- Department of Occupational and Environment Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - Qin Zhu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Hu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xian Zhang
- Department of Occupational and Environment Health, Xiangya School of Public Health, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, China
| |
Collapse
|
50
|
Golin MO, Souza FIS, Paiva LDS, Sarni ROS. "The Value of Clinical Examination in Preterm Newborns after Neonatal Sepsis: A Cross-sectional Observational Study.". Dev Neurorehabil 2022; 25:80-86. [PMID: 34346264 DOI: 10.1080/17518423.2021.1941372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Neonatal sepsis is an important risk factor for lesions in the brain of preterm newborns (PTNB) and the most effective strategies to minimize its deleterious effects are early detection and intervention. AIM To investigate the presence of neurological abnormalities in PTNBs after neonatal sepsis. METHODS This was a prospective cross-sectional study with 100 PTNBs selected at random, 50 of the study group (sepsis) and 50 of the control group (non-sepsis). The neurological evaluation protocol adopted was the Hammersmith Neonatal Neurological Examination (HNNE). RESULTS The PTNBs of the sepsis group had total HNNE scores lower than expected for normality in 86% of the cases, and the non-sepsis group in 26% (p < .001). Higher prevalence levels of altered scores in tone category (p < .001), tone patterns (p = .026), reflexes (p = .002), movements (p < .001), abnormal signs (p < .001) and behavior (p < .001). CONCLUSION The neurological dysfunctions after neonatal sepsis could be identified by clinical neonatal neurological evaluation.
Collapse
Affiliation(s)
- Marina Ortega Golin
- Department of Physiotherapy, ABC Faculty of Medicine, Santo André / SP, Brazil
| | | | - Laércio da Silva Paiva
- Department of Health of the Community, ABC Faculty of Medicine, Santo André / SP, Brazil
| | | |
Collapse
|