1
|
Meer E. Role of Noncoding RNAs in Modulating Microglial Phenotype. Glob Med Genet 2024; 11:304-311. [PMID: 39258255 PMCID: PMC11383642 DOI: 10.1055/s-0044-1790283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024] Open
Abstract
Microglia are immunocompetent cells that are present in the retina and central nervous system, and are involved in the development maintenance and immune functions in these systems. Developing from yolk sac-primitive macrophages, they proliferate in the local tissues during the embryonic period without resorting to the production from the hematopoietic stem cells, and are critical in sustaining homeostasis and performing in disease and injury; they have morphological characteristics and distinct phenotypes according to the microenvironment. Microglia are also present in close association with resident cells in the retina where they engage in synapse formation, support normal functions, as well as immune defense. They are involved in the development of numerous neurodegenerative and ophthalmic diseases and act as diversity shields and triggers. Noncoding ribonucleic acids (ncRNAs) refer to RNA molecules synthesized from the mammalian genome, and these do not have protein-coding capacity. These ncRNAs play a role in the regulation of gene expression patterns. ncRNAs have only been recently identified as vastly significant molecules that are involved in the posttranscriptional regulation. Microglia are crucial for brain health and functions and current studies have focused on the effects caused by ncRNA on microglial types. Thus, the aim of the review was to provide an overview of the current knowledge about the regulation of microglial phenotypes by ncRNAs.
Collapse
Affiliation(s)
- Eiman Meer
- Department of Biological and Health Sciences, Pak-Austria Fachhochschule Institute of Applied Sciences and Technology, Haripur, Pakistan
| |
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 2024:10.1007/s12035-024-04594-7. [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] [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
|
Gai C, Li T, Zhao Y, Cheng Y, Song Y, Luo Q, Liu D, Wang Z. Mesenchymal stromal cells deliver H 2S-enhanced Nrf2 via extracellular vesicles to mediate mitochondrial homeostasis for repairing hypoxia-ischemia brain damage. Free Radic Biol Med 2024; 225:528-545. [PMID: 39427747 DOI: 10.1016/j.freeradbiomed.2024.10.292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Mesenchymal stromal cells (MSCs) are considered a therapeutic approach for neurological diseases via extracellular vesicles (EVs). Modified EVs contain active components with enhanced therapeutic potential. In this study, we aimed to explore the role and underlying mechanism of EVs from MSCs preconditioned by NaHS (an Hydrogen sulfide donor) (H2S-EVs) in hypoxia-ischemia (HI) brain damage. Our results showed that H2S-EVs treatment via the non-invasive intranasal route in HI mice was able to reduce oxidative stress and mitochondrial dysfunction compared to EVs treatment. Mechanistic studies demonstrated that NaHS promoted nuclear factor erythroid-2 related factor 2 (Nrf2) expression in the cytoplasm by inducing Parkinson disease protein 7 (PARK7)-dependent disintegration of Nrf2/Keap-1 complex in MSCs. In particular, the free Nrf2 was loaded into the EVs as a result of its KFERQ motif being recognized by 70-kDa heat shock proteins and lysosomal-associated membrane protein 2A. Subsequently, H2S-EVs were internalized into neurons in the ipsilateral hemisphere, thus delivering abundant Nrf2 to accumulate in the mitochondria and remodeling mitochondrial function following H2S-EVs treatment in HI mice. Moreover, Nrf2 knockdown in MSCs remarkably impaired H2S-EVs-mediated therapeutic effects on HI mice. In brief, the present study for the first time demonstrated that H2S-modified MSCs significantly accumulated higher Nrf2 in EVs via upregulating PARK7 expression, revealing the mechanism through which antioxidant protein Nrf2 delivered by H2S-EVs protect against mitochondrial dysfunction in HI brain damage.
Collapse
Affiliation(s)
- Chengcheng Gai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Tingting Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yijing Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yahong Cheng
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yan Song
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Qian Luo
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
4
|
De Rose DU, Maddaloni C, Ronci S, Bersani I, Martini L, Caoci S, Savarese I, Di Pede A, Campi F, Di Felice G, Berti P, Porzio O, Luciani M, Dotta A. Coagulation profiles and percentiles in neonates with hypoxic-ischemic encephalopathy undergoing therapeutic hypothermia: A step toward more accurate transfusion thresholds. Pediatr Blood Cancer 2024; 71:e31193. [PMID: 39022989 DOI: 10.1002/pbc.31193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND In the literature, there are no studies about the transfusion threshold for neonates with hypoxic-ischemic encephalopathy (HIE) undergoing therapeutic hypothermia (TH). In order to facilitate accurate interpretation of coagulation results in these neonates, we aimed to generate specific reference intervals in this specific population. METHODS This retrospective study included all HIE neonates admitted from 2014 to 2022 to undergo TH. All infants during TH underwent blood exams, including the coagulation profile. Our primary outcome was to assess the estimates of the 3rd, 10th, 25th, 50th, 75th, 90th, and 97th percentiles for each parameter on admission (before transfusion). By the receiver operating characteristic (ROC) analysis, the area under the ROC curve (AUC) and the best cut-off point were used to evaluate the ability of the prothrombin time expressed as international normalized ratio (PT-INR) to predict the risk of any bleeding. RESULTS A total of 143 infants were included in this study. On admission, the median fibrinogen value was 205 mg/dL, prothrombin time 18.6 seconds, PT-INR 1.50, activated partial thromboplastin time 38.3 seconds, thrombin time 18.6 seconds, antithrombin 57.0%. The optimal cut-off of PT-INR in predicting the risk of any bleeding was greater than 1.84 (AUC .623, p = .024). CONCLUSION For the first time, we proposed the percentiles of coagulation parameters in our cohort of neonates with HIE. Furthermore, we found that a PT-INR greater than 1.84 can significantly predict the risk of any bleeding. Further studies are needed to determine if a restrictive versus a liberal transfusion approach can be equally safer for these high-risk infants.
Collapse
Affiliation(s)
| | - Chiara Maddaloni
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Sara Ronci
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Iliana Bersani
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Ludovica Martini
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Stefano Caoci
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Immacolata Savarese
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Alessandra Di Pede
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Francesca Campi
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Giovina Di Felice
- Clinical Biochemistry Laboratory, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Pierpaolo Berti
- Transfusion Medicine Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| | - Ottavia Porzio
- Clinical Biochemistry Laboratory, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
- Transfusion Medicine Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
- Department of Experimental Medicine, "Tor Vergata" University, Rome, Italy
| | - Matteo Luciani
- Hematology and Oncology Unit, "Bambino Gesù" Children Hospital IRCSS, Rome, Italy
| | - Andrea Dotta
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, Rome, Italy
| |
Collapse
|
5
|
Babbo CCR, Mellet J, van Rensburg J, Pillay S, Horn AR, Nakwa FL, Velaphi SC, Kali GTJ, Coetzee M, Masemola MYK, Ballot DE, Pepper MS. Neonatal encephalopathy due to suspected hypoxic ischemic encephalopathy: pathophysiology, current, and emerging treatments. World J Pediatr 2024:10.1007/s12519-024-00836-9. [PMID: 39237728 DOI: 10.1007/s12519-024-00836-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/31/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Neonatal encephalopathy (NE) due to suspected hypoxic-ischemic encephalopathy (HIE), referred to as NESHIE, is a clinical diagnosis in late preterm and term newborns. It occurs as a result of impaired cerebral blood flow and oxygen delivery during the peripartum period and is used until other causes of NE have been discounted and HIE is confirmed. Therapeutic hypothermia (TH) is the only evidence-based and clinically approved treatment modality for HIE. However, the limited efficacy and uncertain benefits of TH in some low- to middle-income countries (LMICs) and the associated need for intensive monitoring have prompted investigations into more accessible and effective stand-alone or additive treatment options. DATA SOURCES This review describes the rationale and current evidence for alternative treatments in the context of the pathophysiology of HIE based on literatures from Pubmed and other online sources of published data. RESULTS The underlining mechanisms of neurotoxic effect, current clinically approved treatment, various categories of emerging treatments and clinical trials for NE are summarized in this review. Melatonin, caffeine citrate, autologous cord blood stem cells, Epoetin alfa and Allopurinal are being tested as potential neuroprotective agents currently. CONCLUSION This review describes the rationale and current evidence for alternative treatments in the context of the pathophysiology of HIE. Neuroprotective agents are currently only being investigated in high- and middle-income settings. Results from these trials will need to be interpreted and validated in LMIC settings. The focus of future research should therefore be on the development of inexpensive, accessible monotherapies and should include LMICs, where the highest burden of NESHIE exists.
Collapse
Affiliation(s)
- Carina Corte-Real Babbo
- SAMRC Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, University of Pretoria, Room 5-64, Level 5, Pathology Building, 15 Bophelo Road (Cnr. Steve Biko and Dr. Savage Streets), Prinshof Campus, Gezina, Pretoria, South Africa
| | - Juanita Mellet
- SAMRC Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, University of Pretoria, Room 5-64, Level 5, Pathology Building, 15 Bophelo Road (Cnr. Steve Biko and Dr. Savage Streets), Prinshof Campus, Gezina, Pretoria, South Africa
| | - Jeanne van Rensburg
- SAMRC Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, University of Pretoria, Room 5-64, Level 5, Pathology Building, 15 Bophelo Road (Cnr. Steve Biko and Dr. Savage Streets), Prinshof Campus, Gezina, Pretoria, South Africa
| | - Shakti Pillay
- Department of Paediatrics and Child Health, Division of Neonatology, Groote Schuur Hospital, University of Cape Town, Neonatal Unit, Cape Town, South Africa
| | - Alan Richard Horn
- Department of Paediatrics and Child Health, Division of Neonatology, Groote Schuur Hospital, University of Cape Town, Neonatal Unit, Cape Town, South Africa
| | - Firdose Lambey Nakwa
- Department of Paediatrics and Child Health, Faculty of Health Sciences, Chris Hani Baragwanath Academic Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | - Sithembiso Christopher Velaphi
- Department of Paediatrics and Child Health, Faculty of Health Sciences, Chris Hani Baragwanath Academic Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Melantha Coetzee
- Department of Paediatrics and Child Health, Division of Neonatology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Mogomane Yvonne Khomotso Masemola
- Department of Paediatrics and Child Health, Faculty of Health Sciences, Kalafong Hospital, University of Pretoria, Pretoria, South Africa
| | - Daynia Elizabeth Ballot
- Department of Paediatrics and Child Health, Faculty of Health Sciences, Charlotte Maxeke Johannesburg Academic Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | - Michael Sean Pepper
- SAMRC Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, University of Pretoria, Room 5-64, Level 5, Pathology Building, 15 Bophelo Road (Cnr. Steve Biko and Dr. Savage Streets), Prinshof Campus, Gezina, Pretoria, South Africa.
| |
Collapse
|
6
|
Reyes-Corral M, Gil-González L, González-Díaz Á, Tovar-Luzón J, Ayuso MI, Lao-Pérez M, Montaner J, de la Puerta R, Fernández-Torres R, Ybot-González P. Pretreatment with oleuropein protects the neonatal brain from hypoxia-ischemia by inhibiting apoptosis and neuroinflammation. J Cereb Blood Flow Metab 2024:271678X241270237. [PMID: 39157939 DOI: 10.1177/0271678x241270237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Hypoxic-ischemic (HI) encephalopathy is a cerebrovascular injury caused by oxygen deprivation to the brain and remains a major cause of neonatal mortality and morbidity worldwide. Therapeutic hypothermia is the current standard of care but it does not provide complete neuroprotection. Our aim was to investigate the neuroprotective effect of oleuropein (Ole) in a neonatal (seven-day-old) mouse model of HI. Ole, a secoiridoid found in olive leaves, has previously shown to reduce damage against cerebral and other ischemia/reperfusion injuries. Here, we administered Ole as a pretreatment prior to HI induction at 20 or 100 mg/kg. A week after HI, Ole significantly reduced the infarct area and the histological damage as well as white matter injury, by preserving myelination, microglial activation and the astroglial reactive response. Twenty-four hours after HI, Ole reduced the overexpression of caspase-3 and the proinflammatory cytokines IL-6 and TNF-α. Moreover, using UPLC-MS/MS we found that maternal supplementation with Ole during pregnancy and/or lactation led to the accumulation of its metabolite hydroxytyrosol in the brains of the offspring. Overall, our results indicate that pretreatment with Ole confers neuroprotection and can prevent HI-induced brain damage by modulating apoptosis and neuroinflammation.
Collapse
Affiliation(s)
- Marta Reyes-Corral
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
| | - Laura Gil-González
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
| | - Ángela González-Díaz
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
| | - Javier Tovar-Luzón
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
| | - María Irene Ayuso
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
- CIBERSAM, ISCIII (Spanish Network for Research in Mental Health), Seville, Spain
| | - Miguel Lao-Pérez
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
| | - Joan Montaner
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
- Department of Neurology, Virgen Macarena University Hospital, Seville, Spain
| | - Rocío de la Puerta
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Rut Fernández-Torres
- Departamento de Química Analítica, Facultad de Química, Universidad de Sevilla, Seville, Spain
| | - Patricia Ybot-González
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
- Spanish National Research Council (CSIC), Spain
| |
Collapse
|
7
|
Aycan N, Demir DÇ, Yürektürk E, Başaranoğlu M, Karaman S, Tuncer O. Oxidative and Antioxidative Biomarker Profiles in Neonatal Hypoxic-Ischemic Encephalopathy: Insights for Pathophysiology and Treatment Strategies. Med Sci Monit 2024; 30:e945045. [PMID: 39152631 PMCID: PMC11337971 DOI: 10.12659/msm.945045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 07/02/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Neonatal hypoxic-ischemic encephalopathy (HIE) is a significant cause of perinatal and postnatal morbidity and mortality worldwide. Catalase (CAT) activity detection is used to determine levels of inflammation and oxidative stress. Glutathione (GSH) is the most critical non-enzymatic endogenous antioxidant. Lipid peroxidation levels marked after hypoxia can be detected based on the level of malondialdehyde (MDA). Ischemia-modified albumin (IMA) is considered a biomarker for cardiac ischemia and is known to increase in the liver, brain, and kidney in states of insufficient oxygenation. We aimed to explain the results and relations between the oxidant and antioxidants to detail oxidant-antioxidant balance and cellular mechanisms. MATERIAL AND METHODS Serum levels of IMA and MDA, as an oxidative stress marker, and CAT and GSH, as antioxidant enzymes, were measured in first blood samples of 59 neonates diagnosed with HIE, with pH <7, base excess >12, and APGAR scores. RESULTS Neonates who were ≥37 weeks of gestation and had hypoxia were included. Compared with healthy newborns (n=32), CAT was statistically significantly lower in the hypoxia group (P=0.0001), while MDA serum levels were significantly higher in neonates with hypoxia (P=0.01). There was no difference between hypoxic and healthy neonates in GSH and IMA measurements (P=0.054, P=0.19 respectively). CONCLUSIONS HIE pathophysiology involves oxidative stress and mitochondrial energy production failure. Explaining the pathways between oxidant-antioxidant balance and cell death, which explains the pathophysiology of HIE, is essential to develop treatment strategies that will minimize the effects of oxygen deprivation on other body organs, especially the brain.
Collapse
Affiliation(s)
- Nur Aycan
- Department of Pediatrics, Yuzuncu Yil University, Van, Türkiye
| | - Derya Çay Demir
- Department of Chemistry, Yuzuncu Yil University, Van, Türkiye
| | - Eyyüp Yürektürk
- Department of Neonatology, Yuzuncu Yil University, Van, Türkiye
| | | | - Serap Karaman
- Department of Neonatology, Yuzuncu Yil University, Van, Türkiye
| | - Oğuz Tuncer
- Department of Neonatology, Yuzuncu Yil University, Van, Türkiye
| |
Collapse
|
8
|
Yang M, Wang K, Liu B, Shen Y, Liu G. Hypoxic-Ischemic Encephalopathy: Pathogenesis and Promising Therapies. Mol Neurobiol 2024:10.1007/s12035-024-04398-9. [PMID: 39073530 DOI: 10.1007/s12035-024-04398-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a brain lesion caused by inadequate blood supply and oxygen deprivation, often occurring in neonates. It has emerged as a grave complication of neonatal asphyxia, leading to chronic neurological damage. Nevertheless, the precise pathophysiological mechanisms underlying HIE are not entirely understood. This paper aims to comprehensively elucidate the contributions of hypoxia-ischemia, reperfusion injury, inflammation, oxidative stress, mitochondrial dysfunction, excitotoxicity, ferroptosis, endoplasmic reticulum stress, and apoptosis to the onset and progression of HIE. Currently, hypothermia therapy stands as the sole standard treatment for neonatal HIE, albeit providing only partial neuroprotection. Drug therapy and stem cell therapy have been explored in the treatment of HIE, exhibiting certain neuroprotective effects. Employing drug therapy or stem cell therapy as adjunctive treatments to hypothermia therapy holds great significance. This article presents a systematic review of the pathogenesis and treatment strategies of HIE, with the goal of enhancing the effect of treatment and improving the quality of life for HIE patients.
Collapse
Affiliation(s)
- Mingming Yang
- Department of Pediatrics, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, P. R. China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China.
| | - Guangliang Liu
- Department of Pediatrics, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, P. R. China.
| |
Collapse
|
9
|
Tayman C, Çakır U, Kurt A, Ertekin Ö, Taskin Turkmenoglu T, Çağlayan M, Işık E. Evaluation of beneficial effects of dexpanthenol on hypoxic-ischemic encephalopathy. Biotech Histochem 2024:1-9. [PMID: 38869860 DOI: 10.1080/10520295.2024.2365231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a cause of serious morbidity and mortality in newborns. Dexpanthenol, which is metabolized into D-pantothenic acid, has antioxidant and other potentially therapeutic properties. We examined some effects of dexpanthenol on the brains of week-old rat pups with HIE induced by obstruction of the right carotid artery followed by keeping in 8% O2 for 2 hours. Dexpanthenol (500 mg/kg) was administered intraperitoneally to 16 of 32 pups with HIE. Protein, DNA, and lipid oxidation degradation products were assayed and hippocampal and cortical cell apoptosis and neuronal cell numbers were evaluated in stained sections. Dexpanthenol application reduced oxidative stress and inflammation. TNF-α and IL-6 cytokine levels in HIE also decreased with dexpanthenol treatment. The numbers of caspase-3 positive cells in the dentate gyrus and CA1/CA2/CA3 regions of the hippocampus was lower, and apoptosis was decreased in the dexpanthenol-treated animals. These findings suggest possible clinical applications of dexpanthenol in human HIE.
Collapse
Affiliation(s)
- Cuneyt Tayman
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Ufuk Çakır
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Abdullah Kurt
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Ömer Ertekin
- Department of Neonatology, SBU Ankara City Hospital, Ankara, Turkey
| | - Tugba Taskin Turkmenoglu
- Department of Pathology, Ankara Dişkapi Yildirim Beyzat Training and Research Hospital, Ankara, Turkey
| | - Murat Çağlayan
- Department of Medical Biochemistry, Dışkapı Yıldırım Beyazıt Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Eray Işık
- Department of Otorhinolaryngology (Ear-Nose-Throat), Ankara Dişkapi Yildirim Beyzat Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
10
|
Shimochi S, Ihalainen J, Parikka V, Kudomi N, Tolvanen T, Hietanen A, Kokkomäki E, Johansson S, Tsuji M, Kanaya S, Yatkin E, Grönroos TJ, Iida H. Small animal PET with spontaneous inhalation of 15O-labelled oxygen gases: Longitudinal assessment of cerebral oxygen metabolism in a rat model of neonatal hypoxic-ischaemic encephalopathy. J Cereb Blood Flow Metab 2024; 44:1024-1038. [PMID: 38112197 PMCID: PMC11318403 DOI: 10.1177/0271678x231220691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/05/2023] [Accepted: 11/12/2023] [Indexed: 12/21/2023]
Abstract
Perinatal hypoxic-ischaemic encephalopathy (HIE) is the leading cause of irreversible brain damage resulting in serious neurological dysfunction among neonates. We evaluated the feasibility of positron emission tomography (PET) methodology with 15O-labelled gases without intravenous or tracheal cannulation for assessing temporal changes in cerebral blood flow (CBF) and cerebral metabolic rate for oxygen (CMRO2) in a neonatal HIE rat model. Sequential PET scans with spontaneous inhalation of 15O-gases mixed with isoflurane were performed over 14 days after the hypoxic-ischaemic insult in HIE pups and age-matched controls. CBF and CMRO2 in the injured hemispheres of HIE pups remarkably decreased 2 days after the insult, gradually recovering over 14 days in line with their increase found in healthy controls according to their natural maturation process. The magnitude of hemispheric tissue loss histologically measured after the last PET scan was significantly correlated with the decreases in CBF and CMRO2.This fully non-invasive imaging strategy may be useful for monitoring damage progression in neonatal HIE and for evaluating potential therapeutic outcomes.
Collapse
Affiliation(s)
- Saeka Shimochi
- Turku PET Centre, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Nara Institute of Science and Technology, Ikoma City, Japan
| | - Jukka Ihalainen
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Medical Physics, Turku University Hospital, Turku, Finland
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Vilhelmiina Parikka
- Turku PET Centre, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Nobuyuki Kudomi
- Department of Medical Physics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Tuula Tolvanen
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Ari Hietanen
- Turku PET Centre, University of Turku, Turku, Finland
| | - Esa Kokkomäki
- Turku PET Centre, University of Turku, Turku, Finland
| | - Stefan Johansson
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | | | - Emrah Yatkin
- Central Animal Laboratory, University of Turku, Turku, Finland
| | - Tove J Grönroos
- Turku PET Centre, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Hidehiro Iida
- Turku PET Centre, University of Turku, Turku, Finland
- Nara Institute of Science and Technology, Ikoma City, Japan
| |
Collapse
|
11
|
Hu Y, Nan Y, Lin H, Zhao Q, Chen T, Tao X, Ding B, Lu L, Chen S, Zhu J, Guo X, Lin Z. Celastrol ameliorates hypoxic-ischemic brain injury in neonatal rats by reducing oxidative stress and inflammation. Pediatr Res 2024:10.1038/s41390-024-03246-9. [PMID: 38763946 DOI: 10.1038/s41390-024-03246-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) is caused by perinatal hypoxia and subsequent reductions in cerebral blood flow and is one of the leading causes of severe disability or death in newborns. Despite its prevalence, we currently lack an effective drug therapy to combat HIE. Celastrol (Cel) is a pentacyclic triterpene extracted from Tripterygium Wilfordi that can protect against oxidative stress, inflammation, and cancer. However, whether Cel can alleviate neonatal hypoxic-ischemic (HI) brain damage remains unclear. METHODS Here, we established both in vitro and in vivo models of HI brain damage using CoCl2-treated PC12 cells and neonatal rats, respectively, and explored the neuroprotective effects of Cel in these models. RESULTS Analyses revealed that Cel administration reduced brain infarction size, microglia activation, levels of inflammation factors, and levels of oxidative stress markers by upregulating levels of p-AMPKα, Nrf2, HO-1, and by downregulating levels of TXNIP and NLRP3. Conversely, these beneficial effects of Cel on HI brain damage were largely inhibited by AMPKα inhibitor Compound C and its siRNA. CONCLUSIONS We present compelling evidence that Cel decreases inflammation and oxidative stress through the AMPKα/Nrf2/TXNIP signaling pathway, thereby alleviating neonatal HI brain injury. Cel therefore represents a promising therapeutic agent for treating HIE. IMPACT We firstly report that celastrol can ameliorate neonatal hypoxic-ischemic brain injury both in in vivo and in vitro, which represents a promising therapeutic agent for treating related brain injuries. Celastrol activates the AMPKα/Nrf2/TXNIP signaling pathway to relieve oxidative stress and inflammation and thereby alleviates neonatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Yingying Hu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Nan
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongzhou Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qianlei Zhao
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tingting Chen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoyue Tao
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bingqing Ding
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liying Lu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shangqin Chen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianghu Zhu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Xiaoling Guo
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Zhenlang Lin
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Key Laboratory of Perinatal Medicine of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
12
|
Yang Y, Li Y, Yang W, Yang X, Luo M, Qin L, Zhu J. Protecting effects of 4-octyl itaconate on neonatal hypoxic-ischemic encephalopathy via Nrf2 pathway in astrocytes. J Neuroinflammation 2024; 21:132. [PMID: 38760862 PMCID: PMC11102208 DOI: 10.1186/s12974-024-03121-8] [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: 12/26/2023] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the most common neurological problems occurring in the perinatal period. However, there still is not a promising approach to reduce long-term neurodevelopmental outcomes of HIE. Recently, itaconate has been found to exhibit anti-oxidative and anti-inflammatory effects. However, the therapeutic efficacy of itaconate in HIE remains inconclusive. Therefore, this study attempts to explore the pathophysiological mechanisms of oxidative stress and inflammatory responses in HIE as well as the potential therapeutic role of a derivative of itaconate, 4-octyl itaconate (4OI). METHODS We used 7-day-old mice to induce hypoxic-ischemic (HI) model by right common carotid artery ligation followed by 1 h of hypoxia. Behavioral experiments including the Y-maze and novel object recognition test were performed on HI mice at P60 to evaluate long-term neurodevelopmental outcomes. We employed an approach combining non-targeted metabolomics with transcriptomics to screen alterations in metabolic profiles and gene expression in the hippocampal tissue of the mice at 8 h after hypoxia. Immunofluorescence staining and RT-PCR were used to evaluate the pathological changes in brain tissue cells and the expression of mRNA and proteins. 4OI was intraperitoneally injected into HI model mice to assess its anti-inflammatory and antioxidant effects. BV2 and C8D1A cells were cultured in vitro to study the effect of 4OI on the expression and nuclear translocation of Nrf2. We also used Nrf2-siRNA to further validate 4OI-induced Nrf2 pathway in astrocytes. RESULTS We found that in the acute phase of HI, there was an accumulation of pyruvate and lactate in the hippocampal tissue, accompanied by oxidative stress and pro-inflammatory, as well as increased expression of antioxidative stress and anti-inflammatory genes. Treatment of 4OI could inhibit activation and proliferation of microglial cells and astrocytes, reduce neuronal death and relieve cognitive dysfunction in HI mice. Furthermore, 4OI enhanced nuclear factor erythroid-2-related factor (Nfe2l2; Nrf2) expression and nuclear translocation in astrocytes, reduced pro-inflammatory cytokine production, and increased antioxidant enzyme expression. CONCLUSION Our study demonstrates that 4OI has a potential therapeutic effect on neuronal damage and cognitive deficits in HIE, potentially through the modulation of inflammation and oxidative stress pathways by Nrf2 in astrocytes.
Collapse
Affiliation(s)
- Yanping Yang
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Li
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wenyi Yang
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xueying Yang
- Department of Physiology, China Medical University, Shenyang, Liaoning, China
| | - Man Luo
- Department of Anesthesiology, Shenzhen Cancer Hospital, Shenzhen, China
| | - Ling Qin
- Department of Physiology, China Medical University, Shenyang, Liaoning, China.
| | - Junchao Zhu
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
13
|
Maïza A, Hamoudi R, Mabondzo A. Targeting the Multiple Complex Processes of Hypoxia-Ischemia to Achieve Neuroprotection. Int J Mol Sci 2024; 25:5449. [PMID: 38791487 PMCID: PMC11121719 DOI: 10.3390/ijms25105449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/06/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a major cause of newborn brain damage stemming from a lack of oxygenated blood flow in the neonatal period. Twenty-five to fifty percent of asphyxiated infants who develop HIE die in the neonatal period, and about sixty percent of survivors develop long-term neurological disabilities. From the first minutes to months after the injury, a cascade of events occurs, leading to blood-brain barrier (BBB) opening, neuronal death and inflammation. To date, the only approach proposed in some cases is therapeutic hypothermia (TH). Unfortunately, TH is only partially protective and is not applicable to all neonates. This review synthesizes current knowledge on the basic molecular mechanisms of brain damage in hypoxia-ischemia (HI) and on the different therapeutic strategies in HI that have been used and explores a major limitation of unsuccessful therapeutic approaches.
Collapse
Affiliation(s)
- Auriane Maïza
- CEA, DMTS, SPI, Neurovascular Unit Research & Therapeutic Innovation Laboratory, Paris-Saclay University, CEDEX 91191 Gif-sur-Yvette, France;
| | - Rifat Hamoudi
- Center of Excellence of Precision Medicine, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates;
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London NW3 2PF, UK
| | - Aloïse Mabondzo
- CEA, DMTS, SPI, Neurovascular Unit Research & Therapeutic Innovation Laboratory, Paris-Saclay University, CEDEX 91191 Gif-sur-Yvette, France;
| |
Collapse
|
14
|
Auriti C, Mondì V, Piersigilli F, Timelli L, Del Pinto T, Prencipe G, Lucignani G, Longo D, Bersani I. Plasmatic profiles of cytokines/chemokines, glial fibrillary acidic protein (GFAP) and MRI brain damage in neonates with hypoxic ischemic encephalopathy (HIE). Cytokine 2024; 177:156565. [PMID: 38442443 DOI: 10.1016/j.cyto.2024.156565] [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: 11/01/2023] [Revised: 02/21/2024] [Accepted: 02/29/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Perinatal hypoxia triggers the release of cytokines and chemokines by neurons, astrocytes and microglia. In response to hypoxia-ischemia resting/ramified microglia proliferate and undergo activation, producing proinflammatory molecules. The brain damage extension seems to be related to both the severity of hypoxia and the balance between pro and anti-inflammatory response and can be explored with neuroimaging. AIMS The aim of this preliminary study was to explore possible relationships between plasma levels of inflammatory cytokines/chemokines and the severe brain damage detectable by Magnetic Resonance Imaging (MRI), performed during the hospitalization. METHODS In 10 full terms neonates with hypoxic ischemic encephalopathy (HIE) undergoing therapeutic hypothermia (TH), divided into cases and controls, according to MRI results, we measured and compared the plasma levels of CCL2/MCP-1, CXCL8, GFAP, IFN y, IL-10, IL-18, IL-6, CCL3, ENOLASE2, GM-CSF, IL-1b, IL-12p70, IL-33, TNFα, collected at four different time points during TH (24, 25-48, 49-72 h of life, and 7-10 days from birth). Five of enrolled babies had pathological brain MRI (cases) and 5 had a normal MRI examination (controls). Cytokines were measured by Magnetic Luminex Assay. MRI images were classified according to Barkovich's score. RESULTS Mean levels of all cytokines and molecules at time T1 were not significantly different in the two groups. Comparing samples paired by day of collection, the greatest differences between cases and controls were found at times T2 and T3, during TH. At T4, levels tended to get closer again (except for IL-6, IL10 and IL18). Infants with worse MRI showed higher plasmatic GFAP levels than those with normal MRI, while their IL-18 was lower. The mean levels of CCL3MIP1alpha, GMCSF, IL1BETA overlapped throughout the observation period in both groups. CONCLUSION In a small number of infants with worse brain MRI, we found higher levels of GFAP and of IL-10 at T4 and a trend toward low IL-18 levels than in infants with normal MRI, considered early biomarker of brain damage and a predictor of adverse outcome, respectively. The greatest, although not significant, difference between the levels of molecules was found in cases and controls at time points T2 and T3, during TH.
Collapse
Affiliation(s)
- Cinzia Auriti
- Unicamillus-Saint Camillus International University of Health Sciences, Rome, Italy; Villa Margherita Private Clinic, Rome, Italy.
| | - Vito Mondì
- Neonatology and Neonatal Intensive Care Unit, Policlinico Casilino, Via Casilina 1049, Rome, Italy
| | - Fiammetta Piersigilli
- Section of Neonatology, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Avenue Hippocrate 10, Bruxelles, Belgium
| | - Laura Timelli
- Neonatal Intensive Care Unit, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Tamara Del Pinto
- Unicamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | - Giusi Prencipe
- Laboratory of Immuno-Rheumatology, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Giulia Lucignani
- Department of Imaging, "Bambino Gesù" Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Daniela Longo
- Department of Imaging, "Bambino Gesù" Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| | - Iliana Bersani
- Neonatal Intensive Care Unit, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165 Rome, Italy
| |
Collapse
|
15
|
Zuo HJ, Wang PX, Ren XQ, Shi HL, Shi JS, Guo T, Wan C, Li JJ. Gastrodin Regulates PI3K/AKT-Sirt3 Signaling Pathway and Proinflammatory Mediators in Activated Microglia. Mol Neurobiol 2024; 61:2728-2744. [PMID: 37930585 DOI: 10.1007/s12035-023-03743-8] [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: 08/01/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023]
Abstract
Activated microglia and their mediated inflammatory responses play an important role in the pathogenesis of hypoxic-ischemic brain damage (HIBD). Therefore, regulating microglia activation is considered a potential therapeutic strategy. The neuroprotective effects of gastrodin were evaluated in HIBD model mice, and in oxygen glucose deprivation (OGD)-treated and lipopolysaccharide (LPS)activated BV-2 microglia cells. The potential molecular mechanism was investigated using western blotting, immunofluorescence labeling, quantitative realtime reverse transcriptase polymerase chain reaction, and flow cytometry. Herein, we found that PI3K/AKT signaling can regulate Sirt3 in activated microglia, but not reciprocally. And gastrodin exerts anti-inflammatory and antiapoptotic effects through the PI3K/AKT-Sirt3 signaling pathway. In addition, gastrodin could promote FOXO3a phosphorylation, and inhibit ROS production in LPSactivated BV-2 microglia. Moreover, the level P-FOXO3a decreased significantly in Sirt3-siRNA group. However, there was no significant change after gastrodin and siRNA combination treatment. Notably, gastrodin might also affect the production of ROS in activated microglia by regulating the level of P-FOXO3a via Sirt3. Together, this study highlighted the neuroprotective role of PI3K/AKT-Sirt3 axis in HIBD, and the anti-inflammatory, anti-apoptotic, and anti-oxidative stress effects of gastrodin on HIBD.
Collapse
Affiliation(s)
- Han-Jun Zuo
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, China
| | - Peng-Xiang Wang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, China
| | - Xue-Qi Ren
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, China
| | - Hao-Long Shi
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, China
| | - Jin-Sha Shi
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, China
| | - Tao Guo
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, China
| | - Cheng Wan
- Department of Medical Imaging, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650031, China
| | - Juan-Juan Li
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, China.
| |
Collapse
|
16
|
Anilkumar S, Wright-Jin E. NF-κB as an Inducible Regulator of Inflammation in the Central Nervous System. Cells 2024; 13:485. [PMID: 38534329 DOI: 10.3390/cells13060485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/09/2024] [Indexed: 03/28/2024] Open
Abstract
The NF-κB (nuclear factor K-light-chain-enhancer of activated B cells) transcription factor family is critical for modulating the immune proinflammatory response throughout the body. During the resting state, inactive NF-κB is sequestered by IκB in the cytoplasm. The proteasomal degradation of IκB activates NF-κB, mediating its translocation into the nucleus to act as a nuclear transcription factor in the upregulation of proinflammatory genes. Stimuli that initiate NF-κB activation are diverse but are canonically attributed to proinflammatory cytokines and chemokines. Downstream effects of NF-κB are cell type-specific and, in the majority of cases, result in the activation of pro-inflammatory cascades. Acting as the primary immune responders of the central nervous system, microglia exhibit upregulation of NF-κB upon activation in response to pathological conditions. Under such circumstances, microglial crosstalk with other cell types in the central nervous system can induce cell death, further exacerbating the disease pathology. In this review, we will emphasize the role of NF-κB in triggering neuroinflammation mediated by microglia.
Collapse
Affiliation(s)
- Sudha Anilkumar
- Neonatal Brain Injury Laboratory, Division of Biomedical Research, Nemours Children's Health, Wilmington, DE 19803, USA
| | - Elizabeth Wright-Jin
- Neonatal Brain Injury Laboratory, Division of Biomedical Research, Nemours Children's Health, Wilmington, DE 19803, USA
- Division of Neurology, Department of Pediatrics, Nemours Children's Health, Wilmington, DE 19803, USA
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
17
|
Toptan HH, Tezel KG, Tezel O, Ataç Ö, Vardar G, Gülcan Kersin S, Özek E. Inflammatory and Hematologic Liver and Platelet (HALP) Scores in Hypothermia-Treated Hypoxic-Ischemic Encephalopathy (HIE). CHILDREN (BASEL, SWITZERLAND) 2024; 11:72. [PMID: 38255385 PMCID: PMC10814453 DOI: 10.3390/children11010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/23/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024]
Abstract
OBJECTIVE This study examined systemic inflammatory indices and "Hemoglobin, Albumin, Lymphocyte, Platelet (HALP) scores" in neonates with hypoxic-ischemic encephalopathy (HIE). METHODS A total of 43 neonates with moderate-to-severe HIE at 36 weeks' gestation were assessed. Systemic inflammatory markers were measured before HT commenced within 0-6 h after birth and between 60 and 72 h during and after therapy or before adjusting for hypothermia. RESULTS Platelet counts, hemoglobin levels, and platelet indices in the HIE group were significantly lower at both time points (p = 0.001). Both the neutrophil-to-lymphocyte ratio (NLR) and monocyte-to-lymphocyte ratio (MLR) decreased in the HIE group after hypothermia therapy (p = 0.001). Seizures, PVL, and kidney injuries were associated with higher HALP scores. The AUCs of NLR, PLR, MLR, SII, SIRI, and platelet, neutrophil, monocyte, and lymphocyte Index (PIV) showed significant sensitivity and specified HIE, with area under the curve (AUC) values of 0.654, 0.751, 0.766, 0.700, 0.722, and 0.749, respectively. CONCLUSIONS A significant difference in systemic inflammatory markers was found between the HIE and control groups after hypothermia treatment, with significant reductions in the MLR and NLR. These markers, particularly MLR, were significant predictors of adverse clinical outcomes including seizures, PVL, and kidney damage.
Collapse
Affiliation(s)
- Handan Hakyemez Toptan
- Department of Pediatrics, Division of Neonatology, School of Medicine, Marmara University, Istanbul 34722, Turkey; (K.G.T.); (O.T.); (G.V.); (S.G.K.); (E.Ö.)
| | - Kübra Gökçe Tezel
- Department of Pediatrics, Division of Neonatology, School of Medicine, Marmara University, Istanbul 34722, Turkey; (K.G.T.); (O.T.); (G.V.); (S.G.K.); (E.Ö.)
| | - Oğuzhan Tezel
- Department of Pediatrics, Division of Neonatology, School of Medicine, Marmara University, Istanbul 34722, Turkey; (K.G.T.); (O.T.); (G.V.); (S.G.K.); (E.Ö.)
| | - Ömer Ataç
- Department of Public Health, International School of Medicine, Istanbul Medipol University, Istanbul 34810, Turkey;
| | - Gonca Vardar
- Department of Pediatrics, Division of Neonatology, School of Medicine, Marmara University, Istanbul 34722, Turkey; (K.G.T.); (O.T.); (G.V.); (S.G.K.); (E.Ö.)
| | - Sinem Gülcan Kersin
- Department of Pediatrics, Division of Neonatology, School of Medicine, Marmara University, Istanbul 34722, Turkey; (K.G.T.); (O.T.); (G.V.); (S.G.K.); (E.Ö.)
| | - Eren Özek
- Department of Pediatrics, Division of Neonatology, School of Medicine, Marmara University, Istanbul 34722, Turkey; (K.G.T.); (O.T.); (G.V.); (S.G.K.); (E.Ö.)
| |
Collapse
|
18
|
Davies MR, Greenberg Z, van Vuurden DG, Cross CB, Zannettino ACW, Bardy C, Wardill HR. More than a small adult brain: Lessons from chemotherapy-induced cognitive impairment for modelling paediatric brain disorders. Brain Behav Immun 2024; 115:229-247. [PMID: 37858741 DOI: 10.1016/j.bbi.2023.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023] Open
Abstract
Childhood is recognised as a period of immense physical and emotional development, and this, in part, is driven by underlying neurophysiological transformations. These neurodevelopmental processes are unique to the paediatric brain and are facilitated by augmented rates of neuroplasticity and expanded neural stem cell populations within neurogenic niches. However, given the immaturity of the developing central nervous system, innate protective mechanisms such as neuroimmune and antioxidant responses are functionally naïve which results in periods of heightened sensitivity to neurotoxic insult. This is highly relevant in the context of paediatric cancer, and in particular, the neurocognitive symptoms associated with treatment, such as surgery, radio- and chemotherapy. The vulnerability of the developing brain may increase susceptibility to damage and persistent symptomology, aligning with reports of more severe neurocognitive dysfunction in children compared to adults. It is therefore surprising, given this intensified neurocognitive burden, that most of the pre-clinical, mechanistic research focuses exclusively on adult populations and extrapolates findings to paediatric cohorts. Given this dearth of age-specific research, throughout this review we will draw comparisons with neurodevelopmental disorders which share comparable pathways to cancer treatment related side-effects. Furthermore, we will examine the unique nuances of the paediatric brain along with the somatic systems which influence neurological function. In doing so, we will highlight the importance of developing in vitro and in vivo paediatric disease models to produce age-specific discovery and clinically translatable research.
Collapse
Affiliation(s)
- Maya R Davies
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia; Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia.
| | - Zarina Greenberg
- South Australian Health and Medical Research Institute (SAHMRI), Laboratory of Human Neurophysiology and Genetics, Adelaide, SA, Australia
| | - Dannis G van Vuurden
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the weNetherlands
| | - Courtney B Cross
- Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Andrew C W Zannettino
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Cedric Bardy
- South Australian Health and Medical Research Institute (SAHMRI), Laboratory of Human Neurophysiology and Genetics, Adelaide, SA, Australia; Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Hannah R Wardill
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia; Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| |
Collapse
|
19
|
Sun Z, Song J, Song Q, Li L, Tian X, Wang L. Recombinant human erythropoietin protects against immature brain damage induced by hypoxic/ischemia insult. Neuroreport 2023; 34:801-810. [PMID: 37938927 PMCID: PMC10609708 DOI: 10.1097/wnr.0000000000001957] [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: 07/17/2023] [Accepted: 09/04/2023] [Indexed: 11/10/2023]
Abstract
To investigate the neuroprotection of recombinant human erythropoietin (rhEPO) against hypoxic/ischemic (HI) insult in three-day-old rats. Postnatal day 3 (PD3) rats were randomly divided into three groups: Sham group, HI group and HI+rhEPO group. Ligation of the right common carotid artery and hypoxia to induce HI brain injury. After HI insult, the rats received intraperitoneal injection of rhEPO (5000 IU/Kg, qod) in HI+rhEPO group or equal saline in other groups. On PD10, damage of brain tissue was examined by hematoxylin-eosin (HE) staining, observation of neuronal apoptosis in the hippocampus and cortex using immunofluorescence assay (marker: TUNEL). Immunohistochemical staining or western blotting was performed to detect the expression of cyclooxygenase-2 (COX-2), Caspase-3 and phosphorylated Akt (p-Akt) protein. On PD28, cognitive ability of rats was assessed by Morris water maze test. HI injury causes brain pathological morphology and cognitive function damage in PD3 rats, which can be alleviated by rhEPO intervention. Compared with the HI group, the HI+rhEPO group showed an increase in platform discovery rate and cross platform frequency, while the search platform time was shortened (P < 0.05). The proportion of TUNEL positive neurons and the expression of COX-2 and Caspase-3 proteins in brain tissue in the hippocampus and cortex was decreased, while the expression of p-Akt protein was upregulated (P < 0.05). RhEPO could protect against the pathological and cognitive impairment of immature brain induced by HI insult. This neuroprotective activity may involve in inhibiting inflammatory and apoptosis by activation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Zhengda Sun
- Department of Neonatology, Jinan Maternity and Child Health Care Hospital
- Department of Neonatology, Shandong Provincial Hospital affiliated to Shandong First Medical University
- Shandong First Medical University
| | - Jiqing Song
- Department of Radiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | | | - Lin Li
- Department of Neonatology, Shandong Provincial Hospital affiliated to Shandong First Medical University
- Shandong First Medical University
| | | | - Lijun Wang
- Department of Neonatology, Shandong Provincial Hospital affiliated to Shandong First Medical University
| |
Collapse
|
20
|
Wu DM, Liu JP, Liu J, Ge WH, Wu SZ, Zeng CJ, Liang J, Liu K, Lin Q, Hong XW, Sun YE, Lu J. Immune pathway activation in neurons triggers neural damage after stroke. Cell Rep 2023; 42:113368. [PMID: 37917581 DOI: 10.1016/j.celrep.2023.113368] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/24/2023] [Accepted: 10/17/2023] [Indexed: 11/04/2023] Open
Abstract
Ischemic brain injury is a severe medical condition with high incidences in elderly people without effective treatment for the resulting neural damages. Using a unilateral mouse stroke model, we analyze single-cell transcriptomes of ipsilateral and contralateral cortical penumbra regions to objectively reveal molecular events with single-cell resolution at 4 h and 1, 3, and 7 days post-injury. Here, we report that neurons are among the first cells that sense the lack of blood supplies by elevated expression of CCAAT/enhancer-binding protein β (C/EBPβ). To our surprise, the canonical inflammatory cytokine gene targets for C/EBPβ, including interleukin-1β (IL-1β) and tumor necrosis factor α (TNF-α), are subsequently induced also in neuronal cells. Neuronal-specific silencing of C/EBPβ or IL-1β and TNF-α substantially alleviates downstream inflammatory injury responses and is profoundly neural protective. Taken together, our findings reveal a neuronal inflammatory mechanism underlying early pathological triggers of ischemic brain injury.
Collapse
Affiliation(s)
- Dong-Mei Wu
- Clinical Medicine Center, Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Guangdong 528000, China
| | - Ji-Ping Liu
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jie Liu
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; Clinical Medicine Center, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China
| | - Wei-Hong Ge
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Su-Zhen Wu
- Clinical Medicine Center, Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Guangdong 528000, China
| | - Chi-Jia Zeng
- Clinical Medicine Center, Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Guangdong 528000, China
| | - Jia Liang
- Life Science Institution, Jinzhou Medical University, Jinzhou 121000, China
| | - KeJian Liu
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Quan Lin
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xiao-Wu Hong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Research Institute of Fudan University in Ningbo, Zhejiang 315336, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Jun Lu
- Clinical Medicine Center, Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Guangdong 528000, China.
| |
Collapse
|
21
|
Zhang M, Liu Z, Zhou W, Shen M, Mao N, Xu H, Wang Y, Xu Z, Li M, Jiang H, Chen Y, Zhu J, Lin W, Yuan J, Lin Z. Ferrostatin-1 attenuates hypoxic-ischemic brain damage in neonatal rats by inhibiting ferroptosis. Transl Pediatr 2023; 12:1944-1970. [PMID: 38130589 PMCID: PMC10730959 DOI: 10.21037/tp-23-189] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 11/02/2023] [Indexed: 12/23/2023] Open
Abstract
Background Hypoxic-ischemic brain damage (HIBD) is a type of brain damage that is caused by perinatal asphyxia and serious damages the central nervous system. At present, there is no effective drug for the treatment of this disease. Besides, the pathogenesis of HIBD remains elusive. While studies have shown that ferroptosis plays an important role in HIBD, its role and mechanism in HIBD are yet to be fully understood. Methods The HIBD model of neonatal rats was established using the Rice-Vannucci method. A complete medium of PC12 cells was adjusted to a low-sugar medium, and the oxygen-glucose deprivation model was established after continuous hypoxia for 12 h. Laser Doppler blood flow imaging was used to detect the blood flow intensity after modeling. 2,3,5-triphenyl tetrazolium chloride staining was employed to detect ischemic cerebral infarction in rat brain tissue, and hematoxylin and eosin staining and transmission electron microscopy were used to observe brain injury and mitochondrial damage. Immunofluorescence was applied to monitor the expression of GFAP. Real-time quantitative polymerase chain reaction, western blot, and immunofluorescence were utilized to detect the expression of messenger RNA and protein. The level of reactive oxygen species (ROS) in cells was detected using the ROS detection kit. Results The results showed that ferrostatin-1 (Fer-1) significantly alleviated the brain injury caused by hypoxia and ischemia. Fer-1 significantly increased the expression of SLC3A2, SLC7A11, ACSL3, GSS, and GPX4 (P<0.05) and dramatically decreased the expressions of GFAP, ACSL4, TFRC, FHC, FLC, 4-HNE, HIF-1α, and ROS (P<0.05). Conclusions Fer-1 inhibits ferroptosis and alleviates HIBD by potentially targeting the GPX4/ACSL3/ACSL4 axis; however, its specific mechanism warrants further exploration.
Collapse
Affiliation(s)
- Min Zhang
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhiming Liu
- Department of Spinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Zhou
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ming Shen
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Niping Mao
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hang Xu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yanan Wang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zidi Xu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Mopu Li
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Haibin Jiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yuetong Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jianghu Zhu
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Lin
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junhui Yuan
- Department of Neonatology, Wenling Maternal and Child Health Care Hospital, Wenling, China
| | - Zhenlang Lin
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
22
|
Ranjan AK, Gulati A. Advances in Therapies to Treat Neonatal Hypoxic-Ischemic Encephalopathy. J Clin Med 2023; 12:6653. [PMID: 37892791 PMCID: PMC10607511 DOI: 10.3390/jcm12206653] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a condition that results in brain damage in newborns due to insufficient blood and oxygen supply during or after birth. HIE is a major cause of neurological disability and mortality in newborns, with over one million neonatal deaths occurring annually worldwide. The severity of brain injury and the outcome of HIE depend on several factors, including the cause of oxygen deprivation, brain maturity, regional blood flow, and maternal health conditions. HIE is classified into mild, moderate, and severe categories based on the extent of brain damage and resulting neurological issues. The pathophysiology of HIE involves different phases, including the primary phase, latent phase, secondary phase, and tertiary phase. The primary and secondary phases are characterized by episodes of energy and cell metabolism failures, increased cytotoxicity and apoptosis, and activated microglia and inflammation in the brain. A tertiary phase occurs if the brain injury persists, characterized by reduced neural plasticity and neuronal loss. Understanding the cellular and molecular aspects of the different phases of HIE is crucial for developing new interventions and therapeutics. This review aims to discuss the pathophysiology of HIE, therapeutic hypothermia (TH), the only approved therapy for HIE, ongoing developments of adjuvants for TH, and potential future drugs for HIE.
Collapse
Affiliation(s)
- Amaresh K Ranjan
- Research and Development, Pharmazz Inc., Willowbrook, IL 60527, USA
| | - Anil Gulati
- Research and Development, Pharmazz Inc., Willowbrook, IL 60527, USA
- Department of Bioengineering, The University of Illinois at Chicago, Chicago, IL 60607, USA
- College of Pharmacy, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
23
|
Gaston-Breton R, Maïza Letrou A, Hamoudi R, Stonestreet BS, Mabondzo A. Brain organoids for hypoxic-ischemic studies: from bench to bedside. Cell Mol Life Sci 2023; 80:318. [PMID: 37804439 PMCID: PMC10560197 DOI: 10.1007/s00018-023-04951-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 10/09/2023]
Abstract
Our current knowledge regarding the development of the human brain mostly derives from experimental studies on non-human primates, sheep, and rodents. However, these studies may not completely simulate all the features of human brain development as a result of species differences and variations in pre- and postnatal brain maturation. Therefore, it is important to supplement the in vivo animal models to increase the possibility that preclinical studies have appropriate relevance for potential future human trials. Three-dimensional brain organoid culture technology could complement in vivo animal studies to enhance the translatability of the preclinical animal studies and the understanding of brain-related disorders. In this review, we focus on the development of a model of hypoxic-ischemic (HI) brain injury using human brain organoids to complement the translation from animal experiments to human pathophysiology. We also discuss how the development of these tools provides potential opportunities to study fundamental aspects of the pathophysiology of HI-related brain injury including differences in the responses between males and females.
Collapse
Affiliation(s)
- Romane Gaston-Breton
- Université Paris Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (DMTS), Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique, 91191, Gif-sur-Yvette Cedex, France
| | - Auriane Maïza Letrou
- Université Paris Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (DMTS), Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique, 91191, Gif-sur-Yvette Cedex, France
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, P. O. 27272, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, P. O. 27272, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, UK
- ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, Sharjah, United Arab Emirates
| | - Barbara S Stonestreet
- Departments of Molecular Biology, Cell Biology and Biochemistry and Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, 101 Dudley Street, Providence, RI, 02905, USA
| | - Aloïse Mabondzo
- Université Paris Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (DMTS), Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique, 91191, Gif-sur-Yvette Cedex, France.
| |
Collapse
|
24
|
Chen Y, Li X, Xiong Q, Du Y, Luo M, Yi L, Pang Y, Shi X, Wang YT, Dong Z. Inhibiting NLRP3 inflammasome signaling pathway promotes neurological recovery following hypoxic-ischemic brain damage by increasing p97-mediated surface GluA1-containing AMPA receptors. J Transl Med 2023; 21:567. [PMID: 37620837 PMCID: PMC10463885 DOI: 10.1186/s12967-023-04452-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/19/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND The nucleotide-binding oligomeric domain (NOD)-like receptor protein 3 (NLRP3) inflammasome is believed to be a key mediator of neuroinflammation and subsequent secondary brain injury induced by ischemic stroke. However, the role and underlying mechanism of the NLRP3 inflammasome in neonates with hypoxic-ischemic encephalopathy (HIE) are still unclear. METHODS The protein expressions of the NLRP3 inflammasome including NLRP3, cysteinyl aspartate specific proteinase-1 (caspase-1) and interleukin-1β (IL-1β), the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionicacid receptor (AMPAR) subunit, and the ATPase valosin-containing protein (VCP/p97), were determined by Western blotting. The interaction between p97 and AMPA glutamate receptor 1 (GluA1) was determined by co-immunoprecipitation. The histopathological level of hypoxic-ischemic brain damage (HIBD) was determined by triphenyltetrazolium chloride (TTC) staining. Polymerase chain reaction (PCR) and Western blotting were used to confirm the genotype of the knockout mice. Motor functions, including myodynamia and coordination, were evaluated by using grasping and rotarod tests. Hippocampus-dependent spatial cognitive function was measured by using the Morris-water maze (MWM). RESULTS We reported that the NLRP3 inflammasome signaling pathway, such as NLRP3, caspase-1 and IL-1β, was activated in rats with HIBD and oxygen-glucose deprivation (OGD)-treated cultured primary neurons. Further studies showed that the protein level of the AMPAR GluA1 subunit on the hippocampal postsynaptic membrane was significantly decreased in rats with HIBD, and it could be restored to control levels after treatment with the specific caspase-1 inhibitor AC-YVAD-CMK. Similarly, in vitro studies showed that OGD reduced GluA1 protein levels on the plasma membrane in cultured primary neurons, whereas AC-YVAD-CMK treatment restored this reduction. Importantly, we showed that OGD treatment obviously enhanced the interaction between p97 and GluA1, while AC-YVAD-CMK treatment promoted the dissociation of p97 from the GluA1 complex and consequently facilitated the localization of GluA1 on the plasma membrane of cultured primary neurons. Finally, we reported that the deficits in motor function, learning and memory in animals with HIBD, were ameliorated by pharmacological intervention or genetic ablation of caspase-1. CONCLUSION Inhibiting the NLRP3 inflammasome signaling pathway promotes neurological recovery in animals with HIBD by increasing p97-mediated surface GluA1 expression, thereby providing new insight into HIE therapy.
Collapse
Affiliation(s)
- Yuxin Chen
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Xiaohuan Li
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Qian Xiong
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yehong Du
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Man Luo
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Lilin Yi
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yayan Pang
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Xiuyu Shi
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yu Tian Wang
- Department of Medicine, Brain Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
| | - Zhifang Dong
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
25
|
Choudhary RC, Shoaib M, Hayashida K, Yin T, Miyara SJ, d’Abramo C, Heuser WG, Shinozaki K, Kim N, Takegawa R, Nishikimi M, Li T, Owens C, Molmenti EP, He M, Vanpatten S, Al-Abed Y, Kim J, Becker LB. Multi-Drug Cocktail Therapy Improves Survival and Neurological Function after Asphyxial Cardiac Arrest in Rodents. Cells 2023; 12:1548. [PMID: 37296668 PMCID: PMC10253071 DOI: 10.3390/cells12111548] [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: 03/31/2023] [Revised: 05/17/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Cardiac arrest (CA) can lead to neuronal degeneration and death through various pathways, including oxidative, inflammatory, and metabolic stress. However, current neuroprotective drug therapies will typically target only one of these pathways, and most single drug attempts to correct the multiple dysregulated metabolic pathways elicited following cardiac arrest have failed to demonstrate clear benefit. Many scientists have opined on the need for novel, multidimensional approaches to the multiple metabolic disturbances after cardiac arrest. In the current study, we have developed a therapeutic cocktail that includes ten drugs capable of targeting multiple pathways of ischemia-reperfusion injury after CA. We then evaluated its effectiveness in improving neurologically favorable survival through a randomized, blind, and placebo-controlled study in rats subjected to 12 min of asphyxial CA, a severe injury model. RESULTS 14 rats were given the cocktail and 14 received the vehicle after resuscitation. At 72 h post-resuscitation, the survival rate was 78.6% among cocktail-treated rats, which was significantly higher than the 28.6% survival rate among vehicle-treated rats (log-rank test; p = 0.006). Moreover, in cocktail-treated rats, neurological deficit scores were also improved. These survival and neurological function data suggest that our multi-drug cocktail may be a potential post-CA therapy that deserves clinical translation. CONCLUSIONS Our findings demonstrate that, with its ability to target multiple damaging pathways, a multi-drug therapeutic cocktail offers promise both as a conceptual advance and as a specific multi-drug formulation capable of combatting neuronal degeneration and death following cardiac arrest. Clinical implementation of this therapy may improve neurologically favorable survival rates and neurological deficits in patients suffering from cardiac arrest.
Collapse
Affiliation(s)
- Rishabh C. Choudhary
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Muhammad Shoaib
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Kei Hayashida
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Tai Yin
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Santiago J. Miyara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030, USA
| | - Cristina d’Abramo
- Litwin-Zucker Center for Research in Alzheimer’s Disease, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - William G. Heuser
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Koichiro Shinozaki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Nancy Kim
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Ryosuke Takegawa
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Mitsuaki Nishikimi
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Timmy Li
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Casey Owens
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | | | - Mingzhu He
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Sonya Vanpatten
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Yousef Al-Abed
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Junhwan Kim
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Lance B. Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; (R.C.C.)
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
- Emergency Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr., Manhasset, NY 11030, USA
| |
Collapse
|
26
|
Aoki Y, Dai H, Furuta F, Akamatsu T, Oshima T, Takahashi N, Goto YI, Oka A, Itoh M. LOX-1 mediates inflammatory activation of microglial cells through the p38-MAPK/NF-κB pathways under hypoxic-ischemic conditions. Cell Commun Signal 2023; 21:126. [PMID: 37268943 DOI: 10.1186/s12964-023-01048-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/14/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Microglial cells play an important role in the immune system in the brain. Activated microglial cells are not only injurious but also neuroprotective. We confirmed marked lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) expression in microglial cells in pathological lesions in the neonatal hypoxic-ischemic encephalopathy (nHIE) model brain. LOX-1 is known to be an activator of cytokines and chemokines through intracellular pathways. Here, we investigated a novel role of LOX-1 and the molecular mechanism of LOX-1 gene transcription microglial cells under hypoxic and ischemic conditions. METHODS We isolated primary rat microglial cells from 3-day-old rat brains and confirmed that the isolated cells showed more than 98% Iba-1 positivity with immunocytochemistry. We treated primary rat microglial cells with oxygen glucose deprivation (OGD) as an in vitro model of nHIE. Then, we evaluated the expression levels of LOX-1, cytokines and chemokines in cells treated with or without siRNA and inhibitors compared with those of cells that did not receive OGD-treatment. To confirm transcription factor binding to the OLR-1 gene promoter under the OGD conditions, we performed a luciferase reporter assay and chromatin immunoprecipitation assay. In addition, we analyzed reactive oxygen species and cell viability. RESULTS We found that defects in oxygen and nutrition induced LOX-1 expression and led to the production of inflammatory mediators, such as the cytokines IL-1β, IL-6 and TNF-α; the chemokines CCL2, CCL5 and CCL3; and reactive oxygen/nitrogen species. Then, the LOX-1 signal transduction pathway was blocked by inhibitors, LOX-1 siRNA, the p38-MAPK inhibitor SB203580 and the NF-κB inhibitor BAY11-7082 suppressed the production of inflammatory mediators. We found that NF-κB and HIF-1α bind to the promoter region of the OLR-1 gene. Based on the results of the luciferase reporter assay, NF-κB has strong transcriptional activity. Moreover, we demonstrated that LOX-1 in microglial cells was autonomously overexpressed by positive feedback of the intracellular LOX-1 pathway. CONCLUSION The hypoxic/ischemic conditions of microglial cells induced LOX-1 expression and activated the immune system. LOX-1 and its related molecules or chemicals may be major therapeutic candidates. Video abstract.
Collapse
Affiliation(s)
- Yoshinori Aoki
- Department of Mental Retardation and Birth Defect Research, National Institute of Neurology, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Pediatrics, Faculty of Medicine, University of Miyazaki, Kiyotake-cho Kihara 5200, Miyazaki, 889-1692, Japan
| | - Hongmei Dai
- Department of Mental Retardation and Birth Defect Research, National Institute of Neurology, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Fumika Furuta
- Department of Mental Retardation and Birth Defect Research, National Institute of Neurology, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Tomohisa Akamatsu
- Department of Mental Retardation and Birth Defect Research, National Institute of Neurology, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, Japan
| | - Takuya Oshima
- Department of Mental Retardation and Birth Defect Research, National Institute of Neurology, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoto Takahashi
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yu-Ichi Goto
- Department of Mental Retardation and Birth Defect Research, National Institute of Neurology, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Akira Oka
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Saitama Children's Medical Center, 1-2 Shintoshin, Chuo-ku, Saitama, Japan
| | - Masayuki Itoh
- Department of Mental Retardation and Birth Defect Research, National Institute of Neurology, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| |
Collapse
|
27
|
Bernis ME, Zweyer M, Maes E, Schleehuber Y, Sabir H. Neutrophil Extracellular Traps Release following Hypoxic-Ischemic Brain Injury in Newborn Rats Treated with Therapeutic Hypothermia. Int J Mol Sci 2023; 24:3598. [PMID: 36835009 PMCID: PMC9966013 DOI: 10.3390/ijms24043598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/01/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
The peripheral immune system plays a critical role in neuroinflammation of the central nervous system after an insult. Hypoxic-ischemic encephalopathy (HIE) induces a strong neuroinflammatory response in neonates, which is often associated with exacerbated outcomes. In adult models of ischemic stroke, neutrophils infiltrate injured brain tissue immediately after an ischemic insult and aggravate inflammation via various mechanisms, including neutrophil extracellular trap (NETs) formation. In this study, we used a neonatal model of experimental hypoxic-ischemic (HI) brain injury and demonstrated that circulating neutrophils were rapidly activated in neonatal blood. We observed an increased infiltration of neutrophils in the brain after exposure to HI. After treatment with either normothermia (NT) or therapeutic hypothermia (TH), we observed a significantly enhanced expression level of the NETosis marker Citrullinated H3 (Cit-H3), which was significantly more pronounced in animals treated with TH than in those treated with NT. NETs and NLR family pyrin domain containing 3 (NLRP-3) inflammasome assembly are closely linked in adult models of ischemic brain injury. In this study, we observed an increase in the activation of the NLRP-3 inflammasome at the time points analyzed, particularly immediately after TH, when we observed a significant increase in NETs structures in the brain. Together, these results suggest the important pathological functions of early arriving neutrophils and NETosis following neonatal HI, particularly after TH treatment, which is a promising starting point for the development of potential new therapeutic targets for neonatal HIE.
Collapse
Affiliation(s)
- Maria E. Bernis
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Margit Zweyer
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Elke Maes
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Yvonne Schleehuber
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Hemmen Sabir
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| |
Collapse
|
28
|
Teo EJ, Chand KK, Miller SM, Wixey JA, Colditz PB, Bjorkman ST. Early evolution of glial morphology and inflammatory cytokines following hypoxic-ischemic injury in the newborn piglet brain. Sci Rep 2023; 13:282. [PMID: 36609414 PMCID: PMC9823001 DOI: 10.1038/s41598-022-27034-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/23/2022] [Indexed: 01/09/2023] Open
Abstract
Neuroinflammation is a hallmark of hypoxic-ischemic injury and can be characterized by the activation of glial cells and the expression of inflammatory cytokines and chemokines. Interleukin (IL)-1β and tumor necrosis factor (TNF)α are among the best-characterized early response cytokines and are often expressed concurrently. Several types of central nervous system cells secrete IL-1β and TNFα, including microglia, astrocytes, and neurons, and these cytokines convey potent pro-inflammatory actions. Chemokines also play a central role in neuroinflammation by controlling inflammatory cell trafficking. Our aim was to characterise the evolution of early neuroinflammation in the neonatal piglet model of hypoxic-ischemic encephalopathy (HIE). Piglets (< 24 h old) were exposed to HI insult, and recovered to 2, 4, 8, 12 or 24H post-insult. Brain tissue from the frontal cortex and basal ganglia was harvested for assessment of glial cell activation profiles and transcription levels of inflammatory markers in HI piglets with comparison to a control group of newborn piglets. Fluorescence microscopy was used to observe microglia, astrocytes, neurons, degenerating neurons and possibly apoptotic cells, and quantitative polymerase chain reaction was used to measure gene expression of several cytokines and chemokines. HI injury was associated with microglial activation and morphological changes to astrocytes at all time points examined. Gene expression analyses of inflammation-related markers revealed significantly higher expression of pro-inflammatory cytokines tumor necrosis factor-α (TNFα) and interleukin 1 beta (IL-1β), chemokines cxc-chemokine motif ligand (CXCL)8 and CXCL10, and anti-inflammatory cytokine transforming growth factor (TGF)β in every HI group, with some region-specific differences noted. No significant difference was observed in the level of C-X-C chemokine receptor (CCR)5 over time. This high degree of neuroinflammation was associated with a reduction in the number of neurons in piglets at 12H and 24H in the frontal cortex, and the putamen at 12H. This reduction of neurons was not associated with increased numbers of degenerating neurons or potentially apoptotic cells. HI injury triggered a robust early neuroinflammatory response associated with a reduction in neurons in cortical and subcortical regions in our piglet model of HIE. This neuroinflammatory response may be targeted using novel therapeutics to reduce neuropathology in our piglet model of neonatal HIE.
Collapse
Affiliation(s)
- Elliot J. Teo
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| | - Kirat. K. Chand
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| | - Stephanie M. Miller
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| | - Julie A. Wixey
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| | - Paul B. Colditz
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| | - S. Tracey. Bjorkman
- grid.1003.20000 0000 9320 7537Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Building 71/918 RBWH Herston, Brisbane City, QLD 4029 Australia ,grid.416100.20000 0001 0688 4634Perinatal Research Centre, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| |
Collapse
|
29
|
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: 0] [Impact Index Per Article: 0] [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
|
30
|
Duve KV, Shkrobot SI. THE NEUROLOGICAL MANIFESTATIONS AND FUNCTIONAL INDEPENDENCE IN PATIENTS WITH ENCEPHALOPATHIES OF DIFFERENT TYPES. POLSKI MERKURIUSZ LEKARSKI : ORGAN POLSKIEGO TOWARZYSTWA LEKARSKIEGO 2023; 51:489-495. [PMID: 38069849 DOI: 10.36740/merkur202305107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
OBJECTIVE Aim: To access the neurological manifestations and activities of daily living in patients with encephalopathy of one of the following types: post-infectious, chronic traumatic encephalopathy, alcohol-induced, and microvascular ischemic disease of the brain. PATIENTS AND METHODS Materials and Methods: In the period of 2021-2022 we examined 520 patients, who signed the informed consent, taking into account their age, sex, occupation, the cause, and the disease duration. Such parameters were evaluated, as the data of neurological manifestations, the activities of daily living (Barthel index), cognitive functioning (MoCA-test), and statistical methods (Statistica 13.0). RESULTS Results: A probable influence of the age factor on the frequency of occurrence of different types of encephalopathies was established (χ2=235.05; p<0.001). The cognitive impairment was diagnosed in 53.79 % of patients with CTE, 66.21% with SVD, and 58.82% with AE. 40% of patients with CTE are dependent on their activities of daily living, among patients with SVD - 31,72 %, among patients with AE - 44.12%, among patients with PIE - 53.91%. 17.97% of patients with PIE had moderate dependence by the Barthel index. Thus, the severity of disability doesn't depend on the age or sex of patients but is correlating with the duration of the disease. CONCLUSION Conclusions: The neurological manifestations in patients with encephalopathies and their activities of daily living were studied profoundly and the data obtained opened new directions in the following research.
Collapse
Affiliation(s)
- Khrystyna V Duve
- I. HORBACHEVSKY TERNOPIL NATIONAL MEDICAL UNIVERSITY OF THE MINISTRY OF HEALTH OF UKRAINE, TERNOPIL, UKRAINE
| | - Svitlana I Shkrobot
- I. HORBACHEVSKY TERNOPIL NATIONAL MEDICAL UNIVERSITY OF THE MINISTRY OF HEALTH OF UKRAINE, TERNOPIL, UKRAINE
| |
Collapse
|
31
|
Leng K, Rose IVL, Kim H, Xia W, Romero-Fernandez W, Rooney B, Koontz M, Li E, Ao Y, Wang S, Krawczyk M, Tcw J, Goate A, Zhang Y, Ullian EM, Sofroniew MV, Fancy SPJ, Schrag MS, Lippmann ES, Kampmann M. CRISPRi screens in human iPSC-derived astrocytes elucidate regulators of distinct inflammatory reactive states. Nat Neurosci 2022; 25:1528-1542. [PMID: 36303069 PMCID: PMC9633461 DOI: 10.1038/s41593-022-01180-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/07/2022] [Indexed: 01/30/2023]
Abstract
Astrocytes become reactive in response to insults to the central nervous system by adopting context-specific cellular signatures and outputs, but a systematic understanding of the underlying molecular mechanisms is lacking. In this study, we developed CRISPR interference screening in human induced pluripotent stem cell-derived astrocytes coupled to single-cell transcriptomics to systematically interrogate cytokine-induced inflammatory astrocyte reactivity. We found that autocrine-paracrine IL-6 and interferon signaling downstream of canonical NF-κB activation drove two distinct inflammatory reactive signatures, one promoted by STAT3 and the other inhibited by STAT3. These signatures overlapped with those observed in other experimental contexts, including mouse models, and their markers were upregulated in human brains in Alzheimer's disease and hypoxic-ischemic encephalopathy. Furthermore, we validated that markers of these signatures were regulated by STAT3 in vivo using a mouse model of neuroinflammation. These results and the platform that we established have the potential to guide the development of therapeutics to selectively modulate different aspects of inflammatory astrocyte reactivity.
Collapse
Affiliation(s)
- Kun Leng
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA.
| | - Indigo V L Rose
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Hyosung Kim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Wenlong Xia
- Departments of Neurology and Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Brendan Rooney
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Mark Koontz
- Department of Ophthalmology, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Emmy Li
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Yan Ao
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shinong Wang
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mitchell Krawczyk
- Interdepartmental PhD Program in Neuroscience, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Julia Tcw
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison Goate
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Erik M Ullian
- Department of Ophthalmology, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Michael V Sofroniew
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Stephen P J Fancy
- Departments of Neurology and Pediatrics, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Matthew S Schrag
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
32
|
Cai Y, Li X, Tan X, Wang P, Zhao X, Zhang H, Song Y. Vitamin D suppresses ferroptosis and protects against neonatal hypoxic-ischemic encephalopathy by activating the Nrf2/HO-1 pathway. Transl Pediatr 2022; 11:1633-1644. [PMID: 36345441 PMCID: PMC9636464 DOI: 10.21037/tp-22-397] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/02/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) is a major cause of neonatal death, and vitamin D (VD) is a neuroprotection nutrition whose deficiency is associated with its risk. However, the mechanism of VD involved in neonatal HIE is not well known. METHODS In this experiment a hypoxic-ischemic brain damage (HIBD) model was established by using the Rice-Vannucci method, rats were intraperitoneally injected with 0.1 µg/kg VD every day for two weeks. The brain damage and mitochondria injury were examined by hematoxylin-eosin (HE) staining and transmission electron microscope (TEM), respectively. The oxidation response and inflammatory factors were determined by enzyme-linked immunosorbent assay (ELISA), and the cell viability was determined by Cell Counting Kit-8 (CCK-8). mRNA and protein expression were detected by quantitative real real-time PCR (qRT-PCR), Western blot, and immunofluorescence. RESULTS The results showed VD effectively ameliorated brain histologic damage and mitochondria injury induced by hypoxic ischemia (HI). VD elevated the expression of Nrf2 and HO-1, which resulted in increased levels of GPX4, superoxide dismutase (SOD), and glutathione (GSH) and reduced content of malondialdehyde (MDA) and reactive oxygen species (ROS), resulting in decreased ferroptosis in HI-treated rats. Moreover, VD reduced the secretion of inflammatory factors, tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-1β. CONCLUSIONS VD suppresses ferroptosis through activation of the Nrf2/HO-1 signaling pathway and exerts a protective role in neonatal HIE.
Collapse
Affiliation(s)
- Yueju Cai
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaolan Li
- Department of Children Healthcare, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xuying Tan
- Department of Children Healthcare, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ping Wang
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaopeng Zhao
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huayan Zhang
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yanyan Song
- Department of Children Healthcare, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
33
|
Foletto VS, da Rosa TF, Serafin MB, Hörner R. Selective serotonin reuptake inhibitor (SSRI) antidepressants reduce COVID-19 infection: prospects for use. Eur J Clin Pharmacol 2022; 78:1601-1611. [PMID: 35943535 PMCID: PMC9360648 DOI: 10.1007/s00228-022-03372-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/30/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE The absence of specific treatments for COVID-19 leads to an intense global effort in the search for new therapeutic interventions and better clinical outcomes for patients. This review aimed to present a selection of accepted studies that reported the activity of antidepressant drugs belonging to the selective serotonin receptor inhibitor (SSRI) class for treating the novel coronavirus. METHODS A search was performed in PubMed and SciELO databases using the following search strategies: [(coronavirus) OR (COVID) OR (SARS-CoV-2) AND (antidepressant) OR (serotonin) OR (selective serotonin receptor inhibitors)]. In the end, eleven articles were included. We also covered information obtained from ClinicalTrials.gov in our research. RESULTS Although several clinical trials are ongoing, only a few drugs have been officially approved to treat the infection. Remdesivir, an antiviral drug, despite favorable preliminary results, has restricted the use due to the risk of toxicity and methodological flaws. Antidepressant drugs were able to reduce the risk of intubation or death related to COVID-19, decrease the need for intensive medical care, and severely inhibit viral titers by up to 99%. Among the SSRIs studied so far, fluoxetine and fluvoxamine have shown to be the most promising against SARS-CoV-2. CONCLUSION If successful, these drugs can substantially reduce hospitalization and mortality rates, as well as allow for fully outpatient treatment for mild-to-moderate infections. Thus, repositioning SSRIs can provide benefits when faced with a rapidly evolving pandemic such as COVID-19.
Collapse
Affiliation(s)
| | - Taciéli Fagundes da Rosa
- Federal University of Santa Maria, Postgraduate Program in Pharmaceutical Sciences, Santa Maria, RS, Brazil
| | - Marissa Bolson Serafin
- Federal University of Santa Maria, Postgraduate Program in Pharmaceutical Sciences, Santa Maria, RS, Brazil
| | - Rosmari Hörner
- Federal University of Santa Maria, Postgraduate Program in Pharmaceutical Sciences, Santa Maria, RS, Brazil.
- Department of Clinical and Toxicological Analysis, Federal University of Santa Maria, UFSM, Building 26, Room 1201, Santa Maria, RS, 97015-900, Brazil.
| |
Collapse
|
34
|
Chen J, Chang Y, Zhu J, Peng Y, Li Z, Zhang K, Zhang Y, Lin C, Lin Z, Pan S, Huang K. Flufenamic acid improves survival and neurologic outcome after successful cardiopulmonary resuscitation in mice. J Neuroinflammation 2022; 19:214. [PMID: 36050694 PMCID: PMC9438280 DOI: 10.1186/s12974-022-02571-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/19/2022] [Indexed: 11/10/2022] Open
Abstract
Background Brain injury is the main cause of high mortality and disability after successful cardiopulmonary resuscitation (CPR) from sudden cardiac arrest (CA). The transient receptor potential M4 (TRPM4) channel is a novel target for ameliorating blood–brain barrier (BBB) disruption and neuroinflammation. Herein, we tested whether flufenamic acid (FFA), which is reported to block TRPM4 with high potency, could confer neuroprotection against brain injury secondary to CA/CPR and whether its action was exerted by blocking the TRPM4 channel. Methods Wild-type (WT) and Trpm4 knockout (Trpm4−/−) mice subjected to 10-min CA/CPR were randomized to receive FFA or vehicle once daily. Post-CA/CPR brain injuries including neurologic deficits, survival rate, histological damage, edema formation, BBB destabilization and neuroinflammation were assessed. Results In WT mice subjected to CA/CPR, FFA was effective in improving survival and neurologic outcome, reducing neuropathological injuries, attenuating brain edema, lessening the leakage of IgG and Evans blue dye, restoring tight junction protein expression and promoting microglia/macrophages from the pro-inflammatory subtype toward the anti-inflammatory subtype. In comparison to WT mice, Trpm4−/− mice exhibited less neurologic deficiency, milder histological impairment, more BBB integrity and more anti-inflammatory microglia/macrophage polarization. As expected, FFA did not provide a benefit of superposition compared with vehicle in the Trpm4−/− mice after CA/CPR. Conclusions FFA mitigates BBB breach and modifies the functional status of microglia/macrophages, thereby improving survival and neurologic deficits following CA/CPR. The neuroprotective effects occur at least partially by interfering with the TRPM4 channel in the neurovascular unit. These results indicate the significant clinical potential of FFA to improve the prognosis for CA victims who are successfully resuscitated. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02571-2.
Collapse
Affiliation(s)
- Jiancong Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Yuan Chang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Juan Zhu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Yuqin Peng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Zheqi Li
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Kunxue Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Yuzhen Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Chuman Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Zhenzhou Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China.
| | - Kaibin Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China.
| |
Collapse
|
35
|
HMGA2 Promotes Brain Injury in Rats with Cerebral Infarction by Activating TLR4/NF-κB Signaling Pathway. Mediators Inflamm 2022; 2022:1376959. [PMID: 35966335 PMCID: PMC9371803 DOI: 10.1155/2022/1376959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/09/2022] [Indexed: 11/18/2022] Open
Abstract
Cerebral infarction is a common disease with a higher disability and fatality rates. The incidence rates of cerebral infarction or cerebral ischemic stroke gradually increase with aging and cerebrovascular disease progression. This study is aimed at evaluating the effects of HMGA2 on cerebral infarction-induced brain tissue damage and its underlying mechanisms. Adult Sprague Dawley rats were pretreated with sh-HMGA2 before cerebral infarction operation. The effect of HMGA2 on the arrangement, distribution, and morphological structure of neurons and the cell apoptosis ratio in brain tissue were detected via hematoxylin and eosin staining, brain-water content, TTC staining, and TUNEL staining. The results from ELISA assay, qPCR, and western blot indicated that downregulation of HMGA2 mitigated inflammatory stress via regulating the expression of TLR4/NF-κB. In addition, results showed that suppressed HMGA2 attenuated the neurological dysfunction of brain injury rats and markedly reduced infarct volume. HMGA2 might be able to alleviate the damage associated with cerebral infarction-induced inflammatory response and cell apoptosis. Moreover, downregulation of HMGA2 had a protective effect on the brain damage derived from cerebral infarction by mediating the TLR4/NF-κB pathway. In conclusion, the current study demonstrated that downregulation of HMGB2 decreased the infarct size, inflammatory responses, and apoptosis in cerebral injury and further had neuroprotective effects against cerebral infarction-induced brain damage. Finally, these results indicated that the downregulation of the TLR4/NF-κB pathway after ischemia by HMGB2 inhibition is a potential mechanism of the neuroprotective effect of cerebral injury.
Collapse
|
36
|
Xia P, Marjan M, Liu Z, Zhou W, Zhang Q, Cheng C, Zhao M, Tao Y, Wang Z, Ye Z. Chrysophanol postconditioning attenuated cerebral ischemia-reperfusion injury induced NLRP3-related pyroptosis in a TRAF6-dependent manner. Exp Neurol 2022; 357:114197. [PMID: 35932799 DOI: 10.1016/j.expneurol.2022.114197] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/28/2022] [Accepted: 07/31/2022] [Indexed: 11/04/2022]
Abstract
Individuals who suffer from post-CA (cardiac arrest) brain injury experience higher mortality and more severe functional disability. Neuroinflammation has been identified as a vital factor in cerebral ischemia-reperfusion injury (CIRI) following CA. Pyroptosis induces neuronal death by triggering an excessive inflammatory injury. Chrysophanol possesses robust anti-inflammatory features, and it is protective against CIRI. The purpose of this research was to assess the effect of Chrysophanol postconditioning on CIRI-induced pyroptotic cell death, and to explore its underlying mechanisms. CIRI was induced in rats by CA and subsequent cardiopulmonary resuscitation, and PC12 cells were exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) to imitate CIRI in vitro. It was found that post-CA brain injury led to a notable cerebral damage revealed by histopathological changes and neurological outcomes. The existence of pyroptosis was also confirmed in in vivo and in vitro CIRI models. Moreover, we further confirmed that Chrysophanol, the main bioactive ingredient of Rhubarb, significantly suppressed expressions of pyroptosis-associated proteins, e.g., NLRP3, ASC, cleaved-caspase-1 and N-terminal GSDMD, and inhibited the expression of tumor necrosis factor receptor-associated factor 6 (TRAF6). Furthermore, NLRP3 overexpression neutralized the neuroprotection of Chrysophanol postconditioning, suggesting that pyroptosis was the major neuronal death pathway modulated by Chrysophanol postconditioning in OGD/R. Additionally, the neuroprotection of Chrysophanol postconditioning was also abolished by gain-of-function analyses of TRAF6. Finally, the results demonstrated that Chrysophanol postconditioning suppressed the interaction between TRAF6 and NLRP3. Taken together, our findings revealed that Chrysophanol postconditioning was protective against CIRI by inhibiting NLRP3-related pyroptosis in a TRAF6-dependent manner.
Collapse
Affiliation(s)
- Pingping Xia
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha 410008, Hunan Province, China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, Hunan Province, China
| | - Murat Marjan
- Department of Anesthesiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054 Xinjiang, Uygur Autonomous Region, China
| | - Zhuoyi Liu
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha 410008, Hunan Province, China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, Hunan Province, China
| | - Wanqing Zhou
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha 410008, Hunan Province, China
| | - Qian Zhang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha 410008, Hunan Province, China
| | - Chen Cheng
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha 410008, Hunan Province, China
| | - Minxi Zhao
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha 410008, Hunan Province, China
| | - Yuanyuan Tao
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha 410008, Hunan Province, China
| | - Zhihua Wang
- Department of Anesthesiology, Hainan General Hospital, Haikou, Hainan Province, China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha 410008, Hunan Province, China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, Hunan Province, China.
| |
Collapse
|
37
|
Ehlting A, Zweyer M, Maes E, Schleehuber Y, Doshi H, Sabir H, Bernis ME. Impact of Hypoxia-Ischemia on Neurogenesis and Structural and Functional Outcomes in a Mild-Moderate Neonatal Hypoxia-Ischemia Brain Injury Model. Life (Basel) 2022; 12:life12081164. [PMID: 36013343 PMCID: PMC9410039 DOI: 10.3390/life12081164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a common type of brain injury caused by a lack of oxygen and blood flow to the brain during the perinatal period. The incidence of HIE is approximately 2−3 cases per 1000 live births in high-income settings; while in low- and middle-income countries, the incidence is 3−10-fold higher. Therapeutic hypothermia (TH) is the current standard treatment for neonates affected by moderate−severe HIE. However, more than 50% of all infants with suspected HIE have mild encephalopathy, and these infants are not treated with TH because of their lower risk of adverse outcomes. Despite this, several analyses of pooled data provide increasing evidence that infants who initially have mild encephalopathy may present signs of more significant brain injury later in life. The purpose of this study was to expand our knowledge about the effect of mild−moderate hypoxia-ischemia (HI) at the cellular, structural, and functional levels. An established rat model of mild−moderate HI was used, where postnatal day (P) 7 rats were exposed to unilateral permanent occlusion of the left carotid artery and 90 min of 8% hypoxia, followed by TH or normothermia (NT) treatment. The extent of injury was assessed using histology (P14 and P42) and MRI (P11 and P32), as well as with short-term and long-term behavioral tests. Neurogenesis was assessed by BrdU staining. We showed that mild−moderate HI leads to a progressive loss of brain tissue, pathological changes in MRI scans, as well as an impairment of long-term motor function. At P14, the median area loss assessed by histology for HI animals was 20% (p < 0.05), corresponding to mild−moderate brain injury, increasing to 55% (p < 0.05) at P42. The data assessed by MRI corroborated our results. HI led to a decrease in neurogenesis, especially in the hippocampus and the lateral ventricle at early time points, with a delayed partial recovery. TH was not neuroprotective at early time points following mild−moderate HI, but prevented the increase in brain damage over time. Additionally, rats treated with TH showed better long-term motor function. Altogether, our results bring more light to the understanding of pathophysiology following mild-moderate HI. We showed that, in the context of mild-moderate HI, TH failed to be significantly neuroprotective. However, animals treated with TH showed a significant improvement in motor, but not cognitive long-term function. These results are in line with what is observed in some cases where neonates with mild HIE are at risk of neurodevelopmental deficits in infancy or childhood. Whether TH should be used as a preventive treatment to reduce adverse outcomes in mild-HIE remains of active interest, and more research has to be carried out in order to address this question.
Collapse
Affiliation(s)
- Anne Ehlting
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany; (A.E.); (M.Z.); (E.M.); (Y.S.); (M.E.B.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany;
| | - Margit Zweyer
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany; (A.E.); (M.Z.); (E.M.); (Y.S.); (M.E.B.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany;
| | - Elke Maes
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany; (A.E.); (M.Z.); (E.M.); (Y.S.); (M.E.B.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany;
| | - Yvonne Schleehuber
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany; (A.E.); (M.Z.); (E.M.); (Y.S.); (M.E.B.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany;
| | - Hardik Doshi
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany;
| | - Hemmen Sabir
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany; (A.E.); (M.Z.); (E.M.); (Y.S.); (M.E.B.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany;
- Correspondence:
| | - Maria Eugenia Bernis
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany; (A.E.); (M.Z.); (E.M.); (Y.S.); (M.E.B.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany;
| |
Collapse
|
38
|
Analysis of Givinostat/ITF2357 Treatment in a Rat Model of Neonatal Hypoxic-Ischemic Brain Damage. Int J Mol Sci 2022; 23:ijms23158287. [PMID: 35955430 PMCID: PMC9368553 DOI: 10.3390/ijms23158287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 01/27/2023] Open
Abstract
The histone deacetylase inhibitor (HDACi) Givinostat/ITF2357 provides neuroprotection in adult models of brain injury; however, its action after neonatal hypoxia-ischemia (HI) is still undefined. The aim of our study was to test the hypothesis that the mechanism of Givinostat is associated with the alleviation of inflammation. For this purpose, we analyzed the microglial response and the effect on molecular mediators (chemokines/cytokines) that are crucial for inducing cerebral damage after neonatal hypoxia-ischemia. Seven-day-old rat pups were subjected to unilateral carotid artery ligation followed by 60 min of hypoxia (7.6% O2). Givinostat (10 mg/kg b/w) was administered in a 5-day regimen. The effects of Givinostat on HI-induced inflammation (cytokine, chemokine and microglial activation and polarization) were assessed with a Luminex assay, immunohistochemistry and Western blot. Givinostat treatment did not modulate the microglial response specific for HI injury. After Givinostat administration, the investigated chemokines and cytokines remained at the level induced by HI. The only immunosuppressive effect of Givinostat may be associated with the decrease in MIP-1α. Neonatal hypoxia-ischemia produces an inflammatory response by activating the proinflammatory M1 phenotype of microglia, disrupting the microglia–neuron (CX3CL1/CX3CR1) axis and elevating numerous proinflammatory cytokines/chemokines. Givinostat/ITF2357 did not prevent an inflammatory reaction after HI.
Collapse
|
39
|
The impact of the histone deacetylase inhibitor sodium butyrate on microglial polarization after oxygen and glucose deprivation. Pharmacol Rep 2022; 74:909-919. [PMID: 35796871 DOI: 10.1007/s43440-022-00384-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Microglia play a major role in the development of brain inflammation after central nervous system injury. On the other hand, microglia also participate in the repair process. The dualistic role of these cells results from the fact that various states of their activation are associated with specific phenotypes. The M1 phenotype is responsible for the production of proinflammatory mediators, whereas the M2 microglia release anti-inflammatory and trophic factors and take part in immunosuppressive and neuroprotective processes. The histone deacetylase inhibitor sodium butyrate (SB) shows anti-inflammatory and neuroprotective effects in some animal models of brain injury. The aim of this study was to examine the effects of sodium butyrate on the proliferation and M1/M2 polarization of primary microglial cells after oxygen and glucose deprivation (OGD) in vitro. METHODS Primary microglial cultures were prepared from 1-day-old rats, subjected to the OGD procedure and treated with SB (0.1 mM, 1 mM and 10 mM). The effect of OGD and SB on microglial proliferation was assessed by double immunofluorescence, and microglial phenotypes were evaluated by qPCR. RESULTS The OGD procedure stimulated the proliferation of microglia after 24 h of culturing, and SB treatment reduced the division of these cells. This effect was inversely proportional to the SB concentration. The OGD procedure increased proinflammatory CD86 and IL1β gene expression and reduced the expression of the anti-inflammatory M2 markers arginase and CD200 in microglia. CONCLUSIONS SB can change the polarization of microglia after OGD from an unfavourable M1 to a beneficial M2 phenotype. Our results show that SB is a potential immunosuppressive agent that can modulate microglial activation stimulated by ischaemic-like conditions.
Collapse
|
40
|
Zhou Y, Yang L, Liu X, Wang H. Lactylation may be a Novel Posttranslational Modification in Inflammation in Neonatal Hypoxic-Ischemic Encephalopathy. Front Pharmacol 2022; 13:926802. [PMID: 35721121 PMCID: PMC9202888 DOI: 10.3389/fphar.2022.926802] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/12/2022] [Indexed: 01/22/2023] Open
Abstract
Perinatal hypoxia-ischemia remains the most common cause of acute neonatal brain injury and is associated with a high death rate and long-term neurological abnormalities such as memory and cognitive deficits and dyskinesia. Hypoxia-ischemia triggers an inflammatory cascade in the brain that is amplified by the activation of immune cells and the influx of peripheral immune cells into the brain parenchyma in response to cellular injury. Thus, acute cerebral hypoxic-ischemic inflammation is a major contributor to the pathogenesis of newborn hypoxic-ischemic brain injury. Lactate is a glycolysis end product that can regulate inflammation through histone lactylation, a unique posttranslational modification that was identified in recent studies. The purpose of this review is to outline the recent improvements in our understanding of microglia-mediated hypoxic-ischemic inflammation and to further discuss how histone lactylation regulates inflammation by affecting macrophage activation. These findings may suggest that epigenetic reprogramming-associated lactate input is linked to disease outcomes such as acute neonatal brain injury pathogenesis and the therapeutic effects of drugs and other strategies in relieving neonatal hypoxic-ischemic brain injury. Therefore, improving our knowledge of the reciprocal relationships between histone lactylation and inflammation could lead to the development of new immunomodulatory therapies for brain damage in newborns.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Pharmacy, Xindu District People's Hospital of Chengdu, Chengdu, China
| | - Li Yang
- Department of Pharmacy, Xindu District People's Hospital of Chengdu, Chengdu, China
| | - Xiaoying Liu
- Department of Pharmacy, Xindu District People's Hospital of Chengdu, Chengdu, China
| | - Hao Wang
- Department of Pharmacy, Xindu District People's Hospital of Chengdu, Chengdu, China
| |
Collapse
|
41
|
Time Course of Changes in the Neurovascular Unit after Hypoxic-Ischemic Injury in Neonatal Rats. Int J Mol Sci 2022; 23:ijms23084180. [PMID: 35456999 PMCID: PMC9027443 DOI: 10.3390/ijms23084180] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 02/07/2023] Open
Abstract
Exposure to hypoxic-ischemic (HI) insults in newborns can predispose them to severe neurological sequela. The mechanisms underlying HI-related brain injury have not been completely elucidated. The neurovascular unit (NVU) is a composite of structures that protect the brain from the influx of detrimental molecules. Changes in the NVU after HI are important because they could reveal endogenous neuroprotective pathways in the cerebral microvasculature. Furthermore, the time course of changes in the NVU after exposure to HI in the newborn remains to be determined. In this study, we examined the effects of severe HI on the time course of changes in the NVU in neonatal rats. Brains were collected from rats exposed to right carotid artery ligation and 2 h of hypoxia on postnatal day 7 with recovery for 6 or 48 h after exposure to sham treatment (Sham) or HI. The right HI and left hypoxic alone sides of the brains were examined by quantitative immunohistochemistry for vascular density (laminin), pericyte vascular coverage (PDGFRβ), astrocyte vascular coverage (GFAP), and claudin-5 expression in the microvasculature of the cerebral cortex, white matter, and hippocampus. HI-related brain injury in neonatal rats was associated with increases in vascular density in the cortex and hippocampus 48 h after HI as well as neurovascular remodeling, including loss of pericyte coverage in the cortex and increases in claudin-5 in the hippocampus 6 h after HI. Astrocyte coverage was not affected by HI injury. The time course of the responses in the different components of the NVU varied after exposure to HI. There were also differential regional responses in the elements of the NVU in response to HI and hypoxia alone.
Collapse
|
42
|
Ding X, Pan T, Tian Q, Huang W, Hayashi LS, Liu Q, Li F, Xu LX, Miao P, Yang X, Sun B, Feng CX, Feng X, Li M, Huang J. Profiling Temporal Changes of the Pineal Transcriptomes at Single Cell Level Upon Neonatal HIBD. Front Cell Dev Biol 2022; 10:794012. [PMID: 35350377 PMCID: PMC8958010 DOI: 10.3389/fcell.2022.794012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022] Open
Abstract
Neonatal hypoxic-ischemic brain damage (HIBD) often results in various neurological deficits. Among them, a common, yet often neglected, symptom is circadian rhythm disorders. Previous studies revealed that the occurrence of cysts in the pineal gland, an organ known to regulate circadian rhythm, is associated with circadian problems in children with HIBD. However, the underlying mechanisms of pineal dependent dysfunctions post HIBD remain largely elusive. Here, by performing 10x single cell RNA sequencing, we firstly molecularly identified distinct pineal cell types and explored their transcriptome changes at single cell level at 24 and 72 h post neonatal HIBD. Bioinformatic analysis of cell prioritization showed that both subtypes of pinealocytes, the predominant component of the pineal gland, were mostly affected. We then went further to investigate how distinct pineal cell types responded to neonatal HIBD. Within pinealocytes, we revealed a molecularly defined β to α subtype conversion induced by neonatal HIBD. Within astrocytes, we discovered that all three subtypes responded to neonatal HIBD, with differential expression of reactive astrocytes markers. Two subtypes of microglia cells were both activated by HIBD, marked by up-regulation of Ccl3. Notably, microglia cells showed substantial reduction at 72 h post HIBD. Further investigation revealed that pyroptosis preferentially occurred in pineal microglia through NLRP3-Caspase-1-GSDMD signaling pathway. Taken together, our results delineated temporal changes of molecular and cellular events occurring in the pineal gland following neonatal HIBD. By revealing pyroptosis in the pineal gland, our study also provided potential therapeutic targets for preventing extravasation of pineal pathology and thus improving circadian rhythm dysfunction in neonates with HIBD.
Collapse
Affiliation(s)
- Xin Ding
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China
| | - Tao Pan
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China
| | - Qiuyan Tian
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Wenxi Huang
- Undergraduate Program, University of Virginia, Charlottesville, VA, United States
| | - Lauren S Hayashi
- IRTA Fellow, National Institutes of Health, Bethesda, MD, United States
| | - Qin Liu
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Fuyong Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Li-Xiao Xu
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Po Miao
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China
| | - Xiaofeng Yang
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China
| | - Bin Sun
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China
| | - Chen-Xi Feng
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Xing Feng
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, China.,Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China.,Undergraduate Program, University of Virginia, Charlottesville, VA, United States.,IRTA Fellow, National Institutes of Health, Bethesda, MD, United States.,School of Basic Medicine and Biological Sciences, Medical College of Soochow University, Suzhou, China
| | - Mei Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, China
| | - Jian Huang
- School of Basic Medicine and Biological Sciences, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
43
|
Xin DQ, Zhao YJ, Li TT, Ke HF, Gai CC, Guo XF, Chen WQ, Liu DX, Wang Z. The delivery of miR-21a-5p by extracellular vesicles induces microglial polarization via the STAT3 pathway following hypoxia-ischemia in neonatal mice. Neural Regen Res 2022; 17:2238-2246. [PMID: 35259844 PMCID: PMC9083169 DOI: 10.4103/1673-5374.336871] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Extracellular vesicles (EVs) from mesenchymal stromal cells (MSCs) have previously been shown to protect against brain injury caused by hypoxia-ischemia (HI). The neuroprotective effects have been found to relate to the anti-inflammatory effects of EVs. However, the underlying mechanisms have not previously been determined. In this study, we induced oxygen-glucose deprivation in BV-2 cells (a microglia cell line), which mimics HI in vitro, and found that treatment with MSCs-EVs increased the cell viability. The treatment was also found to reduce the expression of pro-inflammatory cytokines, induce the polarization of microglia towards the M2 phenotype, and suppress the phosphorylation of selective signal transducer and activator of transcription 3 (STAT3) in the microglia. These results were also obtained in vivo using neonatal mice with induced HI. We investigated the potential role of miR-21a-5p in mediating these effects, as it is the most highly expressed miRNA in MSCs-EVs and interacts with the STAT3 pathway. We found that treatment with MSCs-EVs increased the levels of miR-21a-5p in BV-2 cells, which had been lowered following oxygen-glucose deprivation. When the level of miR-21a-5p in the MSCs-EVs was reduced, the effects on microglial polarization and STAT3 phosphorylation were reduced, for both the in vitro and in vivo HI models. These results indicate that MSCs-EVs attenuate HI brain injury in neonatal mice by shuttling miR-21a-5p, which induces microglial M2 polarization by targeting STAT3.
Collapse
Affiliation(s)
- Dan-Qing Xin
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Yi-Jing Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Ting-Ting Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Hong-Fei Ke
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Cheng-Cheng Gai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiao-Fan Guo
- Department of Neurology, Loma Linda University Health, Loma Linda, CA, USA
| | - Wen-Qiang Chen
- Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - De-Xiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
44
|
Temporal Characterization of Microglia-Associated Pro- and Anti-Inflammatory Genes in a Neonatal Inflammation-Sensitized Hypoxic-Ischemic Brain Injury Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2479626. [PMID: 35281473 PMCID: PMC8906938 DOI: 10.1155/2022/2479626] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/22/2021] [Accepted: 01/27/2022] [Indexed: 02/06/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) mainly affects preterm and term newborns, leading to a high risk of brain damage. Coexisting infection/inflammation and birth asphyxia are key factors associated with intracerebral increase of proinflammatory cytokines linked to HIE. Microglia are key mediators of inflammation during perinatal brain injury, characterized by their phenotypic plasticity, which may facilitate their participation in both the progression and resolution of injury-induced inflammation. The purpose of this study was to investigate the temporal expression of genes associated with pro- and anti-inflammatory cytokines as well as the nucleotide-binding domain, leucine-rich repeat protein (NLRP-3) inflammasome from microglia cells. For this purpose, we used our established neonatal rat model of inflammation-sensitized hypoxic-ischemic (HI) brain injury in seven-day-old rats. We assessed gene expression profiles of 11 cytokines and for NLRP-3 using real-time PCR from sorted CD11b/c microglia of brain samples at different time points (3.5 h after LPS injection and 0, 5, 24, 48, and 72 hours post HI) following different treatments: vehicle, E. coli lipopolysaccharide (LPS), vehicle/HI, and LPS/HI. Our results showed that microglia are early key mediators of the inflammatory response and exacerbate the inflammatory response following HI, polarizing into a predominant proinflammatory M1 phenotype in the early hours post HI. The brains only exposed to HI showed a delay in the expression of proinflammatory cytokines. We also demonstrated that NLRP-3 plays a role in the inflammatory resolution with a high expression after HI insult. The combination of both, a preinfection/inflammation condition and hypoxia-ischemia, resulted in a higher proinflammatory cytokine storm, highlighting the significant contribution of acute inflammation sensitizing prior to a hypoxic insult on the severity of perinatal brain damage.
Collapse
|
45
|
Huang Z, Luo Z, Ovcjak A, Wan J, Chen NH, Hu W, Sun HS, Feng ZP. AD-16 Protects Against Hypoxic-Ischemic Brain Injury by Inhibiting Neuroinflammation. Neurosci Bull 2022; 38:857-870. [PMID: 35072896 PMCID: PMC9352839 DOI: 10.1007/s12264-021-00816-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/19/2021] [Indexed: 11/28/2022] Open
Abstract
Neuroinflammation is a key contributor to the pathogenic cascades induced by hypoxic-ischemic (HI) insult in the neonatal brain. AD-16 is a novel anti-inflammatory compound, recently found to exert potent inhibition of the lipopolysaccharide-induced production of pro-inflammatory and neurotoxic mediators. In this study, we evaluated the effect of AD-16 on primary astrocytes and neurons under oxygen-glucose deprivation (OGD) in vitro and in mice with neonatal HI brain injury in vivo. We demonstrated that AD-16 protected against OGD-induced astrocytic and neuronal cell injury. Single dose post-treatment with AD-16 (1 mg/kg) improved the neurobehavioral outcome and reduced the infarct volume with a therapeutic window of up to 6 h. Chronic administration reduced the mortality rate and preserved whole-brain morphology following neonatal HI. The in vitro and in vivo effects suggest that AD-16 offers promising therapeutic efficacy in attenuating the progression of HI brain injury and protecting against the associated mortality and morbidity.
Collapse
Affiliation(s)
- Zhihua Huang
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Zhengwei Luo
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Andrea Ovcjak
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Jiangfan Wan
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wenhui Hu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
46
|
Plinia trunciflora Extract Administration Prevents HI-Induced Oxidative Stress, Inflammatory Response, Behavioral Impairments, and Tissue Damage in Rats. Nutrients 2022; 14:nu14020395. [PMID: 35057576 PMCID: PMC8779767 DOI: 10.3390/nu14020395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/14/2022] Open
Abstract
The disruption of redox homeostasis and neuroinflammation are key mechanisms in the pathogenesis of brain hypoxia–ischemia (HI); medicinal plants have been studied as a therapeutic strategy, generally associated with the prevention of oxidative stress and inflammatory response. This study evaluates the neuroprotective role of the Plinia trunciflora fruit extract (PTE) in neonatal rats submitted to experimental HI. The HI insult provoked a marked increase in the lipoperoxidation levels and glutathione peroxidase (GPx) activity, accompanied by a decrease in the brain concentration of glutathione (GSH). Interestingly, PTE was able to prevent most of the HI-induced pro-oxidant effects. It was also observed that HI increased the levels of interleukin-1β in the hippocampus, and that PTE-treatment prevented this effect. Furthermore, PTE was able to prevent neuronal loss and astrocyte reactivity induced by HI, as demonstrated by NeuN and GFAP staining, respectively. PTE also attenuated the anxiety-like behavior and prevented the spatial memory impairment caused by HI. Finally, PTE prevented neural tissue loss in the brain hemisphere, the hippocampus, cerebral cortex, and the striatum ipsilateral to the HI. Taken together our results provide good evidence that the PTE extract has the potential to be investigated as an adjunctive therapy in the treatment of brain insult caused by neonatal hypoxia–ischemia.
Collapse
|
47
|
Dong X, Luo S, Hu D, Cao R, Wang Q, Meng Z, Feng Z, Zhou W, Song W. Gallic acid inhibits neuroinflammation and reduces neonatal hypoxic-ischemic brain damages. Front Pediatr 2022; 10:973256. [PMID: 36619526 PMCID: PMC9813953 DOI: 10.3389/fped.2022.973256] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Neuroinflammation is a leading cause of secondary neuronal injury in neonatal hypoxic-ischemic encephalopathy (HIE). Regulation of neuroinflammation may be beneficial for treatment of HIE and its secondary complications. Gallic acid (GA) has been shown to have anti-inflammatory and antioxidant effects. In this report we found that oxygen-glucose deprivation and/reoxygenation (OGD/R)-induced cell death, and the generation of excessive reactive oxygen species (ROS) and inflammatory cytokines by microglia were inhibited by GA treatment. Furthermore, GA treatment reduced neuroinflammation and neuronal loss, and alleviated motor and cognitive impairments in rats with hypoxic-ischemic brain damage (HIBD). Together, our results reveal that GA is an effective regulator of neuroinflammation and has potential as a pharmaceutical intervention for HIE therapy.
Collapse
Affiliation(s)
- Xiangjun Dong
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Shuyue Luo
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Dongjie Hu
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ruixue Cao
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qunxian Wang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zijun Meng
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zijuan Feng
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Weihui Zhou
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Weihong Song
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, China
| |
Collapse
|
48
|
Wang R, Li L, Wang B. Poncirin ameliorates oxygen glucose deprivation/reperfusion injury in cortical neurons via inhibiting NOX4-mediated NLRP3 inflammasome activation. Int Immunopharmacol 2022; 102:107210. [PMID: 34266770 DOI: 10.1016/j.intimp.2020.107210] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/01/2020] [Accepted: 11/11/2020] [Indexed: 12/25/2022]
Abstract
Poncirin, a natural flavonoid present abundantly in citrus fruits, possesses anti-oxidant and anti-inflammatory activities that contribute to neuroprotection, but its roles and mechanisms in neuronal injury is still poorly understood. In this study, an oxygen-glucose deprivation/reoxygenation (OGD/R) model was established in primary cortical neurons to induce neuronal injury in vitro. Poncirin effectively attenuated OGD/R-induced neuronal damage by enhancing cell viability, restraining lactate dehydrogenase release, and reducing apoptosis of neurons. Poncirin restrained mitochondrial dysfunction and oxidative stress by increasing mitochondrial membrane potential, declining reactive oxygen species production, lessening malondialdehyde generation, and increasing the activities of antioxidant enzymes in OGD/R-treated neurons. Poncirin also repressed inflammatory responses by reducing the secretion of pro-inflammatory factors, and inhibiting NLRP3 inflammasome activation. Importantly, poncirin administration notably abolished OGD/R-induced upregulation of NADPH oxidase 4 (NOX4), and overexpression of NOX4 neutralized poncirin-mediated neuroprotection. In conclusion, poncirin protects cortical neurons from OGD/R injury via inhibiting NOX4/ROS/NLRP3 axis.
Collapse
Affiliation(s)
- Ruili Wang
- Department of Pediatrics, Zhoukou Central Hospital, Zhoukou 466000, Henan, China.
| | - Lei Li
- Department of Pediatrics, Zhoukou Central Hospital, Zhoukou 466000, Henan, China
| | - Baogong Wang
- Department of Cardiology, Zhoukou Central Hospital, Zhoukou 466000, Henan, China
| |
Collapse
|
49
|
Ding Y, Zhang M, Li C, Xie B, Zhao G, Sun Y. RETRACTED ARTICLE: A reusable aptasensor based on the dual signal amplification of Ce@AuNRs-PAMAM-Fc and DNA walker for ultrasensitive detection of TNF-α. J Solid State Electrochem 2022. [DOI: 10.1007/s10008-020-04885-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
50
|
Kurt A, Zenciroğlu A, Akduman H. The impact of therapeutic hypothermia on peripheral blood cell in newborns with hypoxic ischemic encephalopathy. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e181053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|