1
|
Barros M, Liang M, Iannucci N, Dickinson R. Xenon and Argon as Neuroprotective Treatments for Perinatal Hypoxic-Ischemic Brain Injury: A Preclinical Systematic Review and Meta-Analysis. Anesth Analg 2024:00000539-990000000-01012. [PMID: 39453983 DOI: 10.1213/ane.0000000000007223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Xenon and argon are currently being evaluated as potential neuroprotective treatments for acquired brain injuries. Xenon has been evaluated clinically as a treatment for brain ischemia with equivocal results in small trials, but argon has not yet undergone clinical evaluation. Several preclinical studies have investigated xenon or argon as treatments in animal models of perinatal hypoxic-ischemic encephalopathy (HIE). A systematic review of MEDLINE and Embase databases was performed. After screening of titles, abstracts, and full text, data were extracted from included studies. A pairwise meta-analysis of neuroprotective efficacy was performed using a random effects model. Heterogeneity was investigated using subgroup analysis, funnel plot asymmetry, and Egger's regression. The protocol was prospectively registered on PROSPERO (CRD42022301986). A total of 21 studies met the inclusion criteria. The data extracted included measurements from 1591 animals, involving models of HIE in mice, rats, and pigs. The meta-analysis found that both xenon and argon had significant (P < .0001) neuroprotective efficacies. The summary estimate for xenon was 39.7% (95% confidence interval [CI], 28.3%-51.1%) and for argon it was 70.3% (95% CI, 59.0%-81.7%). The summary effect for argon was significantly (P < .001) greater than that of xenon. Our results provide evidence supporting further investigation of xenon and argon as neuroprotective treatments for HIE.
Collapse
Affiliation(s)
- Mariana Barros
- From the Anaesthetics, Pain Medicine and Intensive Care Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Min Liang
- Anaesthesiology Research Institute, Department of Anaesthesiology, First Affiliated Hospital of Fujian Medical University, Binhai Campus, Fuzhou, China
| | - Noemi Iannucci
- From the Anaesthetics, Pain Medicine and Intensive Care Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Robert Dickinson
- From the Anaesthetics, Pain Medicine and Intensive Care Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Centre for Blast Injury Studies, Imperial College London, London, United Kingdom
| |
Collapse
|
2
|
Jongruk P, Soontaravarapas N, Angkurawaranon S, Kosarat S, Khuwuthyakorn V, Tantiprabha W, Manopunya S, Boonchooduang N, Louthrenoo O, Likhitweerawong N, Katanyuwong K, Sanguansermsri C, Wiwattanadittakul N. Adjuvant High-Dose Erythropoietin With Delayed Therapeutic Hypothermia in Neonatal Hypoxic-Ischemic Encephalopathy. Pediatr Neurol 2024; 161:268-276. [PMID: 39481349 DOI: 10.1016/j.pediatrneurol.2024.10.003] [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: 03/20/2024] [Revised: 08/19/2024] [Accepted: 10/01/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND To evaluate the benefits of high-dose erythropoietin (EPO) combined with therapeutic hypothermia (TH) on brain magnetic resonance imaging (MRI) scores and neurodevelopmental outcomes in neonates with moderate to severe hypoxic-ischemic-ecephalopathy (HIE), especially in neonates who received TH between six to 12 hours of birth. METHODS This prospective, single-blind, randomized, placebo-controlled trial enrolled term newborns with moderate to severe HIE admitted to neonatal intensive care unit between April 2018 and April 2022. Hypothermia was started within 12 hours of birth. Infants were randomized to receive EPO 1000 U/kg or an equal volume of normal saline (placebo) on days 1, 2, 3, 5, and 7 of age in combination with hypothermia. RESULTS Fifty-seven neonates with moderate to severe HIE were recruited; 10 were excluded. Forty-seven patients were included: 32 received TH within six hours (group I) and in 15 TH was started within six to 12 hours of birth (group II). The clinical characteristics of mothers and infants, maternal complications, and resuscitations performed during the perinatal period showed no statistical differences between EPO group and placebo groups I and II. The MRI scores and brain injury patterns did not differ between the EPO and placebo groups. There is no statistical significance in either group's seizure and severe electroencephalography background (initial and after rewarming) between EPO and placebo in each group. There were no differences in developmental outcomes (abnormal Denver II > 2 area, Gross Motor Function Classification Score >1); Bayley Scales of Infant and Toddler Development, third edition (BSID-III) score (cognitive, language, and motor); or disability (hearing impairment and impaired vision) between the EPO and placebo groups I and II at 12 and 18 months. CONCLUSIONS Among term infants with moderate to severe HIE, TH with EPO administration, compared with TH alone, did not reduce brain injury on MRI or the risk of neurological sequelae both in patients who received TH within six hours and in those who received TH later (six to 12 hours). Further studies on the benefit of EPO injection alone or before TH in situations where TH cannot be performed are required.
Collapse
Affiliation(s)
- Piyathida Jongruk
- Faculty of Medicine, Division of Neonatology, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Nawamon Soontaravarapas
- Faculty of Medicine, Division of Neonatology, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Salita Angkurawaranon
- Faculty of Medicine, Department of Radiology, Chiang Mai University, Chiang Mai, Thailand
| | - Shanika Kosarat
- Faculty of Medicine, Division of Neonatology, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Varangthip Khuwuthyakorn
- Faculty of Medicine, Division of Neonatology, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Watcharee Tantiprabha
- Faculty of Medicine, Division of Neonatology, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Satit Manopunya
- Faculty of Medicine, Division of Neonatology, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Nonglak Boonchooduang
- Faculty of Medicine, Division of Growth and Development, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Orawan Louthrenoo
- Faculty of Medicine, Division of Growth and Development, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Narueporn Likhitweerawong
- Faculty of Medicine, Division of Growth and Development, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Kamornwan Katanyuwong
- Faculty of Medicine, Division of Neurology, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Chinnuwat Sanguansermsri
- Faculty of Medicine, Division of Neurology, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand
| | - Natrujee Wiwattanadittakul
- Faculty of Medicine, Division of Neurology, Department of Pediatrics, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
3
|
Sha S, Jin N, Zhou R, Ruan Y, Ouyang Y. The Activation of PKM2 Induces Pyroptosis in Hippocampal Neurons via the NLRP3/Caspase-1/GSDMD Pathway in Neonatal Rats With Hypoxic-Ischemic Brain Injury. Brain Behav 2024; 14:e70108. [PMID: 39444090 PMCID: PMC11499207 DOI: 10.1002/brb3.70108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/27/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION The presence of hypoxic-ischemic brain damage (HIBD) in neonates triggers a strong neuroinflammatory reaction. Pyroptosis, a programmed cell death mechanism associated with inflammation, plays a crucial role in HIBD. Pyruvate kinase M2 (PKM2) plays a significant role in connecting metabolic processes and inflammatory responses, but whether it affects hippocampus pyroptosis in HIBD is unclear. The aim of this study is to elucidate the role of PKM2 in HIBD and to propose a novel therapeutic approach for neonatal ischemic-hypoxic encephalopathy. METHODS In this study, we employed neonatal 7-day-old Sprague Dawley rats to establish a model of HIBD using the Rice-Vannucci surgical technique and a hypoxia device. To inhibit the elevation of PKM2, we utilized the PKM2 inhibitor shikonin. The rats were categorized into four groups: Sham, Shikonin, HIBD, and Shikonin + HIBD. Behavioral tests, hematoxylin eosin staining, immunofluorescence staining, ELISA (IL-1β, IL-18), and LDH were conducted in each group to evaluate neurological function, hippocampal damage, the occurrence of neuronal pyroptosis, and the neuroinflammation. Western blot was used to assess the expression levels of PKM2, NLRP3, Caspase-1, Cleaved Caspase-1, GSDMD, GSDMDN, and IL-1β. RESULTS The expression of PKM2 elevated in hippocampal tissues of the HIBD model and the localization of PKM2 in the hippocampus was activated in neurons instead of microglia during the HIBD. Meanwhile, the inhibition of PKM2 improved the behavioral test scores and the body weight of rats, the neuronal damage in the CA1 region of hippocampal tissue was also attenuated. In addition, inhibiting PKM2 alleviated neuronal pyroptosis by decreasing the expression of PKM2, NLRP3, Caspase-1, Cleaved Caspase-1, GSDMD, GSDMDN. Furthermore, serum levels of LDH and inflammatory factors IL-1β and IL-18 decrease with PKM2 inhibition. CONCLUSIONS Based on these findings, we can conclude that PKM2 plays a crucial role in regulating hippocampal neuronal pyroptosis of HIBD rats via NLRP3/Caspase-1/GSDMD pathway. Therefore, inhibiting PKM2 could be a promising therapeutic strategy for the treatment of neonatal ischemic-hypoxic encephalopathy.
Collapse
Affiliation(s)
- Sha Sha
- Sun Yat‐sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Ni Jin
- Sun Yat‐sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Ruiyu Zhou
- The Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Yanghao Ruan
- The Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdongChina
| | - Ying Ouyang
- Sun Yat‐sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
4
|
Lew CO, Calabrese E, Chen JV, Tang F, Chaudhari G, Lee A, Faro J, Juul S, Mathur A, McKinstry RC, Wisnowski JL, Rauschecker A, Wu YW, Li Y. Artificial Intelligence Outcome Prediction in Neonates with Encephalopathy (AI-OPiNE). Radiol Artif Intell 2024; 6:e240076. [PMID: 38984984 PMCID: PMC11427921 DOI: 10.1148/ryai.240076] [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: 02/05/2024] [Revised: 05/21/2024] [Accepted: 06/18/2024] [Indexed: 07/11/2024]
Abstract
Purpose To develop a deep learning algorithm to predict 2-year neurodevelopmental outcomes in neonates with hypoxic-ischemic encephalopathy using MRI and basic clinical data. Materials and Methods In this study, MRI data of term neonates with encephalopathy in the High-dose Erythropoietin for Asphyxia and Encephalopathy (HEAL) trial (ClinicalTrials.gov: NCT02811263), who were enrolled from 17 institutions between January 25, 2017, and October 9, 2019, were retrospectively analyzed. The harmonized MRI protocol included T1-weighted, T2-weighted, and diffusion tensor imaging. Deep learning classifiers were trained to predict the primary outcome of the HEAL trial (death or any neurodevelopmental impairment at 2 years) using multisequence MRI and basic clinical variables, including sex and gestational age at birth. Model performance was evaluated on test sets comprising 10% of cases from 15 institutions (in-distribution test set, n = 41) and 10% of cases from two institutions (out-of-distribution test set, n = 41). Model performance in predicting additional secondary outcomes, including death alone, was also assessed. Results For the 414 neonates (mean gestational age, 39 weeks ± 1.4 [SD]; 232 male, 182 female), in the study cohort, 198 (48%) died or had any neurodevelopmental impairment at 2 years. The deep learning model achieved an area under the receiver operating characteristic curve (AUC) of 0.74 (95% CI: 0.60, 0.86) and 63% accuracy in the in-distribution test set and an AUC of 0.77 (95% CI: 0.63, 0.90) and 78% accuracy in the out-of-distribution test set. Performance was similar or better for predicting secondary outcomes. Conclusion Deep learning analysis of neonatal brain MRI yielded high performance for predicting 2-year neurodevelopmental outcomes. Keywords: Convolutional Neural Network (CNN), Prognosis, Pediatrics, Brain, Brain Stem Clinical trial registration no. NCT02811263 Supplemental material is available for this article. © RSNA, 2024 See also commentary by Rafful and Reis Teixeira in this issue.
Collapse
Affiliation(s)
| | | | - Joshua V. Chen
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - Felicia Tang
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - Gunvant Chaudhari
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - Amanda Lee
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - John Faro
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - Sandra Juul
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - Amit Mathur
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - Robert C. McKinstry
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - Jessica L. Wisnowski
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - Andreas Rauschecker
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - Yvonne W. Wu
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| | - Yi Li
- From the Department of Radiology, Duke University Medical Center,
2301 Erwin Rd, Box 3808, Durham, NC 27710 (C.O.L., E.C, A.L., J.F.); Department
of Radiology (J.V.C., F.T., G.C., A.R., Y.L.) and Weill Institute for
Neurosciences (Y.W.W.), University of California San Francisco, San Francisco,
Calif; Department of Pediatrics, University of Washington, Seattle, Wash (S.J.);
Department of Pediatrics, Saint Louis University, St Louis, Mo (A.M.);
Mallinckrodt Institute of Radiology, Washington University School of Medicine,
St Louis, Mo (R.C.M.); and Children’s Hospital Los Angeles, University of
Southern California, Los Angeles, Calif (J.L.W.)
| |
Collapse
|
5
|
Yang M, Wang K, Liu B, Shen Y, Liu G. Hypoxic-Ischemic Encephalopathy: Pathogenesis and Promising Therapies. Mol Neurobiol 2024:10.1007/s12035-024-04398-9. [PMID: 39073530 DOI: 10.1007/s12035-024-04398-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a brain lesion caused by inadequate blood supply and oxygen deprivation, often occurring in neonates. It has emerged as a grave complication of neonatal asphyxia, leading to chronic neurological damage. Nevertheless, the precise pathophysiological mechanisms underlying HIE are not entirely understood. This paper aims to comprehensively elucidate the contributions of hypoxia-ischemia, reperfusion injury, inflammation, oxidative stress, mitochondrial dysfunction, excitotoxicity, ferroptosis, endoplasmic reticulum stress, and apoptosis to the onset and progression of HIE. Currently, hypothermia therapy stands as the sole standard treatment for neonatal HIE, albeit providing only partial neuroprotection. Drug therapy and stem cell therapy have been explored in the treatment of HIE, exhibiting certain neuroprotective effects. Employing drug therapy or stem cell therapy as adjunctive treatments to hypothermia therapy holds great significance. This article presents a systematic review of the pathogenesis and treatment strategies of HIE, with the goal of enhancing the effect of treatment and improving the quality of life for HIE patients.
Collapse
Affiliation(s)
- Mingming Yang
- Department of Pediatrics, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, P. R. China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China.
| | - Guangliang Liu
- Department of Pediatrics, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, P. R. China.
| |
Collapse
|
6
|
Roychaudhuri S, Hannon K, Sunwoo J, Garvey AA, El-Dib M. Quantitative EEG and prediction of outcome in neonatal encephalopathy: a review. Pediatr Res 2024; 96:73-80. [PMID: 38503980 DOI: 10.1038/s41390-024-03138-y] [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: 08/18/2023] [Revised: 02/18/2024] [Accepted: 03/01/2024] [Indexed: 03/21/2024]
Abstract
Electroencephalogram (EEG) is an important biomarker for neonatal encephalopathy (NE) and has significant predictive value for brain injury and neurodevelopmental outcomes. Quantitative analysis of EEG involves the representation of complex EEG data in an objective, reproducible and scalable manner. Quantitative EEG (qEEG) can be derived from both a limited channel EEG (as available during amplitude integrated EEG) and multi-channel conventional EEG. It has the potential to enable bedside clinicians to monitor and evaluate details of cortical function without the necessity of continuous expert input. This is particularly useful in NE, a dynamic and evolving condition. In these infants, continuous, detailed evaluation of cortical function at the bedside is a valuable aide to management especially in the current era of therapeutic hypothermia and possible upcoming neuroprotective therapies. This review discusses the role of qEEG in newborns with NE and its use in informing monitoring and therapy, along with its ability to predict imaging changes and short and long-term neurodevelopmental outcomes. IMPACT: Quantitative representation of EEG data brings the evaluation of continuous brain function, from the neurophysiology lab to the NICU bedside and has a potential role as a biomarker for neonatal encephalopathy. Clinical and research applications of quantitative EEG in the newborn are rapidly evolving and a wider understanding of its utility is valuable. This overview summarizes the role of quantitative EEG at different timepoints, its relevance to management and its predictive value for short- and long-term outcomes in neonatal encephalopathy.
Collapse
Affiliation(s)
- Sriya Roychaudhuri
- Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Katie Hannon
- Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - John Sunwoo
- Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Aisling A Garvey
- Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital, Boston, MA, USA
- INFANT Research Centre, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - Mohamed El-Dib
- Division of Newborn Medicine, Department of Pediatrics, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Aronowitz DI, Geoffrion TR, Piel S, Benson EJ, Morton SR, Starr J, Melchior RW, Gaudio HA, Degani RE, Widmann NJ, Weeks MK, Ko TS, Licht DJ, Hefti M, Gaynor JW, Kilbaugh TJ, Mavroudis CD. Early Impairment of Cerebral Bioenergetics After Cardiopulmonary Bypass in Neonatal Swine. World J Pediatr Congenit Heart Surg 2024; 15:459-466. [PMID: 38646826 DOI: 10.1177/21501351241232077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Objectives: We previously demonstrated cerebral mitochondrial dysfunction in neonatal swine immediately following a period of full-flow cardiopulmonary bypass (CPB). The extent to which this dysfunction persists in the postoperative period and its correlation with other markers of cerebral bioenergetic failure and injury is unknown. We utilized a neonatal swine model to investigate the early evolution of mitochondrial function and cerebral bioenergetic failure after CPB. Methods: Twenty piglets (mean weight 4.4 ± 0.5 kg) underwent 3 h of CPB at 34 °C via cervical cannulation and were followed for 8, 12, 18, or 24 h (n = 5 per group). Markers of brain tissue damage (glycerol) and bioenergetic dysfunction (lactate to pyruvate ratio) were continuously measured in cerebral microdialysate samples. Control animals (n = 3, mean weight 4.1 ± 1.2 kg) did not undergo cannulation or CPB. Brain tissue was extracted immediately after euthanasia to obtain ex-vivo cortical mitochondrial respiration and frequency of cortical microglial nodules (indicative of cerebral microinfarctions) via neuropathology. Results: Both the lactate to pyruvate ratio (P < .0001) and glycerol levels (P = .01) increased in cerebral microdialysate within 8 h after CPB. At 24 h post-CPB, cortical mitochondrial respiration was significantly decreased compared with controls (P = .046). The presence of microglial nodules increased throughout the study period (24 h) (P = .01, R2 = 0.9). Conclusion: CPB results in impaired cerebral bioenergetics that persist for at least 24 h. During this period of bioenergetic impairment, there may be increased susceptibility to secondary injury related to alterations in metabolic delivery or demand, such as hypoglycemia, seizures, and decreased cerebral blood flow.
Collapse
Affiliation(s)
- Danielle I Aronowitz
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tracy R Geoffrion
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sarah Piel
- Resuscitation Science Center of Emphasis, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emilie J Benson
- Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah R Morton
- Resuscitation Science Center of Emphasis, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan Starr
- Resuscitation Science Center of Emphasis, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Richard W Melchior
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hunter A Gaudio
- Resuscitation Science Center of Emphasis, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rinat E Degani
- Resuscitation Science Center of Emphasis, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nicholas J Widmann
- Resuscitation Science Center of Emphasis, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - M Katie Weeks
- Resuscitation Science Center of Emphasis, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tiffany S Ko
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Daniel J Licht
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marco Hefti
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, USA
| | - J William Gaynor
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Todd J Kilbaugh
- Resuscitation Science Center of Emphasis, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Constantine D Mavroudis
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
8
|
Tayman C, Çakır U, Kurt A, Ertekin Ö, Taskin Turkmenoglu T, Çağlayan M, Işık E. Evaluation of beneficial effects of dexpanthenol on hypoxic-ischemic encephalopathy. Biotech Histochem 2024:1-9. [PMID: 38869860 DOI: 10.1080/10520295.2024.2365231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a cause of serious morbidity and mortality in newborns. Dexpanthenol, which is metabolized into D-pantothenic acid, has antioxidant and other potentially therapeutic properties. We examined some effects of dexpanthenol on the brains of week-old rat pups with HIE induced by obstruction of the right carotid artery followed by keeping in 8% O2 for 2 hours. Dexpanthenol (500 mg/kg) was administered intraperitoneally to 16 of 32 pups with HIE. Protein, DNA, and lipid oxidation degradation products were assayed and hippocampal and cortical cell apoptosis and neuronal cell numbers were evaluated in stained sections. Dexpanthenol application reduced oxidative stress and inflammation. TNF-α and IL-6 cytokine levels in HIE also decreased with dexpanthenol treatment. The numbers of caspase-3 positive cells in the dentate gyrus and CA1/CA2/CA3 regions of the hippocampus was lower, and apoptosis was decreased in the dexpanthenol-treated animals. These findings suggest possible clinical applications of dexpanthenol in human HIE.
Collapse
Affiliation(s)
- Cuneyt Tayman
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Ufuk Çakır
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Abdullah Kurt
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Ömer Ertekin
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Tugba Taskin Turkmenoglu
- Department of Pathology, Ankara Dişkapi Yildirim Beyzat Training and Research Hospital, Ankara, Turkey
| | - Murat Çağlayan
- Department of Medical Biochemistry, Dışkapı Yıldırım Beyazıt Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Eray Işık
- Department of Otorhinolaryngology (Ear-Nose-Throat), Ankara Dişkapi Yildirim Beyzat Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
9
|
Poupon-Bejuit L, Geard A, Millicheap N, Rocha-Ferreira E, Hagberg H, Thornton C, Rahim AA. Diabetes drugs activate neuroprotective pathways in models of neonatal hypoxic-ischemic encephalopathy. EMBO Mol Med 2024; 16:1284-1309. [PMID: 38783166 PMCID: PMC11178908 DOI: 10.1038/s44321-024-00079-1] [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: 06/06/2022] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Hypoxic-ischaemic encephalopathy (HIE) arises from diminished blood flow and oxygen to the neonatal brain during labor, leading to infant mortality or severe brain damage, with a global incidence of 1.5 per 1000 live births. Glucagon-like Peptide 1 Receptor (GLP1-R) agonists, used in type 2 diabetes treatment, exhibit neuroprotective effects in various brain injury models, including HIE. In this study, we observed enhanced neurological outcomes in post-natal day 10 mice with surgically induced hypoxic-ischaemic (HI) brain injury after immediate systemic administration of exendin-4 or semaglutide. Short- and long-term assessments revealed improved neuropathology, survival rates, and locomotor function. We explored the mechanisms by which GLP1-R agonists trigger neuroprotection and reduce inflammation following oxygen-glucose deprivation and HI in neonatal mice, highlighting the upregulation of the PI3/AKT signalling pathway and increased cAMP levels. These findings shed light on the neuroprotective and anti-inflammatory effects of GLP1-R agonists in HIE, potentially extending to other neurological conditions, supporting their potential clinical use in treating infants with HIE.
Collapse
Affiliation(s)
- Laura Poupon-Bejuit
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Amy Geard
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Nathan Millicheap
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Eridan Rocha-Ferreira
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claire Thornton
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Ahad A Rahim
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK.
| |
Collapse
|
10
|
Maïza A, Hamoudi R, Mabondzo A. Targeting the Multiple Complex Processes of Hypoxia-Ischemia to Achieve Neuroprotection. Int J Mol Sci 2024; 25:5449. [PMID: 38791487 PMCID: PMC11121719 DOI: 10.3390/ijms25105449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/06/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a major cause of newborn brain damage stemming from a lack of oxygenated blood flow in the neonatal period. Twenty-five to fifty percent of asphyxiated infants who develop HIE die in the neonatal period, and about sixty percent of survivors develop long-term neurological disabilities. From the first minutes to months after the injury, a cascade of events occurs, leading to blood-brain barrier (BBB) opening, neuronal death and inflammation. To date, the only approach proposed in some cases is therapeutic hypothermia (TH). Unfortunately, TH is only partially protective and is not applicable to all neonates. This review synthesizes current knowledge on the basic molecular mechanisms of brain damage in hypoxia-ischemia (HI) and on the different therapeutic strategies in HI that have been used and explores a major limitation of unsuccessful therapeutic approaches.
Collapse
Affiliation(s)
- Auriane Maïza
- CEA, DMTS, SPI, Neurovascular Unit Research & Therapeutic Innovation Laboratory, Paris-Saclay University, CEDEX 91191 Gif-sur-Yvette, France;
| | - Rifat Hamoudi
- Center of Excellence of Precision Medicine, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates;
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London NW3 2PF, UK
| | - Aloïse Mabondzo
- CEA, DMTS, SPI, Neurovascular Unit Research & Therapeutic Innovation Laboratory, Paris-Saclay University, CEDEX 91191 Gif-sur-Yvette, France;
| |
Collapse
|
11
|
Zhao Y, Li T, Jiang Z, Gai C, Yu S, Xin D, Li T, Liu D, Wang Z. The miR-9-5p/CXCL11 pathway is a key target of hydrogen sulfide-mediated inhibition of neuroinflammation in hypoxic ischemic brain injury. Neural Regen Res 2024; 19:1084-1094. [PMID: 37862212 PMCID: PMC10749591 DOI: 10.4103/1673-5374.382860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/05/2022] [Accepted: 07/11/2023] [Indexed: 10/22/2023] Open
Abstract
We previously showed that hydrogen sulfide (H2S) has a neuroprotective effect in the context of hypoxic ischemic brain injury in neonatal mice. However, the precise mechanism underlying the role of H2S in this situation remains unclear. In this study, we used a neonatal mouse model of hypoxic ischemic brain injury and a lipopolysaccharide-stimulated BV2 cell model and found that treatment with L-cysteine, a H2S precursor, attenuated the cerebral infarction and cerebral atrophy induced by hypoxia and ischemia and increased the expression of miR-9-5p and cystathionine β synthase (a major H2S synthetase in the brain) in the prefrontal cortex. We also found that an miR-9-5p inhibitor blocked the expression of cystathionine β synthase in the prefrontal cortex in mice with brain injury caused by hypoxia and ischemia. Furthermore, miR-9-5p overexpression increased cystathionine-β-synthase and H2S expression in the injured prefrontal cortex of mice with hypoxic ischemic brain injury. L-cysteine decreased the expression of CXCL11, an miR-9-5p target gene, in the prefrontal cortex of the mouse model and in lipopolysaccharide-stimulated BV-2 cells and increased the levels of proinflammatory cytokines BNIP3, FSTL1, SOCS2 and SOCS5, while treatment with an miR-9-5p inhibitor reversed these changes. These findings suggest that H2S can reduce neuroinflammation in a neonatal mouse model of hypoxic ischemic brain injury through regulating the miR-9-5p/CXCL11 axis and restoring β-synthase expression, thereby playing a role in reducing neuroinflammation in hypoxic ischemic brain injury.
Collapse
Affiliation(s)
- Yijing Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Tong Li
- Department of Neurosurgery, Qingdao Municipal Hospital, Qingdao, Shandong Province, China
| | - Zige Jiang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Chengcheng Gai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Shuwen Yu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Danqing Xin
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Tingting Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao University, Jinan, Shandong Province, China
| |
Collapse
|
12
|
Andorka C, Barta H, Sesztak T, Nyilas N, Kovacs K, Dunai L, Rudas G, Jermendy A, Szabo M, Szakmar E. The predictive value of MRI scores for neurodevelopmental outcome in infants with neonatal encephalopathy. Pediatr Res 2024:10.1038/s41390-024-03189-1. [PMID: 38637693 DOI: 10.1038/s41390-024-03189-1] [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: 07/23/2023] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND MRI scoring systems are utilized to quantify brain injury and predict outcome in infants with neonatal encephalopathy (NE). Our aim was to evaluate the predictive accuracy of total scores, white matter (WM) and grey matter (GM) subscores of Barkovich and Weeke scoring systems for neurodevelopmental outcome at 2 years of age in infants receiving therapeutic hypothermia for NE. METHODS Data of 162 infants were analyzed in this retrospective cohort study. DeLong tests were used to compare areas under the curve of corresponding items of the two scoring systems. LASSO logistic regression was carried out to evaluate the association between MRI scores and adverse composite (death or severe disabilities), motor and cognitive outcomes (Bayley developmental index <70). RESULTS Weeke scores predicted each outcome measure with greater accuracy than the corresponding items of Barkovich system (DeLong tests p < 0.03). Total scores, GM and cerebellum involvement were associated with increased odds for adverse outcomes, in contrast to WM injury, after adjustment to 5' Apgar score, first postnatal lactate and aEEG normalization within 48 h. CONCLUSION A more detailed scoring system had better predictive value for adverse outcome. GM injury graded on both scoring systems was an independent predictor of each outcome measure. IMPACT STATEMENTS A more detailed MRI scoring system had a better predictive value for motor, cognitive and composite outcomes. While hypoxic-ischemic brain injuries in the deep grey matter and cerebellum were predictive of adverse outcome, white matter injury including cortical involvement was not associated with any of the outcome measures at 2 years of age. Structured MRI evaluation based on validated scores may aid future clinical research, as well as inform parents and caregivers to optimize care beyond the neonatal period.
Collapse
Affiliation(s)
- Csilla Andorka
- Division of Neonatology, Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Hajnalka Barta
- Division of Neonatology, Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Timea Sesztak
- Division of Neonatology, Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
- Department of Neuroradiology, Medical Imaging Center, Semmelweis University, Budapest, Hungary
| | - Nora Nyilas
- Department of Neuroradiology, Medical Imaging Center, Semmelweis University, Budapest, Hungary
| | - Kata Kovacs
- Division of Neonatology, Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Ludovika Dunai
- Division of Neonatology, Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Gabor Rudas
- Department of Neuroradiology, Medical Imaging Center, Semmelweis University, Budapest, Hungary
| | - Agnes Jermendy
- Division of Neonatology, Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Miklos Szabo
- Division of Neonatology, Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary
| | - Eniko Szakmar
- Division of Neonatology, Pediatric Center, MTA Center of Excellence, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
13
|
Fei Q, Wang D, Yuan T. Comparison of Different Adjuvant Therapies for Hypothermia in Neonates with Hypoxic-Ischemic Encephalopathy: A Systematic Review and Network Meta-Analysis. Indian J Pediatr 2024; 91:235-241. [PMID: 37199820 DOI: 10.1007/s12098-023-04563-3] [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: 11/17/2022] [Accepted: 03/10/2023] [Indexed: 05/19/2023]
Abstract
OBJECTIVES Neonatal hypoxic-ischemic encephalopathy is a major cause of perinatal death and neurodevelopmental impairment (NDI). Hypothermia (HT) is the standard of care; however, additional neuroprotective agents are required to improve prognosis. The authors searched for all drugs in combination with HT and compared their effects using a network meta-analysis. METHODS The authors searched PubMed, Embase, and Cochrane Library until September 24, 2022 for articles assessing mortality, NDI, seizures, and abnormal brain imaging findings in neonates with hypoxic-ischemic encephalopathy. Direct pairwise comparisons and a network meta-analysis was performed under random effects. RESULTS Thirteen randomized clinical trials enroled 902 newborns treated with six combination therapies: erythropoietin magnesium sulfate, melatonin (MT), topiramate, xenon, and darbepoetin alfa. The results of all comparisons were not statistically significant, except for NDI, HT vs. MT+HT: odds ratio = 6.67, 95% confidence interval = 1.14-38.83; however, the overall evidence quality was low for the small sample size. CONCLUSIONS Currently, no combination therapy can reduce mortality, seizures, or abnormal brain imaging findings in neonatal hypoxic-ischemic encephalopathy. According to low quality evidence, HT combined with MT may reduce NDI.
Collapse
Affiliation(s)
- Qiang Fei
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Binsheng Rd, No.3333, Hangzhou, 310003, China
| | - Dandan Wang
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Binsheng Rd, No.3333, Hangzhou, 310003, China
| | - Tianming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Binsheng Rd, No.3333, Hangzhou, 310003, China.
| |
Collapse
|
14
|
Hayashi S, Seki-Omura R, Yamada S, Kamata T, Sato Y, Oe S, Koike T, Nakano Y, Iwashita H, Hirahara Y, Tanaka S, Sekijima T, Ito T, Yasukochi Y, Higasa K, Kitada M. OLIG2 translocates to chromosomes during mitosis via a temperature downshift: A novel neural cold response of mitotic bookmarking. Gene 2024; 891:147829. [PMID: 37748631 DOI: 10.1016/j.gene.2023.147829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/09/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023]
Affiliation(s)
- Shinichi Hayashi
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan.
| | - Ryohei Seki-Omura
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Shintaro Yamada
- Department of Functional Neuroscience, Institute of Biomedical Science, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Taito Kamata
- Graduate School of Science and Technology, Niigata University, 8050 Ikarashi 2-nocho, Niigata, Japan; Faculty of Agriculture, Niigata University, 8050 Ikarashi 2-nocho, Niigata, Japan
| | - Yuki Sato
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Souichi Oe
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Taro Koike
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Yousuke Nakano
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Hikaru Iwashita
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Yukie Hirahara
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan; Faculty of Nursing, Kansai Medical University, Shinmachi 2-2-2, Hirakata, Osaka, Japan
| | - Susumu Tanaka
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan; Department of Anatomy and Physiology, Faculty of Nursing and Nutrition, University of Nagasaki, Manabino 1-1-1, Nagasaki, Japan
| | - Tsuneo Sekijima
- Faculty of Agriculture, Niigata University, 8050 Ikarashi 2-nocho, Niigata, Japan
| | - Takeshi Ito
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Yoshiki Yasukochi
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Koichiro Higasa
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan
| | - Masaaki Kitada
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, Japan.
| |
Collapse
|
15
|
Tian J, Zhu J, Fan Q, Luo X, Nie Q, Yu J, Wu X, Tang Y, Liu T, Yin H. Interleukin-33 improves the neurogenesis of neural stem cells in perinatal brain after hypoxia-ischemia. Int Immunopharmacol 2023; 123:110778. [PMID: 37573691 DOI: 10.1016/j.intimp.2023.110778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/30/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
Perinatal hypoxia-ischemia (HI) insult is an important cause of neonatal encephalopathy, and the effective therapeutic approaches are currently limited. Interleukin (IL)-33 acts as a member of the IL-1 superfamily and has been shown to be neuroprotective following experimental neonatal HI and adult stroke. Here, we explore the effect of IL-33 and its specific receptor ST2 axis on endogenous neurogenesis in neonatal brain after HI. ST2 was found on the surface of NSCs, and the expression of ST2 was further enhanced after HI challenge. Delivery of IL-33 obviously repopulated the size of NSC pool, whereas ST2 deficiency worsened the neurogenesis of NSCs in neonatal brain post HI insult. Further in vivo and in vitro studies showed IL-33 regulates the survival, proliferation and differentiation of NSCs through ST2 signaling pathways. Intriguingly, IL-33 facilitated translocation of Nrf2 from the cytoplasm to the nucleus, which is involved in neural differentiation of NSCs. These data demonstrate a critical role of IL-33/ST2 axis in regulation of endogenous neurogenesis of NSCs via activation of the Nrf2 signaling, which provide a new insight into the effect of IL-33 in neonatal brain following HI injury.
Collapse
Affiliation(s)
- Jing Tian
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jieqiong Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qiuxiang Fan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaotian Luo
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qianying Nie
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jingwei Yu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaoyong Wu
- School of Food Science, Guangdong Pharmaceutical University, Zhongshan 528453, China
| | - Yanli Tang
- Department of Pediatrics, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen 518172, China
| | - Tao Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Hui Yin
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
16
|
Ueda K, Sato Y, Shimizu S, Suzuki T, Onoda A, Miura R, Go S, Mimatsu H, Kitase Y, Yamashita Y, Irie K, Tsuji M, Mishima K, Mizuno M, Takahashi Y, Dezawa M, Hayakawa M. Systemic administration of clinical-grade multilineage-differentiating stress-enduring cells ameliorates hypoxic-ischemic brain injury in neonatal rats. Sci Rep 2023; 13:14958. [PMID: 37696826 PMCID: PMC10495445 DOI: 10.1038/s41598-023-41026-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/21/2023] [Indexed: 09/13/2023] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are endogenous reparative pluripotent stem cells present in the bone marrow, peripheral blood, and organ connective tissues. We assessed the homing and therapeutic effects of systemically administered nafimestrocel, a clinical-grade human Muse cell-based product, without immunosuppressants in a neonatal hypoxic-ischemic (HI) rat model. HI injury was induced on postnatal day 7 (P7) and was confirmed by T2-weighted magnetic resonance imaging on P10. HI rats received a single dose nafimestrocel (1 × 106 cells/body) or Hank's balanced salt solution (vehicle group) intravenously at either three days (on P10; M3 group) or seven days (on P14; M7 group) after HI insult. Radioisotope experiment demonstrated the homing of chromium-51-labeled nafimestrocel to the both cerebral hemispheres. The cylinder test (M3 and M7 groups) and open-field test (M7 group) showed significant amelioration of paralysis and hyperactivity at five weeks of age compared with those in the vehicle group. Nafimestrocel did not cause adverse events such as death or pathological changes in the lung at ten weeks in the both groups. Nafimestrocel attenuated the production of tumor necrosis factor-α and inducible nitric oxide synthase from activated cultured microglia in vitro. These results demonstrate the potential therapeutic benefits and safety of nafimestrocel.
Collapse
Affiliation(s)
- Kazuto Ueda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan.
| | - Shinobu Shimizu
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Toshihiko Suzuki
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| | - Atsuto Onoda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
- Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi, Japan
| | - Ryosuke Miura
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| | - Shoji Go
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| | - Haruka Mimatsu
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| | - Yuma Kitase
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| | - Yuta Yamashita
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Keiichi Irie
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Masahiro Tsuji
- Department of Food and Nutrition, Faculty of Home Economics, Kyoto Women's University, Kyoto, Japan
| | - Kenichi Mishima
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Masaaki Mizuno
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| |
Collapse
|
17
|
ERGON EY, ÇELİK A, DİNİZ G, ÇOLAK R, ÖZDEMİR SA, ÇALKAVUR Ş, YILMAZ O. Evaluation of syringin's neuroprotective effect in a model of neonatal hypoxic-ischemic brain injury. Turk J Med Sci 2023; 53:1312-1320. [PMID: 38813032 PMCID: PMC10763795 DOI: 10.55730/1300-0144.5697] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 10/26/2023] [Accepted: 06/21/2023] [Indexed: 05/31/2024] Open
Abstract
Background/aim A significant cause of mortality and morbidity in the neonatal era is hypoxic-ischemic encephalopathy (HIE). This study examined the histopathological analysis and neuroprotective impact of syringin (SYR) in an experimental HIE rat model. Material and methods On the 7th postnatal day, 24 Wistar albino rats were evaluated in 3 groups using the HIE model under gas anesthesia. In the experiment, Group A received 10 mg/kg SYR plus dimethyl sulfoxide (DMSO), Group B received DMSO only, and Group C served as a sham group. Immunohistochemical techniques were used to assess apoptotic cell measurement and proinflammatory cytokines (TNF-α and IL-1β primary antibodies). Results Rats suffering from hypoxic-ischemic brain damage had their apoptosis assessed. The SYR and sham groups had statistically fewer cells undergoing apoptosis (p < 0.001). There was no difference between the groups in terms of IL-1β and TNF-α during immunohistochemical staining. Neuronal degeneration was significantly lower in the histological evaluation of the hippocampus in the SYR group (p = 0.01). A statistically significant difference (p = 0.01) was observed between the SYR and the control groups regarding pericellular and perivascular edema. Conclusion SYR reduced apoptosis, perivascular and pericellular edema, and neuronal degeneration in rat cerebral tissue. These results raise the possibility that SYR may have a neuroprotective effect on the harm brought on by HIE. This is the first investigation of SYR's function within the HIE paradigm.
Collapse
Affiliation(s)
- Ezgi Yangın ERGON
- Neonatal Intensive Care Unit, Pediatric Division, Dr Behçet Uz Children’s Education and Research Hospital, İzmir,
Turkiye
| | - Aslı ÇELİK
- Department of Laboratory Animal Science, Faculty of Health Sciences, Dokuz Eylül University, İzmir,
Turkiye
| | - Gülden DİNİZ
- Department of Pathology, Medical Faculty, İzmir Democracy University, İzmir,
Turkiye
| | - Rüya ÇOLAK
- Neonatal Intensive Care Unit, Pediatric Division, Medikal Park Florya Hospital, Aydın University Medical Faculty, İstanbul,
Turkiye
| | - Senem Alkan ÖZDEMİR
- Neonatal Intensive Care Unit, Pediatric Division, Dr Behçet Uz Children’s Education and Research Hospital, İzmir,
Turkiye
| | - Şebnem ÇALKAVUR
- Neonatal Intensive Care Unit, Pediatric Division, Dr Behçet Uz Children’s Education and Research Hospital, İzmir,
Turkiye
| | - Osman YILMAZ
- Department of Laboratory Animal Science, Faculty of Health Sciences, Dokuz Eylül University, İzmir,
Turkiye
| |
Collapse
|
18
|
Lin C, Li L, Xu Q, Xu S, Tang C. Yap1-Usp14 Axis Inhibits Neuronal Mitophagy During Neonatal Hypoxia-Ischemia Encephalopathy by Regulation of Beclin-1 Ubiquitination in Mouse. Mol Neurobiol 2023:10.1007/s12035-023-03344-5. [PMID: 37062801 DOI: 10.1007/s12035-023-03344-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/07/2023] [Indexed: 04/18/2023]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) that results from perinatal cerebral hypoxia-ischemia has become one of the leading causes of acute mortality and chronic disability in infants and children. Despite that neuronal mitophagy and subsequent clearance of damaged neurons exert protective effect, the pathogenesis of HIE and effective treatment strategies for intervention of HIE remain poorly understood. Here, we report that ubiquitin-specific protease 14 (Usp14, a deubiquitinating enzyme) is closely associated with HIE progression by its negative regulation in neuronal mitophagy in mouse. The expression of Usp14 is elevated in both an oxygen-glucose deprivation (OGD) mouse neuronal cell line culture model in vitro and a HIE mouse model in vivo. Mechanistically, OGD treatment activates Hippo signaling that enhances Yap1 phosphorylation levels at Ser-127 but inhibits Yap1 protein level, which potentiates Usp14 transcription and leads to the downregulated ubiquitination at Lys-63 of Beclin-1, a key molecule in autophagy, resulting in the suppressed neuronal mitophagy, subsequent failure in the clearance of damaged neurons, and finally possible dysregulation in brain functions. Thus, our results provide with Usp14 as a novel target and treatment strategy for intervention of HIE, which may help diagnose and treat HIE in clinic.
Collapse
Affiliation(s)
- Chao Lin
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
- Department of Neurosurgery, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Lin Li
- Depanrtment of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China
| | - Qiang Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Shouying Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Chao Tang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
| |
Collapse
|
19
|
Girolamo F, Lim YP, Virgintino D, Stonestreet BS, Chen XF. Inter-Alpha Inhibitor Proteins Modify the Microvasculature after Exposure to Hypoxia-Ischemia and Hypoxia in Neonatal Rats. Int J Mol Sci 2023; 24:6743. [PMID: 37047713 PMCID: PMC10094872 DOI: 10.3390/ijms24076743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
Microvasculature develops during early brain development. Hypoxia-ischemia (HI) and hypoxia (H) predispose to brain injury in neonates. Inter-alpha inhibitor proteins (IAIPs) attenuate injury to the neonatal brain after exposure to HI. However, the effects of IAIPs on the brain microvasculature after exposure to HI have not been examined in neonates. Postnatal day-7 rats were exposed to sham treatment or right carotid artery ligation and 8% oxygen for 90 min. HI comprises hypoxia (H) and ischemia to the right hemisphere (HI-right) and hypoxia to the whole body, including the left hemisphere (H-left). Human IAIPs (hIAIPs, 30 mg/kg) or placebo were injected immediately, 24 and 48 h after HI/H. The brains were analyzed 72 h after HI/H to determine the effects of hIAIPs on the microvasculature by laminin immunohistochemistry and calculation of (1) the percentage area stained by laminin, (2) cumulative microvessel length, and (3) density of tunneling nanotubes (TNTs), which are sensitive indicators of the earliest phases of neo-vascularization/collateralization. hIAIPs mainly affected the percent of the laminin-stained area after HI/H, cumulative vessel length after H but not HI, and TNT density in females but not males. hIAIPs modify the effects of HI/H on the microvasculature after brain injury in neonatal rats and exhibit sex-related differential effects. Our findings suggest that treatment with hIAIPs after exposure to H and HI in neonatal rats affects the laminin content of the vessel basal lamina and angiogenic responses in a sex-related fashion.
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Translational Biomedicines and Neuroscience (DiBraiN), University of Bari School of Medicine, 70124 Bari, Italy
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02905, USA
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Daniela Virgintino
- Department of Translational Biomedicines and Neuroscience (DiBraiN), University of Bari School of Medicine, 70124 Bari, Italy
| | - Barbara S. Stonestreet
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Xiaodi F. Chen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| |
Collapse
|
20
|
Narayanamurthy R, Armstrong EA, Yang JLJ, Yager JY, Unsworth LD. Administration of selective brain hypothermia using a simple cooling device in neonatal rats. J Neurosci Methods 2023; 390:109838. [PMID: 36933705 DOI: 10.1016/j.jneumeth.2023.109838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The interruption of oxygen and blood supply to the newborn brain around the time of birth is a risk factor for hypoxic-ischemic encephalopathy and may lead to infant mortality or lifelong neurological impairments. Currently, therapeutic hypothermia, the cooling of the infant's head or entire body, is the only treatment to curb the extent of brain damage. NEW METHOD In this study, we designed a focal brain cooling device that circulates cooled water at a steady state temperature of 19 ± 1 °C through a coil of tubing fitted onto the neonatal rat's head. We tested its ability to selectively decrease brain temperature and offer neuroprotection in a neonatal rat model of hypoxic-ischemic brain injury. RESULTS Our method cooled the brain to 30-33 °C in conscious pups, while keeping the core body temperature approximately 3.2 °C warmer. Furthermore, the application of the cooling device to the neonatal rat model demonstrated a reduction in brain volume loss compared to pups maintained at normothermia and achieved a level of brain tissue protection the same as that of whole-body cooling. COMPARISON WITH EXISTING METHODS Prevailing methods of selective brain hypothermia are designed for adult animal models rather than for immature animals such as the rat as a conventional model of developmental brain pathology. Contrary to existing methods, our method of cooling does not require surgical manipulation or anaesthesia. CONCLUSION Our simple, economical, and effective method of selective brain cooling is a useful tool for rodent studies in neonatal brain injury and adaptive therapeutic interventions.
Collapse
Affiliation(s)
- Rukhmani Narayanamurthy
- Department of Pediatrics, Division of Pediatric Neurosciences, University of Alberta, 11405 87 Avenue, Edmonton, Alberta T6G 1C9, Canada
| | - Edward A Armstrong
- Department of Pediatrics, Division of Pediatric Neurosciences, University of Alberta, 11405 87 Avenue, Edmonton, Alberta T6G 1C9, Canada
| | - Jung-Lynn Jonathan Yang
- Department of Chemical and Materials Engineering, University of Alberta, 11487 89 Avenue, Edmonton, Alberta T6G 2M7, Canada
| | - Jerome Y Yager
- Department of Pediatrics, Division of Pediatric Neurosciences, University of Alberta, 11405 87 Avenue, Edmonton, Alberta T6G 1C9, Canada
| | - Larry D Unsworth
- Department of Chemical and Materials Engineering, University of Alberta, 11487 89 Avenue, Edmonton, Alberta T6G 2M7, Canada.
| |
Collapse
|
21
|
Tan X, Zhang T, Ding X, Zhao X, Liu Q, Xia Z, Cao Q, Yan F, Chen L, Zhu M, Tang Y, Song Y. Iron overload facilitates neonatal hypoxic-ischemic brain damage via SLC7A11-mediated ferroptosis. J Neurosci Res 2023; 101:1107-1124. [PMID: 36929608 DOI: 10.1002/jnr.25184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 12/26/2022] [Accepted: 01/09/2023] [Indexed: 03/18/2023]
Abstract
Oxidative damage and cell death are involved in the pathogenesis of hypoxic-ischemic brain damage (HIBD). Ferroptosis is a newly identified mode of cell death that results from the oxidative damage induced by excessive iron. In HIBD, iron accumulates in brain tissues due to the massive destruction of red blood cells and increased permeability of the blood brain barrier vasculature, which can trigger ferroptosis. Ferroptosis is implicated in various diseases involving neuronal injury; however, the roles of iron and ferroptosis in HIBD have not been identified. In the present study, we investigated the role of iron overload in neuronal ferroptosis both in HIBD rat models and in oxygen- and glucose-deprived (OGD) SH-SY5Y cells. We observed that iron deposition in the cerebral cortex was significantly increased in HIBD rats. Features of ferroptosis such as shrunken mitochondria, increased MDA (malondialdehyde) levels, and reduced solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) expression were observed in the cerebral cortex of HIBD rats. Administration of an iron chelator in HIBD rats upregulated SLC7A11 expression and alleviated neuronal ferroptosis in cerebral cortex tissue. Additionally, overexpression of SLC7A11 in SH-SY5Y cells increased cell viability and attenuated OGD-induced ferroptosis. Our results demonstrate that iron overload induces neuronal ferroptosis by inhibiting SLC7A11 expression in HIBD. Inhibition of neuronal ferroptosis may be a promising strategy to alleviate brain damage in HIBD.
Collapse
Affiliation(s)
- Xuying Tan
- Department of Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Ting Zhang
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Xuejiao Ding
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Xiaopeng Zhao
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Qianjun Liu
- Department of Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Zhenglong Xia
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Qihua Cao
- Department of Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Feng Yan
- Department of Delivery Room, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Li Chen
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Mingwei Zhu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Yaping Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Yanyan Song
- Department of Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| |
Collapse
|
22
|
Pluta R, Furmaga-Jabłońska W, Januszewski S, Tarkowska A. Melatonin: A Potential Candidate for the Treatment of Experimental and Clinical Perinatal Asphyxia. Molecules 2023; 28:1105. [PMID: 36770769 PMCID: PMC9919754 DOI: 10.3390/molecules28031105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Perinatal asphyxia is considered to be one of the major causes of brain neurodegeneration in full-term newborns. The worst consequence of perinatal asphyxia is neurodegenerative brain damage, also known as hypoxic-ischemic encephalopathy. Hypoxic-ischemic encephalopathy is the leading cause of mortality in term newborns. To date, due to the complex mechanisms of brain damage, no effective or causal treatment has been developed that would ensure complete neuroprotection. Although hypothermia is the standard of care for hypoxic-ischemic encephalopathy, it does not affect all changes associated with encephalopathy. Therefore, there is a need to develop effective treatment strategies, namely research into new agents and therapies. In recent years, it has been pointed out that natural compounds with neuroprotective properties, such as melatonin, can be used in the treatment of hypoxic-ischemic encephalopathy. This natural substance with anti-inflammatory, antioxidant, anti-apoptotic and neurofunctional properties has been shown to have pleiotropic prophylactic or therapeutic effects, mainly against experimental brain neurodegeneration in hypoxic-ischemic neonates. Melatonin is a natural neuroprotective hormone, which makes it promising for the treatment of neurodegeneration after asphyxia. It is supposed that melatonin alone or in combination with hypothermia may improve neurological outcomes in infants with hypoxic-ischemic encephalopathy. Melatonin has been shown to be effective in the last 20 years of research, mainly in animals with perinatal asphyxia but, so far, no clinical trials have been performed on a sufficient number of newborns. In this review, we summarize the advantages and limitations of melatonin research in the treatment of experimental and clinical perinatal asphyxia.
Collapse
Affiliation(s)
- Ryszard Pluta
- Ecotech-Complex Analytical and Programme Centre for Advanced Environmentally-Friendly Technologies, Marie Curie-Skłodowska University in Lublin, 20-612 Lublin, Poland
| | - Wanda Furmaga-Jabłońska
- Department of Neonate and Infant Pathology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Sławomir Januszewski
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Agata Tarkowska
- Department of Neonate and Infant Pathology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
23
|
The relationship between miRNA-210 and SCN1B in fetal rats with hypoxic-ischemic brain injury. Biosci Rep 2023; 43:232346. [PMID: 36541246 PMCID: PMC9842947 DOI: 10.1042/bsr20222016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/02/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Hypoxic-ischemic brain injury contributes to major neurodevelopmental disorders and is one of the leading causes of seizures, which substantially results in neurodevelopmental impairments with long-lasting outcomes and is one of the main causes of death in neonates. We aimed to investigate the correlation between miRNA-210 and SCN1B, a voltage-gated sodium channel gene, in brain tissue of fetal rats with hypoxic-ischemic brain injury. We found that after 10 min of hypoxia-ischemia, all reperfusion groups showed different degrees of damage. The degree of the injury increased in all the groups after 30 min of hypoxia-ischemia. Those changes include changes in the pericellular lumen, capillaries in the cortex, erythrocytes, enlarged pericellular lumen, the enlarged pericapillary lumen in the cortex, edema around glial cells, enlarged gap to form multiple necrotic foci, deformation of neurons, and loss of cell structure. The expression levels of HIF-1α, miRNA-210, and HIF-1α mRNA were higher in the hypoxic-ischemic groups than that in the control groups, among which the expression levels in the severe group were higher than that in mild group. SCN1B is down-regulated in both the mild and severe groups, and the lowest level was found at 30 min after hypoxia in both groups. MiRNA-210 plays a role in the development of hypoxic-ischemic encephalopathy (HIE) by regulating the expression changes of SCN1B. The brain tissue of fetal rats in the hypoxic-ischemic animal model showed pathological changes of brain injury.
Collapse
|
24
|
Repurposed Edaravone, Metformin, and Perampanel as a Potential Treatment for Hypoxia-Ischemia Encephalopathy: An In Vitro Study. Biomedicines 2022; 10:biomedicines10123043. [PMID: 36551799 PMCID: PMC9775340 DOI: 10.3390/biomedicines10123043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Hypoxia-ischemia encephalopathy results from the interruption of oxygen delivery and blood flow to the brain. In the developing brain, it can lead to a brain injury, which is associated with high mortality rates and comorbidities. The hippocampus is one of the brain regions that may be affected by hypoxia-ischemia with consequences on cognition. Unfortunately, clinically approved therapeutics are still scarce and limited. Therefore, in this study, we aimed to test three repurposed drugs with good pharmacological properties to evaluate if they can revert, or at least attenuate, the deleterious effects of hypoxia-ischemia in an in vitro model. Edaravone, perampanel, and metformin are used for the treatment of stroke and amyotrophic lateral sclerosis, some forms of epileptic status, and diabetes type 2, respectively. Through cell viability assays, morphology analysis, and detection of reactive oxygen species (ROS) production, in two different cell lines (HT-22 and SH-SY5Y), we found that edaravone and low concentrations of perampanel are able to attenuate cell damage induced by hypoxia and oxygen-glucose deprivation. Metformin did not attenuate hypoxic-induced events, at least in the initial phase. Among these repurposed drugs, edaravone emerged as the most efficient in the attenuation of events induced by hypoxia-ischemia, and the safest, since it did not exhibit significant cytotoxicity, even in high concentrations, and induced a decrease in ROS. Our results also reinforce the view that ROS and overexcitation play an important role in the pathophysiology of hypoxia-ischemia brain injury.
Collapse
|
25
|
Hashish M, Bassiouny MR. Neonatal seizures: stepping outside the comfort zone. Clin Exp Pediatr 2022; 65:521-528. [PMID: 35381172 PMCID: PMC9650361 DOI: 10.3345/cep.2022.00115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/22/2022] [Indexed: 11/27/2022] Open
Abstract
Seizures are the most common neurological disorders in newborns. Managing neonatal seizures is challenging, especially for neurologists who are not neonatal specialists. Acute brain injury during ischemic insult is a key component of seizure occurrence, while genetic and metabolic disorders play less prevalent but more severe roles. The diagnosis of neonatal seizure is ambiguous, as the subjective differentiation between seizure and nonepileptic events is difficult; therefore, electrographic recording is the gold standard for diagnosis. The detection of electrographic seizures by neonatologists is currently facilitated by amplitude-integrated electroencephalography availability in many neonatal intensive care units. Although it is less sensitive than conventional electroencephalography, it is better to record all risky neonates to filter the abnormal events as early as possible to enable the initiation of dedicated therapy at proper dose and time and facilitate the initial response to antiepileptic drugs. This, in turn, helps maintain the balance between unnecessary drug use and their neurotoxic effects. Moreover, the early treatment of electrographic seizures plays a vital role in the suppression of subsequent abnormal brain electricity (status epilepticus) and shortening the hospital stay. An explicit understanding of seizure etiology and pathophysiology should direct attention to the proper prescription of short- and long-term antiepileptic medications to solve the challenging issue of whether neonatal seizures progress to postneonatal epilepsy and long-term cognitive deficits. This review addresses recent updates in different aspects of neonatal seizures, particularly electrographic discharge, including their definition, etiology, classification, diagnosis, management, and neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Menna Hashish
- Neonatal Intensive Care Unit, Mansoura University Children's Hospital, Mansoura, Egypt
| | | |
Collapse
|
26
|
Transcriptional regulation of NRF1 on metabotropic glutamate receptors in a neonatal hypoxic‑ischemic encephalopathy rat model. Pediatr Res 2022:10.1038/s41390-022-02353-9. [PMID: 36280709 DOI: 10.1038/s41390-022-02353-9] [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: 05/16/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Neonatal hypoxic-ischemic encephalopathy (HIE) is a kind of brain injury that causes severe neurological disorders in newborns. Metabotropic glutamate receptors (mGluRs) and ionotropic glutamate receptors (iGluRs) are significantly associated with HIE and are involved in ischemia-induced excitotoxicity. This study aimed to investigate the upstream mechanisms of mGluRs and the transcriptional regulation by nuclear respiratory factor 1 (NRF1). METHODS The rat model of neonatal HIE was created using unilateral carotid artery ligation and in vitro oxygen-glucose deprivation paradigm. We used western blot, immunofluorescence, Nissl staining, and Morris water maze to investigate the impact of NRF1 on brain damage and learning memory deficit by HIE. We performed ChIP and luciferase activities to identify the transcriptional regulation of NRF1 on mGluRs. RESULTS The neuronal NRF1 and some glutamatergic genes expression synchronously declined in infarcted tissues. The NRF1 overexpression effectively restored the expression of some glutamatergic genes and improved cognitive performance. NRF1 regulated some members of mGluRs and iGluRs in hypoxic-ischemic neurons. Finally, NRF1 is bound to the promoter regions of Grm1, Grm2, and Grm8 to activate their transcription. CONCLUSIONS NRF1 is involved in the pathology of the neonatal HIE rat model, suggesting a novel therapeutic approach to neonatal HIE. IMPACT NRF1 and some glutamatergic genes were synchronously downregulated in the infarcted brain of the neonatal HIE rat model. NRF1 overexpression could rescue cognitive impairment caused by the neonatal HIE rat model. NRF1 regulated the expressions of Grm1, Grm2, and Grm8, which activated their transcription by binding to the promoter regions.
Collapse
|
27
|
Li J, Feng Y, Zhao J, Fang Z, Liu H. Telomerase reverse transcriptase promotes angiogenesis in neonatal rats after hypoxic-ischemic brain damage. PeerJ 2022; 10:e14220. [PMID: 36299510 PMCID: PMC9590416 DOI: 10.7717/peerj.14220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/20/2022] [Indexed: 01/24/2023] Open
Abstract
Background Angiogenesis is an endogenous repair mechanism following hypoxic-ischemic brain damage (HIBD). Interestingly, recent studies have shown that angiogenesis can be regulated by telomerase reverse transcriptase (TERT), a critical component of telomerase. As telomerase reverse transcriptase can promote angiogenesis after stroke, we hypothesized that it could also promote angiogenesis after HIBD. To test this hypothesis, we developed in vivo and in vitro HIBD models in neonatal rats. Methods TERT was overexpressed by lentivirus and adenovirus infection, and levels were measured using quantitative real-time polymerase chain reaction. We used a cell counting kit to quantify the proliferation rate of brain microvascular endothelial cells (BMECs), and immunofluorescence staining to measure CD34 expression levels. A microvessel formation assay was used to evaluate angiogenesis. Blood-brain barrier (BBB) integrity was assessed using immunohistochemical staining for ZO-1 and Evans Blue staining. Lastly, the expression level of Notch-1 was measured by western blotting. Results Overexpression of TERT promoted the proliferation of BMECs after hypoxic-ischemic damage in vitro. TERT overexpression increased the formation of microvessels in the neonatal brain after HIBD both in vivo and in vitro. Overexpression of TERT improved BBB integrity in the brains of neonatal rats after HIBD. In addition, the expression level of Notch-1 was increased in BMECs following oxygen glucose deprivation, and overexpression of TERT further increased Notch-1 expression levels in BMECs following oxygen glucose deprivation. Discussion Our results reveal that telomerase reverse transcriptase promotes angiogenesis and maintains the integrity of the blood-brain barrier after neonatal hypoxic-ischemic brain damage. Furthermore, the Notch-1 signaling pathway appears to contribute to the angiogenic function of telomerase reverse transcriptase. This protective effect of telomerase reverse transcriptase opens new horizons for future investigations aimed at uncovering the full potential of telomerase reverse transcriptase as a promising new target for the treatment of hypoxic-ischemic encephalopathy.
Collapse
Affiliation(s)
- Jiao Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Feng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jing Zhao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhi Fang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Haiting Liu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Wang Q, Lv H, Wu S, Song J, Li J, Huo H, Ren P, Li L. Effect of Hypothermia on Serum Myelin Basic Protein and Tumor Necrosis Factor-α in Neonatal Hypoxic-Ischemic Encephalopathy. Am J Perinatol 2022; 39:1367-1374. [PMID: 33454948 DOI: 10.1055/s-0040-1722601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Multiple randomized controlled trials have shown that hypothermia is a safe and effective treatment for neonatal moderate or severe hypoxic-ischemic encephalopathy (HIE). The neuroprotective mechanisms of hypothermia need further study. The aim of this study was to investigate the effect of hypothermia on the serum levels of myelin basic protein (MBP) and tumor necrosis factor-α (TNF-α) as well as neurodevelopmental outcomes in neonatal HIE. STUDY DESIGN Eighty-five neonates with moderate-to-severe HIE were divided into a hypothermia group (n = 49) and a control group (n = 36). Serum levels of MBP and TNF-α within 6 hours after birth and after 3 days of treatment were determined by enzyme-linked immunosorbent assay, and neurodevelopmental outcome at the age of 12 to 15 months was assessed by using the Gesell development scale. RESULTS After 3 days of treatment, serum levels of MBP and TNF-α in the control group were not significantly different from levels before treatment (p > 0.05), and serum levels of MBP and TNF-α in the hypothermia group were significantly lower than levels before treatment (p < 0.05). Serum levels of MBP and TNF-α were significantly negatively correlated with developmental quotient (DQ; r = - 0.7945, p = 0.0000; r = - 0.7035, p = 0.0000, respectively). Serum levels of MBP and TNF-α in neurodevelopmentally impaired infants were significantly higher than those in infants with suspected neurodevelopmental impairment and those in neurodevelopmentally normal infants (both p < 0.01). The rate of reduction of neurodevelopmental impairment was higher among infants in the hypothermia group than among those in the control group (χ2 = 16.3900, p < 0.05). CONCLUSION Hypothermia can reduce serum levels of MBP and TNF-α in neonates with HIE. Inhibiting the release of TNF-α may be one of the mechanisms by which hypothermia protects the myelin sheath. KEY POINTS · Hypothermia can reduce serum levels of MBP and TNF-α in neonatal HIE.. · Hypothermia improves neurodevelopmental outcomes and reduces the rate of neurodevelopmental impairment.. · Hypothermia is a feasible and effective treatment for neonates with moderate or severe HIE..
Collapse
Affiliation(s)
- Qiuli Wang
- Department of Neonatology, Handan Maternal and Child Health Care Hospital, Handan, Hebei Province, People's Republic of China
| | - Hongyan Lv
- Department of Neonatology, Handan Maternal and Child Health Care Hospital, Handan, Hebei Province, People's Republic of China.,Department of Neonatal Pathology, Handan Maternal and Child Health Care Hospital, Handan, Hebei Province, People's Republic of China
| | - Sujing Wu
- Department of Neonatology, Handan Maternal and Child Health Care Hospital, Handan, Hebei Province, People's Republic of China
| | - Junxia Song
- Department of Neonatology, Handan Maternal and Child Health Care Hospital, Handan, Hebei Province, People's Republic of China
| | - Junqin Li
- Department of Neonatology, Handan Maternal and Child Health Care Hospital, Handan, Hebei Province, People's Republic of China
| | - Haihua Huo
- Department of Neonatology, Handan Maternal and Child Health Care Hospital, Handan, Hebei Province, People's Republic of China
| | - Pengshun Ren
- Department of Neonatology, Handan Maternal and Child Health Care Hospital, Handan, Hebei Province, People's Republic of China
| | - Lianxiang Li
- Department of Neonatal Pathology, Handan Maternal and Child Health Care Hospital, Handan, Hebei Province, People's Republic of China.,Department of Neural Development and Neural Pathology, Hebei University of Engineering School of Medicine, Handan, Hebei Province, People's Republic of China
| |
Collapse
|
29
|
Remifentanil alleviates hypoxic-ischemic brain damage-induced cognitive impairment via BACH1. Neurosci Lett 2022; 786:136802. [PMID: 35853564 DOI: 10.1016/j.neulet.2022.136802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 11/20/2022]
Abstract
Hypoxia-ischemia (HI) is among the most frequent causes of death and disability in neonates. We aimed here to examine the neuroprotective effects of Remifentanil (RE) and the underlying mechanisms in a rat model of hypoxic-ischemic brain damage (HIBD). We found that RE improved the learning memory ability, reduced neuronal cell damage and apoptosis, reduced inflammation induced by suppressing the expression of BTB domain and CNC homolog 1 (BACH1) in rats with HIBD. BACH1 attenuated the alleviating effect of RE on cognitive impairment in HIBD rats. Moreover, RE inhibited TRAF3 expression by downregulating BACH1, and TRAF3 attenuated the therapeutic effect of RE on cognitive impairment by activating the NF-κB signaling. In conclusion, our findings demonstrated that RE inhibits the expression of BACH1, which in turn inhibits the NF-κB signaling pathway by suppressing TRAF3. RE may be a promising therapeutic agent to attenuate HIBD-induced cognitive impairment.
Collapse
|
30
|
Bäcke P, Bruschettini M, Blomqvist YT, Olsson E. Interventions for the management of Pain and Sedation in Newborns undergoing Therapeutic hypothermia for hypoxic-ischemic encephalopathy (IPSNUT): protocol of a systematic review. Syst Rev 2022; 11:101. [PMID: 35606836 PMCID: PMC9128112 DOI: 10.1186/s13643-022-01982-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Clinical research has shown that therapeutic hypothermia after neonatal hypoxic-ischemic injury improves survival without disability. There is no consensus regarding pain relief or sedation during therapeutic hypothermia in newborns; however, therapeutic hypothermia seems to be associated with pain and stress, and adequate analgesia and sedation are central to maximize the effect of therapeutic hypothermia. Pain needs to be adequately managed in all patients, especially the newborn infant due to the potential short- and long-term negative effects of inadequately treated pain in this population. METHODS We will perform a systematic review of pharmacological and non-pharmacological interventions for the management of pain and sedation in newborn infants undergoing therapeutic hypothermia for hypoxic-ischemic encephalopathy. We will include randomized, quasi-randomized controlled trials and observational studies. The use of pharmacological or non-pharmacological interventions will be compared to other pharmacological and or non-pharmacological interventions or no intervention/placebo. The primary outcomes for this review will be analgesia and sedation assessed with validated pain scales, circulatory instability, mortality to discharge, and moderate-to-severe neurodevelopmental disability. We will search the following databases: CINAHL, ClinicalTrials.gov , Cochrane Library, Embase, PubMed, Scopus, and Web of Science. Two independent researchers will screen the records for inclusion, extract data using a data extraction form, and assess the risk of bias in the included trials. DISCUSSION The result of this review will summarize the knowledge regarding the management of pain and sedation in infants treated with therapeutic hypothermia and potentially provide clinicians with guidance on the effective and safe methods. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42020205755.
Collapse
Affiliation(s)
- Pyrola Bäcke
- University Hospital, Neonatal Intensive Care Unit, Uppsala, Sweden.,Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Matteo Bruschettini
- Department of Pediatrics, Lund University, Lund, Sweden.,Cochrane Sweden, Research and Development, Skåne University Hospital, Lund, Sweden
| | - Ylva Thernström Blomqvist
- University Hospital, Neonatal Intensive Care Unit, Uppsala, Sweden.,Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Emma Olsson
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, Örebro, Sweden. .,Faculty of Medicine and Health, School of Health Sciences, Örebro University, 701 82, Örebro, Sweden.
| |
Collapse
|
31
|
Zhong W, Cheng J, Yang X, Liu W, Li Y. Heliox Preconditioning Exerts Neuroprotective Effects on Neonatal Ischemia/Hypoxia Injury by Inhibiting Necroptosis Induced by Ca 2+ Elevation. Transl Stroke Res 2022; 14:409-424. [PMID: 35445968 DOI: 10.1007/s12975-022-01021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/11/2022] [Accepted: 04/09/2022] [Indexed: 10/18/2022]
Abstract
Our previous studies have indicated that heliox preconditioning (HePC) may exert neuroprotective effects on neonatal hypoxic-ischemic encephalopathy (HIE). The present study was to investigate whether HePC alleviates neonatal HIE by inhibiting necroptosis and explore the potential mechanism. Seven-day-old rat pups were randomly divided into Sham group, HIE group, HIE + HePC group, HIE + Dantrolene (DAN) group, and HIE + Necrostatin-1 (Nec-1) group. HIE was induced by common carotid artery ligation and subsequent hypoxia exposure. The neurological function, brain injury, and molecular mechanism were evaluated by histological staining, neurobehavioral test, Western blotting, Ca2+, immunofluorescence staining, co-immunoprecipitation (Co-IP), and transmission electron microscopy (TEM). Results supported that the expression of necroptosis markers and p-RyR2 in the brain increased significantly after HIE. HePC, DAN, or Nec-1 was found to improve the neurological deficits after H/I and inhibit neuronal necroptosis. Interestingly, both HePC and DAN inhibited the increases in cytoplasmic Ca2+ and CaMK-II phosphorylation in the brain secondary to HIE, but Nec-1 failed to affect Ca2+. In conclusion, our results suggest HePC may alleviate cytoplasmic Ca2+ overload by regulating p-RyR2, which inhibits the necroptosis in the brain, exerting neuroprotective effects on HIE.
Collapse
Affiliation(s)
- Weijie Zhong
- Department of Neurosurgery, Ninth People Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Juan Cheng
- Department of Ultrasound, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaosheng Yang
- Department of Neurosurgery, Ninth People Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Wenwu Liu
- Naval Characteristic Medical Center Diving and Hyperbaric Medicine Research Laboratory, Shanghai, 200433, People's Republic of China.
| | - Yi Li
- Department of Neurosurgery, Ninth People Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
32
|
Pyk2 inhibition attenuates hypoxic-ischemic brain injury in neonatal mice. Acta Pharmacol Sin 2022; 43:797-810. [PMID: 34226665 PMCID: PMC8976000 DOI: 10.1038/s41401-021-00694-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Newborns suffering from hypoxia-ischemia (HI) brain injury still lack effective treatment. Proline-rich tyrosine kinase 2 (Pyk2) is a non-receptor tyrosine kinase, which is highly correlated with transient ischemic brain injury in adult. In this study, we investigated the role of Pyk2 in neonatal HI brain injury. HI was induced in postnatal day 7 mouse pups by unilateral common carotid artery ligation followed by hypoxic exposure. Pyk2 interference lentivirus (LV-Pyk2 shRNA) was constructed and injected into unilateral cerebral ventricle of neonatal mice before HI. Infarct volume, pathological changes, and neurological behaviors were assessed on postnatal day 8-14. We showed that the phosphorylation level of Pyk2 was significantly increased in neonatal brain after HI, whereas LV-Pyk2 shRNA injection significantly attenuated acute HI brain damage and improved neurobehavioral outcomes. In oxygen-glucose deprivation-treated cultured cortical neurons, Pyk2 inhibition significantly alleviated NMDA receptor-mediated excitotoxicity; similar results were also observed in neonatal HI brain injury. We demonstrated that Pyk2 inhibition contributes to the long-term cerebrovascular recovery assessed by laser speckle contrast imaging, but cognitive function was not obviously improved as evaluated in Morris water maze and novel object recognition tests. Thus, we constructed lentiviral LV-HIF-Pyk2 shRNA, through which HIF-1α promoter-mediated interference of Pyk2 would occur during the anoxic environment. Intracerebroventricular injection of LV-HIF-Pyk2 shRNA significantly improved long-term recovery of cognitive function in HI-treated neonatal mice. In conclusion, this study demonstrates that Pyk2 interference protects neonatal brain from hypoxic-ischemic injury. HIF-1α promoter-mediated hypoxia conditional control is a useful tool to distinguish between hypoxic period and normal period. Pyk2 is a promising drug target for potential treatment of neonatal HI brain injury.
Collapse
|
33
|
BMS-470539 Attenuates Oxidative Stress and Neuronal Apoptosis via MC1R/cAMP/PKA/Nurr1 Signaling Pathway in a Neonatal Hypoxic-Ischemic Rat Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4054938. [PMID: 35140838 PMCID: PMC8820941 DOI: 10.1155/2022/4054938] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 12/28/2021] [Indexed: 12/25/2022]
Abstract
Neuronal apoptosis induced by oxidative stress plays an important role in the pathogenesis and progression of hypoxic-ischemic encephalopathy (HIE). Previous studies reported that activation of melanocortin-1 receptor (MC1R) exerts antioxidative stress, antiapoptotic, and neuroprotective effects in various neurological diseases. However, whether MC1R activation can attenuate oxidative stress and neuronal apoptosis after hypoxic-ischemic- (HI-) induced brain injury remains unknown. Herein, we have investigated the role of MC1R activation with BMS-470539 in attenuating oxidative stress and neuronal apoptosis induced by HI and the underlying mechanisms. 159 ten-day-old unsexed Sprague-Dawley rat pups were used. HI was induced by right common carotid artery ligation followed by 2.5 h of hypoxia. The novel-selective MC1R agonist BMS-470539 was administered intranasally at 1 h after HI induction. MC1R CRISPR KO plasmid and Nurr1 CRISPR KO plasmid were administered intracerebroventricularly at 48 h before HI induction. Percent brain infarct area, short-term neurobehavioral tests, Western blot, immunofluorescence staining, Fluoro-Jade C staining, and MitoSox Staining were performed. We found that the expression of MC1R and Nurr1 increased, peaking at 48 h post-HI. MC1R and Nurr1 were expressed on neurons at 48 h post-HI. BMS-470539 administration significantly attenuated short-term neurological deficits and infarct area, accompanied by a reduction in cleaved caspase-3-positive neurons at 48 h post-HI. Moreover, BMS-470539 administration significantly upregulated the expression of MC1R, cAMP, p-PKA, Nurr1, HO-1, and Bcl-2. However, it downregulated the expression of 4-HNE and Bax, as well as reduced FJC-positive cells, MitoSox-positive cells, and 8-OHdG-positive cells at 48 h post-HI. MC1R CRISPR and Nurr1 CRISPR abolished the antioxidative stress, antiapoptotic, and neuroprotective effects of BMS-470539. In conclusion, our findings demonstrated that BMS-470539 administration attenuated oxidative stress and neuronal apoptosis and improved neurological deficits in a neonatal HI rat model, partially via the MC1R/cAMP/PKA/Nurr1 signaling pathway. Early administration of BMS-470539 may be a novel therapeutic strategy for infants with HIE.
Collapse
|
34
|
Li P, Lu X, Hu J, Dai M, Yan J, Tan H, Yu P, Chen X, Zhang C. Human amniotic fluid derived-exosomes alleviate hypoxic encephalopathy by enhancing angiogenesis in neonatal mice after hypoxia. Neurosci Lett 2022; 768:136361. [PMID: 34826550 DOI: 10.1016/j.neulet.2021.136361] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/11/2021] [Accepted: 11/19/2021] [Indexed: 12/17/2022]
Abstract
Neonatal hypoxic encephalopathy is a type of central nervous system dysfunction manifested by high mortality and morbidity. Exosomes play a crucial role in neuroprotection by enhancing angiogenesis. The objective of this study was to investigate the effect of human amniotic fluid-derived exosomes (hAFEXOs) on functional recovery in neonatal hypoxic encephalopathy. The transwell assay, scratch wound healing assay, and tube formation assay were used to evaluate the effect of hAFEXOs on the angiogenesis of human umbilical vein endothelial cells (HUVECs) after oxygen and glucose deprivation (OGD). The angiogenesis of microvascular endothelial cells (MECs) in the cortex was tested in neonatal mice treated with hAFEXOs or phosphate-buffered saline (PBS) after hypoxia. Expressions of hypoxia-inducible factor 1 α (HIF-1α) and vascular endothelial growth factor (VEGF) in the cerebral cortex were also tested by western blot. The Morris Water Maze Test (MWM) was carried out to detect the performance of spatial memory after processing with hAFEXOs or PBS. The results indicated that hAFEXOs favored tubing formation and migration of HUVECs after in vitro OGD. The hAFEXOs also favored the expression of CD31 in neonatal mice following hypoxia. The expressions of both HIF-1α and VEGF were significantly augmented in the cerebral cortex of neonatal mice which were treated with hAFEXOs. Moreover, the MWM test results showed that the performance of the spatial memory was better in the hAFEXO-treated group than in the PBS-treated group. Our study indicates that hAFEXOs alleviated hypoxic encephalopathy and enhanced angiogenesis in neonatal mice after hypoxia. In addition, hAFEXOs promoted migration and tube formation of HUVECs after OGD in vitro. These findings confirm that hAFEXOs show great potential for further studies aimed at developing therapeutic agents for hypoxic encephalopathy.
Collapse
Affiliation(s)
- Ping Li
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha 410008, China; Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha 410008, China
| | - Xiaoxu Lu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiajia Hu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Minhui Dai
- Department of Clinical Dietitian, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jianqin Yan
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Huiling Tan
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Anesthesiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Peilin Yu
- School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xuliang Chen
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Chengliang Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
35
|
Khan MH, Ann QU, Khan MS, Ahmad N, Ahmed M. Efficacy of Magnesium Sulfate in Addition to Melatonin Therapy in Neonates With Hypoxic-Ischemic Encephalopathy. Cureus 2022; 14:e21163. [PMID: 35165613 PMCID: PMC8833294 DOI: 10.7759/cureus.21163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2022] [Indexed: 11/17/2022] Open
Abstract
Background: One of the most important causes of neonatal deaths in developing nations is birth asphyxia. Though the probability of a complete recovery is very low, hypoxic-ischemic encephalopathy (HIE) associated with asphyxia can be managed with multiple treatment options. The study evaluated the efficacy of the addition of magnesium sulfate (MgSO4) to melatonin therapy in neonates with HIE. Methodology: A prospective, observational study was conducted in the department of neonatal intensive care, Pakistan Institute of Medical Sciences Hospital, Islamabad, Pakistan from October 2020 to March 2021. All neonates with an Appearance, Pulse, Grimace, Activity, and Respiration (APGAR) score of less than five at five minutes, umbilical blood pH of less than 7.0, and moderate neonatal encephalopathy as detected on the modified Sarnat score which is a clinical tool used for the assessment of the severity of HIE were included in the study. Neonates with congenital abnormalities, intrauterine growth retardation, neonatal sepsis, and infants born to mothers with diabetes mellitus type 2 were excluded from the study. The infants were randomly assigned to either of the groups, i.e., i) group 1 included neonates who were administered at least three doses of magnesium sulfate (MgSO4) infusion in addition to melatonin, or ii) group 2 included neonates who were administered melatonin only. Blood samples of all neonates were evaluated and compared between the two groups. Results: A total of 90 neonates with HIE were included in the study. There was a predominance of female neonates. The mean ages of babies in group 1 and group 2 were 37.2 ± 0.43 and 37.3 ± 0.59 weeks, respectively. The mean weight on the term was 2.88 ± 0.11 and 2.89 ± 0.10, respectively. The Apgar score at 5 mins in group 1 was 1.73 ± 0.81 while in group 2, 1.82 ± 0.94. It was found that there was a more significant improvement in pH after 3 days and after one week of treatment in group 1 as compared to group 2. The mean pH in babies after three days of intervention was 7.23 ± 0.03 in group 1 which was significantly better than group 2 (p<0.0001). After seven days, the mean normalized to 7.39 ± 0.04 in group 1 (p < 0.0001). It was found that in patients in group 1, the mortality was lower than in group 2 (p < 0.0001). Conclusion: HIE patients who were administered melatonin in combination with magnesium sulfate yielded better patient outcomes. Thus, it was concluded that the use of magnesium sulfate as dual therapy with melatonin improved patient outcomes for HIE. However, it is recommended that similar studies are conducted with a wider range of parameters, such as duration of hospital stay and assessment of the neurological outcomes of the patients.
Collapse
|
36
|
Wang R, Li L, Wang B. Poncirin ameliorates oxygen glucose deprivation/reperfusion injury in cortical neurons via inhibiting NOX4-mediated NLRP3 inflammasome activation. Int Immunopharmacol 2022; 102:107210. [PMID: 34266770 DOI: 10.1016/j.intimp.2020.107210] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/01/2020] [Accepted: 11/11/2020] [Indexed: 12/25/2022]
Abstract
Poncirin, a natural flavonoid present abundantly in citrus fruits, possesses anti-oxidant and anti-inflammatory activities that contribute to neuroprotection, but its roles and mechanisms in neuronal injury is still poorly understood. In this study, an oxygen-glucose deprivation/reoxygenation (OGD/R) model was established in primary cortical neurons to induce neuronal injury in vitro. Poncirin effectively attenuated OGD/R-induced neuronal damage by enhancing cell viability, restraining lactate dehydrogenase release, and reducing apoptosis of neurons. Poncirin restrained mitochondrial dysfunction and oxidative stress by increasing mitochondrial membrane potential, declining reactive oxygen species production, lessening malondialdehyde generation, and increasing the activities of antioxidant enzymes in OGD/R-treated neurons. Poncirin also repressed inflammatory responses by reducing the secretion of pro-inflammatory factors, and inhibiting NLRP3 inflammasome activation. Importantly, poncirin administration notably abolished OGD/R-induced upregulation of NADPH oxidase 4 (NOX4), and overexpression of NOX4 neutralized poncirin-mediated neuroprotection. In conclusion, poncirin protects cortical neurons from OGD/R injury via inhibiting NOX4/ROS/NLRP3 axis.
Collapse
Affiliation(s)
- Ruili Wang
- Department of Pediatrics, Zhoukou Central Hospital, Zhoukou 466000, Henan, China.
| | - Lei Li
- Department of Pediatrics, Zhoukou Central Hospital, Zhoukou 466000, Henan, China
| | - Baogong Wang
- Department of Cardiology, Zhoukou Central Hospital, Zhoukou 466000, Henan, China
| |
Collapse
|
37
|
Xing Z, Zhen T, Jie F, Jie Y, Shiqi L, Kaiyi Z, Zhicui O, Mingyan H. Early Toll-like receptor 4 inhibition improves immune dysfunction in the hippocampus after hypoxic-ischemic brain damage. Int J Med Sci 2022; 19:142-151. [PMID: 34975308 PMCID: PMC8692118 DOI: 10.7150/ijms.66494] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/21/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Toll-like receptor 4 (TLR4) is implicated in neonatal hypoxic-ischemic brain damage (HIBD), but the underlying mechanism is unclear. Hypothesis: We hypothesized that TLR4 mediates brain damage after hypoxic ischemia (HI) by inducing abnormal neuroimmune responses, including activation of immune cells and expression disorder of immune factors, while early inhibition of TLR4 can alleviate the neuroimmune dysfunction. Method: Postnatal day 7 rats were randomized into control, HI, and HI+TAK-242 (TAK-242) groups. The HIBD model was developed using the Rice-Vannucci method (the left side was the ipsilateral side of HI). TAK-242 (0.5 mg/kg) was given to rat pups in the TAK-242 group at 30 min before modeling. Immunofluorescence, immunohistochemistry, and western blotting were used to determine the TLR4 expression; the number of Iba-1+, GFAP+, CD161+, MPO+, and CD3+ cells; ICAM-1 and C3a expression; and interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and IL-10 expression in the hippocampal CA1 region. Result: Significantly increased TLR4 expression was observed in the left hippocampus, and was alleviated by TAK-242. The significant increases in Iba-1+, MPO+, and CD161+ cells at 24 h and 7 days after HI and in GFAP+ and CD3+ T cells at 7 days after HI were also counteracted by TAK-242, but no significant differences were observed among groups at 24 h after HI. ICAM-1 expression increased 24 h after HI, while C3a expression decreased; TAK-242 also alleviated these changes. TNF-α and IL-1β expression increased, while IL-10 expression decreased at 24 h and 7 days after HI; TAK-242 counteracted the increased TNF-α and IL-1β expression at 24 h and the changes in IL-1β and IL-10 at 7 days, but induced no significant differences in IL-10 expression at 24 h and TNF-α expression at 7 days. Conclusion: Early TLR4 inhibition can alleviate hippocampal immune dysfunction after neonatal HIBD.
Collapse
Affiliation(s)
- Zhu Xing
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - Tang Zhen
- Department of Neonatology, Affiliated Hospital of Guilin Medical College, Guilin, Guangxi, 541001 China.,Department of Pediatrics, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013 China
| | - Fan Jie
- Department of Neonatology, East Hospital of Shaoyang Central Hospital, Shaoyang, Hunan, 422000 China
| | - Yu Jie
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - Liu Shiqi
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - Zhu Kaiyi
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - OuYang Zhicui
- Department of Neonatology, Affiliated Hospital of Guilin Medical College, Guilin, Guangxi, 541001 China
| | - Hei Mingyan
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| |
Collapse
|
38
|
Lin L, Liu W, Mu J, Zhan E, Wei H, Hong S, Hua Z. Effect of neonatal neuronal intensive care unit on neonatal encephalopathy. PLoS One 2021; 16:e0261837. [PMID: 34972144 PMCID: PMC8719725 DOI: 10.1371/journal.pone.0261837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/10/2021] [Indexed: 11/19/2022] Open
Abstract
Prophylaxis of brain injury in newborns has been a main concern since the first neonatal neuronal intensive care unit (NNICU) was established in the world in 2008. The aim of this study was to outline and evaluate the unit's development by analyzing the demographics of the patients, the services delivered, the short-term outcomes before and after the establishment of NNICU. During the two investigation periods, 384 newborns were diagnosed or suspected as "neonatal encephalopathy", among which 185 patients admitted to NNICU between 2011.03.01 and 2012.09.30 before the establishment of NNICU were enrolled in the pre-NNICU group, another 199 neonates hospitalized during 2018.03.01 to 2019.09.30 were included in the post-NNICU group. Patients in the post-NNICU group were more likely to have seizures (P = 0.001), incomplete or absent primitive reflexes (P = 0.002), therapeutic hypothermia (P<0.001) and liquid control (P<0.001) in acute phase. Meanwhile, amplitude-integrated electro encephalogram (aEEG) monitoring (P<0.001) and cranial ultrasound (P<0.001) were more often used in NNICU. Both of the follow-up rate in brain MRI and the assessment of neurodevelopment at 3 months were higher in the post-NNICU group (P<0.001). In conclusion, the NNICU focused on the neonatal neurocritical care for the babies susceptible to NE with the guidance of evidence-based medicine, the establishment of NNICU is gradually improving and standardizing the neuroprotective therapy and clinical follow-up to improve neurodevelopmental prognosis of the NE patients in CHCMU.
Collapse
Affiliation(s)
- Lu Lin
- Department of Neonatology, 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 Key Laboratory of Child Health and Nutrition, Children’s Hospital of Chongqing Medical University, Chongqing, P.R China
| | - Weiqin Liu
- Department of Neonatology, 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 Key Laboratory of Child Health and Nutrition, Children’s Hospital of Chongqing Medical University, Chongqing, P.R China
| | - Jing Mu
- Department of Neonatology, 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 Key Laboratory of Child Health and Nutrition, Children’s Hospital of Chongqing Medical University, Chongqing, P.R China
| | - Enmei Zhan
- Department of Neonatology, 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 Key Laboratory of Child Health and Nutrition, Children’s Hospital of Chongqing Medical University, Chongqing, P.R China
| | - Hong Wei
- Department of Neonatology, 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 Key Laboratory of Child Health and Nutrition, Children’s Hospital of Chongqing Medical University, Chongqing, P.R China
| | - Siqi Hong
- 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 Key Laboratory of Child Health and Nutrition, Children’s Hospital of Chongqing Medical University, Chongqing, P.R China
- Department of Neurology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Ziyu Hua
- Department of Neonatology, 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 Key Laboratory of Child Health and Nutrition, Children’s Hospital of Chongqing Medical University, Chongqing, P.R China
| |
Collapse
|
39
|
Wyckoff MH, Sawyer T, Lakshminrusimha S, Collins A, Ohls RK, Leone TA. Resuscitation 2020: Proceedings From the NeoHeart 2020 International Conference. World J Pediatr Congenit Heart Surg 2021; 13:77-88. [PMID: 34919486 DOI: 10.1177/21501351211038835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Resuscitation guidelines are developed and revised by medical societies throughout the world. These guidelines are increasingly based on evidence from preclinical and clinical research. The International Liaison Committee on Resuscitation reviews evidence for each resuscitation practice and provides summary consensus statements that inform resuscitation guideline committees. A similar process is used for different populations including neonatal, pediatric, and adult resuscitation. The NeoHeart 2020 Conference brought together experts in resuscitation to discuss recent evidence and guidelines for resuscitation practices. This review summarizes the main focus of discussion from this symposium.
Collapse
Affiliation(s)
| | - Taylor Sawyer
- 12353University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA, USA
| | | | - Amélie Collins
- 12294Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Robin K Ohls
- 266111University of Utah, Salt Lake City, UT, USA
| | - Tina A Leone
- 12294Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
40
|
Yin C, Ji Y, Ma N, Chen K, Zhang W, Bai D, Jia X, Xia S, Yin H. RNA-seq analysis reveals potential molecular mechanisms of ZNF580/ZFP580 promoting neuronal survival and inhibiting apoptosis after Hypoxic-ischemic brain damage. Neuroscience 2021; 483:52-65. [PMID: 34929337 DOI: 10.1016/j.neuroscience.2021.12.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 10/19/2022]
Abstract
Neonatal hypoxic-ischemic brain damage (HIBD) is one of the main causes of neonatal acute death and chronic nervous system impairment, but still lacks effective treatments. ZNF580/ZFP580, reported in our previous studies, may be a newly identified member of the Krüppel-like factor (KLF) family, and has anti-apoptotic effects during ischemic myocardial injury. In the present study, we showed that the expression levels of both ZFP580/ZNF580 mRNA and protein increased significantly in neonatal HIBD rats and oxygen-glucose deprivation (OGD) SH-SY5Y cell models. ZNF580 overexpression promoted neuron survival and suppressed neuron apoptosis after OGD in neuron-like SH-SY5Y cells, while interference with ZNF580 resulted in the opposite results. RNA-seq analysis identified 248 differentially-expressed genes (DEGs) between ZNF580 overexpression SH-SY5Y cells and interference-expressed SH-SY5Y cells. Gene Ontology functional enrichment analysis showed that these DEGs played significant roles in the growth, development, and regeneration of axons, DNA biosynthetic processes, DNA replication, and apoptosis. Kyoto Encyclopedia of Genes and Genomes enrichment analysis indicated that these DEGs were found in some pathways, including ferroptosis, glutamatergic synapses, protein processing in the endoplasmic reticulum, estrogen signaling pathways, the TGF-beta signaling pathway, and the longevity regulating pathway. The qRT-PCR validation results were consistent with RNA-seq results, which showed that HSPA5, IGFBP3, NTN4, and KLF9 increased in ZNF580-overexpressed SH-SY5Y cells and decreased in interference-expressed SH-SY5Y cells, when compared with normal cells. Together, the results suggested that ZNF580 targeted these genes to inhibit neuronal apoptosis.
Collapse
Affiliation(s)
- Chongjuan Yin
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yansu Ji
- Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, Hebei, China
| | - Ning Ma
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Kai Chen
- Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, Hebei, China
| | - Wencheng Zhang
- Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, Hebei, China
| | - Dan Bai
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaojun Jia
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shihai Xia
- Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, Hebei, China.
| | - Huaiqing Yin
- First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
41
|
MicroRNA-21-5p Reduces Hypoxia/Reoxygenation-Induced Neuronal Cell Damage through Negative Regulation of CPEB3. Anal Cell Pathol (Amst) 2021; 2021:5543212. [PMID: 34900520 PMCID: PMC8660214 DOI: 10.1155/2021/5543212] [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: 01/13/2021] [Revised: 09/04/2021] [Accepted: 11/07/2021] [Indexed: 11/26/2022] Open
Abstract
Objectives To explore the role of microRNA-21-5p (miR-21-5p) in hypoxia/reoxygenation- (H/R-) induced HT22 cell damage. Methods The hypoxia/reoxygenation (H/R) model was established in mouse neuronal cells HT22. Cell Counting Kit-8 (CCK-8) and qRT-PCR were used to determine the effects of H/R treatment on cell viability and miR-21-5p expression. HT22 cells were transfected with miR-21-5p mimic or negative control (NC) followed by the induction of H/R; cell viability, apoptosis, and SOD, MDA, and LDH activities were detected. Besides, the apoptosis-related proteins including BAX, BCL2, cleaved caspase-3, and caspase-3 as well as proteins of EGFR/PI3K/AKT signaling pathways were measured by Western blot. To verify the target relation between cytoplasmic polyadenylation element binding protein 3 (CPEB3) and miR-21-5p, luciferase reporter gene experiment was performed. After cotransfection with miR-21-5p mimic and CPEB3 plasmids, the reversal effects of CPEB3 on miR-21-5p in H/R damage were studied. Results H/R treatment could significantly reduce the cell viability (P < 0.05) and miR-21-5p levels (P < 0.05) in HT22 cells. After overexpressing miR-21-5p, cell viability was increased (P < 0.05) under H/R treatment, and the apoptosis rate and the levels of apoptosis-related proteins were suppressed (all P < 0.05). Furthermore, SOD activity was increased (P < 0.05), while MDA and LDH activity was decreased (both P < 0.05). Besides, miR-21-5p could restore the activation of the EGFR/PI3K/AKT signaling pathway inhibited by H/R treatment (all P < 0.05). The luciferase reporter gene experiment verified that CPEB3 is the target of miR-21-5p (P < 0.05). When coexpressing miR-21-5p mimic and CPEB3 in the cells, the protective effects of miR-21-5p under H/R were reversed (all P < 0.05), and the activation of the EGFR/PI3K/AKT pathway was also inhibited (all P < 0.05). Conclusion This study showed that miR-21-5p may regulate the EGFR/PI3K/AKT signaling pathway by targeting CPEB3 to reduce H/R-induced cell damage and apoptosis.
Collapse
|
42
|
Abdel-Aziz SM, Rahman MSMA, Shoreit AH, Din MEE, Hamed EA, Gad EF. Outcome of Infants with Hypoxic-Ischemic Encephalopathy Treated by Whole Body Cooling and Magnesium Sulfate. JOURNAL OF CHILD SCIENCE 2021. [DOI: 10.1055/s-0041-1736562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
AbstractTherapeutic hypothermia (TH) either by selective head cooling or whole-body cooling decreases brain damage and provide neuroprotection and reduced mortality rate in cases of moderate-to-severe hypoxia-ischemia encephalopathy (HIE) of newborns, especially if started at first 6 hours after birth. Also, management with adjuvant therapies like magnesium sulfate (MS) provides more neuroprotection. The interventional randomized controlled research aimed to assess short-term actions of TH as sole therapy and in combination with MS as a neuroprotective agent for the treatment of HIE newborn infants. A total of 36 full-terms and near-term infants delivered at Assiut University Children's Hospital and fulfilled HIE criteria were enrolled. They were divided equally into three groups; Group 1 (n = 12) received whole body cooling during first 6 hours of life as a sole therapy; Group 2 (n = 12) received whole body cooling in addition to MS as adjuvant therapy; Group 3 (n = 12) received supportive intensive care measures as a control. TH plus MS group (group 2) had a significantly good short-term outcomes as short period of respiratory support and mechanical ventilation (p-value =0.001), less in incidence of convulsion (p-value = 0.001) and early in feeding initiation (p-value = 0.009), compared with other groups managed by TH (group 1) or by supportive treatment (group 3). In conclusion, whole body cooling in addition to MS as adjunctive therapy for the treatment of HIE neonates is safe therapy that improves short-term outcome both clinically and radiologically.
Collapse
Affiliation(s)
- Safwat M. Abdel-Aziz
- Department of Pediatrics and Neonatology, Assiut University Children's Hospital, Assiut, Egypt
| | | | - Asmaa H. Shoreit
- Department of Pediatrics and Neonatology, Assiut University Children's Hospital, Assiut, Egypt
| | - Moustafa Ez El Din
- Department of Radiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Enas A. Hamed
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Eman Fathalla Gad
- Department of Pediatrics and Neonatology, Assiut University Children's Hospital, Assiut, Egypt
| |
Collapse
|
43
|
Noh J, Jeong J, Park S, Jin Jung K, Lee B, Kim W, Han J, Cho M, Sung DK, Ahn SY, Chang YS, Son H, Jeong EJ. Preclinical assessment of thrombin-preconditioned human Wharton's jelly-derived mesenchymal stem cells for neonatal hypoxic-ischaemic brain injury. J Cell Mol Med 2021; 25:10430-10440. [PMID: 34651412 PMCID: PMC8581315 DOI: 10.1111/jcmm.16971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/23/2021] [Accepted: 09/09/2021] [Indexed: 01/17/2023] Open
Abstract
Hypoxic-ischaemic encephalopathy (HIE) is a type of brain injury affecting approximately 1 million newborn babies per year worldwide, the only treatment for which is therapeutic hypothermia. Thrombin-preconditioned mesenchymal stem cells (MSCs) exert neuroprotective effects by enriching cargo contents and boosting exosome biogenesis, thus showing promise as a new therapeutic strategy for HIE. This study was conducted to evaluate the tissue distribution and potential toxicity of thrombin-preconditioned human Wharton's jelly-derived mesenchymal stem cells (th-hWJMSCs) in animal models before the initiation of clinical trials. We investigated the biodistribution, tumorigenicity and general toxicity of th-hWJMSCs. MSCs were administered the maximum feasible dose (1 × 105 cells/10 µL/head) once, or at lower doses into the cerebral ventricle. To support the clinical use of th-hWJMSCs for treating brain injury, preclinical safety studies were conducted in newborn Sprague-Dawley rats and BALB/c nude mice. In addition, growth parameters were evaluated to assess the impact of th-hWJMSCs on the growth of newborn babies. Our results suggest that th-hWJMSCs are non-toxic and non-tumorigenic in rodent models, survive for up to 7 days in the brain and hold potential for HIE therapy.
Collapse
Affiliation(s)
- Jung‐Ho Noh
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
- College of Veterinary MedicineChungnam National UniversityDaejeonRepublic of Korea
| | - Ji‐Seong Jeong
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Sang‐Jin Park
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Kyung Jin Jung
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Byoung‐Seok Lee
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Woo‐Jin Kim
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Ji‐Seok Han
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Min‐Kyung Cho
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Dong Kyung Sung
- Stem Cell and Regenerative Medicine InstituteSamsung Medical CenterSamsung Biomedical Research InstituteSeoulRepublic of Korea
| | - So Yoon Ahn
- Stem Cell and Regenerative Medicine InstituteSamsung Medical CenterSamsung Biomedical Research InstituteSeoulRepublic of Korea
| | - Yun Sil Chang
- Stem Cell and Regenerative Medicine InstituteSamsung Medical CenterSamsung Biomedical Research InstituteSeoulRepublic of Korea
- Department of PediatricsSamsung Medical CenterSungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Hwa‐Young Son
- College of Veterinary MedicineChungnam National UniversityDaejeonRepublic of Korea
| | - Eun Ju Jeong
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| |
Collapse
|
44
|
Shan W, Wu Y, Han X, Chen Q, Wu J. The mechanism of sevoflurane post-treatment alleviating hypoxic-ischemic encephalopathy by affecting histone methyltransferase G9a in rats. Bioengineered 2021; 12:9790-9805. [PMID: 34672892 PMCID: PMC8810117 DOI: 10.1080/21655979.2021.1995105] [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: 12/30/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is recognized as the main cause of neonatal death, and efficient treatment strategies remain limited. This study aims to investigate the mechanism of sevoflurane (SF) post-treatment in alleviating HIE in rats. The HIE rat model and oxygen-glucose deprivation (OGD) cell model were established, and adeno-associated virus (AAV)-histone-lysine N-methyltransferase EHMT2 (G9a) was transfected after SF treatment. The learning and memory ability and the levels of nerve growth factor (NGF)/brain-derived neurotrophic factor (BDNF) were evaluated and determined. The levels of G9a/histone H3 lysine 9 dimethylation (H3K9me2) and the enrichment level of H3K9me2 in the promoter region of BDNF gene were analyzed. After SF post-treatment, the neurons in cerebral cortex, the learning and memory skills and the contents of NGF/BDNF were increased, while the apoptosis and G9a/H3K9me2 levels were reduced. After overexpression of G9a in vitro/vivo, the enrichment levels of H3K9me2 in the promoter region of BDNF were increased, the levels of BDNF were decreased, the neurons were damaged and the learning and memory abilities of HIE rats were impaired. The conclusion is that SF post-treatment can promote the expression of BDNF by inhibiting H3K9me2 on the BDNF gene promoter and inhibiting G9a, thus alleviating HIE in rats.
Collapse
Affiliation(s)
- Weifeng Shan
- Department of Anesthesiology, Lishui City People's Hospital, Lishui, China
| | - Yini Wu
- Department of Anesthesiology, Lishui City People's Hospital, Lishui, China
| | - Xin Han
- Department of Anesthesiology, Lishui City People's Hospital, Lishui, China
| | - Qin Chen
- Department of Anesthesiology, Lishui City People's Hospital, Lishui, China
| | - Jimin Wu
- Department of Anesthesiology, Lishui City People's Hospital, Lishui, China
| |
Collapse
|
45
|
Metallinou D, Lazarou E, Lykeridou A. Pharmacological and Non-Pharmacological Brain-Focused Clinical Practices for Premature Neonates at High Risk of Neuronal Injury. MÆDICA 2021; 16:281-290. [PMID: 34621352 DOI: 10.26574/maedica.2020.16.2.281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective:Disruption of smooth intrauterine brain development is a significant consequence of premature birth that may lead to adverse neurological outcomes. Although noteworthy progress has been made in the management of prematurity, the rates of neonatal morbidity and neurodevelopmental disorders remain high, underlining the need to find clinical practices that particularly protect the central nervous system. Aim:To identify recent articles regarding pharmacological and non-pharmacological brain-focused clinical practices (BFCP) for premature neonates at high risk of neuronal injury. Material and methods:We did an extensive search of PubMed and Google Scholar for relevant research published between 2000 and 2020. Results:Nineteen full-length original research papers fulfilled the inclusion criteria and were selected for the purpose of the present review. Non-pharmacological BFCP intend to improve the neonate's experience in the NICU environment and can be applied by a multidisciplinary team, while pharmacological ones are related to novel molecules that aim to quell apoptosis and inflammation or promote neurogenesis. Conclusion:In the future, a combination of pharmacological and non-pharmacological BFCP might be considered as the most promising protection and/or treatment provided in clinical practice to premature neonates at high risk of neuronal injury.
Collapse
Affiliation(s)
- Dimitra Metallinou
- Department of Midwifery, Faculty of Health and Caring Sciences, University of West Attica, Greece
| | | | - Aikaterini Lykeridou
- Department of Midwifery, Faculty of Health and Caring Sciences, University of West Attica, Greece
| |
Collapse
|
46
|
Cortical Visual Impairment in Childhood: 'Blindsight' and the Sprague Effect Revisited. Brain Sci 2021; 11:brainsci11101279. [PMID: 34679344 PMCID: PMC8533908 DOI: 10.3390/brainsci11101279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/14/2021] [Accepted: 09/24/2021] [Indexed: 11/29/2022] Open
Abstract
The paper discusses and provides support for diverse processes of brain plasticity in visual function after damage in infancy and childhood in comparison with injury that occurs in the adult brain. We provide support and description of neuroplastic mechanisms in childhood that do not seemingly exist in the same way in the adult brain. Examples include the ability to foster the development of thalamocortical connectivities that can circumvent the lesion and reach their cortical destination in the occipital cortex as the developing brain is more efficient in building new connections. Supporting this claim is the fact that in those with central visual field defects we can note that the extrastriatal visual connectivities are greater when a lesion occurs earlier in life as opposed to in the neurologically mature adult. The result is a significantly more optimized system of visual and spatial exploration within the ‘blind’ field of view. The discussion is provided within the context of “blindsight” and the “Sprague Effect”.
Collapse
|
47
|
In vivo MRI evaluation of early postnatal development in normal and impaired rat eyes. Sci Rep 2021; 11:15513. [PMID: 34330952 PMCID: PMC8324881 DOI: 10.1038/s41598-021-93991-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 07/01/2021] [Indexed: 11/08/2022] Open
Abstract
This study employed in vivo 7-T magnetic resonance imaging (MRI) to evaluate the postnatal ocular growth patterns under normal development or neonatal impairments in Sprague-Dawley rats. Using T2-weighted imaging on healthy rats from postnatal day (P) 1 (newborn) to P60 (adult), the volumes of the anterior chamber and posterior chamber (ACPC), lens, and vitreous humor increased logistically with ACPC expanding by 33-fold and the others by fivefold. Intravitreal potassium dichromate injection at P1, P7, and P14 led to T1-weighted signal enhancement in the developing retina by 188-289%. Upon unilateral hypoxic-ischemic encephalopathy at P7, monocular deprivation at P15, and monocular enucleation at P1, T2-weighted imaging of the adult rats showed decreased ocular volumes to different extents. In summary, in vivo high-field MRI allows for non-invasive evaluation of early postnatal development in the normal and impaired rat eyes. Chromium-enhanced MRI appeared effective in examining the developing retina before natural eyelid opening at P14 with relevance to lipid metabolism. The reduced ocular volumes upon neonatal visual impairments provided evidence to the emerging problems of why some impaired visual outcomes cannot be solely predicted by neurological assessments and suggested the need to look into both the eye and the brain under such conditions.
Collapse
|
48
|
[Effect of astragaloside IV on the expression of NOD-like receptor protein 3 inflammasome in neonatal rats with hypoxic-ischemic brain damage]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23. [PMID: 33840414 PMCID: PMC8050542 DOI: 10.7499/j.issn.1008-8830.2010053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To study the effect of astragaloside IV (AS-IV) on NOD-like receptor protein 3 (NLRP3) inflammasome in neonatal rats with hypoxic-ischemic brain damage (HIBD). METHODS A total of 24 Sprague-Dawley rats, aged 7 days, were randomly divided into a sham-operation group, an HIBD group, and an AS-IV treatment group, with 8 rats in each group. After 24 hours of modeling, brain tissue was collected for hematoxylin-eosin staining, yo-PRO-1 staining, and EthD-2 immunofluorescent staining in order to observe the cerebral protection effect of AS-IV in vivo. HT22 cells were used to prepare a model of oxygen-glycogen deprivation (OGD), and a concentration gradient (50-400 μmol/L) was established for AS-IV. CCK-8 assay was used to measure the viability of HT22 cells. RT-PCR and Western blot were used to observe the effect of different concentrations of AS-IV on the mRNA and protein expression of NLRP3, gasdermin D (GSDMD), caspase-1, and interleukin-1β (IL-1β). RESULTS Yo-Pro-1 and EthD-2 staining showed that compared with the sham-operation group, the HIBD group had an increase in pyroptotic cells with a small number of necrotic cells, and the AS-IV group had reductions in both pyroptotic and necrotic cells. Compared with the sham-operation group, the HIBD group had significantly higher protein expression levels of NLRP3, IL-1β, caspase-1, and GSDMD (P < 0.05). Compared with the HIBD group, the AS-IV group had significant reductions in the protein expression levels of NLRP3, caspase-1, and GSDMD (P < 0.05). HT22 cell experiment showed that compared with the OGD group, the AS-IV group had inhibited mRNA and protein expression of NLRP3, GSDMD, caspase-1, and IL-1β, with the best therapeutic effect at the concentration of 200 μmol/L (P < 0.05). CONCLUSIONS AS-IV may alleviate HIBD in neonatal rats by inhibiting the expression of NLRP3, GSDMD, caspase-1, and IL-1β.
Collapse
|
49
|
Kimura T, Toriuchi K, Kakita H, Tamura T, Takeshita S, Yamada Y, Aoyama M. Hypothermia Attenuates Neuronal Damage via Inhibition of Microglial Activation, Including Suppression of Microglial Cytokine Production and Phagocytosis. Cell Mol Neurobiol 2021; 41:459-468. [PMID: 32382852 DOI: 10.1007/s10571-020-00860-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
Although therapeutic hypothermia (TH) provides neuroprotection, the cellular mechanism underlying the neuroprotective effect of TH has not yet been fully elucidated. In the present study, we investigated the effect of TH on microglial activation to determine whether hypothermia attenuates neuronal damage via microglial activation. After lipopolysaccharide (LPS) stimulation, BV-2 microglia cells were cultured under normothermic (37 °C) or hypothermic (33.5 °C) conditions. Under hypothermic conditions, expression of pro-inflammatory cytokines and inducible nitric oxide synthase (iNOS) was suppressed. In addition, phagocytosis of latex beads was significantly suppressed in BV-2 cells under hypothermic conditions. Moreover, nuclear factor-κB signaling was inhibited under hypothermic conditions. Finally, neuronal damage was attenuated following LPS stimulation in neurons co-cultured with BV-2 cells under hypothermic conditions. In conclusion, hypothermia attenuates neuronal damage via inhibition of microglial activation, including microglial iNOS and pro-inflammatory cytokine expression and phagocytic activity. Investigating the mechanism of microglial activation regulation under hypothermic conditions could contribute to the development of novel neuroprotective therapies.
Collapse
Affiliation(s)
- Tomoka Kimura
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Mizohoku Tanabedori, Nagoya, Aichi, 467-8603, Japan
| | - Kohki Toriuchi
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Mizohoku Tanabedori, Nagoya, Aichi, 467-8603, Japan
| | - Hiroki Kakita
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Mizohoku Tanabedori, Nagoya, Aichi, 467-8603, Japan
- Department of Perinatal and Neonatal Medicine, Aichi Medical University, 1-1 Yazako, Karimata, Nagakute, Aichi, 480-1195, Japan
| | - Tetsuya Tamura
- Department of Anesthesiology and Intensive Care Medicine, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Aichi, 467-0001, Japan
| | - Satoru Takeshita
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Mizohoku Tanabedori, Nagoya, Aichi, 467-8603, Japan
- Department of Perinatal and Neonatal Medicine, Aichi Medical University, 1-1 Yazako, Karimata, Nagakute, Aichi, 480-1195, Japan
| | - Yasumasa Yamada
- Department of Perinatal and Neonatal Medicine, Aichi Medical University, 1-1 Yazako, Karimata, Nagakute, Aichi, 480-1195, Japan
| | - Mineyoshi Aoyama
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Mizohoku Tanabedori, Nagoya, Aichi, 467-8603, Japan.
| |
Collapse
|
50
|
Dumbuya JS, Chen L, Wu JY, Wang B. The role of G-CSF neuroprotective effects in neonatal hypoxic-ischemic encephalopathy (HIE): current status. J Neuroinflammation 2021; 18:55. [PMID: 33612099 PMCID: PMC7897393 DOI: 10.1186/s12974-021-02084-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/14/2021] [Indexed: 12/23/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is an important cause of permanent damage to central nervous system (CNS) that may result in neonatal death or manifest later as mental retardation, epilepsy, cerebral palsy, or developmental delay. The primary cause of this condition is systemic hypoxemia and/or reduced cerebral blood flow with long-lasting neurological disabilities and neurodevelopmental impairment in neonates. About 20 to 25% of infants with HIE die in the neonatal period, and 25-30% of survivors are left with permanent neurodevelopmental abnormalities. The mechanisms of hypoxia-ischemia (HI) include activation and/or stimulation of myriad of cascades such as increased excitotoxicity, oxidative stress, N-methyl-D-aspartic acid (NMDA) receptor hyperexcitability, mitochondrial collapse, inflammation, cell swelling, impaired maturation, and loss of trophic support. Different therapeutic modalities have been implicated in managing neonatal HIE, though translation of most of these regimens into clinical practices is still limited. Therapeutic hypothermia, for instance, is the most widely used standard treatment in neonates with HIE as studies have shown that it can inhibit many steps in the excito-oxidative cascade including secondary energy failure, increases in brain lactic acid, glutamate, and nitric oxide concentration. Granulocyte-colony stimulating factor (G-CSF) is a glycoprotein that has been implicated in stimulation of cell survival, proliferation, and function of neutrophil precursors and mature neutrophils. Extensive studies both in vivo and ex vivo have shown the neuroprotective effect of G-CSF in neurodegenerative diseases and neonatal brain damage via inhibition of apoptosis and inflammation. Yet, there are still few experimentation models of neonatal HIE and G-CSF's effectiveness, and extrapolation of adult stroke models is challenging because of the evolving brain. Here, we review current studies and/or researches of G-CSF's crucial role in regulating these cytokines and apoptotic mediators triggered following neonatal brain injury, as well as driving neurogenesis and angiogenesis post-HI insults.
Collapse
Affiliation(s)
- John Sieh Dumbuya
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Lu Chen
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Jang-Yen Wu
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Bin Wang
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China.
| |
Collapse
|