1
|
Wang L, Gu Y, Shen C. Transcriptome analysis and lncRNA expression profile in brain tissues of neonatal hypoxic-ischemic brain damage rat model. Gene 2025; 952:149363. [PMID: 40064305 DOI: 10.1016/j.gene.2025.149363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/21/2025]
Abstract
BACKGROUND AND OBJECTIVE Neonatal hypoxic-ischemic encephalopathy (HIE) remains a critical challenge in perinatal medicine. This study aimed to elucidate the transcriptomic landscape, focusing on long non-coding RNAs (lncRNAs) expression patterns in the brain tissues of a neonatal rat model of HIE. METHODOLOGY We employed a modified Rice-Vannucci model to induce HIE in postnatal day 4 (P4) rats. The experimental groups were subjected to either 5 or 7 min of hypoxia (0 % O2, 100 % N2), while control animals were exposed to normoxic conditions. RESULTS RNA sequencing revealed a complex transcriptomic landscape in HIE brains, with approximately 80 million differentially expressed lncRNAs compared to controls. ELISA results demonstrated a significant upregulation of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) and a concomitant decrease in anti-inflammatory IL-10 levels in brain tissue of HIE rats. qRT-PCR analysis revealed aberrant expression of several miRNAs. Biochemical assays indicated a marked reduction in superoxide dismutase (SOD) activity and an increase in malondialdehyde (MDA) content in HIE brain tissues. CONCLUSIONS This study highlights the potential regulatory roles of lncRNAs in HIE brains. The intricate interplay between lncRNAs, miRNAs, and mRNAs and alterations in inflammatory and oxidative stress markers suggests a complex regulatory network governing HIE pathogenesis.
Collapse
Affiliation(s)
- Limin Wang
- Department of Pediatrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Yanni Gu
- Department of Fever Emergency, Shanghai TCM-Integrated Hospital, Shanghai 200082, China
| | - Chaobin Shen
- Department of Pediatrics, Shanghai TCM-Integrated Hospital, Shanghai 200082, China.
| |
Collapse
|
2
|
Koehn LM, Nguyen KV, Tucker R, Lim YP, Chen X, Stonestreet BS. Inter-alpha Inhibitor Proteins Modulate Microvascular Endothelial Components and Cytokines After Exposure to Hypoxia-Ischemia in Neonatal Rats. Mol Neurobiol 2025; 62:5057-5072. [PMID: 39505805 DOI: 10.1007/s12035-024-04594-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024]
Abstract
Inter-alpha inhibitor proteins (IAIPs) are neuroprotective and attenuate lipopolysaccharide (LPS)-mediated blood-brain barrier (BBB) disruption in neonatal rodents. We investigated some mechanism(s) fundamental to neuroprotection by IAIPs including changes in cerebral endothelial components and inflammation. Postnatal day-7 rats exposed to sham surgery and placebo or carotid ligation plus 8% FiO2 (90 min) were given IAIPs (30 or 60 mg/kg) or placebo and were killed 6, 12, 24, or 36 h after hypoxia-ischemia (HI). Proteins regulating BBB permeability to leukocytes (vascular cell adhesion molecule 1, VCAM-1), lipid-soluble (P-glycoprotein, PGP), and lipid-insoluble molecules (zonula occludens-1, ZO-1) were measured by immunoblot, and cytokines were measured in serum and cortex. HI resulted in reductions in ZO-1 and increases in VCAM-1, PGP, interferon-γ (IFN-γ), interleukin-12 (IL-12), vascular endothelial growth factor (VEGF), IL-α, and macrophage colony-stimulating factor (M-CSF) in cortex and increases in IL-4, IL-5, IL-10, and granulocyte colony-stimulating factor (G-CSF) in serum. IAIPs attenuated the reductions in ZO-1 and delayed increases in VCAM-1 and PGP in cortex and attenuated increases in cytokines in serum (IL-4, IL-5, IL-10, IFN-γ, G-CSF) and cortex (IL-1α, IL-12, IFN-γ, VEGF, M-CSF) after HI. We conclude that vascular endothelial proteins and cytokines exhibit sequential changes after HI and IAIPs modulate some of these HI-related changes in neonatal rats.
Collapse
Affiliation(s)
- Liam M Koehn
- Department of Pediatrics, Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
- Present Address: Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Kevin V Nguyen
- Department of Pediatrics, Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Richard Tucker
- Department of Pediatrics, Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Yow-Pin Lim
- ProThera Biologics Inc, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, The Alpert Medical School of Brown University, Providence, RI, USA
| | - Xiaodi Chen
- Department of Pediatrics, Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA.
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|
3
|
Lin TI, Hsieh PY, Lin HJ, Chiang CK, Sheu JJC, Chang WT, Liau I, Hsu HY. Soy Protein-Cultured Mesenchymal Stem Cell-Secreted Extracellular Vesicles Target the Neurovascular Unit: Insights from a Zebrafish Brain Injury Model. ACS Biomater Sci Eng 2025; 11:1432-1444. [PMID: 40000145 PMCID: PMC11897944 DOI: 10.1021/acsbiomaterials.4c02304] [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: 12/05/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025]
Abstract
Cerebral vascular disorders often accompany hypoxia-induced brain injury. In this study, we develop a zebrafish model of hypoxia-induced cerebral vascular injury to replicate the associated phenotypic changes, including cerebrovascular damage, neuronal apoptosis, and neurological dysfunction. We then explored the therapeutic potential of extracellular vesicles derived from Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) cultured on soy protein-coated surfaces. These vesicles demonstrated superior recovery efficacy, especially in restoring the blood-brain barrier integrity and improving neurological function. Our findings suggest that these potent therapeutic extracellular vesicles, easily produced from WJ-MSCs cultured in the presence of soy proteins, may mitigate hypoxia-induced brain injury by decreasing the severity of vascular disorder caused by oxidative stress. Protein-protein interactome analysis further suggests that multiple signaling pathways are likely involved in restoring normal neurovascular unit function.
Collapse
Affiliation(s)
- Tai-I Lin
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Pei-Ying Hsieh
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Hui-Jen Lin
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Cheng-Kang Chiang
- Department
of Chemistry, National Dong Hwa University, Hualien 974301, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute
of Biomedical Sciences, National Sun Yat-Sen
University, Kaohsiung 804201, Taiwan
| | - Wei-Tien Chang
- National
Taiwan University Hospital/National Taiwan University, Taipei 100233, Taiwan
| | - Ian Liau
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
- Center
for Emergent Functional Matter Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| | - Hsin-Yun Hsu
- Department
of Applied Chemistry and Institute of Molecular Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
- Center
for Emergent Functional Matter Science, National Yang-Ming Chiao-Tung University, Hsinchu 300093, Taiwan
| |
Collapse
|
4
|
Al-Ward H, Chen W, Gao W, Zhang C, Yang X, Xiong Y, Wang X, Agila R, Xu H, Sun YE. Can miRNAs in MSCs-EVs Offer a Potential Treatment for Hypoxic-ischemic Encephalopathy? Stem Cell Rev Rep 2025; 21:236-253. [PMID: 39503828 DOI: 10.1007/s12015-024-10803-6] [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] [Accepted: 10/09/2024] [Indexed: 01/26/2025]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a critical condition resulting from impaired oxygen and blood flow to the brain during birth, leading to neuroinflammation, neuronal apoptosis, and long-term neurological deficits. Despite the use of therapeutic hypothermia, current treatments remain inadequate in fully preventing brain damage. Recent advances in mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) offer a novel, cell-free therapeutic approach, as these EVs can cross the blood-brain barrier (BBB) and deliver functional microRNAs (miRNAs) to modulate key pathways involved in inflammation and neuroprotection. This review examines how specific miRNAs encapsulated in MSC-EVs-including miR-21, miR-124, miR-146, and the miR-17-92 cluster-target the complex inflammatory responses that drive HIE pathology. By modulating pathways such as NF-κB, STAT3, and PI3K/Akt, these miRNAs influence neuroinflammatory processes, reduce neuronal apoptosis, and promote tissue repair. The aim is to assess the therapeutic potential of miRNA-loaded MSC-EVs in mitigating inflammation and neuronal damage, thus addressing the limitations of current therapies like therapeutic hypothermia.
Collapse
Affiliation(s)
- Hisham Al-Ward
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wei Chen
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenxia Gao
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunxue Zhang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xueyan Yang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yao Xiong
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyi Wang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rafeq Agila
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hui Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
5
|
Hong SJ, De Souza BJ, Penberthy KK, Hwang L, Procaccini DE, Kheir JN, Bembea MM. Plasma brain-related biomarkers and potential therapeutic targets in pediatric ECMO. Neurotherapeutics 2025; 22:e00521. [PMID: 39765416 PMCID: PMC11840354 DOI: 10.1016/j.neurot.2024.e00521] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025] Open
Abstract
Extracorporeal membrane oxygenation (ECMO) is a technique used to support severe cardiopulmonary failure. Its potential life-saving benefits are tempered by the significant risk for acute brain injury (ABI), from both primary pathophysiologic factors and ECMO-related complications through central nervous system cellular injury, blood-brain barrier dysfunction (BBB), systemic inflammation and neuroinflammation, and coagulopathy. Plasma biomarkers are an emerging tool used to stratify risk for and diagnose ABI, and prognosticate neurofunctional outcomes. Components of the neurovascular unit have been rational targets for this inquiry in ECMO. Central nervous system (CNS) neuronal and astroglial cellular-derived neuron-specific enolase (NSE), tau, glial fibrillary acidic protein (GFAP) and S100β elevations have been detected in ABI and are associated with poorer outcomes. Evidence of BBB breakdown through peripheral blood detection of CNS cellular components NSE, GFAP, and S100β, as well as evidence of elevated BBB components vWF and PDGFRβ are associated with higher mortality and worse neurofunctional outcomes. Higher concentrations of pro-inflammatory cytokines (IL-1β, IL-6, IFN-γ, TNF-α) are associated with abnormal neuroimaging, and proteomic expression panels reveal different coagulation and inflammatory responses. Abnormal coagulation profiles are common in ECMO with ongoing studies attempting to describe specific abnormalities either being causal or associated with neurologic outcomes; vWF has shown some promise. Understanding these mechanisms of injury through biomarker analysis supports potential neuroprotective strategies such as individualized blood pressure targets, judicious hypercarbia and hypoxemia correction, and immunomodulation (inhaled hydrogen and N-acetylcysteine). Further research continues to elucidate the role of biomarkers as predictors, prognosticators, and therapeutic targets.
Collapse
Affiliation(s)
- Sue J Hong
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bradley J De Souza
- Department of Critical Care Medicine, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kristen K Penberthy
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lisa Hwang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - John N Kheir
- Department of Cardiology, Boston Children's Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Melania M Bembea
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
6
|
Iwata H, Katoh T, Truong SK, Sato T, Kawashima S, Mimuro S, Nakajima Y. Hydrogen attenuates endothelial glycocalyx damage associated with partial cardiopulmonary bypass in rats. PLoS One 2023; 18:e0295862. [PMID: 38113214 PMCID: PMC10729991 DOI: 10.1371/journal.pone.0295862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/30/2023] [Indexed: 12/21/2023] Open
Abstract
Cardiopulmonary bypass (CPB) causes systemic inflammation and endothelial glycocalyx damage. Hydrogen has anti-oxidant and anti-inflammatory properties; therefore, we hypothesized that hydrogen would alleviate endothelial glycocalyx damage caused by CPB. Twenty-eight male Sprague-Dawley rats were randomly divided into four groups (n = 7 per group), as follows: sham, control, 2% hydrogen, and 4% hydrogen. The rats were subjected to 90 minutes of partial CPB followed by 120 minutes of observation. In the hydrogen groups, hydrogen was administered via the ventilator and artificial lung during CPB, and via the ventilator for 60 minutes after CPB. After observation, blood collection, lung extraction, and perfusion fixation were performed, and the heart, lung, and brain endothelial glycocalyx thickness was measured by electron microscopy. The serum syndecan-1 concentration, a glycocalyx component, in the 4% hydrogen group (5.7 ± 4.4 pg/mL) was lower than in the control (19.5 ± 6.6 pg/mL) and 2% hydrogen (19.8 ± 5.0 pg/mL) groups (P < 0.001 for each), but it was not significantly different from the sham group (6.2 ± 4.0 pg/mL, P = 0.999). The endothelial glycocalyces of the heart and lung in the 4% hydrogen group were thicker than in the control group. The 4% hydrogen group had lower inflammatory cytokine concentrations (interleukin-1β and tumor necrosis factor-α) in serum and lung tissue, as well as a lower serum malondialdehyde concentration, than the control group. The 2% hydrogen group showed no significant difference in the serum syndecan-1 concentration compared with the control group. However, non-significant decreases in serum and lung tissue inflammatory cytokine concentrations, as well as in serum malondialdehyde concentration, were observed. Administration of 4% hydrogen via artificial and autologous lungs attenuated endothelial glycocalyx damage caused by partial CPB in rats, which might be mediated by the anti-inflammatory and anti-oxidant properties of hydrogen.
Collapse
Affiliation(s)
- Hiroki Iwata
- Department of Anesthesiology and Intensive Care, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takasumi Katoh
- Department of Anesthesiology and Intensive Care, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Sang Kien Truong
- Department of Anesthesiology and Surgical Critical Care, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | - Tsunehisa Sato
- Institute for Physiological Sciences, Justus-Liebig-University, Giessen, Germany
| | - Shingo Kawashima
- Department of Anesthesiology and Intensive Care, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Soichiro Mimuro
- Department of Anesthesiology and Intensive Care, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yoshiki Nakajima
- Department of Anesthesiology and Intensive Care, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
7
|
Htun Y, Nakamura S, Nakao Y, Mitsuie T, Ohta K, Arioka M, Yokota T, Inoue E, Inoue K, Tsuchiya T, Koyano K, Konishi Y, Miki T, Ueno M, Kusaka T. Conflicting findings on the effectiveness of hydrogen therapy for ameliorating vascular leakage in a 5-day post hypoxic-ischemic survival piglet model. Sci Rep 2023; 13:10486. [PMID: 37380745 DOI: 10.1038/s41598-023-37577-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 06/23/2023] [Indexed: 06/30/2023] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a major cause of morbidity and mortality in newborns in both high- and low-income countries. The important determinants of its pathophysiology are neural cells and vascular components. In neonatal HIE, increased vascular permeability due to damage to the blood-brain barrier is associated with seizures and poor outcomes in both translational and clinical studies. In our previous studies, hydrogen gas (H2) improved the neurological outcome of HIE and ameliorated the cell death. In this study, we used albumin immunohistochemistry to assess if H2 inhalation effectively reduced the cerebral vascular leakage. Of 33 piglets subjected to a hypoxic-ischemic insult, 26 piglets were ultimately analyzed. After the insult, the piglets were grouped into normothermia (NT), H2 ventilation (H2), therapeutic hypothermia (TH), and H2 combined with TH (H2-TH) groups. The ratio of albumin stained to unstained areas was analyzed and found to be lower in the H2 group than in the other groups, although the difference was not statistically significant. In this study, H2 therapy did not significantly improve albumin leakage despite the histological images suggesting signs of improvement. Further investigations are warranted to study the efficacy of H2 gas for vascular leakage in neonatal HIE.
Collapse
Affiliation(s)
- Yinmon Htun
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Yasuhiro Nakao
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Tsutomu Mitsuie
- Medical Engineering Equipment Management Center, Kagawa University Hospital, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Kenichi Ohta
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Makoto Arioka
- Maternal and Perinatal Center, Kagawa University Hospital, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Takayuki Yokota
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Eri Inoue
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Kota Inoue
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Toi Tsuchiya
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Kosuke Koyano
- Maternal and Perinatal Center, Kagawa University Hospital, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Takanori Miki
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan.
| |
Collapse
|
8
|
Paz AA, González-Candia A. Potential pharmacological target of tight junctions to improve the BBB permeability in neonatal Hypoxic-Ischemic encephalopathy Diseases. Biochem Pharmacol 2023; 207:115356. [PMID: 36455671 DOI: 10.1016/j.bcp.2022.115356] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
Neonatal encephalopathy (NE) is a pathological condition that describes a neurocognitive malfunction in the newborn that arises from fetal, peripartum, or intrapartum events of multifactorial nature, having a poor prognosis and accounting for an incidence of 5-8 per 1000 live births. Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the most studied paradigms of NE, caused by a scarce cerebral perfusion and oxygen supply during perinatal life. The cerebral hypoxic-ischemic insult promotes a loss of permeability of the blood-brain barrier (BBB), an essential structural intermediary of blood-brain communication. This permeability disruption is associated with an increase in inflammatory cytokines, an increase of adhesion molecules, and oxidative stress which disturb the tight junction (TJ) performance and enable transcytosis and paracellular leakage, ultimately leading to death from brain cells. In this context, TJs proteins are essential to preserving the barrier mechanical stability and signaling that modulates the brain-blood vessel multicellular domains, known as neurovascular units (NVU). Recent studies have proposed different strategies with neuroprotective effects that allow for maintaining or restoring the integrity and permeability of the BBB. This review identifies and discusses regulator mechanisms and novel aspects of TJs in the BBB disruption induced by cerebral hypoxic insults during the perinatal period, evaluating potential pharmacological strategies to safeguard BBB integrity.
Collapse
Affiliation(s)
- Adolfo A Paz
- Institute of Health Sciences, University O'Higgins, Rancagua, Chile
| | | |
Collapse
|
9
|
Delavogia E, Ntentakis DP, Cortinas JA, Fernandez-Gonzalez A, Alex Mitsialis S, Kourembanas S. Mesenchymal Stromal/Stem Cell Extracellular Vesicles and Perinatal Injury: One Formula for Many Diseases. Stem Cells 2022; 40:991-1007. [PMID: 36044737 PMCID: PMC9707037 DOI: 10.1093/stmcls/sxac062] [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: 06/08/2022] [Accepted: 08/05/2022] [Indexed: 11/12/2022]
Abstract
Over the past decades, substantial advances in neonatal medical care have increased the survival of extremely premature infants. However, there continues to be significant morbidity associated with preterm birth with common complications including bronchopulmonary dysplasia (BPD), necrotizing enterocolitis (NEC), neuronal injury such as intraventricular hemorrhage (IVH) or hypoxic ischemic encephalopathy (HIE), as well as retinopathy of prematurity (ROP). Common developmental immune and inflammatory pathways underlie the pathophysiology of such complications providing the opportunity for multisystem therapeutic approaches. To date, no single therapy has proven to be effective enough to prevent or treat the sequelae of prematurity. In the past decade mesenchymal stem/stromal cell (MSC)-based therapeutic approaches have shown promising results in numerous experimental models of neonatal diseases. It is now accepted that the therapeutic potential of MSCs is comprised of their secretome, and several studies have recognized the small extracellular vesicles (sEVs) as the paracrine vector. Herein, we review the current literature on the MSC-EVs as potential therapeutic agents in neonatal diseases and comment on the progress and challenges of their translation to the clinical setting.
Collapse
Affiliation(s)
- Eleni Delavogia
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Dimitrios P Ntentakis
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - John A Cortinas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Malondialdehyde and Neutrophil Gelatinase-Associated Lipocalin as Markers of Oxidative Stress in Small for Gestational Age Newborns from Hypertensive and Preeclamptic Pregnancies. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9246233. [PMID: 35224102 PMCID: PMC8881126 DOI: 10.1155/2022/9246233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 02/05/2022] [Indexed: 11/18/2022]
Abstract
Introduction. It is speculated that preeclampsia and hypertension during pregnancy are associated with an imbalance of the placental antioxidant defence, which results in the overproduction of reactive oxygen species and fetal growth restriction. Many research implied that oxidant stress in utero may be an important determinant of mortality and morbidity in neonates. Moreover, the authors demonstrated the reduced number of nephrons and a higher prevalence of renal injury in neonates with growth restriction, including small for gestational age (SGA) neonates. Alas, it remains unclear whether basal antioxidant status is altered in the kidneys of SGA newborns. Materials and Methods. In this study, we assessed neutrophil gelatinase-associated lipocalin (NGAL) and malondialdehyde (MDA) levels in samples collected from umbilical blood and 12 hours after delivery in neonates born by mothers suffering from preeclampsia or hypertension during pregnancy and those from physiological pregnancies. Additionally, the authors evaluated levels of the aforementioned biomarkers regarding the occurrence of growth restriction in newborns. For this study, we enrolled 27 newborns, which fulfilled inclusion criteria for SGA diagnosis (SGA group), while 21 were appropriate for gestational age neonates, as the AGA group. Results. In the presented study, we have found significant differences in umbilical cord MDA and NGAL concentration between the SGA and AGA groups. Such dependencies were not found in blood samples from neonates collected in the first 12 hours of life for MDA and NGAL concentrations. Additionally, we have observed differences in umbilical MDA and NGAL levels between newborns of preeclamptic or hypertensive mothers compared to healthy ones. A significant correlation between the occurrence of hypertension during pregnancy and umbilical MDA and NGAL concentrations was also found. Conclusions. Small for gestational age newborns or those born by preeclamptic and hypertensive mothers had significantly higher MDA and NGAL levels as compared to healthy ones. Further investigation is needed to understand the pathophysiologic influence of hypertension in pregnancy and oxidative stress injury in newborns with growth restriction.
Collapse
|
11
|
Martini S, Castellini L, Parladori R, Paoletti V, Aceti A, Corvaglia L. Free Radicals and Neonatal Brain Injury: From Underlying Pathophysiology to Antioxidant Treatment Perspectives. Antioxidants (Basel) 2021; 10:2012. [PMID: 34943115 PMCID: PMC8698308 DOI: 10.3390/antiox10122012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 01/23/2023] Open
Abstract
Free radicals play a role of paramount importance in the development of neonatal brain injury. Depending on the pathophysiological mechanisms underlying free radical overproduction and upon specific neonatal characteristics, such as the GA-dependent maturation of antioxidant defenses and of cerebrovascular autoregulation, different profiles of injury have been identified. The growing evidence on the detrimental effects of free radicals on the brain tissue has led to discover not only potential biomarkers for oxidative damage, but also possible neuroprotective therapeutic approaches targeting oxidative stress. While a more extensive validation of free radical biomarkers is required before considering their use in routine neonatal practice, two important treatments endowed with antioxidant properties, such as therapeutic hypothermia and magnesium sulfate, have become part of the standard of care to reduce the risk of neonatal brain injury, and other promising therapeutic strategies are being tested in clinical trials. The implementation of currently available evidence is crucial to optimize neonatal neuroprotection and to develop individualized diagnostic and therapeutic approaches addressing oxidative brain injury, with the final aim of improving the neurological outcome of this population.
Collapse
Affiliation(s)
- Silvia Martini
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.A.); (L.C.)
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Laura Castellini
- School of Medicine and Surgery, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy;
| | - Roberta Parladori
- Specialty School of Pediatrics, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy;
| | - Vittoria Paoletti
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Arianna Aceti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.A.); (L.C.)
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Luigi Corvaglia
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.A.); (L.C.)
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| |
Collapse
|
12
|
Li J, Xia Y, Huang Z, Zhao Y, Xiong R, Li X, Huang Q, Shan F. Novel HIF-1-target gene isthmin1 contributes to hypoxia-induced hyperpermeability of pulmonary microvascular endothelial cells monolayers. Am J Physiol Cell Physiol 2021; 321:C671-C680. [PMID: 34469202 DOI: 10.1152/ajpcell.00124.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/19/2021] [Indexed: 11/22/2022]
Abstract
Hypoxia-induced pulmonary microvascular endothelial cell (PMVEC) monolayers hyperpermeability is vital for vascular leakage, which participates in vascular diseases, such as acute lung injury (ALI) and high-altitude pulmonary edema (HAPE). We previously observed that PMVEC permeability was markedly elevated in hypoxia when cocultured with primary type II alveolar epithelial cells (AECII) in which isthmin1 (ISM1) was highly upregulated. However, whether the upregulation of ISM1 plays a role in hypoxia-induced PMVEC hyperpermeability is unclear. In this study, we assessed the role of AECII-derived ISM1 in hypoxia-induced PMVEC hyperpermeability with an AECII/PMVEC coculture system and uncovered the underlying mechanism whereby hypoxia stimulates ISM1 gene expression. We found that ISM1 gene expression was upregulated in cultured AECII cells exposed to hypoxia (3% O2) and that AECII-derived ISM1 participated in hypoxia-induced hyperpermeability of PMVEC monolayers, as small interference RNA (siRNA)-mediated knockdown of ISM1 in AECII markedly attenuated the increase in PMVEC permeability in coculture system under hypoxia. In addition, we confirmed that ISM1 was regulated by hypoxia-inducible factor-1α (HIF1α) according to the evidence that silencing of HIF1α inhibited the hypoxia-mediated upregulation of ISM1. Mechanismly, overexpression of HIF1α transcriptionally activated ISM1 gene expression by directly binding to the conserved regulatory elements upstream of the ism1 locus. We identified a novel HIF-1-target gene ISM1, which involves in hyperpermeability of pulmonary microvascular endothelial cell monolayers under hypoxia. Our in vitro cell experiments implied that the upregulated ISM1 derived from alveolar epithelium might be a vital modulator in hypoxia-induced endothelial hyperpermeability and thereby implicates with hypoxic pulmonary-related diseases.
Collapse
Affiliation(s)
- Junxia Li
- State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yiming Xia
- State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Zhizhong Huang
- State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yan Zhao
- State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Renping Xiong
- State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Xiaoxu Li
- College of High Altitude Military Medicine, Army Medical University, Chongqing, People's Republic of China
| | - Qingyuan Huang
- College of High Altitude Military Medicine, Army Medical University, Chongqing, People's Republic of China
| | - Fabo Shan
- State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| |
Collapse
|
13
|
Effects of Hypothermia and Allopurinol on Oxidative Status in a Rat Model of Hypoxic Ischemic Encephalopathy. Antioxidants (Basel) 2021; 10:antiox10101523. [PMID: 34679658 PMCID: PMC8533154 DOI: 10.3390/antiox10101523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Hypoxic ischemic encephalopathy (HIE) is one of the main causes of morbidity and mortality during the neonatal period, despite treatment with hypothermia. There is evidence that oxidative damage plays an important role in the pathophysiology of hypoxic-ischemic (HI) brain injury. Our aim was to investigate whether postnatal allopurinol administration in combination with hypothermia would reduce oxidative stress (OS) biomarkers in an animal model of HIE. Postnatal 10-day rat pups underwent unilateral HI of moderate severity. Pups were randomized into: Sham operated, hypoxic-ischemic (HI), HI + allopurinol (HIA), HI + hypothermia (HIH), and HI + hypothermia + allopurinol (HIHA). Biomarkers of OS and antioxidants were evaluated: GSH/GSSG ratio and carbonyl groups were tested in plasma. Total antioxidant capacity (TAC) was analyzed in plasma and cerebrospinal fluid, and 8-iso-prostaglandin F2α was measured in brain tissue. Plasma 2,2′–azinobis-(3-ethyl-benzothiazoline-6-sulfonic acid) (ABTS) levels were preserved in those groups that received allopurinol and dual therapy. In cerebrospinal fluid, only the HIA group presented normal ferric reducing ability of plasma (FRAP) levels. Protein oxidation and lipid peroxidation were significantly reduced in all groups treated with hypothermia and allopurinol, thus enhancing neuroprotection in HIE.
Collapse
|
14
|
Gamage TKJB, Fraser M. The Role of Extracellular Vesicles in the Developing Brain: Current Perspective and Promising Source of Biomarkers and Therapy for Perinatal Brain Injury. Front Neurosci 2021; 15:744840. [PMID: 34630028 PMCID: PMC8498217 DOI: 10.3389/fnins.2021.744840] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
This comprehensive review focuses on our current understanding of the proposed physiological and pathological functions of extracellular vesicles (EVs) in the developing brain. Furthermore, since EVs have attracted great interest as potential novel cell-free therapeutics, we discuss advances in the knowledge of stem cell- and astrocyte-derived EVs in relation to their potential for protection and repair following perinatal brain injury. This review identified 13 peer-reviewed studies evaluating the efficacy of EVs in animal models of perinatal brain injury; 12/13 utilized mesenchymal stem cell-derived EVs (MSC-EVs) and 1/13 utilized astrocyte-derived EVs. Animal model, method of EV isolation and size, route, timing, and dose administered varied between studies. Notwithstanding, EV treatment either improved and/or preserved perinatal brain structures both macroscopically and microscopically. Additionally, EV treatment modulated inflammatory responses and improved brain function. Collectively this suggests EVs can ameliorate, or repair damage associated with perinatal brain injury. These findings warrant further investigation to identify the optimal cell numbers, source, and dosage regimens of EVs, including long-term effects on functional outcomes.
Collapse
|
15
|
Shen G, Hu S, Zhao Z, Zhang L, Ma Q. C-Type Natriuretic Peptide Ameliorates Vascular Injury and Improves Neurological Outcomes in Neonatal Hypoxic-Ischemic Brain Injury in Mice. Int J Mol Sci 2021; 22:ijms22168966. [PMID: 34445671 PMCID: PMC8396645 DOI: 10.3390/ijms22168966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 01/01/2023] Open
Abstract
C-type natriuretic peptide (CNP) is an important vascular regulator that is present in the brain. Our previous study demonstrated the innate neuroprotectant role of CNP in the neonatal brain after hypoxic-ischemic (HI) insults. In this study, we further explored the role of CNP in cerebrovascular pathology using both in vivo and in vitro models. In a neonatal mouse HI brain injury model, we found that intracerebroventricular administration of recombinant CNP dose-dependently reduces brain infarct size. CNP significantly decreases brain edema and immunoglobulin G (IgG) extravasation into the brain tissue, suggesting a vasculoprotective effect of CNP. Moreover, in primary brain microvascular endothelial cells (BMECs), CNP dose-dependently protects BMEC survival and monolayer integrity against oxygen-glucose deprivation (OGD). The vasculoprotective effect of CNP is mediated by its innate receptors NPR2 and NPR3, in that inhibition of either NPR2 or NPR3 counteracts the protective effect of CNP on IgG leakage after HI insult and BMEC survival under OGD. Of importance, CNP significantly ameliorates brain atrophy and improves neurological deficits after HI insults. Altogether, the present study indicates that recombinant CNP exerts vascular protection in neonatal HI brain injury via its innate receptors, suggesting a potential therapeutic target for the treatment of neonatal HI brain injury.
Collapse
Affiliation(s)
- Guofang Shen
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (G.S.); (S.H.); (L.Z.)
| | - Shirley Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (G.S.); (S.H.); (L.Z.)
| | - Zhen Zhao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (G.S.); (S.H.); (L.Z.)
| | - Qingyi Ma
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (G.S.); (S.H.); (L.Z.)
- Correspondence: ; Tel.: +1-909-558-4325; Fax: +1-909-558-4029
| |
Collapse
|
16
|
Nathan NO, Hedegaard M, Karlsson G, Knudsen LE, Mathiesen L. Intrapartum transfer of oxytocin across the human placenta: An ex vivo perfusion experiment. Placenta 2021; 112:105-110. [PMID: 34329968 DOI: 10.1016/j.placenta.2021.07.289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/24/2021] [Accepted: 07/19/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Investigation of the maternal to fetal transfer of oxytocin across the dually perfused term human placenta. METHODS Human placentae obtained from term singleton pregnancies were utilized in a dual recirculating model of ex vivo placental perfusion. Six placentae from women delivering by elective cesarean at term were perfused, one blank and five with the test substance synthetic oxytocin (0.8 ng/mL) (OX) added to the maternal perfusate for 180 min. Antipyrine was used as positive control to validate overlap of the maternal and fetal circuits. The concentration of OX was determined by radioimmunoassay. RESULTS A fall in maternal concentration of OX was seen throughout the experiment. At 90 min of perfusion a state of equilibrium was reached between maternal and fetal concentrations; however after 180 min the fetal concentration of OX was higher than that of the maternal. 31 % of the test substance was accounted for at the end of the experiment - suggesting OX protein binding and a high degree of oxytocinase activity. DISCUSSION The ex vivo perfusion experiments revealed low transfer of OX to the fetal circuit below physiologically relevant concentrations.
Collapse
Affiliation(s)
- Nina Olsén Nathan
- The Research Unit Women's and Children's Health, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, Rigshospitalet, Tagensvej 22, 2200, Copenhagen, Denmark; Department of Obstetrics, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Morten Hedegaard
- Department of Obstetrics, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Gösta Karlsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Lisbeth E Knudsen
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Øster Farimagsgade 5, Building 9, 1014, Copenhagen, Denmark
| | - Line Mathiesen
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Øster Farimagsgade 5, Building 9, 1014, Copenhagen, Denmark.
| |
Collapse
|
17
|
Wang L, Xu L, Du J, Zhao X, Liu M, Feng J, Hu K. Nose-to-brain delivery of borneol modified tanshinone IIA nanoparticles in prevention of cerebral ischemia/reperfusion injury. Drug Deliv 2021; 28:1363-1375. [PMID: 34180761 PMCID: PMC8245080 DOI: 10.1080/10717544.2021.1943058] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Targeted treatment of cerebral ischemia/reperfusion injury (CIRI) remains a problem due to the difficulty in drug delivery across the blood–brain barrier (BBB). In this study, we developed Bo-TSA-NP, a novel tanshinone IIA (TSA) loaded nanoparticles modified by borneol, which has long been proved with the ability to enhance other drugs’ transport across the BBB. The Bo-TSA-NP, with a particle size of about 160 nm, drug loading of 3.6%, showed sustained release and P-glycoprotein (P-gp) inhibition property. It demonstrated a significantly higher uptake by 16HBE cells in vitro through the clathrin/caveolae-mediated endocytosis and micropinocytosis. Following intranasal (IN) administration, Bo-TSA-NP significantly improved the preventive effect on a rat model of CIRI with improved neurological scores, decreased cerebral infarction areas and a reduced content of malondialdehyde (MDA) and increased activity of superoxide dismutase (SOD) in rat brain. In conclusion, these results indicate that Bo-TSA-NP is a promising nose-to-brain delivery system that can enhance the prevention effect of TSA on CIRI.
Collapse
Affiliation(s)
- Luting Wang
- Institute of Interdisciplinary Integrative Medicine Research, Murad Research Center for Modernized Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.,Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Lin Xu
- Institute of Interdisciplinary Integrative Medicine Research, Murad Research Center for Modernized Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.,Institute of Interdisciplinary Integrative Medicine Research, The Center for TCM Standardization, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Junfeng Du
- Institute of Interdisciplinary Integrative Medicine Research, Murad Research Center for Modernized Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Xiao Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Murad Research Center for Modernized Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Mei Liu
- Institute of Interdisciplinary Integrative Medicine Research, Murad Research Center for Modernized Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Jianfang Feng
- Institute of Interdisciplinary Integrative Medicine Research, Murad Research Center for Modernized Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.,School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, People's Republic of China
| | - Kaili Hu
- Institute of Interdisciplinary Integrative Medicine Research, Murad Research Center for Modernized Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| |
Collapse
|
18
|
Andersson EA, Mallard C, Ek CJ. Circulating tight-junction proteins are potential biomarkers for blood-brain barrier function in a model of neonatal hypoxic/ischemic brain injury. Fluids Barriers CNS 2021; 18:7. [PMID: 33568200 PMCID: PMC7877092 DOI: 10.1186/s12987-021-00240-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Background Neonatal encephalopathy often leads to lifelong disabilities with limited treatments currently available. The brain vasculature is an important factor in many neonatal neurological disorders but there is a lack of diagnostic tools to evaluate the brain vascular dysfunction of neonates in the clinical setting. Measurement of blood–brain barrier tight-junction (TJ) proteins have shown promise as biomarkers for brain injury in the adult. Here we tested the biomarker potential of tight-junctions in the context of neonatal brain injury. Methods The levels of TJ-proteins (occluding, claudin-5, and zonula occludens protein 1) in both blood plasma and cerebrospinal fluid (CSF) as well as blood–brain barrier function via 14C-sucrose (342 Da) and Evans blue extravasation were measured in a hypoxia/ischemia brain-injury model in neonatal rats. Results Time-dependent changes of occludin and claudin-5 levels could be measured in blood and CSF after hypoxia/ischemia with males generally having higher levels than females. The levels of claudin-5 in CSF correlated with the severity of the brain injury at 24 h post- hypoxia/ischemia. Simultaneously, we detected early increase in blood–brain barrier-permeability at 6 and 24 h after hypoxia/ischemia. Conclusions Levels of circulating claudin-5 and occludin are increased after hypoxic/ischemic brain injuries and blood–brain barrier-impairment and have promise as early biomarkers for cerebral vascular dysfunction and as a tool for risk assessment of neonatal brain injuries.
Collapse
Affiliation(s)
- E Axel Andersson
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 11, 413 90, Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 11, 413 90, Gothenburg, Sweden
| | - C Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 11, 413 90, Gothenburg, Sweden.
| |
Collapse
|
19
|
Soul JS, Bergin AM, Stopp C, Hayes B, Singh A, Fortuno CR, O'Reilly D, Krishnamoorthy K, Jensen FE, Rofeberg V, Dong M, Vinks AA, Wypij D, Staley KJ. A Pilot Randomized, Controlled, Double-Blind Trial of Bumetanide to Treat Neonatal Seizures. Ann Neurol 2021; 89:327-340. [PMID: 33201535 PMCID: PMC8122513 DOI: 10.1002/ana.25959] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 01/19/2023]
Abstract
OBJECTIVE In the absence of controlled trials, treatment of neonatal seizures has changed minimally despite poor drug efficacy. We tested bumetanide added to phenobarbital to treat neonatal seizures in the first trial to include a standard-therapy control group. METHODS A randomized, double-blind, dose-escalation design was employed. Neonates with postmenstrual age 33 to 44 weeks at risk of or with seizures were eligible. Subjects with electroencephalography (EEG)-confirmed seizures after ≥20 and <40mg/kg phenobarbital were randomized to receive additional phenobarbital with either placebo (control) or 0.1, 0.2, or 0.3mg/kg bumetanide (treatment). Continuous EEG monitoring data from ≥2 hours before to ≥48 hours after study drug administration (SDA) were analyzed for seizures. RESULTS Subjects were randomized to treatment (n = 27) and control (n = 16) groups. Pharmacokinetics were highly variable among subjects and altered by hypothermia. The only statistically significant adverse event was diuresis in treated subjects (48% vs 13%, p = 0.02). One treated (4%) and 3 control subjects died (19%, p = 0.14). Among survivors, 2 of 26 treated subjects (8%) and 0 of 13 control subjects had hearing impairment, as did 1 nonrandomized subject. Total seizure burden varied widely, with much higher seizure burden in treatment versus control groups (median = 3.1 vs 1.2 min/h, p = 0.006). There was significantly greater reduction in seizure burden 0 to 4 hours and 2 to 4 hours post-SDA (both p < 0.01) compared with 2-hour baseline in treatment versus control groups with adjustment for seizure burden. INTERPRETATION Although definitive proof of efficacy awaits an appropriately powered phase 3 trial, this randomized, controlled, multicenter trial demonstrated an additional reduction in seizure burden attributable to bumetanide over phenobarbital without increased serious adverse effects. Future trials of bumetanide and other drugs should include a control group and balance seizure severity. ANN NEUROL 2021;89:327-340.
Collapse
Affiliation(s)
- Janet S Soul
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ann M Bergin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christian Stopp
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Breda Hayes
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Avantika Singh
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carmen R Fortuno
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Deirdre O'Reilly
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kalpathy Krishnamoorthy
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Frances E Jensen
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Valerie Rofeberg
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Min Dong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David Wypij
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kevin J Staley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Li X, Yu J, Ma D, Weng X. Edaravone Improves the Post-traumatic Brain Injury Dysfunction in Learning and Memory by Modulating Nrf2/ARE Signal Pathway. Clinics (Sao Paulo) 2021; 76:e3131. [PMID: 34878029 PMCID: PMC8610218 DOI: 10.6061/clinics/2021/e3131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES To investigate the molecular mechanism of edaravone (EDA) in improving the post-traumatic brain injury (TBI) dysfunction in learning and memory. METHODS In vitro and in vivo TBI models were established using hydrogen peroxide (H2O2) treatment for hippocampal nerve stem cells (NSCs) and surgery for rats, followed by EDA treatment. WST 1 measurement, methylthiazol tetrazolium assay, and flow cytometry were performed to determine the activity, proliferation, and apoptosis of NSCs, and malondialdehyde (MDA), lactic dehydrogenase (LDH), and reactive oxygen species (ROS) detection kits were used to analyze the oxides in NSCs. RESULTS Following EDA pretreatment, NSCs presented with promising resistance to H2O2-induced oxidative stress, whereas NSCs manifested significant increases in activity and proliferation and a decrease in apoptosis. Meanwhile, for NSCs, EDA pretreatment reduced the levels of MDA, LDH, and ROS, with a significant upregulation of Nrf2/antioxidant response element (ARE) signaling pathway, whereas for EDA-treated TBI rats, a significant reduction was observed in the trauma area and injury to the hippocampus, with improvement in memory and learning performance and upregulation of Nrf2/ARE signaling pathway. CONCLUSIONS EDA, by regulating the activity of Nrf2/ARE signal pathway, can improve the TBI-induced injury to NSCs and learning and memory dysfunction in rats.
Collapse
Affiliation(s)
| | - Jing Yu
- Corresponding author. E-mail:
| | | | | |
Collapse
|
21
|
LI H, YANG M, ZHAO Z, ZHOU X, LIU Y, ZHUANG Y, SHAN Y. MMP-9 and MMP-3 gene polymorphisms in neonatal hypoxic ischemic encephalopathy. J Neurosurg Sci 2021; 64:579-581. [DOI: 10.23736/s0390-5616.19.04797-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
22
|
Feng L, Li M, Ren J, Li Y, Wang Q, Zhang P, Zhang X, Wang T, Li Y. Proteomic Analysis Reveals that Di Dang Decoction Protects Against Acute Intracerebral Hemorrhage Stroke in Rats by Regulating S100a8, S100a9 Col1a1, and Col1a2. Neuropsychiatr Dis Treat 2021; 17:3301-3314. [PMID: 34785900 PMCID: PMC8591113 DOI: 10.2147/ndt.s331688] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/30/2021] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE The present study aimed to explore the neuroprotective mechanism of Di Dang decoction (DDD) during acute intracerebral hemorrhage (AICH) stroke in Sprague Dawley rats through proteomic analysis. METHODS A total of 135 healthy Sprague Dawley rats were randomly divided into five groups: control (n = 27), model (n = 27), DDD low-dose (n = 27), DDD medium-dose (n = 27), and DDD high-dose (n = 27). AICH stroke in rats was induced by injecting autologous blood into the caudate nucleus. The modified Neurological Severity Score (mNSS) was used to evaluate the cerebral nerve function deficit. Hematoxylin and eosin (HE) staining was performed to observe the brain tissue at the lesion site. Albumin concentration was assessed on obvious blood-brain barrier damaged and brain water content was used to evaluate the brain injury. For quantitative proteomics, proteins were extracted from the cerebral cortices. Target proteins were identified using mass spectrometer-based targeted proteomic quantification. RESULTS mNSS score, HE staining results, albumin concentration, and brain water content showed the most significant improvements in the neuroprotective in the high-dose group 7 days after DDD exposure. Furthermore, quantitative proteomics analysis showed that, relative to the control group, S100a8 and S100a9 were downregulated by 0.614 (p = 0.033702) and 0.506 times (p = 0.000024) in the high-dose group. Compared with the control group, Col1a1 and Col1a2 were upregulated by 1.319 (p = 0.000184) and 1.348 (p = 0.014097) times in the high-dose group. These results were confirmed using mass spectrometer-based targeted proteomic quantification. CONCLUSION Application of a high-dose DDD for 7 days in AICH stroke rats showed the most significant improvements in neuroprotective. Mechanistically, this effect was mediated by S100a8 and S100a9 protein downregulation and Col1a1 and Col1a2 upregulation.
Collapse
Affiliation(s)
- Lina Feng
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, People's Republic of China
| | - Mingquan Li
- Neurology Department, Third Affiliated Clinical Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin Province, People's Republic of China
| | - Jixiang Ren
- Preclinical Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin Province, People's Republic of China
| | - Yujuan Li
- Ultrasonic Diagnosis Department, Third Affiliated Clinical Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin Province, People's Republic of China
| | - Qi Wang
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, People's Republic of China
| | - Pengqi Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, People's Republic of China
| | - Xinyue Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, People's Republic of China
| | - Tianye Wang
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, People's Republic of China
| | - Yunqiang Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, People's Republic of China
| |
Collapse
|
23
|
Koehn LM, Chen X, Logsdon AF, Lim YP, Stonestreet BS. Novel Neuroprotective Agents to Treat Neonatal Hypoxic-Ischemic Encephalopathy: Inter-Alpha Inhibitor Proteins. Int J Mol Sci 2020; 21:E9193. [PMID: 33276548 PMCID: PMC7731124 DOI: 10.3390/ijms21239193] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 02/02/2023] Open
Abstract
Perinatal hypoxia-ischemia (HI) is a major cause of brain injury and mortality in neonates. Hypoxic-ischemic encephalopathy (HIE) predisposes infants to long-term cognitive deficits that influence their quality of life and place a large burden on society. The only approved treatment to protect the brain after HI is therapeutic hypothermia, which has limited effectiveness, a narrow therapeutic time window, and is not considered safe for treatment of premature infants. Alternative or adjunctive therapies are needed to improve outcomes of full-term and premature infants after exposure to HI. Inter-alpha inhibitor proteins (IAIPs) are immunomodulatory molecules that are proposed to limit the progression of neonatal inflammatory conditions, such as sepsis. Inflammation exacerbates neonatal HIE and suggests that IAIPs could attenuate HI-related brain injury and improve cognitive outcomes associated with HIE. Recent studies have shown that intraperitoneal treatment with IAIPs can decrease neuronal and non-neuronal cell death, attenuate glial responses and leukocyte invasion, and provide long-term behavioral benefits in neonatal rat models of HI-related brain injury. The present review summarizes these findings and outlines the remaining experimental analyses necessary to determine the clinical applicability of this promising neuroprotective treatment for neonatal HI-related brain injury.
Collapse
Affiliation(s)
- Liam M. Koehn
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI 02905, USA; (L.M.K.); (X.C.)
| | - Xiaodi Chen
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI 02905, USA; (L.M.K.); (X.C.)
| | - Aric F. Logsdon
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA;
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02903, USA;
- Department of Pathology and Laboratory Medicine, The Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Barbara S. Stonestreet
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI 02905, USA; (L.M.K.); (X.C.)
| |
Collapse
|
24
|
Murden S, Borbélyová V, Laštůvka Z, Mysliveček J, Otáhal J, Riljak V. Gender differences involved in the pathophysiology of the perinatal hypoxic-ischemic damage. Physiol Res 2020; 68:S207-S217. [PMID: 31928039 DOI: 10.33549/physiolres.934356] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a neonatal condition that occurs as a consequence of perinatal asphyxia, which is caused by a number of factors, commonly via compression of the umbilical cord, placental abruption, severe meconium aspiration, congenital cardiac or pulmonary anomalies and birth trauma. Experimental studies have confirmed that male rat pups show a higher resistance to HIE treatment. Moreover, the long-term consequences of hypoxia in male are more severe in comparison to female rat pups. These sex differences can be attributed to the pathophysiology of hypoxia-ischemia, whereby studies are beginning to establish such gender-specific distinctions. The current and sole treatment for HIE is hypothermia, in which a reduction in temperature prevents long-term effects, such as cerebral palsy or seizures. However, in most cases hypothermia is not a sufficient treatment as indicated by a high mortality rate. In the present review, we discuss the gender differences within the pathophysiology of hypoxia-ischemia and delve into the role of gender in the incidence, progression and severity of the disease. Furthermore, this may result in the development of potential novel treatment approaches for targeting and preventing the long-term consequences of HIE.
Collapse
Affiliation(s)
- S Murden
- Department of Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
25
|
DISDIER C, STONESTREET BS. Hypoxic-ischemic-related cerebrovascular changes and potential therapeutic strategies in the neonatal brain. J Neurosci Res 2020; 98:1468-1484. [PMID: 32060970 PMCID: PMC7242133 DOI: 10.1002/jnr.24590] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/20/2020] [Accepted: 01/28/2020] [Indexed: 12/11/2022]
Abstract
Perinatal hypoxic-ischemic (HI)-related brain injury is an important cause of morbidity and long-standing disability in newborns. The only currently approved therapeutic strategy available to reduce brain injury in the newborn is hypothermia. Therapeutic hypothermia can only be used to treat HI encephalopathy in full-term infants and survivors remain at high risk for a wide spectrum of neurodevelopmental abnormalities as a result of residual brain injury. Therefore, there is an urgent need for adjunctive therapeutic strategies. Inflammation and neurovascular damage are important factors that contribute to the pathophysiology of HI-related brain injury and represent exciting potential targets for therapeutic intervention. In this review, we address the role of each component of the neurovascular unit (NVU) in the pathophysiology of HI-related injury in the neonatal brain. Disruption of the blood-brain barrier (BBB) observed in the early hours after an HI-related event is associated with a response at the basal lamina level, which comprises astrocytes, pericytes, and immune cells, all of which could affect BBB function to further exacerbate parenchymal injury. Future research is required to determine potential drugs that could prevent or attenuate neurovascular damage and/or augment repair. However, some studies have reported beneficial effects of hypothermia, erythropoietin, stem cell therapy, anti-cytokine therapy and metformin in ameliorating several different facets of damage to the NVU after HI-related brain injury in the perinatal period.
Collapse
Affiliation(s)
- Clémence DISDIER
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Barbara S STONESTREET
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI 02905, USA
| |
Collapse
|
26
|
Martini S, Austin T, Aceti A, Faldella G, Corvaglia L. Free radicals and neonatal encephalopathy: mechanisms of injury, biomarkers, and antioxidant treatment perspectives. Pediatr Res 2020; 87:823-833. [PMID: 31655487 DOI: 10.1038/s41390-019-0639-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/19/2019] [Accepted: 09/22/2019] [Indexed: 12/11/2022]
Abstract
Neonatal encephalopathy (NE), most commonly a result of the disruption of cerebral oxygen delivery, is the leading cause of neurologic disability in term neonates. Given the key role of free radicals in brain injury development following hypoxia-ischemia-reperfusion, several oxidative biomarkers have been explored in preclinical and clinical models of NE. Among these, antioxidant enzyme activity, uric acid excretion, nitric oxide, malondialdehyde, and non-protein-bound iron have shown promising results as possible predictors of NE severity and outcome. Owing to high costs and technical complexity, however, their routine use in clinical practice is still limited. Several strategies aimed at reducing free radical production or upregulating physiological scavengers have been proposed for NE. Room-air resuscitation has proved to reduce oxidative stress following perinatal asphyxia and is now universally adopted. A number of medications endowed with antioxidant properties, such as melatonin, erythropoietin, allopurinol, or N-acetylcysteine, have also shown potential neuroprotective effects in perinatal asphyxia; nevertheless, further evidence is needed before these antioxidant approaches could be implemented as standard care.
Collapse
Affiliation(s)
- Silvia Martini
- Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.
| | - Topun Austin
- Neonatal Intensive Care Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Arianna Aceti
- Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giacomo Faldella
- Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Luigi Corvaglia
- Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
27
|
Bell AH, Miller SL, Castillo-Melendez M, Malhotra A. The Neurovascular Unit: Effects of Brain Insults During the Perinatal Period. Front Neurosci 2020; 13:1452. [PMID: 32038147 PMCID: PMC6987380 DOI: 10.3389/fnins.2019.01452] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022] Open
Abstract
The neurovascular unit (NVU) is a relatively recent concept in neuroscience that broadly describes the relationship between brain cells and their blood vessels. The NVU incorporates cellular and extracellular components involved in regulating cerebral blood flow and blood-brain barrier function. The NVU within the adult brain has attracted strong research interest and its structure and function is well described, however, the NVU in the developing brain over the fetal and neonatal period remains much less well known. One area of particular interest in perinatal brain development is the impact of known neuropathological insults on the NVU. The aim of this review is to synthesize existing literature to describe structure and function of the NVU in the developing brain, with a particular emphasis on exploring the effects of perinatal insults. Accordingly, a brief overview of NVU components and function is provided, before discussion of NVU development and how this may be affected by perinatal pathologies. We have focused this discussion around three common perinatal insults: prematurity, acute hypoxia, and chronic hypoxia. A greater understanding of processes affecting the NVU in the perinatal period may enable application of targeted therapies, as well as providing a useful basis for research as it expands further into this area.
Collapse
Affiliation(s)
- Alexander H. Bell
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Suzanne L. Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
28
|
Shen G, Ma Q. MicroRNAs in the Blood-Brain Barrier in Hypoxic-Ischemic Brain Injury. Curr Neuropharmacol 2020; 18:1180-1186. [PMID: 32348227 PMCID: PMC7770646 DOI: 10.2174/1570159x18666200429004242] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/31/2020] [Accepted: 04/24/2020] [Indexed: 12/29/2022] Open
Abstract
Hypoxic-ischemic (HI) brain injury is a leading cause of acute mortality and chronic disability in newborns. Current evidence shows that cerebral microvascular response and compromised blood-brain barrier (BBB) integrity occur rapidly and could primarily be responsible for the brain injury observed in many infants with HI brain injury. MicroRNAs (miRNAs) are a type of highly conserved non-coding RNAs (ncRNAs), which consist of 21-25 nucleotides in length and usually lead to suppression of target gene expression. Growing evidence has revealed that brainenriched miRNAs act as versatile regulators of BBB dysfunctions in various neurological disorders including neonatal HI brain injury. In the present review, we summarize the current findings regarding the role of miRNAs in BBB impairment after hypoxia/ischemia brain injury. Specifically, we focus on the recent progress of miRNAs in the pathologies of neonatal HI brain injury. These findings can not only deepen our understanding of the role of miRNAs in BBB impairment in HI brain injury, but also provide insight into the development of new therapeutic strategies for preservation of BBB integrity under pathological conditions.
Collapse
Affiliation(s)
- Guofang Shen
- Department of Basic Sciences, The Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA92350, USA
| | - Qingyi Ma
- Department of Basic Sciences, The Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA92350, USA
| |
Collapse
|
29
|
Extracellular Vesicles as a Potential Therapy for Neonatal Conditions: State of the Art and Challenges in Clinical Translation. Pharmaceutics 2019; 11:pharmaceutics11080404. [PMID: 31405234 PMCID: PMC6723449 DOI: 10.3390/pharmaceutics11080404] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/27/2019] [Accepted: 07/31/2019] [Indexed: 12/15/2022] Open
Abstract
Despite advances in intensive care, several neonatal conditions typically due to prematurity affect vital organs and are associated with high mortality and long-term morbidities. Current treatment strategies for these babies are only partially successful or are effective only in selected patients. Regenerative medicine has been shown to be a promising option for these conditions at an experimental level, but still warrants further exploration for the development of optimal treatment. Although stem cell-based therapy has emerged as a treatment option, studies have shown that it is associated with potential risks and hazards, especially in the fragile population of babies. Recently, extracellular vesicles (EVs) have emerged as an attractive therapeutic alternative that holds great regenerative potential and is cell-free. EVs are nanosized particles endogenously produced by cells that mediate intercellular communication through the transfer of their cargo. Currently, EVs are garnering considerable attention as they are the key effectors of stem cell paracrine signaling and can epigenetically regulate target cell genes through the release of RNA species, such as microRNA. Herein, we review the emerging literature on the therapeutic potential of EVs derived from different sources for the treatment of neonatal conditions that affect the brain, retinas, spine, lungs, and intestines and discuss the challenges for the translation of EVs into clinical practice.
Collapse
|
30
|
Lan XB, Wang Q, Yang JM, Ma L, Zhang WJ, Zheng P, Sun T, Niu JG, Liu N, Yu JQ. Neuroprotective effect of Vanillin on hypoxic-ischemic brain damage in neonatal rats. Biomed Pharmacother 2019; 118:109196. [PMID: 31310955 DOI: 10.1016/j.biopha.2019.109196] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/25/2022] Open
Abstract
Neonatal hypoxic-ischemic brain damage (HIBD) is a leading cause of death and perpetual neurological dysfunction in neonates. Vanillin (Van), a natural phenolic compound with neuroprotective properties, exerts neuroprotection on a gerbil model of global ischemia by inhibiting oxidative damage. This study aimed to explore the potential neuroprotective roles of Van in neonatal rats suffering from hypoxic-ischemic (HI). An HI model of 7-day-old SD rats was induced by left carotid artery ligation followed by exposure to 8% oxygen (balanced with nitrogen) for 2.5 h at 37 °C. At 48 h after intraperitoneal injection with Van (20, 40, and 80 mg/kg) or saline, neurobehavioral function, cerebral infract volume, brain water content, and histomorphological changes were performed to evaluate brain injury. Transmission electron microscopy and immunoglobulin G (IgG) staining were conducted to evaluate the integrity of the blood-brain barrier (BBB). The levels of oxidative stress and tight junction proteins, as well as the activities of matrix metalloproteinases (MMPs), were also determined in the ipsilateral hemisphere. Results showed that Van post-treatment significantly ameliorated early neurobehavioral deficits, decreased infarct volume and brain edema, as well as attenuated histopathologic injury and IgG extravasation. Furthermore, Van markedly increased the activities of endogenous antioxidant enzymes and decreased malondialdehyde content. Meanwhile, the activation of MMP-2 and MMP-9 induced by HI was partially blocked by Van. Finally, Van obviously increased the expression of ZO-1, Occludin, and Claudin-5 compared with the HI group. Collectively, Van can provide neuroprotective effects against neonatal HIBD possibly by attenuating oxidative damage and preserving BBB integrity.
Collapse
Affiliation(s)
- Xiao-Bing Lan
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China
| | - Qing Wang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China
| | - Jia-Mei Yang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China
| | - Lin Ma
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China
| | - Wen-Jin Zhang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China
| | - Ping Zheng
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China
| | - Jian-Guo Niu
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China.
| | - Ning Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China.
| | - Jian-Qiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China; Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, People's Republic of China.
| |
Collapse
|
31
|
Goasdoue K, Chand KK, Miller SM, Lee KM, Colditz PB, Wixey JA, Bjorkman ST. Seizures Are Associated with Blood-Brain Barrier Disruption in a Piglet Model of Neonatal Hypoxic-Ischaemic Encephalopathy. Dev Neurosci 2019; 40:1-16. [PMID: 31048585 DOI: 10.1159/000499365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/04/2019] [Indexed: 11/19/2022] Open
Abstract
Seizures in the neonatal period are most often symptomatic of central nervous system (CNS) dysfunction and the most common cause is hypoxic-ischaemic encephalopathy (HIE). Seizures are associated with poor long-term outcomes and increased neuropathology. Blood-brain barrier (BBB) disruption and inflammation may contribute to seizures and increased neuropathology but are incompletely understood in neonatal HIE. The aim of this study was to investigate the impact of seizures on BBB integrity in a preclinical model of neonatal hypoxic-ischaemic (HI) injury. Piglets (age: <24 h) were subjected to a 30-min HI insult followed by recovery to 72 h post-insult. Amplitude-integrated electroencephalography (aEEG) was performed and seizure burden and background aEEG pattern were analysed. BBB disruption was evaluated in the parietal cortex and hippocampus by means of immunohistochemistry and Western blot. mRNA and protein expression of tight-junction proteins (zonula-occludens 1 [ZO1], occludin [OCLN], and claudin-5 [CLDN5]) was assessed using quantitative polymerase chain reaction (qPCR) and Western blot. In addition, mRNA from genes associated with BBB disruption vascular endothelial growth factor (VEGF) and matrix metalloproteinase 2 (MMP2) as well as inflammatory cytokines and chemokines was assessed with qPCR. Piglets that developed seizures following HI (HI-Sz) had significantly greater injury, as demonstrated by poorer aEEG background pattern scores, lower neurobehavioural scores, and greater histopathology. HI-Sz animals had severe IgG extravasation into brain tissue and uptake into neurons as well as significantly greater levels of IgG in both brain regions as assessed by Western blot. IgG protein in both brain regions was significantly associated with seizure burden, aEEG pattern scores, and neurobehavioural scores. There was no difference in mRNA expression of the tight junctions, however a significant loss of ZO1 and OCLN protein was observed in the parietal cortex. The inflammatory genes TGFβ, IL1β, IL8, IL6, and TNFα were significantly upregulated in HI-Sz animals. MMP2 was significantly increased in animals with seizures compared with animals without seizures. Increasing our understanding of neuropathology associated with seizure is vital because of the association between seizure and poor outcomes. Investigating the BBB is a major untapped area of research and a potential avenue for novel treatments.
Collapse
Affiliation(s)
- Kate Goasdoue
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Kirat Kishore Chand
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Stephanie Melita Miller
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Kah Meng Lee
- Institute of Health Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Queensland, Australia
| | - Paul Bernard Colditz
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Julie Anne Wixey
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Stella Tracey Bjorkman
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia,
| |
Collapse
|
32
|
Gussenhoven R, Klein L, Ophelders DRMG, Habets DHJ, Giebel B, Kramer BW, Schurgers LJ, Reutelingsperger CPM, Wolfs TGAM. Annexin A1 as Neuroprotective Determinant for Blood-Brain Barrier Integrity in Neonatal Hypoxic-Ischemic Encephalopathy. J Clin Med 2019; 8:jcm8020137. [PMID: 30682787 PMCID: PMC6406389 DOI: 10.3390/jcm8020137] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/08/2019] [Accepted: 01/20/2019] [Indexed: 12/13/2022] Open
Abstract
Blood-brain barrier (BBB) disruption is associated with hypoxia-ischemia (HI) induced brain injury and life-long neurological pathologies. Treatment options are limited. Recently, we found that mesenchymal stem/stromal cell derived extracellular vesicles (MSC-EVs) protected the brain in ovine fetuses exposed to HI. We hypothesized that Annexin A1 (ANXA1), present in MSC-EVs, contributed to their therapeutic potential by targeting the ANXA1/Formyl peptide receptor (FPR), thereby preventing loss of the BBB integrity. Cerebral ANXA1 expression and leakage of albumin into the fetal ovine brain parenchyma after HI were analyzed by immunohistochemistry. For mechanistic insights, barrier integrity of primary fetal endothelial cells was assessed after oxygen-glucose deprivation (OGD) followed by treatment with MSC-EVs or human recombinant ANXA1 in the presence or absence of FPR inhibitors. Our study revealed that BBB integrity was compromised after HI which was improved by MSC-EVs containing ANXA1. Treatment with these MSC-EVs or ANXA1 improved BBB integrity after OGD, an effect abolished by FPR inhibitors. Furthermore, endogenous ANXA1 was depleted within 24 h after induction of HI in cerebovasculature and ependyma and upregulated 72 h after HI in microglia. Targeting ANXA1/FPR with ANXA1 in the immature brain has great potential in preventing BBB loss and concomitant brain injury following HI.
Collapse
Affiliation(s)
- Ruth Gussenhoven
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands.
- School for Mental Health and Neuroscience (MHeNs), Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands.
| | - Luise Klein
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands.
- School for Mental Health and Neuroscience (MHeNs), Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands.
| | - Daan R M G Ophelders
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands.
- School of Oncology and Developmental Biology (GROW), Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands.
| | - Denise H J Habets
- School of Oncology and Developmental Biology (GROW), Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands.
- Department of Obstetrics and Gynecology, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands.
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany.
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands.
- School for Mental Health and Neuroscience (MHeNs), Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands.
- School of Oncology and Developmental Biology (GROW), Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands.
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands.
| | - Chris P M Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands.
| | - Tim G A M Wolfs
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands.
- School of Oncology and Developmental Biology (GROW), Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands.
- Department of Biomedical Engineering (BMT), School for Cardiovascular Diseases (CARIM), Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
33
|
Huun MU, Garberg HT, Buonocore G, Longini M, Belvisi E, Bazzini F, Proietti F, Saugstad OD, Solberg R. Regional differences of hypothermia on oxidative stress following hypoxia-ischemia: a study of DHA and hypothermia on brain lipid peroxidation in newborn piglets. J Perinat Med 2018; 47:82-89. [PMID: 30110254 DOI: 10.1515/jpm-2017-0355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 04/11/2018] [Indexed: 12/11/2022]
Abstract
Background Oxidative stress plays an important part in the pathophysiology of hypoxic-ischemic encephalopathy (HIE) and is reliably measured through prostanoids following lipid peroxidation of polyunsaturated fatty acids (PUFAs). The aim of the study is to measure oxidative stress in the prefrontal cortex, white matter and hippocampus in the brains of hypoxic-ischemic piglets treated with docosahexaenoic acid (DHA) and therapeutic hypothermia (TH) and investigate the additive effects of DHA on hypothermia by factorial design. Methods Fifty-five piglets were randomized as having severe global hypoxia (n=48) or not (sham, n=7). Hypoxic piglets were further randomized: vehicle (VEH), DHA, VEH+hypothermia (HT) or HT+DHA. A total of 5 mg/kg DHA was given intravenously 210 min after the end of hypoxia. Brain tissues were analyzed using liquid chromatography triple quadrupole mass spectrometry technique (LC-MS). A two-way analysis of variance (ANOVA) was performed with DHA and HT as main effects. Results In the white matter, we found main effects of DHA on DH-isoprostanes (P=0.030) and a main effect of HT on F4-neuroprostanes (F4-NeuroPs) (P=0.007), F2-isoprostanes (F2-IsoPs) (P=0.043) and DH-isoprostanes (P=0.023). In the cortex, the ANOVA analysis showed the interactions of main effects between DHA and HT for neurofuranes (NeuroFs) (P=0.092) and DH-isoprostanes (P=0.015) as DHA significantly reduced lipid peroxidation in the absence of HT. DHA compared to VEH significantly reduced NeuroFs (P=0.019) and DH-isoprostanes (P=0.010). No differences were found in the hippocampus. Conclusion After severe hypoxia, HT reduced lipid peroxidation in the white matter but not in the cortical gray matter. HT attenuated the reducing effect of DHA on lipid peroxidation in the cortex. Further studies are needed to determine whether DHA can be an effective add-on therapy for TH.
Collapse
Affiliation(s)
- Marianne U Huun
- Department of Pediatric Research, Institute of Surgical Research, University of Oslo, Oslo University Hospital Rikshospitalet, Postboks 4950 Nydalen, 0424 Oslo, Norway, Tel.: +47 97060117
| | - Håvard T Garberg
- Department of Pediatric Research, Institute of Surgical Research, University of Oslo, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Mariangela Longini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Elisa Belvisi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Francesco Bazzini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Fabrizio Proietti
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ola Didrik Saugstad
- Department of Pediatric Research, Institute of Surgical Research, University of Oslo, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Rønnaug Solberg
- Department of Pediatric Research, Institute of Surgical Research, University of Oslo, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Pediatrics, Vestfold Hospital Trust, Tønsberg, Norway
| |
Collapse
|
34
|
Liu J, Zhou S, Zhang Y, Li X, Qian X, Tao W, Jin L, Zhao J. Bax inhibitor-1 suppresses early brain injury following experimental subarachnoid hemorrhage in rats. Int J Mol Med 2018; 42:2891-2902. [PMID: 30226536 DOI: 10.3892/ijmm.2018.3858] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 09/04/2018] [Indexed: 11/05/2022] Open
Abstract
Early brain injury (EBI) following subarachnoid hemorrhage (SAH) is an important cause of high mortality and poor prognosis in SAH. B‑cell lymphoma 2‑associated X protein inhibitor‑1 (BI‑1) is an evolutionarily conserved antiapoptotic protein that is primarily located in the membranes of endoplasmic reticulum (ER). BI‑1 has been studied in certain nervous system‑associated diseases, but the role of this protein in SAH remains unclear. In the present study, the role of BI‑1 in EBI following SAH was investigated in rat models and its associated mechanisms were examined. The SAH rat model was generated by inserting nylon cords into the internal carotid artery from the external carotid artery. Samples were assessed using neurological scores, brain water content measurements, hematoxylin and eosin (H&E) staining, blood‑brain barrier (BBB) permeability, terminal deoxynucleotidyl transferase‑mediated dUTP nick‑end labeling and quantitative polymerase chain reaction assays, and western blot analyses. It was identified that the mRNA and protein levels of BI‑1 decreased markedly and were lowest at 24 h after SAH. BI‑1 overexpression and small hairpin RNA (shRNA)‑mediated silencing markedly suppressed or severely exacerbated EBI following SAH, respectively. BI‑1 overexpression in the SAH model improved neurological scores and decreased the brain water content, BBB permeability and levels of apoptosis compared with the control and sham groups following SAH. BI‑1 shRNA in the SAH model demonstrated contrary results. In addition, the mRNA or protein expression levels of ER stress‑associated genes (glucose regulated protein, 78 kDa, C/EBP homologous protein, Serine/threonine‑protein kinase/endoribonuclease IRE1, c‑Jun N terminal kinases and apoptotic signaling kinase‑1) were markedly suppressed or increased following BI‑1 overexpression and shRNA‑mediated silencing, respectively. The present study suggested that BI‑1 serves a neuroprotective role in EBI following SAH by attenuating BBB disruption, brain edema and apoptosis mediated by ER stress.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Environmental Science, Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650504, P.R. China
| | - Shuai Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Yueting Zhang
- Very Important Person Ward, The Second Affiliated Hospital of Kunming Medical College, Kunming, Yunnan 650032, P.R. China
| | - Xiuying Li
- Department of Pharmacology, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650504, P.R. China
| | - Xiying Qian
- Department of Neurosurgery, The First Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Weihua Tao
- Department of Neurosurgery, The First Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Lide Jin
- Department of Neurosurgery, The First Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Jianhua Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
35
|
Charriaut-Marlangue C, Baud O. A Model of Perinatal Ischemic Stroke in the Rat: 20 Years Already and What Lessons? Front Neurol 2018; 9:650. [PMID: 30131764 PMCID: PMC6090994 DOI: 10.3389/fneur.2018.00650] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/19/2018] [Indexed: 12/18/2022] Open
Abstract
Neonatal hypoxia-ischemia (HI) and ischemia are a common cause of neonatal brain injury resulting in cerebral palsy with subsequent learning disabilities and epilepsy. Recent data suggest a higher incidence of focal ischemia-reperfusion located in the middle cerebral artery (MCA) territory in near-term and newborn babies. Pre-clinical studies in the field of cerebral palsy research used, and still today, the classical HI model in the P7 rat originally described by Rice et al. (1). At the end of the 90s, we designed a new model of focal ischemia in the P7 rat to explore the short and long-term pathophysiology of neonatal arterial ischemic stroke, particularly the phenomenon of reperfusion injury and its sequelae (reported in 1998). Cerebral blood-flow and cell death/damage correlates have been fully characterized. Pharmacologic manipulations have been applied to the model to test therapeutic targets. The model has proven useful for the study of seizure occurrence, a clinical hallmark for neonatal ischemia in babies. Main pre-clinical findings obtained within these 20 last years are discussed associated to clinical pattern of neonatal brain damage.
Collapse
Affiliation(s)
| | - Olivier Baud
- INSERM U1141 PROTECT, Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France.,Division of Neonatology and Pediatric Intensive Care, Children's Hospital, Geneva University Hospitals (HUG), University of Geneva, Geneva, Switzerland
| |
Collapse
|
36
|
Neuroprotective Effects of Mitochondria-Targeted Plastoquinone in a Rat Model of Neonatal Hypoxic⁻Ischemic Brain Injury. Molecules 2018; 23:molecules23081871. [PMID: 30060443 PMCID: PMC6222533 DOI: 10.3390/molecules23081871] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/13/2018] [Accepted: 07/25/2018] [Indexed: 12/29/2022] Open
Abstract
Neonatal hypoxia⁻ischemia is one of the main causes of mortality and disability of newborns. To study the mechanisms of neonatal brain cell damage, we used a model of neonatal hypoxia⁻ischemia in seven-day-old rats, by annealing of the common carotid artery with subsequent hypoxia of 8% oxygen. We demonstrate that neonatal hypoxia⁻ischemia causes mitochondrial dysfunction associated with high production of reactive oxygen species, which leads to oxidative stress. Targeted delivery of antioxidants to the mitochondria can be an effective therapeutic approach to treat the deleterious effects of brain hypoxia⁻ischemia. We explored the neuroprotective properties of the mitochondria-targeted antioxidant SkQR1, which is the conjugate of a plant plastoquinone and a penetrating cation, rhodamine 19. Being introduced before or immediately after hypoxia⁻ischemia, SkQR1 affords neuroprotection as judged by the diminished brain damage and recovery of long-term neurological functions. Using vital sections of the brain, SkQR1 has been shown to reduce the development of oxidative stress. Thus, the mitochondrial-targeted antioxidant derived from plant plastoquinone can effectively protect the brain of newborns both in pre-ischemic and post-stroke conditions, making it a promising candidate for further clinical studies.
Collapse
|
37
|
Lespay-Rebolledo C, Perez-Lobos R, Tapia-Bustos A, Vio V, Morales P, Herrera-Marschitz M. Regionally Impaired Redox Homeostasis in the Brain of Rats Subjected to Global Perinatal Asphyxia: Sustained Effect up to 14 Postnatal Days. Neurotox Res 2018; 34:660-676. [PMID: 29959728 DOI: 10.1007/s12640-018-9928-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/30/2022]
Abstract
The present report evaluates the effect of global perinatal asphyxia on several parameters of oxidative stress and cell viability in rat brain tissue sampled at an extended neonatal period up to 14 days, a period characterised by intensive neuritogenesis, synaptogenesis, synaptic consolidation, pruning and delayed cell death. Perinatal asphyxia was induced by immersing foetus-containing uterine horns removed by a caesarean section from on term rat dams into a water bath at 37 °C for 21 min. Asphyxia-exposed and sibling caesarean-delivered foetuses were manually resucitated and nurtured by surrogate dams for 1 to 14 postnatal (P) days. Brain samples (mesencephalon, telencephalon and hippocampus) were assayed for glutathione (reduced and oxidated levels; spectrophotometry), tissue reducing capacity (potassium ferricyanide reducing assay, FRAP), catalase (the key enzyme protecting against oxidative stress and reactive oxygen species, Western blots and ELISA) and cleaved caspase-3 (the key executioner of apoptosis, Western blots) levels. It was found that global PA produced a regionally specific and sustained increase in GSSG/GSH ratio, a regionally specific decrease in tissue reducing capacity and a regionally and time specific decrease of catalase activity and increase of cleaved caspase-3 levels. The present study provides evidence for regionally impaired redox homeostasis in the brain of rats subjected to global PA, an effect observed up to P14, mainly affecting mesencephalon and hippocampus, suggesting a sustained oxidative stress after the posthypoxia period. The oxidative stress observed postnatally can in part be associated to a respiratory apneic-like deficit, since there was a statistically significant decrease in respiration frequency in AS compared to CS neonates, also up to P14, together with the signs of a decreased peripheral blood perfusion (pink-blue skin colour in AS, compared to the pink colour observed in all CS neonates). It is proposed that PA implies a long-term metabolic insult, triggered by the length of hypoxia, the resuscitation/reoxigenation manoevres, but also by the developmental stage of the affected brain regions, and the integrity of cardiovascular and respiratory physiological functions, which are fundamental for warrantying a proper development.
Collapse
Affiliation(s)
- Carolyne Lespay-Rebolledo
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Ronald Perez-Lobos
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Andrea Tapia-Bustos
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Valentina Vio
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Paola Morales
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
- Department Neuroscience, Medical Faculty, University of Chile, Santiago, Chile
| | - Mario Herrera-Marschitz
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile.
| |
Collapse
|
38
|
Mallard C, Ek CJ, Vexler ZS. The myth of the immature barrier systems in the developing brain: role in perinatal brain injury. J Physiol 2018. [PMID: 29528501 DOI: 10.1113/jp274938] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Central nervous system homeostasis is maintained by cellular barriers that protect the brain from external environmental changes and protect the CNS from harmful molecules and pathogens in the blood. Historically, for many years these barriers were thought of as immature, with limited functions, during brain development. In this review, we will present advances in the understanding of the barrier systems during development and evidence to show that in fact the barriers serve many important neurodevelopmental functions and that fetal and newborn brains are well protected. We will also discuss how ischaemic injury or systemic inflammation may breach the integrity of the barriers in the developing brain.
Collapse
Affiliation(s)
- Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - C Joakim Ek
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, USA
| |
Collapse
|
39
|
Effect of serotonin depletion on cortical spreading depression evoked cerebrovascular changes. ASIAN BIOMED 2018. [DOI: 10.2478/abm-2010-0095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract Background: The cortical spreading depression (CSD) is a phenomenon associated with several pathological conditions including migraine. It can induce alterations in both neural and vascular compartments. Serotonin (5-HT) depletion is known as a condition involved in migraine pathophysiology. The hyper-excitability of the cortical neurons to the CSD activation in the low 5-HT state has been previously reported. However, the cerebrovascular responses to CSD activation in this condition have never been studied yet. Objectives: Determine the effect of 5-HT depletion on the cerebrovascular responses to CSD activation. Methods: Wistar rats (weighing 250-300 grams) were divided into three groups: control, CSD, and low 5-HT with CSD group (five rats per group). To induce the low 5-HT state, the para-chlorophenylalanine was injected intraperitoneally into the rats three days before the experiment. CSD was induced by the application of solid KCl (3 mg) on the parietal cortex. NaCl instead of KCl was applied to the control group. Cerebral cortical blood flow was monitored using Laser Doppler flowmetry. The ultrastructure of cerebral microvessels was examined using electron microscopy to determine the cerebral microcirculatory responses to CSD. Results: Depletion of serotonin induced a significant increase in the peak amplitude of CSD-evoked cerebral hyperaemia. This condition also enhanced the development of CSD-induced endothelial pinocytosis and microvillus formation in cerebrocortical microvessels. Conclusion: 5-HT was an important neurotransmitter involved in the control of cerebrovascular responses to CSD activation. The hypersensitivity of the cerebrovascular responses observed in the 5-HT depleted state may explain the relationship between headache and 5-HT depletion.
Collapse
|
40
|
Liu F, Sun X, Zhang Y, Zhao G, Liu Y, Zhang Y. Curative effects of GM1 in the treatment of severe ischemic brain injury and its effects on serum TNF-α and NDS. Exp Ther Med 2018; 15:4851-4855. [PMID: 29805504 PMCID: PMC5952089 DOI: 10.3892/etm.2018.5995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 01/05/2018] [Indexed: 11/06/2022] Open
Abstract
The curative effects of monosialotetrahexosyl ganglioside (GM1) in the treatment of severe ischemic brain injury and its effects on tumor necrosis factor-α (TNF-α) and neuropathy disability score (NDS). Sixty patients with severe ischemic brain injury admitted to The First People's Hospital of Jining (Jining, China) from June 2014 to March 2016 were selected. They were randomly divided into the control group (n=30) and the experimental group (n=30). The patients in the control group were treated with routine therapy while those in the experimental group were treated with GM1. The level of TNF-α in the serum was measured by the enzyme-linked immunosorbent assay. The NDS was used to grade the two groups; Pearson's correlation coefficient was applied to analyze the correlation between the content of TNF-α and NDS; the content of superoxide dismutase (SOD) was detected using xanthine oxidase assay, and the content of malondialdehyde (MDA) was detected by thiobarbituric acid method. The clinical recovery time of two groups of patients was recorded. At 14 days after GM1 treatment, the serum TNF-α content and the NDS in the experimental group were significantly lower than those in the control group (P<0.05). The content of TNF-α in the patients was positively correlated with the NDS. After treatment, the serum MDA content of patients in the experimental group was lower, while the SOD content was significantly higher than that in the control group (P<0.05). After GM1 treatment, hemodynamic parameters of patients in the experimental group were significantly improved compared with those in the control group. The total effective rate of GM1 treatment in the experimental group was higher than that in the control group (P<0.05). GM1 has a good clinical significance in the treatment of patients with severe ischemic brain injury and is worthy of clinical promotion and application.
Collapse
Affiliation(s)
- Fang Liu
- Department of Neurosurgery Medicine, Yantai Yuhuangding Hospital of Shandong, Yantai, Shandong 264000, P.R. China
| | - Xiaoling Sun
- Department of Neurosurgery Medicine, Yantai Yuhuangding Hospital of Shandong, Yantai, Shandong 264000, P.R. China
| | - Yunxia Zhang
- Department of Neurosurgery Medicine, Yantai Yuhuangding Hospital of Shandong, Yantai, Shandong 264000, P.R. China
| | - Guoliang Zhao
- Department of Emergency Medicine, The First People's Hospital of Jining, Jining, Shandong 272011, P.R. China
| | - Yancang Liu
- Department of Emergency Medicine, The First People's Hospital of Jining, Jining, Shandong 272011, P.R. China
| | - Yiwu Zhang
- Department of Emergency Medicine, The First People's Hospital of Jining, Jining, Shandong 272011, P.R. China
| |
Collapse
|
41
|
Huun MU, Garberg HT, Escobar J, Chafer C, Vento M, Holme IM, Saugstad OD, Solberg R. DHA reduces oxidative stress following hypoxia-ischemia in newborn piglets: a study of lipid peroxidation products in urine and plasma. J Perinat Med 2018. [PMID: 28632497 DOI: 10.1515/jpm-2016-0334] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Lipid peroxidation mediated by reactive oxygen species is a major contributor to oxidative stress. Docosahexaenoic acid (DHA) has anti-oxidant and neuroprotective properties. Our objective was to assess how oxidative stress measured by lipid peroxidation was modified by DHA in a newborn piglet model of hypoxia-ischemia (HI). METHODS Fifty-five piglets were randomized to (i) hypoxia, (ii) DHA, (iii) hypothermia, (iv) hypothermia+DHA or (v) sham. All groups but sham were subjected to hypoxia by breathing 8% O2. DHA was administered 210 min after end of hypoxia and the piglets were euthanized 9.5 h after end of hypoxia. Urine and blood were harvested at these two time points and analyzed for F4-neuroprostanes, F2-isoprostanes, neurofuranes and isofuranes using UPLC-MS/MS. RESULTS F4-neuroprostanes in urine were significantly reduced (P=0.006) in groups receiving DHA. Hypoxia (median, IQR 1652 nM, 610-4557) vs. DHA (440 nM, 367-738, P=0.016) and hypothermia (median, IQR 1338 nM, 744-3085) vs. hypothermia+DHA (356 nM, 264-1180, P=0.006). The isoprostane compound 8-iso-PGF2α was significantly lower (P=0.011) in the DHA group compared to the hypoxia group. No significant differences were found between the groups in blood. CONCLUSION DHA significantly reduces oxidative stress by measures of lipid peroxidation following HI in both normothermic and hypothermic piglets.
Collapse
Affiliation(s)
- Marianne Ullestad Huun
- Department of Pediatric Research, Institute of Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway, Tel.: +47-23-07-27-90
| | - Håvard T Garberg
- Department of Pediatric Research, Institute of Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Javier Escobar
- Department of Pediatric Research, Institute of Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Consuelo Chafer
- Neonatal Research Unit, Health Research Institute Hospital La Fé, Valencia, Spain
| | - Maximo Vento
- Neonatal Research Unit, Health Research Institute Hospital La Fé, Valencia, Spain
| | - Ingar M Holme
- Department of Sports Medicine, Norwegian School of Sports Sciences, Oslo, Norway
| | - Ola Didrik Saugstad
- Department of Pediatric Research, Institute of Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Rønnaug Solberg
- Department of Pediatric Research, Institute of Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
42
|
El-Gamasy MA, Alarabawy R. Relation of Serum Creatinine to Sarnat Scoring and Brain Computerized Tomography of Neonates with Hypoxic Ischemic Encephalopathy. A Single-Center Experience. J Pediatr Neurosci 2018; 13:437-442. [PMID: 30937085 PMCID: PMC6413597 DOI: 10.4103/jpn.jpn_64_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background It is not easy to suspect whether newly born infants diagnosed as hypoxic ischemic encephalopathy (HIE) will develop impairment of renal function, so there is an urge to scientifically research about correlations of severity of HIE, which is represented by Sarnat scoring and brain computerized axial tomography (CAT) and serum creatinine level in these newly born infants. Aim To evaluate renal function in the form of serum creatinine levels in full-term neonates with HIE and their correlation with severity degree of HIE, which is represented by Sarnat scoring and CAT. Aim To evaluate renal function in the form of serum creatinine levels in full-term neonates with HIE and their correlation with severity degree of HIE, which is represented by Sarnat scoring and CAT. Subjects and Methods This study was a case-control type. It was conducted on 72 full-term neonates who were classified into group 1, which included 36 full-term neonates who were diagnosed as HIE according to the definition of the World Health Organization and group 2, which included 36 full-term neonates who were matched for age and sex and who served as the control group. Serum creatinine levels were measured at days 1 and 7 postnatally. CAT scans were carried out for cases only. Results Serum creatinine levels were elevated in group 1 when compared to the control group at days 1 and 7 postnatally. They were significantly correlated to the Sarnat scoring system of HIE, meaning that serum creatinine levels gradually increased with the increase in severity of HIE according to Sarnat and Sarnat staging. A statistically significant difference was observed between serum creatinine levels in patients with different findings of brain CAT, meaning that more elevation in serum creatinine levels were reported with more severe cases represented by marked changes in brain CAT. Conclusion Serum creatinine levels correlate with the severity of HIE of neonates according to Sarnat scoring and brain CAT.
Collapse
Affiliation(s)
- Mohamed A El-Gamasy
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Redha Alarabawy
- Department of Diagnostic Radiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
43
|
Dou Y, Shen H, Feng D, Li H, Tian X, Zhang J, Wang Z, Chen G. Tumor necrosis factor receptor-associated factor 6 participates in early brain injury after subarachnoid hemorrhage in rats through inhibiting autophagy and promoting oxidative stress. J Neurochem 2017; 142:478-492. [PMID: 28543180 DOI: 10.1111/jnc.14075] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/24/2017] [Accepted: 05/17/2017] [Indexed: 12/21/2022]
Abstract
Tumor necrosis factor receptor-associated factor 6 (TRAF6) is a member of the TRAF family and an important multifunctional intracellular adaptin of the tumor necrosis factor superfamily and toll/IL-1 receptor (TIR) superfamily. TRAF6 has been studied in several central nervous system diseases, including ischemic stroke, traumatic brain injury, and neurodegenerative diseases, but its role in subarachnoid hemorrhage (SAH) has not been fully illustrated. This study was designed to explore changes of expression level and potential roles and mechanisms of TRAF6 in early brain injury (EBI) after SAH using a Sprague-Dawley rat model of SAH induced in 0.3 mL non-heparinized autologous arterial blood injected into the pre-chiasmatic cistern. First, compared with the sham group, we found that the expression levels of TRAF6 increased gradually and peaked at 24 h after SAH. Second, the results showed that application of TRAF6 over-expression plasmid and genetic silencing siRNA could increase or decrease expression of TRAF6, respectively, and severely exacerbate or relieve EBI after SAH, including neuronal death, brain edema, and blood-brain barrier injury. Meanwhile, the levels of autophagy and oxidative stress were reduced and increased separately. Finally, GFP-TRAF6-C70A, which is a TRAF6 mutant that lacks E3 ubiquitin ligase activity, was used to explore the mechanism of TRAF6 in SAH, and the results showed that EBI and oxidative stress were reduced, but the levels of autophagy were increased under this condition. Collectively, these results indicated that TRAF6 affected the degree of EBI after SAH by inhibiting autophagy and promoting oxidative stress.
Collapse
Affiliation(s)
- Yang Dou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | | | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaodi Tian
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jian Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
44
|
Wang Z, Zhou F, Dou Y, Tian X, Liu C, Li H, Shen H, Chen G. Melatonin Alleviates Intracerebral Hemorrhage-Induced Secondary Brain Injury in Rats via Suppressing Apoptosis, Inflammation, Oxidative Stress, DNA Damage, and Mitochondria Injury. Transl Stroke Res 2017; 9:74-91. [PMID: 28766251 PMCID: PMC5750335 DOI: 10.1007/s12975-017-0559-x] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/20/2017] [Accepted: 07/23/2017] [Indexed: 12/12/2022]
Abstract
Intracerebral hemorrhage (ICH) is a cerebrovascular disease with high mortality and morbidity, and the effective treatment is still lacking. We designed this study to investigate the therapeutic effects and mechanisms of melatonin on the secondary brain injury (SBI) after ICH. An in vivo ICH model was induced via autologous whole blood injection into the right basal ganglia in Sprague-Dawley (SD) rats. Primary rat cortical neurons were treated with oxygen hemoglobin (OxyHb) as an in vitro ICH model. The results of the in vivo study showed that melatonin alleviated severe brain edema and behavior disorders induced by ICH. Indicators of blood-brain barrier (BBB) integrity, DNA damage, inflammation, oxidative stress, apoptosis, and mitochondria damage showed a significant increase after ICH, while melatonin reduced their levels. Meanwhile, melatonin promoted further increasing of expression levels of antioxidant indicators induced by ICH. Microscopically, TUNEL and Nissl staining showed that melatonin reduced the numbers of ICH-induced apoptotic cells. Inflammation and DNA damage indicators exhibited an identical pattern compared to those above. Additionally, the in vitro study demonstrated that melatonin reduced the apoptotic neurons induced by OxyHb and protected the mitochondrial membrane potential. Collectively, our investigation showed that melatonin ameliorated ICH-induced SBI by impacting apoptosis, inflammation, oxidative stress, DNA damage, brain edema, and BBB damage and reducing mitochondrial membrane permeability transition pore opening, and melatonin may be a potential therapeutic agent of ICH.
Collapse
Affiliation(s)
- Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Feng Zhou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Yang Dou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Xiaodi Tian
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Chenglin Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| |
Collapse
|
45
|
Lai JCY, Rocha-Ferreira E, Ek CJ, Wang X, Hagberg H, Mallard C. Immune responses in perinatal brain injury. Brain Behav Immun 2017; 63:210-223. [PMID: 27865947 DOI: 10.1016/j.bbi.2016.10.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/28/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
The perinatal period has often been described as immune deficient. However, it has become clear that immune responses in the neonate following exposure to microbes or as a result of tissue injury may be substantial and play a role in perinatal brain injury. In this article we will review the immune cell composition under normal physiological conditions in the perinatal period, both in the human and rodent. We will summarize evidence of the inflammatory responses to stimuli and discuss how neonatal immune activation, both in the central nervous system and in the periphery, may contribute to perinatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Jacqueline C Y Lai
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Eridan Rocha-Ferreira
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - C Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Xiaoyang Wang
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Henrik Hagberg
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden.
| |
Collapse
|
46
|
Ma Q, Dasgupta C, Li Y, Huang L, Zhang L. MicroRNA-210 Suppresses Junction Proteins and Disrupts Blood-Brain Barrier Integrity in Neonatal Rat Hypoxic-Ischemic Brain Injury. Int J Mol Sci 2017; 18:ijms18071356. [PMID: 28672801 PMCID: PMC5535849 DOI: 10.3390/ijms18071356] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 12/16/2022] Open
Abstract
Cerebral edema, primarily caused by disruption of the blood-brain barrier (BBB), is one of the serious complications associated with brain injury in neonatal hypoxic-ischemic encephalopathy (HIE). Our recent study demonstrated that the hypoxic-ischemic (HI) treatment significantly increased microRNA-210 (miR-210) in the neonatal rat brain and inhibition of miR-210 provided neuroprotection in neonatal HI brain injury. The present study aims to determine the role of miR-210 in the regulation of BBB integrity in the developing brain. miR-210 mimic was administered via intracerebroventricular injection (i.c.v.) into the brain of rat pups. Forty-eight hours after the injection, a modified Rice-Vannucci model was conducted to produce HI brain injury. Post-assays included cerebral edema analysis, western blotting, and immunofluorescence staining for serum immunoglobulin G (IgG) leakage. The results showed that miR-210 mimic exacerbated cerebral edema and IgG leakage into the brain parenchyma. In contrast, inhibition of miR-210 with its complementary locked nucleic acid oligonucleotides (miR-210-LNA) significantly reduced cerebral edema and IgG leakage. These findings suggest that miR-210 negatively regulates BBB integrity i n the neonatal brain. Mechanistically, the seed sequences of miR-210 were identified complementary to the 3' untranslated region (3' UTR) of the mRNA transcripts of tight junction protein occludin and adherens junction protein β-catenin, indicating downstream targets of miR-210. This was further validated by in vivo data showing that miR-210 mimic significantly reduced the expression of these junction proteins in rat pup brains. Of importance, miR-210-LNA preserved the expression of junction proteins occludin and β-catenin from neonatal HI insult. Altogether, the present study reveals a novel mechanism of miR-210 in impairing BBB integrity that contributes to cerebral edema formation after neonatal HI insult, and provides new insights in miR-210-LNA mediated neuroprotection in neonatal HI brain injury.
Collapse
Affiliation(s)
- Qingyi Ma
- Center for Neonatal Biology, Division of Pharmacology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Chiranjib Dasgupta
- Center for Neonatal Biology, Division of Pharmacology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Yong Li
- Center for Neonatal Biology, Division of Pharmacology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Lei Huang
- Center for Neonatal Biology, Division of Pharmacology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Lubo Zhang
- Center for Neonatal Biology, Division of Pharmacology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA.
| |
Collapse
|
47
|
Rat umbilical cord blood cells attenuate hypoxic-ischemic brain injury in neonatal rats. Sci Rep 2017; 7:44111. [PMID: 28281676 PMCID: PMC5345001 DOI: 10.1038/srep44111] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 02/03/2017] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence has suggested that human umbilical cord blood cells (hUCBC) have a favorable effect on hypoxic-ischemic (HI) brain injury. However, the efficacy of using hUCBCs to treat this injury has been variable and the underlying mechanism remains elusive. Here, we investigated its effectiveness using stereological analysis in an allogeneic system to examine whether intraperitoneal injection of cells derived from UCBCs of green fluorescent protein (GFP)-transgenic rats could ameliorate brain injury in neonatal rats. Three weeks after the HI event, the estimated residual brain volume was larger and motor function improved more in the cell-injected rats than in the control (PBS-treated) rats. The GFP-positive cells were hardly detectable in the brain (0.0057% of injected cells) 9 days after injection. Although 60% of GFP-positive cells in the brain were Iba1-positive, none of these were positive for NeuroD or DCX. While the number of proliferating cells increased in the hippocampus, that of activated microglia/macrophages decreased and a proportion of M2 microglia/macrophages increased in the ipsilateral hemisphere of cell-injected rats. These results suggest that intraperitoneal injection of cells derived from UCBCs could ameliorate HI injury, possibly through an endogenous response and not by supplying differentiated neurons derived from the injected stem cells.
Collapse
|
48
|
Pfisterer U, Khodosevich K. Neuronal survival in the brain: neuron type-specific mechanisms. Cell Death Dis 2017; 8:e2643. [PMID: 28252642 PMCID: PMC5386560 DOI: 10.1038/cddis.2017.64] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/24/2017] [Accepted: 01/31/2017] [Indexed: 12/19/2022]
Abstract
Neurogenic regions of mammalian brain produce many more neurons that will eventually survive and reach a mature stage. Developmental cell death affects both embryonically produced immature neurons and those immature neurons that are generated in regions of adult neurogenesis. Removal of substantial numbers of neurons that are not yet completely integrated into the local circuits helps to ensure that maturation and homeostatic function of neuronal networks in the brain proceed correctly. External signals from brain microenvironment together with intrinsic signaling pathways determine whether a particular neuron will die. To accommodate this signaling, immature neurons in the brain express a number of transmembrane factors as well as intracellular signaling molecules that will regulate the cell survival/death decision, and many of these factors cease being expressed upon neuronal maturation. Furthermore, pro-survival factors and intracellular responses depend on the type of neuron and region of the brain. Thus, in addition to some common neuronal pro-survival signaling, different types of neurons possess a variety of 'neuron type-specific' pro-survival constituents that might help them to adapt for survival in a certain brain region. This review focuses on how immature neurons survive during normal and impaired brain development, both in the embryonic/neonatal brain and in brain regions associated with adult neurogenesis, and emphasizes neuron type-specific mechanisms that help to survive for various types of immature neurons. Importantly, we mainly focus on in vivo data to describe neuronal survival specifically in the brain, without extrapolating data obtained in the PNS or spinal cord, and thus emphasize the influence of the complex brain environment on neuronal survival during development.
Collapse
Affiliation(s)
- Ulrich Pfisterer
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
49
|
Arteaga O, Álvarez A, Revuelta M, Santaolalla F, Urtasun A, Hilario E. Role of Antioxidants in Neonatal Hypoxic-Ischemic Brain Injury: New Therapeutic Approaches. Int J Mol Sci 2017; 18:E265. [PMID: 28134843 PMCID: PMC5343801 DOI: 10.3390/ijms18020265] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 01/14/2017] [Accepted: 01/19/2017] [Indexed: 01/08/2023] Open
Abstract
Hypoxic-ischemic brain damage is an alarming health and economic problem in spite of the advances in neonatal care. It can cause mortality or detrimental neurological disorders such as cerebral palsy, motor impairment and cognitive deficits in neonates. When hypoxia-ischemia occurs, a multi-faceted cascade of events starts out, which can eventually cause cell death. Lower levels of oxygen due to reduced blood supply increase the production of reactive oxygen species, which leads to oxidative stress, a higher concentration of free cytosolic calcium and impaired mitochondrial function, triggering the activation of apoptotic pathways, DNA fragmentation and cell death. The high incidence of this type of lesion in newborns can be partly attributed to the fact that the developing brain is particularly vulnerable to oxidative stress. Since antioxidants can safely interact with free radicals and terminate that chain reaction before vital molecules are damaged, exogenous antioxidant therapy may have the potential to diminish cellular damage caused by hypoxia-ischemia. In this review, we focus on the neuroprotective effects of antioxidant treatments against perinatal hypoxic-ischemic brain injury, in the light of the most recent advances.
Collapse
Affiliation(s)
- Olatz Arteaga
- Department of Cell Biology & Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
| | - Antonia Álvarez
- Department of Cell Biology & Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
| | - Miren Revuelta
- Department of Cell Biology & Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
| | - Francisco Santaolalla
- Department of Otorhinolaryngology, Basurto University Hospital, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
| | - Andoni Urtasun
- Department of Neuroscience, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
- Neurogenomiks Laboratory, Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, 48170 Zamudio, Spain.
| | - Enrique Hilario
- Department of Cell Biology & Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
| |
Collapse
|
50
|
Najjar S, Pahlajani S, De Sanctis V, Stern JNH, Najjar A, Chong D. Neurovascular Unit Dysfunction and Blood-Brain Barrier Hyperpermeability Contribute to Schizophrenia Neurobiology: A Theoretical Integration of Clinical and Experimental Evidence. Front Psychiatry 2017; 8:83. [PMID: 28588507 PMCID: PMC5440518 DOI: 10.3389/fpsyt.2017.00083] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 04/28/2017] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia is a psychotic disorder characterized by delusions, hallucinations, negative symptoms, as well as behavioral and cognitive dysfunction. It is a pathoetiologically heterogeneous disorder involving complex interrelated mechanisms that include oxidative stress and neuroinflammation. Neurovascular endothelial dysfunction and blood-brain barrier (BBB) hyperpermeability are established mechanisms in neurological disorders with comorbid psychiatric symptoms such as epilepsy, traumatic brain injury, and Alzheimer's disease. Schizophrenia is frequently comorbid with medical conditions associated with peripheral vascular endothelial dysfunction, such as metabolic syndrome, cardiovascular disease, and diabetes mellitus. However, the existence and etiological relevance of neurovascular endothelial dysfunction and BBB hyperpermeability in schizophrenia are still not well recognized. Here, we review the growing clinical and experimental evidence, indicating that neurovascular endotheliopathy and BBB hyperpermeability occur in schizophrenia patients. We present a theoretical integration of human and animal data linking oxidative stress and neuroinflammation to neurovascular endotheliopathy and BBB breakdown in schizophrenia. These abnormalities may contribute to the cognitive and behavioral symptoms of schizophrenia via several mechanisms involving reduced cerebral perfusion and impaired homeostatic processes of cerebral microenvironment. Furthermore, BBB disruption can facilitate interactions between brain innate and peripheral adaptive immunity, thereby perpetuating harmful neuroimmune signals and toxic neuroinflammatory responses, which can also contribute to the symptoms of schizophrenia. Taken together, these findings support the "mild encephalitis" hypothesis of schizophrenia. If neurovascular abnormalities prove to be etiologically relevant to the neurobiology of schizophrenia, then targeting these abnormalities may represent a promising therapeutic strategy.
Collapse
Affiliation(s)
- Souhel Najjar
- Department of Neurology, Hofstra Northwell School of Medicine, New York, NY, USA.,Neuroinflammation Division, Department of Neurology, Lenox Hill Hospital, New York, NY, USA
| | - Silky Pahlajani
- Neuroinflammation Division, Department of Neurology, Lenox Hill Hospital, New York, NY, USA
| | - Virginia De Sanctis
- Neuroinflammation Division, Department of Neurology, Lenox Hill Hospital, New York, NY, USA
| | - Joel N H Stern
- Department of Neurology, Hofstra Northwell School of Medicine, New York, NY, USA.,Neuroinflammation Division, Department of Neurology, Lenox Hill Hospital, New York, NY, USA
| | - Amanda Najjar
- Department of Psychology and Human Development, Peabody College, Vanderbilt University, Nashville, TN, USA
| | - Derek Chong
- Department of Neurology, Hofstra Northwell School of Medicine, New York, NY, USA
| |
Collapse
|