1
|
Yang M, Wang K, Liu B, Shen Y, Liu G. Hypoxic-Ischemic Encephalopathy: Pathogenesis and Promising Therapies. Mol Neurobiol 2025; 62:2105-2122. [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] [MESH Headings] [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
|
2
|
Mohammadi A, Balduini W, Carloni S. Melatonin modulates the Notch1 signaling pathway and Sirt3 in the hippocampus of hypoxic-ischemic neonatal rats. Sci Rep 2024; 14:25069. [PMID: 39443594 PMCID: PMC11500095 DOI: 10.1038/s41598-024-76307-y] [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: 12/29/2023] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
The Notch1 signaling pathway plays a crucial role in the development of the central nervous system, governing pivotal functional activities in the brain, such as neurogenesis. Sirt3 is instrumental in managing mitochondrial homeostasis and is essential to cell survival. Dysregulation of these signaling pathways is implicated in the pathogenesis of a wide range of diseases, including neurodegenerative disorders such as stroke. We have previously shown that melatonin significantly improved the perinatal brain damage caused by hypoxia-ischemia (HI) through the activation of several protective mechanisms such as restoring mitochondria status and increasing the hippocampal cell proliferation. This study assessed whether melatonin affects the Notch1 signaling pathway and Sirt3 after neonatal HI. Results show that HI significantly increased Notch1 expression both in hippocampal neurons and glial cells as well as the expression of the key proteins of the pathway NICD, HES1, and c-Myc. Melatonin significantly prevented the Notch1 signaling pathway activation induced by HI, maintaining NICD and HES1 expression to control levels. In the same neurons, melatonin also prevents the Sirt3 depletion caused by HI. In summary, this study provides new insights into the effects of melatonin on the Notch1 signaling pathway and Sirt3 in in vivo neonatal brain ischemia. We suggest that the rapid modulation of the Notch1 signaling pathway and Sirt3 induced by melatonin may support neuronal survival during ischemia.
Collapse
Affiliation(s)
- Atefeh Mohammadi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via S. Chiara 27, 61029, Urbino (PU), Italy
| | - Water Balduini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via S. Chiara 27, 61029, Urbino (PU), Italy
| | - Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via S. Chiara 27, 61029, Urbino (PU), Italy.
| |
Collapse
|
3
|
Erdos T, Masuda M, Venketaraman V. Glutathione in HIV-Associated Neurocognitive Disorders. Curr Issues Mol Biol 2024; 46:5530-5549. [PMID: 38921002 PMCID: PMC11202908 DOI: 10.3390/cimb46060330] [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: 03/21/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
A large portion of patients with Human Immunodeficiency Virus (HIV) have neurologic sequelae. Those with better-controlled HIV via antiretroviral therapies generally have less severe neurologic symptoms. However, for many patients, antiretrovirals do not adequately resolve symptoms. Since much of the pathogenesis of HIV/AIDS (Autoimmune Deficiency Syndrome) involves oxidative stress either directly, through viral interaction, or indirectly, through inflammatory mechanisms, we have reviewed relevant trials of glutathione supplementation in each of the HIV-associated neurocognitive diseases and have found disease-specific results. For diseases for which trials have not been completed, predicted responses to glutathione supplementation are made based on relevant mechanisms seen in the literature. It is not sufficient to conclude that all HIV-associated neurocognitive disorders (HAND) will benefit from the antioxidant effects of glutathione supplementation. The potential effects of glutathione supplementation in patients with HAND are likely to differ based on the specific HIV-associated neurocognitive disease.
Collapse
Affiliation(s)
| | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (T.E.); (M.M.)
| |
Collapse
|
4
|
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: 1.0] [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
|
5
|
Saini KK, Upadhyay RK, Kant R, Vajpayee A, Jain K, Kumar A, Kumar LS, Kumar R. Design, synthesis, molecular docking and DFT studies on novel melatonin and isatin based azole derivatives. RSC Adv 2023; 13:27525-27534. [PMID: 37720826 PMCID: PMC10500251 DOI: 10.1039/d3ra05531k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023] Open
Abstract
In order to address the pressing demand for newer broad-spectrum antifungal medicines with enhanced activity, computer modelling was utilised to rationally develop newer antifungal azole-based drugs. Based on the drug and active sites of the Lanosterol 14 alpha-Demethylases (LAD) of the prominent fungal pathogen Candida albicans interaction, Novel triazole-linked melatonin and isatin derivatives 7a-d and 8a-d were synthesised using bioisosterism. Besides the experimental synthesis and subsequent characterization, the present study focused on obtaining optimised geometries, frequency calculations, and TD-DFT studies of the synthesised molecules. We also performed molecular docking studies to explore the inhibitory ability of the synthesised compounds against the active sites of the Lanosterol 14 alpha-Demethylases (LAD) of the prominent fungal pathogen Candida albicans. The binding interactions resulted in positive findings, demonstrating the involvement of the synthesised compounds in the suppression of fungal growth. Comparative analysis of the binding potential of the synthesised molecules and commercially available drug fluconazole revealed a remarkable note: the docking scores for the designed drugs 7b, 7c, and 8c are much greater than those of the fluconazole molecule. The in silico study of the designed series of drug molecules serves as an important guideline for further exploration in the quest for potent antifungal agents.
Collapse
Affiliation(s)
- Keshav Kumar Saini
- Department of Chemistry, University of Delhi Delhi 110007 India
- Department of Chemistry, Dyal Singh College, University of Delhi Lodhi Road New Delhi 110003 India
| | - Ravindra Kumar Upadhyay
- Department of Chemistry, University of Delhi Delhi 110007 India
- Department of Chemistry, Sri Venkateswara College, University of Delhi New Delhi 110021 India
| | - Ravi Kant
- Department of Chemistry, Government Post Graduate College G.B. Nagar Noida UP 201301 India
| | - Arpita Vajpayee
- Department of Physics, Dyal Singh College, University of Delhi Lodhi Road New Delhi 110003 India
| | - Kalpana Jain
- Department of Physics, D. J. College Baraut UP 250611 India
| | - Amit Kumar
- Department of Chemistry, Dyal Singh College, University of Delhi Lodhi Road New Delhi 110003 India
| | - Lalita S Kumar
- Chemistry Discipline, School of Sciences, Indira Gandhi National Open University New Delhi 110068 India
| | - Rakesh Kumar
- Department of Chemistry, University of Delhi Delhi 110007 India
| |
Collapse
|
6
|
Chen X, Zhang J, Lin Y, Li Y, Wang H, Wang Z, Liu H, Hu Y, Liu L. Mechanism, prevention and treatment of cognitive impairment caused by high altitude exposure. Front Physiol 2023; 14:1191058. [PMID: 37731540 PMCID: PMC10507266 DOI: 10.3389/fphys.2023.1191058] [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: 03/21/2023] [Accepted: 06/05/2023] [Indexed: 09/22/2023] Open
Abstract
Hypobaric hypoxia (HH) characteristics induce impaired cognitive function, reduced concentration, and memory. In recent years, an increasing number of people have migrated to high-altitude areas for work and study. Headache, sleep disturbance, and cognitive impairment from HH, severely challenges the physical and mental health and affects their quality of life and work efficiency. This review summarizes the manifestations, mechanisms, and preventive and therapeutic methods of HH environment affecting cognitive function and provides theoretical references for exploring and treating high altitude-induced cognitive impairment.
Collapse
Affiliation(s)
- Xin Chen
- Department of Clinical Laboratory Medicine, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jiexin Zhang
- Department of Clinical Laboratory Medicine, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
- Faculty of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Yuan Lin
- Sichuan Xincheng Biological Co., LTD., Chengdu, Sichuan, China
| | - Yan Li
- Department of General Surgery, The 77th Army Hospital, Leshan, Sichuan, China
| | - Han Wang
- Department of Cardiology, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Zhanhao Wang
- Department of Clinical Laboratory Medicine, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Huawei Liu
- Department of Clinical Laboratory Medicine, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Yonghe Hu
- Faculty of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Lei Liu
- Medical Research Center, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Purcell E, Nguyen T, Smith M, Penny T, Paton MCB, Zhou L, Jenkin G, Miller SL, McDonald CA, Malhotra A. Factors Influencing the Efficacy of Umbilical Cord Blood-Derived Cell Therapy for Perinatal Brain Injury. Stem Cells Transl Med 2023; 12:125-139. [PMID: 36847059 PMCID: PMC10021495 DOI: 10.1093/stcltm/szad006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/16/2023] [Indexed: 03/01/2023] Open
Abstract
INTRODUCTION We have previously described preclinical literature which supports umbilical cord blood-derived cell (UCBC) therapy as an efficacious treatment for perinatal brain injury. However, efficacy of UCBCs may be influenced by different patient population and intervention characteristics. OBJECTIVES To systematically review the effects of UCBCs on brain outcomes in animal models of perinatal brain injury across subgroups to better understand the contribution of model type (preterm versus term), brain injury type, UCB cell type, route of administration, timing of intervention, cell dosage, and number of doses. METHODS A systematic search of MEDLINE and Embase databases was performed to identify studies using UCBC therapy in animal models of perinatal brain injury. Subgroup differences were measured by chi2 test where possible. RESULTS Differential benefits of UCBCs were seen across a number of subgroup analyses including intraventricular hemorrhage (IVH) vs. hypoxia ischemia (HI) model (apoptosis white matter (WM): chi2 = 4.07; P = .04, neuroinflammation-TNF-α: chi2 = 5.99; P = .01), UCB-derived mesenchymal stromal cells (MSCs) vs. UCB-derived mononuclear cells (MNCs) (oligodendrocyte WM: chi2 = 5.01; P = .03, neuroinflammation-TNF-α: chi2 = 3.93; P = .05, apoptosis grey matter (GM), astrogliosis WM), and intraventricular/intrathecal vs. systemic routes of administration (microglial activation GM: chi2 = 7.51; P = .02, astrogliosis WM: chi2 = 12.44; P = .002). We identified a serious risk of bias and overall low certainty of evidence. CONCLUSIONS Preclinical evidence suggests UCBCs to show greater efficacy in the injury model of IVH compared to HI, the use of UCB-MSCs compared to UCB-MNCs and the use of local administrative routes compared to systemic routes in animal models of perinatal brain injury. Further research is needed to improve certainty of evidence and address knowledge gaps.
Collapse
Affiliation(s)
| | | | - Madeleine Smith
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Tayla Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Madison C B Paton
- Cerebral Palsy Alliance Research Institute, & Speciality of Child and Adolescent Health, The University of Sydney, Sydney, Australia
| | - Lindsay Zhou
- Department of Paediatrics, Monash University, Melbourne, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | | | - Atul Malhotra
- Corresponding author: Atul Malhotra, Department of Paediatrics, Monash University, 246 Clayton Road, Clayton, VIC 3168, Australia.
| |
Collapse
|
8
|
Okazaki K, Nakamura S, Koyano K, Konishi Y, Kondo M, Kusaka T. Neonatal asphyxia as an inflammatory disease: Reactive oxygen species and cytokines. Front Pediatr 2023; 11:1070743. [PMID: 36776908 PMCID: PMC9911547 DOI: 10.3389/fped.2023.1070743] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Neonatologists resuscitate asphyxiated neonates by every available means, including positive ventilation, oxygen therapy, and drugs. Asphyxiated neonates sometimes present symptoms that mimic those of inflammation, such as fever and edema. The main pathophysiology of the asphyxia is inflammation caused by hypoxic-ischemic reperfusion. At birth or in the perinatal period, neonates may suffer several, hypoxic insults, which can activate inflammatory cells and inflammatory mediator production leading to the release of larger quantities of reactive oxygen species (ROS). This in turn triggers the production of oxygen stress-induced high mobility group box-1 (HMGB-1), an endogenous damage-associated molecular patterns (DAMPs) protein bound to toll-like receptor (TLR) -4, which activates nuclear factor-kappa B (NF-κB), resulting in the production of excess inflammatory mediators. ROS and inflammatory mediators are produced not only in activated inflammatory cells but also in non-immune cells, such as endothelial cells. Hypothermia inhibits pro-inflammatory mediators. A combination therapy of hypothermia and medications, such as erythropoietin and melatonin, is attracting attention now. These medications have both anti-oxidant and anti-inflammatory effects. As the inflammatory response and oxidative stress play a critical role in the pathophysiology of neonatal asphyxia, these drugs may contribute to improving patient outcomes.
Collapse
Affiliation(s)
- Kaoru Okazaki
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kosuke Koyano
- Maternal Perinatal Center, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Masatoshi Kondo
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
9
|
Pluta R, Furmaga-Jabłońska W, Januszewski S, Tarkowska A. Melatonin: A Potential Candidate for the Treatment of Experimental and Clinical Perinatal Asphyxia. Molecules 2023; 28:1105. [PMID: 36770769 PMCID: PMC9919754 DOI: 10.3390/molecules28031105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Perinatal asphyxia is considered to be one of the major causes of brain neurodegeneration in full-term newborns. The worst consequence of perinatal asphyxia is neurodegenerative brain damage, also known as hypoxic-ischemic encephalopathy. Hypoxic-ischemic encephalopathy is the leading cause of mortality in term newborns. To date, due to the complex mechanisms of brain damage, no effective or causal treatment has been developed that would ensure complete neuroprotection. Although hypothermia is the standard of care for hypoxic-ischemic encephalopathy, it does not affect all changes associated with encephalopathy. Therefore, there is a need to develop effective treatment strategies, namely research into new agents and therapies. In recent years, it has been pointed out that natural compounds with neuroprotective properties, such as melatonin, can be used in the treatment of hypoxic-ischemic encephalopathy. This natural substance with anti-inflammatory, antioxidant, anti-apoptotic and neurofunctional properties has been shown to have pleiotropic prophylactic or therapeutic effects, mainly against experimental brain neurodegeneration in hypoxic-ischemic neonates. Melatonin is a natural neuroprotective hormone, which makes it promising for the treatment of neurodegeneration after asphyxia. It is supposed that melatonin alone or in combination with hypothermia may improve neurological outcomes in infants with hypoxic-ischemic encephalopathy. Melatonin has been shown to be effective in the last 20 years of research, mainly in animals with perinatal asphyxia but, so far, no clinical trials have been performed on a sufficient number of newborns. In this review, we summarize the advantages and limitations of melatonin research in the treatment of experimental and clinical perinatal asphyxia.
Collapse
Affiliation(s)
- Ryszard Pluta
- Ecotech-Complex Analytical and Programme Centre for Advanced Environmentally-Friendly Technologies, Marie Curie-Skłodowska University in Lublin, 20-612 Lublin, Poland
| | - Wanda Furmaga-Jabłońska
- Department of Neonate and Infant Pathology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Sławomir Januszewski
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Agata Tarkowska
- Department of Neonate and Infant Pathology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
10
|
Essawy AE, Mohamed AI, Ali RG, Ali AM, Abdou HM. Analysis of Melatonin-Modulating Effects Against Tartrazine-Induced Neurotoxicity in Male Rats: Biochemical, Pathological and Immunohistochemical Markers. Neurochem Res 2023; 48:131-141. [PMID: 36018437 PMCID: PMC9823072 DOI: 10.1007/s11064-022-03723-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 07/20/2022] [Accepted: 08/06/2022] [Indexed: 01/11/2023]
Abstract
Tartrazine (E-102) is one of the most widely used artificial food azo-colors that can be metabolized to highly sensitizing aromatic amines such as sulphanilic acid. These metabolites are oxidized to N-hydroxy derivatives that cause neurotoxicity. Melatonin is a neurohormone. That possesses a free-radical scavenging effect. The present work was mainly designed to evaluate the possible ameliorative role of melatonin against tartrazine induced neurotoxicity in cerebral cortex and cerebellum of male rats. Adult male rats were administered orally with tartrazine (7.5 mg/kg) with or without melatonin (10 mg/kg) daily for four weeks. The data revealed that tartrazine induced redox disruptions as measured by significant (p < 0.05) increased malondialdehyde (MDA) level and inhibition of (GSH) concentration and catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx) antioxidant enzyme activities. Besides, brain acetyl cholin (Ach) and gamma-aminobutyric acid (GABA) were elevated while, dopamine (DA) was depleted in trtrazine -treated rats. Moreover, tartrazine caused a significant (p < 0.05) increase in the brain interleukin-6 (IL-6), interleukin-1β (IL-1 β) and tumor necrosis factor-α (TNFα). At the tissue level, tartrazine caused severe histopathological changes in the cerebellum and cerebral cortex of rats. The immunohistochemical results elucidated strong positive expression for Caspase-3 and GFAP and weak immune reaction for BcL2 and synaptophysin in tatrazine- treated rats. The administration of melatonin to tartrazine -administered rats remarkably alleviated all the aforementioned tartrzine-induced effects. It could be concluded that, melatonin has a potent ameliorative effect against tartrazine induced neurotoxicity via the attenuation of oxidative/antioxidative responses.
Collapse
Affiliation(s)
- Amina E Essawy
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | | | - Rania Gaber Ali
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Awatef M Ali
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Heba Mohamed Abdou
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
11
|
Duranti A, Beldarrain G, Álvarez A, Sbriscia M, Carloni S, Balduini W, Alonso-Alconada D. The Endocannabinoid System as a Target for Neuroprotection/Neuroregeneration in Perinatal Hypoxic-Ischemic Brain Injury. Biomedicines 2022; 11:biomedicines11010028. [PMID: 36672536 PMCID: PMC9855621 DOI: 10.3390/biomedicines11010028] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
The endocannabinoid (EC) system is a complex cell-signaling system that participates in a vast number of biological processes since the prenatal period, including the development of the nervous system, brain plasticity, and circuit repair. This neuromodulatory system is also involved in the response to endogenous and environmental insults, being of special relevance in the prevention and/or treatment of vascular disorders, such as stroke and neuroprotection after neonatal brain injury. Perinatal hypoxia-ischemia leading to neonatal encephalopathy is a devastating condition with no therapeutic approach apart from moderate hypothermia, which is effective only in some cases. This overview, therefore, gives a current description of the main components of the EC system (including cannabinoid receptors, ligands, and related enzymes), to later analyze the EC system as a target for neonatal neuroprotection with a special focus on its neurogenic potential after hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence: (A.D.); (D.A.-A.); Tel.: +39-0722-303501 (A.D.); +34-946-013294 (D.A.-A.)
| | - Gorane Beldarrain
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Antonia Álvarez
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Matilde Sbriscia
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Walter Balduini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Daniel Alonso-Alconada
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Correspondence: (A.D.); (D.A.-A.); Tel.: +39-0722-303501 (A.D.); +34-946-013294 (D.A.-A.)
| |
Collapse
|
12
|
Ferroptosis: A Promising Therapeutic Target for Neonatal Hypoxic-Ischemic Brain Injury. Int J Mol Sci 2022; 23:ijms23137420. [PMID: 35806425 PMCID: PMC9267109 DOI: 10.3390/ijms23137420] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023] Open
Abstract
Ferroptosis is a type of programmed cell death caused by phospholipid peroxidation that has been implicated as a mechanism in several diseases resulting from ischemic-reperfusion injury. Most recently, ferroptosis has been identified as a possible key injury mechanism in neonatal hypoxic-ischemic brain injury (HIBI). This review summarizes the current literature regarding the different ferroptotic pathways, how they may be activated after neonatal HIBI, and which current or investigative interventions may attenuate ferroptotic cell death associated with neonatal HIBI.
Collapse
|
13
|
Delayed Therapeutic Administration of Melatonin Enhances Neuronal Survival Through AKT and MAPK Signaling Pathways Following Focal Brain Ischemia in Mice. J Mol Neurosci 2022; 72:994-1007. [PMID: 35307786 DOI: 10.1007/s12031-022-01995-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 03/01/2022] [Indexed: 10/18/2022]
Abstract
Melatonin has a role in the cell survival signaling pathways as a candidate for secondary stroke prevention. Therefore, in the present study, the coordination of ipsilateral and contralateral hemispheres to evaluate delayed post-acute effect of melatonin was examined on recovery of the cell survival and apoptosis after stroke. Melatonin was administered (4 mg/kg/day) intraperitoneally for 45 days, starting 3 days after 30 min of middle cerebral artery occlusion. The genes and proteins related to the cell survival and apoptosis were investigated by immunofluorescence, western blotting, and RT-PCR techniques after behavioral experiments. Melatonin produced delayed neurological recovery by improving motor coordination on grip strength and rotarod tests. This neurological recovery was also reflected by high level of NeuN positive cells and low level of TUNEL-positive cells suggesting enhanced neuronal survival and reduced apoptosis at the fifty-fifth day of stroke. The increase of NGF, Nrp1, c-jun; activation of AKT; and dephosphorylation of ERK and JNK at the fifty-fifth day showed that cell survival and apoptosis signaling molecules compete to contribute to the remodeling of brain. Furthermore, an increase in the CREB and Atf-1 expressions suggested the melatonin's strong reformative effect on neuronal regeneration. The contralateral hemisphere was more active at the latter stages of the molecular and functional regeneration which provides a further proof of principle about melatonin's action on the promotion of brain plasticity and recovery after stroke.
Collapse
|
14
|
Lee BL, Glass HC. Cognitive outcomes in late childhood and adolescence of neonatal hypoxic-ischemic encephalopathy. Clin Exp Pediatr 2021; 64:608-618. [PMID: 34044480 PMCID: PMC8650814 DOI: 10.3345/cep.2021.00164] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/18/2021] [Indexed: 12/04/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is the most common cause of neonatal encephalopathy with a global incidence of approximately 1 to 8 per 1,000 live births. Neonatal encephalopathy can cause neurodevelopmental and cognitive impairments in survivors of hypoxic-ischemic insults with and without functional motor deficits. Normal neurodevelopmental outcomes in early childhood do not preclude cognitive and behavioral difficulties in late childhood and adolescence because cognitive functions are not yet fully developed at this early age. Therapeutic hypothermia has been shown to significantly reduced death and severe disabilities in term newborns with HIE. However, children treated with hypothermia therapy remain at risk for cognitive impairments and follow-up is necessary throughout late childhood and adolescence. Novel adjunctive neuroprotective therapies combined with therapeutic hypothermia may enhance the survival and neurodevelopmental outcomes of infants with HIE. The extent and severity of brain injury on magnetic resonance imaging might predict neurodevelopmental outcomes and lead to targeted interven tions in children with a history of neonatal encephalopathy. We provide a summary of the long-term cognitive outcomes in late childhood and adolescence in children with a history of HIE and the association between pattern of brain injury and neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Bo Lyun Lee
- Department of Pediatrics, Inje University Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Hannah C Glass
- Department of Neurology and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.,Department of Pediatrics, Benioff Children's Hospital, University of California San Francisco, San Francisco, CA, USA.,Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
15
|
Wesselhoeft R, Rasmussen L, Jensen PB, Jennum PJ, Skurtveit S, Hartz I, Reutfors J, Damkier P, Bliddal M, Pottegård A. Use of hypnotic drugs among children, adolescents, and young adults in Scandinavia. Acta Psychiatr Scand 2021; 144:100-112. [PMID: 34021908 DOI: 10.1111/acps.13329] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 01/11/2023]
Abstract
BACKGROUND Hypnotic use in children and adolescents is controversial. OBJECTIVE To describe the use of hypnotic drugs (melatonin, z-drugs, and sedating antihistamines) among 5- to 24-year-old Scandinavians during 2012 to 2018. METHODS Aggregate-level data were obtained from public data sources in Sweden, Norway, and Denmark. We calculated annual prevalence (users/1000 inhabitants) stratified by age group, sex, and country. Quantity of use (Defined Daily Dose (DDD)/user/day) was estimated for Norway and Denmark. RESULTS Melatonin was the most commonly used hypnotic, and its use increased markedly from 2012 to 2018, particularly among females and 15- to 24-year-old individuals. Sweden had the highest increase in use (6.5 to 25/1000) compared with Norway (10-20/1000) and Denmark (5.7-12/1000). The annual prevalence of sedating antihistamine use was also highest in Sweden, reaching 13/1000 in 2018 in comparison to 7.5/1000 in Norway and 2.5/1000 in Denmark. Z-drug use decreased in all countries toward 2018, dropping to 3.5/1000 in Sweden, 4.4/1000 in Norway, and 1.7/1000 in Denmark. The quantity of hypnotic use in Norway and Denmark was 0.8-1.0 DDD/user/day for melatonin in 2018, as compared to 0.1-0.3 for z-drugs and antihistamines. CONCLUSION The use of melatonin and sedating antihistamines increased among young Scandinavians during 2012-2018, and the increase was twice as high in Sweden compared with Norway and Denmark. In addition, Sweden had the highest use of sedating antihistamines. The Scandinavian variation of hypnotic use could reflect differences in frequency of sleep problems between populations or variation of healthcare access or clinical practice between countries.
Collapse
Affiliation(s)
- Rikke Wesselhoeft
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark.,Mental Health Services in the Region of Southern Denmark, Odense, Denmark
| | - Lotte Rasmussen
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Peter Bjødstrup Jensen
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | | | - Svetlana Skurtveit
- Department of Mental Disorders, Norwegian Institute of Public Health, Oslo, Norway
| | - Ingeborg Hartz
- Department of Mental Disorders, Norwegian Institute of Public Health, Oslo, Norway.,Innlandet Hospital Trust, Hedmark, Norway
| | - Johan Reutfors
- Centre for Pharmacoepidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Per Damkier
- Department of Clinical Biochemistry & Pharmacology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mette Bliddal
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark.,OPEN - Open Patient data Explorative Network, Department of Clinical Research, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Anton Pottegård
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
16
|
Reyes-Corral M, Sola-Idígora N, de la Puerta R, Montaner J, Ybot-González P. Nutraceuticals in the Prevention of Neonatal Hypoxia-Ischemia: A Comprehensive Review of their Neuroprotective Properties, Mechanisms of Action and Future Directions. Int J Mol Sci 2021; 22:2524. [PMID: 33802413 PMCID: PMC7959318 DOI: 10.3390/ijms22052524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/22/2022] Open
Abstract
Neonatal hypoxia-ischemia (HI) is a brain injury caused by oxygen deprivation to the brain due to birth asphyxia or reduced cerebral blood perfusion, and it often leads to lifelong limiting sequelae such as cerebral palsy, seizures, or mental retardation. HI remains one of the leading causes of neonatal mortality and morbidity worldwide, and current therapies are limited. Hypothermia has been successful in reducing mortality and some disabilities, but it is only applied to a subset of newborns that meet strict inclusion criteria. Given the unpredictable nature of the obstetric complications that contribute to neonatal HI, prophylactic treatments that prevent, rather than rescue, HI brain injury are emerging as a therapeutic alternative. Nutraceuticals are natural compounds present in the diet or used as dietary supplements that have antioxidant, anti-inflammatory, or antiapoptotic properties. This review summarizes the preclinical in vivo studies, mostly conducted on rodent models, that have investigated the neuroprotective properties of nutraceuticals in preventing and reducing HI-induced brain damage and cognitive impairments. The natural products reviewed include polyphenols, omega-3 fatty acids, vitamins, plant-derived compounds (tanshinones, sulforaphane, and capsaicin), and endogenous compounds (melatonin, carnitine, creatine, and lactate). These nutraceuticals were administered before the damage occurred, either to the mothers as a dietary supplement during pregnancy and/or lactation or to the pups prior to HI induction. To date, very few of these nutritional interventions have been investigated in humans, but we refer to those that have been successful in reducing ischemic stroke in adults. Overall, there is a robust body of preclinical evidence that supports the neuroprotective properties of nutraceuticals, and these may represent a safe and inexpensive nutritional strategy for the prevention of neonatal HI encephalopathy.
Collapse
Affiliation(s)
- Marta Reyes-Corral
- Neurodevelopment Research Group, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain; (M.R.-C.); (N.S.-I.); (P.Y.-G.)
| | - Noelia Sola-Idígora
- Neurodevelopment Research Group, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain; (M.R.-C.); (N.S.-I.); (P.Y.-G.)
| | - Rocío de la Puerta
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain;
| | - Joan Montaner
- Neurovascular Research Lab, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain
- Department of Neurology and Neurophysiology, Hospital Universitario Virgen Macarena, 41009 Seville, Spain
| | - Patricia Ybot-González
- Neurodevelopment Research Group, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain; (M.R.-C.); (N.S.-I.); (P.Y.-G.)
- Department of Neurology and Neurophysiology, Hospital Universitario Virgen Macarena, 41009 Seville, Spain
| |
Collapse
|
17
|
Garofoli F, Longo S, Pisoni C, Accorsi P, Angelini M, Aversa S, Caporali C, Cociglio S, De Silvestri A, Fazzi E, Rizzo V, Tzialla C, Zecca M, Orcesi S. Oral melatonin as a new tool for neuroprotection in preterm newborns: study protocol for a randomized controlled trial. Trials 2021; 22:82. [PMID: 33482894 PMCID: PMC7820522 DOI: 10.1186/s13063-021-05034-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/08/2021] [Indexed: 11/12/2022] Open
Abstract
Background Prevention of neurodevelopmental impairment due to preterm birth is a major health challenge. Despite advanced obstetric and neonatal care, to date there are few neuroprotective molecules available. Melatonin has been shown to have anti-oxidant/anti-inflammatory effects and to reduce brain damage, mainly after hypoxic ischemic encephalopathy. The planned study will be the first aiming to evaluate the capacity of melatonin to mitigate brain impairment due to premature birth. Method In our planned prospective, multicenter, double-blind, randomized vs placebo study, we will recruit, within 96 h of birth, 60 preterm newborns with a gestational age ≤ 29 weeks + 6 days; these infants will be randomly allocated to oral melatonin, 3 mg/kg/day, or placebo for 15 days. After the administration period, we will measure plasma levels of malondialdehyde, a lipid peroxidation product considered an early biological marker of melatonin treatment efficacy (primary outcome). At term-equivalent age, we will evaluate neurological status (through cerebral ultrasound, cerebral magnetic resonance imaging, vision and hearing evaluations, clinical neurological assessment, and screening for retinopathy of prematurity) as well as the incidence of bronchodysplasia and sepsis. We will also monitor neurodevelopmental outcome during the first 24 months of corrected age (using the modified Fagan Test of Infant Intelligence at 4–6 months and standardized neurological and developmental assessments at 24 months). Discussion Preterm birth survivors often present long-term neurodevelopmental sequelae, such as motor, learning, social-behavioral, and communication problems. We aim to assess the role of melatonin as a neuroprotectant during the first weeks of extrauterine life, when preterm infants are unable to produce it spontaneously. This approach is based on the supposition that its anti-oxidant mechanism could be useful in preventing neurodevelopmental impairment. Considering the short- and long-term morbidities related to preterm birth, and the financial and social costs of the care of preterm infants, both at birth and over time, we suggest that melatonin administration could lead to considerable saving of resources. This would be the first study addressing the role of melatonin in very low birth weight preterm newborns, and it could provide a basis for further studies on melatonin as a neuroprotection strategy in this vulnerable population. Trial registration ClinicalTrials.gov NCT04235673. Prospectively registered on 22 January 2020.
Collapse
Affiliation(s)
- Francesca Garofoli
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Piazzale Golgi 1, 27100, Pavia, Italy
| | - Stefania Longo
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Piazzale Golgi 1, 27100, Pavia, Italy
| | - Camilla Pisoni
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Piazzale Golgi 1, 27100, Pavia, Italy.
| | - Patrizia Accorsi
- Child and Adolescence Neuropsychiatry Unit, Children's Hospital, ASST Spedali Civili of Brescia, 25123, Brescia, Italy
| | - Micol Angelini
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Piazzale Golgi 1, 27100, Pavia, Italy
| | - Salvatore Aversa
- Neonatal Unit and Neonatal Intensive Care Unit, Children's Hospital, ASST Spedali Civili of Brescia, 25123, Brescia, Italy
| | - Camilla Caporali
- Child Neurology and Psychiatry Unit, Department of Brain and Behavioral Sciences, University of Pavia, 27100, Pavia, Italy.,Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, 27100, Pavia, Italy
| | - Sara Cociglio
- Child Neurology and Psychiatry Unit, Department of Brain and Behavioral Sciences, University of Pavia, 27100, Pavia, Italy
| | - Annalisa De Silvestri
- Unit of Clinical Epidemiology & Biometry, Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Elisa Fazzi
- Child and Adolescence Neuropsychiatry Unit, Children's Hospital, ASST Spedali Civili of Brescia, 25123, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, 25123, Brescia, Italy
| | - Vittoria Rizzo
- Clinical Chemistry Laboratory and Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Chryssoula Tzialla
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Piazzale Golgi 1, 27100, Pavia, Italy
| | - Marco Zecca
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Piazzale Golgi 1, 27100, Pavia, Italy
| | - Simona Orcesi
- Child Neurology and Psychiatry Unit, Department of Brain and Behavioral Sciences, University of Pavia, 27100, Pavia, Italy.,Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, 27100, Pavia, Italy
| |
Collapse
|
18
|
Mohsenpour H, Pesce M, Patruno A, Bahrami A, Pour PM, Farzaei MH. A Review of Plant Extracts and Plant-Derived Natural Compounds in the Prevention/Treatment of Neonatal Hypoxic-Ischemic Brain Injury. Int J Mol Sci 2021; 22:E833. [PMID: 33467663 PMCID: PMC7830094 DOI: 10.3390/ijms22020833] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Neonatal hypoxic-ischemic (HI) brain injury is one of the major drawbacks of mortality and causes significant short/long-term neurological dysfunction in newborn infants worldwide. To date, due to multifunctional complex mechanisms of brain injury, there is no well-established effective strategy to completely provide neuroprotection. Although therapeutic hypothermia is the proven treatment for hypoxic-ischemic encephalopathy (HIE), it does not completely chang outcomes in severe forms of HIE. Therefore, there is a critical need for reviewing the effective therapeutic strategies to explore the protective agents and methods. In recent years, it is widely believed that there are neuroprotective possibilities of natural compounds extracted from plants against HIE. These natural agents with the anti-inflammatory, anti-oxidative, anti-apoptotic, and neurofunctional regulatory properties exhibit preventive or therapeutic effects against experimental neonatal HI brain damage. In this study, it was aimed to review the literature in scientific databases that investigate the neuroprotective effects of plant extracts/plant-derived compounds in experimental animal models of neonatal HI brain damage and their possible underlying molecular mechanisms of action.
Collapse
Affiliation(s)
- Hadi Mohsenpour
- Department of Pediatrics, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah 75333–67427, Iran;
| | - Mirko Pesce
- Department of Medicine and Aging Sciences, University G. d’Annunzio, 66100 Chieti, Italy
| | - Antonia Patruno
- Department of Medicine and Aging Sciences, University G. d’Annunzio, 66100 Chieti, Italy
| | - Azam Bahrami
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 67158-47141, Iran;
| | - Pardis Mohammadi Pour
- Department of Pharmacognosy, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran;
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 67158-47141, Iran;
| |
Collapse
|
19
|
Rodriguez J, Li T, Xu Y, Sun Y, Zhu C. Role of apoptosis-inducing factor in perinatal hypoxic-ischemic brain injury. Neural Regen Res 2021; 16:205-213. [PMID: 32859765 PMCID: PMC7896227 DOI: 10.4103/1673-5374.290875] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Perinatal complications, such as asphyxia, can cause brain injuries that are often associated with subsequent neurological deficits, such as cerebral palsy or mental retardation. The mechanisms of perinatal brain injury are not fully understood, but mitochondria play a prominent role not only due to their central function in metabolism but also because many proteins with apoptosis-related functions are located in the mitochondrion. Among these proteins, apoptosis-inducing factor has already been shown to be an important factor involved in neuronal cell death upon hypoxia-ischemia, but a better understanding of the mechanisms behind these processes is required for the development of more effective treatments during the early stages of perinatal brain injury. In this review, we focus on the molecular mechanisms of hypoxic-ischemic encephalopathy, specifically on the importance of apoptosis-inducing factor. The relevance of apoptosis-inducing factor is based not only because it participates in the caspase-independent apoptotic pathway but also because it plays a crucial role in mitochondrial energetic functionality, especially with regard to the maintenance of electron transport during oxidative phosphorylation and in oxidative stress, acting as a free radical scavenger. We also discuss all the different apoptosis-inducing factor isoforms discovered, focusing especially on apoptosis-inducing factor 2, which is only expressed in the brain and the functions of which are starting now to be clarified. Finally, we summarized the interaction of apoptosis-inducing factor with several proteins that are crucial for both apoptosis-inducing factor functions (pro-survival and pro-apoptotic) and that are highly important in order to develop promising therapeutic targets for improving outcomes after perinatal brain injury.
Collapse
Affiliation(s)
- Juan Rodriguez
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tao Li
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yiran Xu
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yanyan Sun
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Anatomy, School of Basic Medical Science, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Changlian Zhu
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Pisani F, Fusco C, Nagarajan L, Spagnoli C. Acute symptomatic neonatal seizures, brain injury, and long-term outcome: The role of neuroprotective strategies. Expert Rev Neurother 2020; 21:189-203. [PMID: 33176104 DOI: 10.1080/14737175.2021.1848547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Neonatal seizures are frequent but underdiagnosed manifestations of acute brain dysfunction and an important contributor to unfavorable outcomes. Etiology and severity of brain injury are the single strongest outcome determinants. AREAS COVERED The authors will discuss the prognostic role of acute symptomatic seizures versus brain injury and the main neuroprotective and neurorestorative strategies for full-term and preterm infants. EXPERT OPINION Prolonged acute symptomatic seizures likely contribute to long-term outcomes by independently adding further brain injury to initial insults. Correct timing and dosing of therapeutic interventions, depending on etiology and gestational ages, need careful evaluation. Although promising strategies are under study, the only standard of care is whole-body therapeutic hypothermia in full-term newborns with hypoxic-ischemic encephalopathy.
Collapse
Affiliation(s)
- Francesco Pisani
- Child Neuropsychiatric Unit, Medicine and Surgery Department, University of Parma , Parma, Italy
| | - Carlo Fusco
- Child Neurology Unit, Department of Paediatrics, Azienda USL-IRCCS Di Reggio Emilia , Reggio Emilia, Italy
| | - Lakshmi Nagarajan
- Department of Neurology, Perth Children's Hospital, University of Western Australia , Perth, Australia
| | - Carlotta Spagnoli
- Child Neurology Unit, Department of Paediatrics, Azienda USL-IRCCS Di Reggio Emilia , Reggio Emilia, Italy
| |
Collapse
|
21
|
Sun R, Wang Y, He H, Wan Y, Li L, Sha J, Jiang G, Li Y, Li T, Ren B. Solubility measurement, solubility behavior analysis and thermodynamic modelling of melatonin in twelve pure solvents from 278.15 K to 323.15 K. J Mol Liq 2020. [DOI: 10.1016/j.molliq.2020.114139] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
22
|
Hurley T, O'Dea M, Aslam S, Aly H, Robertson N, Molloy E. Melatonin treatment for newborns with hypoxic ischaemic encephalopathy. Hippokratia 2020. [DOI: 10.1002/14651858.cd013754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Tim Hurley
- Department of Paediatrics; Trinity College Dublin; Dublin Ireland
| | - Mary O'Dea
- Paediatric and Child Health; Trinity College Dublin; Dublin Ireland
| | - Saima Aslam
- Neonatology; National Maternity Hospital; Dublin Ireland
| | - Hany Aly
- Neonatology; Cleveland Clinic Children’s Hospital; Cleveland OH USA
| | - Nikki Robertson
- Obstetrics and Gynaecology; University College London; London UK
| | - Eleanor Molloy
- Paediatric and Child Health; Trinity College Dublin; Dublin Ireland
- Department of Paediatrics; The National Children’s Hospital, Tallaght; Dublin Ireland
| |
Collapse
|
23
|
Zhang X, Peng K, Zhang X. The Function of the NMDA Receptor in Hypoxic-Ischemic Encephalopathy. Front Neurosci 2020; 14:567665. [PMID: 33117117 PMCID: PMC7573650 DOI: 10.3389/fnins.2020.567665] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/28/2020] [Indexed: 12/17/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is one of the main forms of neonatal brain injury which could lead to neonatal disability or even cause neonatal death. Therefore, HIE strongly affects the health of newborns and brings heavy burden to the family and society. It has been well studied that N-methyl-D-aspartate (NMDA) receptors are involved in the excitotoxicity induced by hypoxia ischemia in adult brain. Recently, it has been shown that the NMDA receptor also plays important roles in HIE. In the present review, we made a summary of the molecular mechanism of NMDA receptor in the pathological process of HIE, focusing on the distinct role of GluN2A- and GluN2B-containing NMDA receptor subtypes and aiming to provide some insights into the clinical treatment and drug development of HIE.
Collapse
|
24
|
Adjunctive Neuroprotective Therapies for Treatment of Hypoxic-Ischemic Encephalopathy (HIE). CURRENT PEDIATRICS REPORTS 2020. [DOI: 10.1007/s40124-020-00231-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
25
|
Abstract
OBJECTIVES To investigate the effect of adding melatonin to hypothermia treatment on neurodevelopmental outcomes in asphyctic newborns. DESIGN Pilot multicenter, randomized, controlled, double-blind clinical trial. Statistical comparison of results obtained in two intervention arms: hypothermia plus placebo and hypothermia plus melatonin. SETTING Level 3 neonatal ICU. PATIENTS Twenty-five newborns were recruited. INTERVENTIONS The hypothermia plus melatonin patients received a daily dose of IV melatonin, 5 mg per kg body weight, for 3 days. General laboratory variables were measured both at neonatal ICU admission and after intervention. All infants were studied with amplitude-integrated electroencephalography and brain MRI within the first week of life. The neurodevelopmental Bayley III test, the Gross Motor Function Classification System, and the Tardieu scale were applied at the ages of 6 and 18 months. MEASUREMENTS AND MAIN RESULTS Clinical characteristics, laboratory evaluations, MRI findings, and amplitude-integrated electroencephalography background did not differ between the treatment groups. The newborns in the hypothermia plus melatonin group achieved a significantly higher composite score for the cognitive section of the Bayley III test at 18 months old, with respect to the hypothermia plus placebo group (p = 0.05). There were no differences between the groups according to the Gross Motor Function Classification System and Tardieu motor assessment scales. CONCLUSIONS The early addition of IV melatonin to asphyctic neonates is feasible and may improve long-term neurodevelopment. To our knowledge, this is the first clinical trial to analyze the administration of IV melatonin as an adjuvant therapy to therapeutic hypothermia.
Collapse
|
26
|
D’Angelo G, Chimenz R, Reiter RJ, Gitto E. Use of Melatonin in Oxidative Stress Related Neonatal Diseases. Antioxidants (Basel) 2020; 9:antiox9060477. [PMID: 32498356 PMCID: PMC7346173 DOI: 10.3390/antiox9060477] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species have a crucial role in the pathogenesis of perinatal diseases. Exposure to inflammation, infections, or high oxygen concentrations is frequent in preterm infants, who have high free iron levels that enhance toxic radical generation and diminish antioxidant defense. The peculiar susceptibility of newborns to oxidative stress supports the prophylactic use of melatonin in preventing or decreasing oxidative stress-mediated diseases. Melatonin, an effective direct free-radical scavenger, easily diffuses through biological membranes and exerts pleiotropic activity everywhere. Multiple investigations have assessed the effectiveness of melatonin to reduce the “oxygen radical diseases of newborn” including perinatal brain injury, sepsis, chronic lung disease (CLD), and necrotizing enterocolitis (NEC). Further studies are still awaited to test melatonin activity during perinatal period.
Collapse
Affiliation(s)
- Gabriella D’Angelo
- Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
- Correspondence: ; Tel.: +39-090-221-3100; Fax: +39-090-221-3876
| | - Roberto Chimenz
- Unit of Pediatric Nephrology and Rheumatology with Dialysis, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 40729, USA;
| | - Eloisa Gitto
- Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
27
|
Wang SY, Shi XC, Laborda P. Indole-based melatonin analogues: Synthetic approaches and biological activity. Eur J Med Chem 2020; 185:111847. [DOI: 10.1016/j.ejmech.2019.111847] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/01/2019] [Accepted: 11/01/2019] [Indexed: 12/11/2022]
|
28
|
Tan HY, Ng KY, Koh RY, Chye SM. Pharmacological Effects of Melatonin as Neuroprotectant in Rodent Model: A Review on the Current Biological Evidence. Cell Mol Neurobiol 2020; 40:25-51. [PMID: 31435851 PMCID: PMC11448813 DOI: 10.1007/s10571-019-00724-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/07/2019] [Indexed: 12/21/2022]
Abstract
The progressive loss of structure and functions of neurons, including neuronal death, is one of the main factors leading to poor quality of life. Promotion of functional recovery of neuron after injury is a great challenge in neuroregenerative studies. Melatonin, a hormone is secreted by pineal gland and has antioxidative, anti-inflammatory, and anti-apoptotic properties. Besides that, melatonin has high cell permeability and is able to cross the blood-brain barrier. Apart from that, there are no reported side effects associated with long-term usage of melatonin at both physiological and pharmacological doses. Thus, in this review article, we summarize the pharmacological effects of melatonin as neuroprotectant in central nervous system injury, ischemic-reperfusion injury, optic nerve injury, peripheral nerve injury, neurotmesis, axonotmesis, scar formation, cell degeneration, and apoptosis in rodent models.
Collapse
Affiliation(s)
- Hui Ying Tan
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Rhun Yian Koh
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia.
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
29
|
Abstract
Advances in neonatology have led to unprecedented improvements in neonatal survival such that those born as early as 22 weeks of gestation now have some chance of survival, and over 70% of those born at 24 weeks of gestation survive. Up to 50% of infants born extremely preterm develop poor outcomes involving long-term neurodevelopmental impairments affecting cognition and learning, or motor problems such as cerebral palsy. Poor outcomes arise because the preterm brain is vulnerable both to direct injury (by events such as intracerebral hemorrhage, infection, and/or hypoxia), or indirect injury due to disruption of normal development. This neonatal brain injury and/or dysmaturation is called "encephalopathy of prematurity". Current and future strategies to improve outcomes in this population include prevention of preterm birth, and pre-, peri-, and postnatal approaches to protect the developing brain. This review will describe mechanisms of preterm brain injury, and current and upcoming therapies in the antepartum and postnatal period to improve preterm encephalopathy.
Collapse
Affiliation(s)
- Pratik Parikh
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, WA.
| | - Sandra E Juul
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, WA.
| |
Collapse
|
30
|
Albrecht M, Zitta K, Groenendaal F, van Bel F, Peeters-Scholte C. Neuroprotective strategies following perinatal hypoxia-ischemia: Taking aim at NOS. Free Radic Biol Med 2019; 142:123-131. [PMID: 30818057 DOI: 10.1016/j.freeradbiomed.2019.02.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/07/2019] [Accepted: 02/19/2019] [Indexed: 12/13/2022]
Abstract
Perinatal asphyxia is characterized by oxygen deprivation and lack of perfusion in the perinatal period, leading to hypoxic-ischemic encephalopathy and sequelae such as cerebral palsy, mental retardation, cerebral visual impairment, epilepsy and learning disabilities. On cellular level PA is associated with a decrease in oxygen and glucose leading to ATP depletion and a compromised mitochondrial function. Upon reoxygenation and reperfusion, the renewed availability of oxygen gives rise to not only restoration of cell function, but also to the activation of multiple detrimental biochemical pathways, leading to secondary energy failure and ultimately, cell death. The formation of reactive oxygen species, nitric oxide and peroxynitrite plays a central role in the development of subsequent neurological damage. In this review we give insight into the pathophysiology of perinatal asphyxia, discuss its clinical relevance and summarize current neuroprotective strategies related to therapeutic hypothermia, ischemic postconditioning and pharmacological interventions. The review will also focus on the possible neuroprotective actions and molecular mechanisms of the selective neuronal and inducible nitric oxide synthase inhibitor 2-iminobiotin that may represent a novel therapeutic agent for the treatment of hypoxic-ischemic encephalopathy, both in combination with therapeutic hypothermia in middle- and high-income countries, as well as stand-alone treatment in low-income countries.
Collapse
Affiliation(s)
- Martin Albrecht
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Karina Zitta
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Floris Groenendaal
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands; Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Frank van Bel
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands; Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cacha Peeters-Scholte
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands; Neurophyxia BV, 's Hertogenbosch, the Netherlands.
| |
Collapse
|
31
|
Shimomura ET, Briones AJ, Gordon CJ, Warren WS, Jackson GF. Case report of sudden death in a twin infant given melatonin supplementation: A challenging interpretation of postmortem toxicology. Forensic Sci Int 2019; 304:109962. [PMID: 31610334 DOI: 10.1016/j.forsciint.2019.109962] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/12/2019] [Accepted: 09/19/2019] [Indexed: 11/17/2022]
Abstract
Melatonin (MEL) is a neurohormone in humans produced in a number of locations. Starting with the amino acid tryptophan, MEL is produced through a number of enzymatic steps that includes serotonin as an intermediate compound. The primary production of MEL is in the pineal gland located in the brain. It is directly associated with the the suprachiasmatic nucleus (SCN) located in the hypothalamus. In young and adult humans, the blood levels of MEL are typically in the picogram levels and produced in a cyclic schedule highly regulated by light detected in the retina by intrinsically photosensitive retinal ganglion cells (ipRGCs), resulting in production primarily during periods of darkness. During periods of light, MEL levels are typically very low or undetectable. Basal levels of MEL in infants have been observed to be either undetectable or also in the picogram levels, although some medical treatment has involved administration of exogenous MEL resulting in peak levels in the nanogram range. MEL is considered to be well tolerated and there have been limited reports of toxicity. In this case, an infant was found unresponsive and cause of death was ruled as Undetermined. Melatonin was detected in the peripheral blood at a concentration of 1,400ng/mL.
Collapse
Affiliation(s)
- Eric T Shimomura
- Division of Forensic Toxicology, Armed Forces Medical Examiner System, 115 Purple Heart Drive, Dover AFB, DE, 19902, United States
| | - Alice J Briones
- Office of the Armed Forces Medical Examiner, Armed Forces Medical Examiner System, 115 Purple Heart Drive, Dover AFB, DE, 19902, United States.
| | - Christopher J Gordon
- 71st Medical Group, 527 Gott Road, Building 810, Vance AFB, OK, 73705, United States
| | - Wendy S Warren
- Office of the Armed Forces Medical Examiner, Armed Forces Medical Examiner System, 115 Purple Heart Drive, Dover AFB, DE, 19902, United States
| | - George F Jackson
- Division of Forensic Toxicology, Armed Forces Medical Examiner System, 115 Purple Heart Drive, Dover AFB, DE, 19902, United States.
| |
Collapse
|
32
|
Liu F, Wang Y, Yao W, Xue Y, Zhou J, Liu Z. Geniposide attenuates neonatal mouse brain injury after hypoxic-ischemia involving the activation of PI3K/Akt signaling pathway. J Chem Neuroanat 2019; 102:101687. [PMID: 31562918 DOI: 10.1016/j.jchemneu.2019.101687] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 12/22/2022]
Abstract
Perinatal hypoxic-ischemia (HI) is a leading cause of acute mortality and neurologic complications in newborns. Geniposide, a natural product extracted from the herb Gardenia jasminoides, has been shown to possess neuroprotective effects in neurologic deficits. This study aims to investigate whether Geniposide has therapeutic potential to HI brain injury and the underlying mechanisms. C57/bl6 mice were subjected to HI insult on postnatal day 10. Geniposide (20 mg/kg b.w.) was administered intragastrically every day after HI insult for 7 successional days. Then mice at P18 were sacrificed and brain tissues were collected for further analysis. Geniposide treatment significantly inhibited cell apoptosis, reduced serum IgG leakage into brain tissue, attenuated astrogliosis and microgliosis, prevented loss of pericytes, loss of tight junction and adherens junction proteins. The PI3K/Akt signaling pathway, which related proteins were downregulated after HI insult, was activated by Geniposide treatment. Geniposide treatment after neonatal HI insult attenuated HI-induced cell apoptosis, IgG leakage, microgliosis, astrogliosis, pericytes loss and junction protein degradation. Geniposide could protect against HI-induced brain injury, which might be through the activation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Fang Liu
- Department of Pediatrics, Zibo Central Hospital, Zibo 255000, Shandong, China
| | - Yanxia Wang
- Department of Pediatrics, Zibo Central Hospital, Zibo 255000, Shandong, China.
| | - Wenjing Yao
- Department of Pediatrics, Zibo Central Hospital, Zibo 255000, Shandong, China
| | - Yuanyuan Xue
- Department of Pediatrics, Zibo Central Hospital, Zibo 255000, Shandong, China
| | - Jianqin Zhou
- Department of Pediatrics, Zibo Central Hospital, Zibo 255000, Shandong, China
| | - Zhaohong Liu
- Department of Pediatrics, Zibo Central Hospital, Zibo 255000, Shandong, China
| |
Collapse
|
33
|
Tsang JKW, Liu J, Lo ACY. Vascular and Neuronal Protection in the Developing Retina: Potential Therapeutic Targets for Retinopathy of Prematurity. Int J Mol Sci 2019; 20:E4321. [PMID: 31484463 PMCID: PMC6747312 DOI: 10.3390/ijms20174321] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/21/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a common retinal disease in preterm babies. To prolong the lives of preterm babies, high oxygen is provided to mimic the oxygen level in the intrauterine environment for postnatal organ development. However, hyperoxia-hypoxia induced pathological events occur when babies return to room air, leading to ROP with neuronal degeneration and vascular abnormality that affects retinal functions. With advances in neonatal intensive care, it is no longer uncommon for increased survival of very-low-birth-weight preterm infants, which, therefore, increased the incidence of ROP. ROP is now a major cause of preventable childhood blindness worldwide. Current proven treatment for ROP is limited to invasive retinal ablation, inherently destructive to the retina. The lack of pharmacological treatment for ROP creates a great need for effective and safe therapies in these developing infants. Therefore, it is essential to identify potential therapeutic agents that may have positive ROP outcomes, especially in preserving retinal functions. This review gives an overview of various agents in their efficacy in reducing retinal damages in cell culture tests, animal experiments and clinical studies. New perspectives along the neuroprotective pathways in the developing retina are also reviewed.
Collapse
Affiliation(s)
- Jessica K W Tsang
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jin Liu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Amy C Y Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
34
|
|
35
|
Cánovas-Ahedo M, Alonso-Alconada D. [Combined therapy in neonatal hypoxic-ischaemic encephalopathy]. An Pediatr (Barc) 2019; 91:59.e1-59.e7. [PMID: 31109785 DOI: 10.1016/j.anpedi.2019.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 01/01/2023] Open
Abstract
Neonatal hypoxic-ischaemic encephalopathy due to the lack of oxygen at birth can have severe neurological consequences, such as cerebral palsy, or even the death of the asphyxiated newborn. Hypothermia is currently the only therapy included in intensive care neonatal units. This shows a clinical benefit in neonates suffering from hypoxic-ischaemic encephalopathy, mainly because of its ability to decrease the accumulation of excitatory amino acids and its anti-inflammatory, antioxidant, and anti-apoptotic effects. However, hypothermia is not effective in half of the cases, making it necessary to search for new, or to optimize current therapies, with the aim on reducing asphyxia-derived neurological consequences, either as single treatments or in combination with cooling. Within current potential therapies, melatonin, allopurinol, and erythropoietin stand out among the others, with clinical trials on the way. While, stem cells, N-acetylcysteine and noble gases have obtained promising pre-clinical results. Melatonin produces a powerful antioxidant and anti-inflammatory effect, acting as free radical scavenger and regulating pro-inflammatory mediators. Through the inhibition of xanthine oxidase, allopurinol can decrease oxidative stress. Erythropoietin has cell death and neurogenesis as its main therapeutic targets. Keeping in mind the whole scenario of current therapies, management of neonates suffering from neonatal asphyxia could rely on the combination of one or some of these treatments, together with therapeutic hypothermia.
Collapse
Affiliation(s)
- María Cánovas-Ahedo
- Departamento de Biología Celular e Histología, Facultad de Medicina y Enfermería, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Bizkaia, España
| | - Daniel Alonso-Alconada
- Departamento de Biología Celular e Histología, Facultad de Medicina y Enfermería, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Bizkaia, España.
| |
Collapse
|
36
|
Qin X, Cheng J, Zhong Y, Mahgoub OK, Akter F, Fan Y, Aldughaim M, Xie Q, Qin L, Gu L, Jian Z, Xiong X, Liu R. Mechanism and Treatment Related to Oxidative Stress in Neonatal Hypoxic-Ischemic Encephalopathy. Front Mol Neurosci 2019; 12:88. [PMID: 31031592 PMCID: PMC6470360 DOI: 10.3389/fnmol.2019.00088] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/21/2019] [Indexed: 12/24/2022] Open
Abstract
Hypoxic ischemic encephalopathy (HIE) is a type of neonatal brain injury, which occurs due to lack of supply and oxygen deprivation to the brain. It is associated with a high morbidity and mortality rate. There are several therapeutic strategies that can be used to improve outcomes in patients with HIE. These include cell therapies such as marrow mesenchymal stem cells (MSCs) and umbilical cord blood stem cells (UCBCs), which are being incorporated into the new protocols for the prevention of ischemic brain damage. The focus of this review is to discuss the mechanism of oxidative stress in HIE and summarize the current available treatments for HIE. We hope that a better understanding of the relationship between oxidative stress and HIE will provide new insights on the potential therapy of this devastating condition.
Collapse
Affiliation(s)
- Xingping Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Neurosurgery, Harvard Medical School, Boston, MA, United States
| | - Jing Cheng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Omer Kamal Mahgoub
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Farhana Akter
- Department of Neurosurgery, Harvard Medical School, Boston, MA, United States.,Department of Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Yanqin Fan
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mohammed Aldughaim
- Department of Neurosurgery, Harvard Medical School, Boston, MA, United States
| | - Qiurong Xie
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lingxia Qin
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Renzhong Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
37
|
Melatonin as a master regulator of cell death and inflammation: molecular mechanisms and clinical implications for newborn care. Cell Death Dis 2019; 10:317. [PMID: 30962427 PMCID: PMC6453953 DOI: 10.1038/s41419-019-1556-7] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 03/19/2019] [Indexed: 12/11/2022]
Abstract
Melatonin, more commonly known as the sleep hormone, is mainly secreted by the pineal gland in dark conditions and regulates the circadian rhythm of the organism. Its intrinsic properties, including high cell permeability, the ability to easily cross both the blood–brain and placenta barriers, and its role as an endogenous reservoir of free radical scavengers (with indirect extra activities), confer it beneficial uses as an adjuvant in the biomedical field. Melatonin can exert its effects by acting through specific cellular receptors on the plasma membrane, similar to other hormones, or through receptor-independent mechanisms that involve complex molecular cross talk with other players. There is increasing evidence regarding the extraordinary beneficial effects of melatonin, also via exogenous administration. Here, we summarize molecular pathways in which melatonin is considered a master regulator, with attention to cell death and inflammation mechanisms from basic, translational and clinical points of view in the context of newborn care.
Collapse
|
38
|
Paprocka J, Kijonka M, Rzepka B, Sokół M. Melatonin in Hypoxic-Ischemic Brain Injury in Term and Preterm Babies. Int J Endocrinol 2019; 2019:9626715. [PMID: 30915118 PMCID: PMC6402213 DOI: 10.1155/2019/9626715] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/23/2019] [Accepted: 01/30/2019] [Indexed: 12/05/2022] Open
Abstract
Melatonin may serve as a potential therapeutic free radical scavenger and broad-spectrum antioxidant. It shows neuroprotective properties against hypoxic-ischemic brain injury in animal models. The authors review the studies focusing on the neuroprotective potential of melatonin and its possibility of treatment after perinatal asphyxia. Melatonin efficacy, low toxicity, and ability to readily cross through the blood-brain barrier make it a promising molecule. A very interesting thing is the difference between the half-life of melatonin in preterm neonates (15 hours) and adults (45-60 minutes). Probably, the use of synergic strategies-hypothermia coupled with melatonin treatment-may be promising in improving antioxidant action. The authors discuss and try to summarize the evidence surrounding the use of melatonin in hypoxic-ischemic events in term and preterm babies.
Collapse
Affiliation(s)
- Justyna Paprocka
- Department of Pediatric Neurology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marek Kijonka
- Department of Medical Physics, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Poland
| | - Beata Rzepka
- Students' Scientific Society, Department Pediatric Neurology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Maria Sokół
- Department of Medical Physics, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Poland
| |
Collapse
|
39
|
Luo C, Yang Q, Liu Y, Zhou S, Jiang J, Reiter RJ, Bhattacharya P, Cui Y, Yang H, Ma H, Yao J, Lawler SE, Zhang X, Fu J, Rozental R, Aly H, Johnson MD, Chiocca EA, Wang X. The multiple protective roles and molecular mechanisms of melatonin and its precursor N-acetylserotonin in targeting brain injury and liver damage and in maintaining bone health. Free Radic Biol Med 2019; 130:215-233. [PMID: 30315933 DOI: 10.1016/j.freeradbiomed.2018.10.402] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/01/2018] [Accepted: 10/02/2018] [Indexed: 12/20/2022]
Abstract
Melatonin is a neurohormone associated with sleep and wakefulness and is mainly produced by the pineal gland. Numerous physiological functions of melatonin have been demonstrated including anti-inflammation, suppressing neoplastic growth, circadian and endocrine rhythm regulation, and its potent antioxidant activity as well as its role in regeneration of various tissues including the nervous system, liver, bone, kidney, bladder, skin, and muscle, among others. In this review, we summarize the recent advances related to the multiple protective roles of melatonin receptor agonists, melatonin and N-acetylserotonin (NAS), in brain injury, liver damage, and bone health. Brain injury, including traumatic brain injury, ischemic stroke, intracerebral hemorrhage, subarachnoid hemorrhage, and newborn perinatal hypoxia-ischemia encephalopathy, is a major cause of mortality and disability. Liver disease causes serious public health problems and various factors including alcohol, chemical pollutants, and drugs induce hepatic damage. Osteoporosis is the most common bone disease in humans. Due in part to an aging population, both the cost of care of fracture patients and the annual fracture rate have increased steadily. Despite the discrepancy in the pathophysiological processes of these disorders, time frames and severity, they may share several common molecular mechanisms. Oxidative stress is considered to be a critical factor in these pathogeneses. We update the current state of knowledge related to the molecular processes, mainly including anti-oxidative stress, anti-apoptosis, autophagy dysfunction, and anti-inflammation as well as other properties of melatonin and NAS. Particularly, the abilities of melatonin and NAS to directly scavenge oxygen-centered radicals and toxic reactive oxygen species, and indirectly act through antioxidant enzymes are disscussed. In this review, we summarize the similarities and differences in the protection provided by melatonin and/or NAS in brain, liver and bone damage. We analyze the involvement of melatonin receptor 1A (MT1), melatonin receptor 1B (MT2), and melatonin receptor 1C (MT3) in the protection of melatonin and/or NAS. Additionally, we evaluate their potential clinical applications. The multiple mechanisms of action and multiple organ-targeted properties of melatonin and NAS may contribute to development of promising therapies for clinical trials.
Collapse
Affiliation(s)
- Chengliang Luo
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Qiang Yang
- Hubei Provincial Key Lab for Quality and Safety of Traditional Chinese Medicine Health Food, Jing Brand Research Institute, Daye, Hubei, China
| | - Yuancai Liu
- Hubei Provincial Key Lab for Quality and Safety of Traditional Chinese Medicine Health Food, Jing Brand Research Institute, Daye, Hubei, China
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiying Jiang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University Texas Health Science Center, San Antonio, TX, USA
| | - Pallab Bhattacharya
- National Institute of Pharmaceutical Education and Research, Ahmedabad, India
| | - Yongchun Cui
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hongwei Yang
- Department of Neurosurgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - He Ma
- Third Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiemin Yao
- Third Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Sean E Lawler
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xinmu Zhang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jianfang Fu
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Renato Rozental
- Lab Neuroproteção & Estratégias Regenerativas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Hany Aly
- Department of Neonatology, Cleveland Clinic Children's Hospital, Cleveland, OH, USA
| | - Mark D Johnson
- Department of Neurosurgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Metwally MMM, Ebraheim LLM, Galal AAA. Potential therapeutic role of melatonin on STZ-induced diabetic central neuropathy: A biochemical, histopathological, immunohistochemical and ultrastructural study. Acta Histochem 2018; 120:828-836. [PMID: 30268437 DOI: 10.1016/j.acthis.2018.09.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/19/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Abstract
The aim of the present study was to assess the therapeutic potential of melatonin (Mel) in diabetic central neuropathy in a rat model of streptozotocin (STZ)-induced diabetes. The rats were injected with 60 mg/kg STZ and diabetes was confirmed by blood glucose levels (BGL) ≥ 250 mg/dL. Mel treatment (50 mg/kg) was started 72 h before the STZ injection and continued for 45 days. In addition, normal control, vehicle (5% ethanol) control, and Mel-treated non-diabetic control were also included. STZ induced a diabetic phenotype with persistent hyperglycemia and elevated oxidative stress in the brain, liver, and kidneys compared to the control groups. In addition, the diabetic rats showed severe β-cell necrosis with reduced insulin levels, cerebral neuronopathy, myelinopathy, axonopathy, microglial and astroglial activation, and vascular damage. While Mel treatment did not prevent the development of STZ-induced diabetes mellitus and had no significant effect on the BGLs of the diabetic rats, it significantly ameliorated the diabetes-induced oxidative stress and neurodegeneration. Taken together, Mel showed potent therapeutic effects against the neurological complications of hyperglycemia and therefore can be used to treat diabetic neuropathy.
Collapse
Affiliation(s)
- Mohamed M M Metwally
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Lamiaa L M Ebraheim
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Azza A A Galal
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt.
| |
Collapse
|
41
|
Rodrigues Helmo F, Etchebehere RM, Bernardes N, Meirelles MF, Galvão Petrini C, Penna Rocha L, Gonçalves Dos Reis Monteiro ML, Souza de Oliveira Guimarães C, de Paula Antunes Teixeira V, Dos Reis MA, Machado JR, Miranda Corrêa RR. Melatonin treatment in fetal and neonatal diseases. Pathol Res Pract 2018; 214:1940-1951. [PMID: 30377024 DOI: 10.1016/j.prp.2018.10.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/19/2018] [Accepted: 10/19/2018] [Indexed: 01/01/2023]
Abstract
This literature review aims to address the main scientific findings on oxidative stress activity in different gestational disorders, as well as the function and application of melatonin in the treatment of fetal and neonatal changes. Oxidative stress has been associated with the etiopathogenesis of recurrent miscarriages, preeclampsia, intrauterine growth restriction, and stillbirth. Both, the exacerbated consumption of the antioxidant enzymes superoxide dismutase, catalase and glutathione peroxidase, and the increased synthesis of reactive oxygen species, such as superoxide, peroxynitrite, and hydrogen peroxide, induce phospholipid peroxidation and endothelial dysfunction, impaired invasion and death of trophoblast cells, impaired decidualization, and remodeling of maternal spiral arteries. It has been postulated that melatonin induces specific biochemical responses that regulate cell proliferation in fetuses, and that its antioxidant action promotes bioavailability of nitric oxide and, thus, placental perfusion and also fetal nutrition and oxygenation. Therefore, the therapeutic action of melatonin has been the subject of major studies that aim to minimize or prevent different injuries affecting this pediatric age group, such as intrauterine growth restriction, encephalopathy, chronic lung diseases, retinopathy of prematurity Conclusion: the results antioxidant and indicate that melatonin is an important therapy for the clinical treatment of these diseases.
Collapse
Affiliation(s)
- Fernanda Rodrigues Helmo
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Renata Margarida Etchebehere
- Surgical Pathology Service, Clinical Hospital, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Natália Bernardes
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Maria Flávia Meirelles
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Caetano Galvão Petrini
- Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Laura Penna Rocha
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | | | - Vicente de Paula Antunes Teixeira
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Marlene Antônia Dos Reis
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Juliana Reis Machado
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Rosana Rosa Miranda Corrêa
- Discipline of General Pathology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil.
| |
Collapse
|
42
|
Kuthati Y, Lin SH, Chen IJ, Wong CS. Melatonin and their analogs as a potential use in the management of Neuropathic pain. J Formos Med Assoc 2018; 118:1177-1186. [PMID: 30316678 DOI: 10.1016/j.jfma.2018.09.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/02/2018] [Accepted: 09/19/2018] [Indexed: 12/14/2022] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine), secreted by the pineal gland is known to perform multiple functions including, antioxidant, anti-hypertensive, anti-cancerous, immunomodulatory, sedative and tranquilizing functions. Melatonin is also known to be involved in the regulation of body mass index, control the gastrointestinal system and play an important role in cardioprotection, thermoregulation, and reproduction. Recently, several studies have reported the efficacy of Melatonin in treating various pain syndromes. The current paper reviews the studies on Melatonin and its analogs, particularly in Neuropathic pain. Here, we first briefly summarized research in preclinical studies showing the possible mechanisms through which Melatonin and its analogs induce analgesia in Neuropathic pain. Second, we reviewed research indicating the role of Melatonin in attenuating analgesic tolerance. Finally, we discussed the recent studies that reported novel Melatonin agonists, which were proven to be effective in treating Neuropathic pain.
Collapse
Affiliation(s)
- Yaswanth Kuthati
- Department of Anesthesiology, Cathay General Hospital, Taipei, Taiwan
| | - Sheng-Hsiung Lin
- Planning and Management Office, Tri-Service General Hospital, National Defense Medical Center, Taiwan
| | - Ing-Jung Chen
- Department of Anesthesiology, Cathay General Hospital, Taipei, Taiwan
| | - Chih-Shung Wong
- Department of Anesthesiology, Cathay General Hospital, Taipei, Taiwan; Planning and Management Office, Tri-Service General Hospital, National Defense Medical Center, Taiwan; Institute of Medical Sciences, National Defense Medical Center, Taiwan; Department of Anesthesiology, Tri-Service General Hospital, Taiwan.
| |
Collapse
|
43
|
Shao M, Shen Y, Sun H, Meng D, Huo W, Qi X. Protectiveness of Artesunate Given Prior Ischemic Cerebral Infarction Is Mediated by Increased Autophagy. Front Neurol 2018; 9:634. [PMID: 30174640 PMCID: PMC6107698 DOI: 10.3389/fneur.2018.00634] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/13/2018] [Indexed: 01/20/2023] Open
Abstract
Background: Ischemic cerebral infarction is a severe clinical condition that can cause serious mortality. Artesunate, an anti-malarial drug that is widely used in cancer treatment, is known to facilitate accelerated cell apoptosis. The aim of this study is to explore the possible neuroprotective effects of artesunate on hypoxic-ischemic cells in rats. Methods: Middle cerebral artery occlusion (MCAO) rats were treated with artesunate in different doses to observe their survival rate. Primary hippocampal neurons were deprived of oxygen-glucose to induce ischemia symptoms. Western blot was performed to determine the protein expressions of p-mTOR, Beclin-1, and Mcl-1. A five-point scale was used to detect neurological deficit. Cell apoptosis was measured using a TUNEL assay. Results: Artesunate supplementation protected MCAO rats from death and ameliorated brain injury among them. Artesunate administration decreased the expression of p-mTOR, increased the expressions of Beclin-1 and Mcl-1, and decreased the activity of caspase-3 in both the rats' ischemia cerebral cortices and their primary ischemia hippocampal neurons when compared with artesunate-absent ischemic brains and cells. The neuroprotective effects of artesunate were abolished by either leucine (LEU) or 3-MA, while the effects of rapamycin were reversed by 3-MA. In vivo experiments verified the protective effects of artesunate on brain-infarct rats. Conclusion: The results indicate the protectiveness of artesunate against ischemic cerebral infarction, whereas the protectiveness might increase autophagy through regulating the activity of mTOR.
Collapse
Affiliation(s)
- Ming Shao
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Shen
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongjing Sun
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Delong Meng
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Huo
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xu Qi
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
44
|
Melatonin protects against blood-brain barrier damage by inhibiting the TLR4/ NF-κB signaling pathway after LPS treatment in neonatal rats. Oncotarget 2018; 8:31638-31654. [PMID: 28404943 PMCID: PMC5458236 DOI: 10.18632/oncotarget.15780] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 01/25/2017] [Indexed: 12/31/2022] Open
Abstract
Hypoxic-ischemic and inflammatory (HII) induces the disruption of blood–brain barrier (BBB) which leads to inflammatory responses and neuronal cell death, resulting in brain secondary damage. Previous studies showed that melatonin produced potent neuroprotective effects in neonatal hypoxic-ischaemic models. However, the relationship between BBB disruption and melatonin in HII was still unclear. The present study therefore investigated the beneficial effects of melatonin on BBB after HII and the underlying mechanisms. HII animal model was conducted by receiving lipopolysaccharide followed by 90 min hypoxia-ischaemia in postnatal day 2 Sprague–Dawley rat pups. Melatonin was injected intraperitoneally 1 h before lipopolysaccharide injection and then once a day for 1 week to evaluate the long-term effects. In this study, we demonstrated that melatonin administration inhibited the disruption of BBB permeability and improved the white matter recovery in HII model rats. Melatonin significantly attenuated the degradation of junction proteins and the neuroprotective role was related to the inhibition of microglial toll-like receptor 4/ nuclear factor-kappa B signaling pathway both in vivo and in vitro. Taken together, our data demonstrated that therapeutic strategies targeting inflammation might be suitable for the therapy of preserving BBB integrity after HII.
Collapse
|
45
|
Taniguti EH, Ferreira YS, Stupp IJV, Fraga-Junior EB, Mendonça CB, Rossi FL, Ynoue HN, Doneda DL, Lopes L, Lima E, Buss ZS, Vandresen-Filho S. Neuroprotective effect of melatonin against lipopolysaccharide-induced depressive-like behavior in mice. Physiol Behav 2018; 188:270-275. [PMID: 29458118 DOI: 10.1016/j.physbeh.2018.02.034] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/09/2018] [Accepted: 02/15/2018] [Indexed: 12/18/2022]
Abstract
Accumulating evidence indicates an interaction between inflammation and depression since increased levels of pro-inflammatory cytokines are associated with depression-related symptoms. Melatonin is a hormone synthesized and secreted by the pineal gland with antioxidant, anti-inflammatory and antidepressant-like effects. In this way, it would be interesting to evaluate the putative antidepressant-like effect of melatonin treatment in an acute inflammation mice model of depression. The present study aimed to investigate the effect of melatonin treatment on lipopolysaccharide (LPS) induced depressive-like behavior, neuroinflammation, oxidative stress and alteration on brain-derived neurotrophic fator (BDNF) levels. Mice were treated with melatonin (10 mg/kg, i.p.) 30 min before LPS (0.5 mg/kg, i.p.) injection. Twenty-four hours after LPS infusion, mice were submitted to the behavioral tests and, thereafter, biochemical determinations were performed. Melatonin treatment prevented LPS-induced depressive-like behavior in the forced swim and tail suspension tests with no alterations in locomotor activity evaluated in the open field test. Melatonin attenuated LPS-induced increase in tumor necrosis factor-α (TNF-α) and reduction of BDNF levels in the hippocampus. Treatment with melatonin also prevented LPS-induced increase in lipid peroxidation and the reduction of glutathione levels in the hippocampus. In conclusion, the present study suggests that melatonin treatment exerted neuroprotective effects against LPS-induced depressive-like behavior which may be related to reduction of TNF-α release, oxidative stress and modulation of BDNF expression.
Collapse
Affiliation(s)
- E H Taniguti
- Laboratório de Fisiologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - Y S Ferreira
- Laboratório de Fisiologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - I J V Stupp
- Laboratório de Fisiologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil; Laboratório de Imunologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - E B Fraga-Junior
- Laboratório de Fisiologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - C B Mendonça
- Laboratório de Imunologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - F L Rossi
- Laboratório de Imunologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - H N Ynoue
- Laboratório de Imunologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - D L Doneda
- Laboratório de Fisiologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - L Lopes
- Laboratório de Fisiologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - E Lima
- Laboratório de Fisiologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - Z S Buss
- Laboratório de Imunologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil
| | - S Vandresen-Filho
- Laboratório de Fisiologia, Departamento de Ciências Básicas em Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Boa Esperança, 78060900 Cuiabá, MT, Brazil.
| |
Collapse
|
46
|
Zhu H, Jin Q, Li Y, Ma Q, Wang J, Li D, Zhou H, Chen Y. Melatonin protected cardiac microvascular endothelial cells against oxidative stress injury via suppression of IP3R-[Ca 2+]c/VDAC-[Ca 2+]m axis by activation of MAPK/ERK signaling pathway. Cell Stress Chaperones 2018; 23:101-113. [PMID: 28669047 PMCID: PMC5741585 DOI: 10.1007/s12192-017-0827-4] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/14/2017] [Accepted: 06/21/2017] [Indexed: 10/19/2022] Open
Abstract
The cardiac microvascular reperfusion injury is characterized by the microvascular endothelial cells (CMECs) oxidative damage which is responsible for the progression of cardiac dysfunction. However, few strategies are available to reverse such pathologies. This study aimed to explore the mechanism by which oxidative stress induced CMECs death and the beneficial actions of melatonin on CMECs survival, with a special focused on IP3R-[Ca2+]c/VDAC-[Ca2+]m damage axis and the MAPK/ERK survival signaling. We found that oxidative stress induced by H2O2 significantly activated cAMP response element binding protein (CREB) that enhanced IP3R and VDAC transcription and expression, leading to [Ca2+]c and [Ca2+]m overload. High concentration of [Ca2+]m suppressed ΔΨm, opened mPTP, and released cyt-c into cytoplasm where it activated mitochondria-dependent death pathway. However, melatonin could protect CMECs against oxidative stress injury via stimulation of MAPK/ERK that inactivated CREB and therefore blocked IP3R/VDAC upregulation and [Ca2+]c/[Ca2+]m overload, sustaining mitochondrial structural and function integrity and ultimately blockading mitochondrial-mediated cellular death. In summary, these findings confirmed the mechanisms by which oxidative injury induced CMECs mitochondrial-involved death and provided an attractive and effective way to enhance CMECs survival.
Collapse
Affiliation(s)
- Hang Zhu
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Rd, Beijing, 100853, China
| | - Qinhua Jin
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Rd, Beijing, 100853, China
| | - Yang Li
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Rd, Beijing, 100853, China
| | - Qiang Ma
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Rd, Beijing, 100853, China
| | - Jing Wang
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Rd, Beijing, 100853, China
| | - Dandan Li
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Rd, Beijing, 100853, China
| | - Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Rd, Beijing, 100853, China.
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Rd, Beijing, 100853, China.
| |
Collapse
|
47
|
Sinha B, Wu Q, Li W, Tu Y, Sirianni AC, Chen Y, Jiang J, Zhang X, Chen W, Zhou S, Reiter RJ, Manning SM, Patel NJ, Aziz-Sultan AM, Inder TE, Friedlander RM, Fu J, Wang X. Protection of melatonin in experimental models of newborn hypoxic-ischemic brain injury through MT1 receptor. J Pineal Res 2018; 64. [PMID: 28796402 DOI: 10.1111/jpi.12443] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 08/04/2017] [Indexed: 12/19/2022]
Abstract
The function of melatonin as a protective agent against newborn hypoxic-ischemic (H-I) brain injury is not yet well studied, and the mechanisms by which melatonin causes neuroprotection in neurological diseases are still evolving. This study was designed to investigate whether expression of MT1 receptors is reduced in newborn H-I brain injury and whether the protective action of melatonin is by alterations of the MT1 receptors. We demonstrated that there was significant reduction in MT1 receptors in ischemic brain of mouse pups in vivo following H-I brain injury and that melatonin offers neuroprotection through upregulation of MT1 receptors. The role of MT1 receptors was further supported by observation of increased mortality in MT1 knockout mice following H-I brain injury and the reversal of the inhibitory role of melatonin on mitochondrial cell death pathways by the melatonin receptor antagonist, luzindole. These data demonstrate that melatonin mediates its neuroprotective effect in mouse models of newborn H-I brain injury, at least in part, by the restoration of MT1 receptors, the inhibition of mitochondrial cell death pathways and the suppression of astrocytic and microglial activation.
Collapse
MESH Headings
- Animals
- Astrocytes/cytology
- Blotting, Western
- Cells, Cultured
- Female
- Genotype
- Hippocampus/cytology
- Hypoxia-Ischemia, Brain/drug therapy
- Hypoxia-Ischemia, Brain/metabolism
- Immunohistochemistry
- Male
- Melatonin/therapeutic use
- Membrane Potentials/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Theoretical
- Receptor, Melatonin, MT1/genetics
- Receptor, Melatonin, MT1/metabolism
Collapse
Affiliation(s)
- Bharati Sinha
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Division of Neonatology, Boston University School of Medicine, Boston, MA, USA
| | - Qiaofeng Wu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Li
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yanyang Tu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana C Sirianni
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yanchun Chen
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Jiying Jiang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Xinmu Zhang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wu Chen
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Clinical Laboratory, Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University Texas Health Science Center, San Antonio, TX, USA
| | - Simon M Manning
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nirav J Patel
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ali M Aziz-Sultan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Terrie E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert M Friedlander
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jianfang Fu
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- The Joslin Beth Israel Deaconess Foot Center, Harvard Medical School, Boston, MA, USA
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
48
|
Parikh P, Juul SE. Neuroprotective Strategies in Neonatal Brain Injury. J Pediatr 2018; 192:22-32. [PMID: 29031859 DOI: 10.1016/j.jpeds.2017.08.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/26/2017] [Accepted: 08/15/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Pratik Parikh
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, WA
| | - Sandra E Juul
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, WA.
| |
Collapse
|
49
|
Hasan M, Genovese S, Fiorito S, Epifano F, Witt-Enderby PA. Oxyprenylated Phenylpropanoids Bind to MT1 Melatonin Receptors and Inhibit Breast Cancer Cell Proliferation and Migration. JOURNAL OF NATURAL PRODUCTS 2017; 80:3324-3329. [PMID: 29144746 DOI: 10.1021/acs.jnatprod.7b00853] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Oxyprenylated compounds (i.e., ferulic acid and coumarin derivatives) demonstrate neuroprotection and anticancer properties as reported in previous studies. We have tested the affinity of oxyprenylated ferulic acid (1-4) and umbelliferone derivatives (5-11) to melatonin receptors as well as their antiproliferation and antimigratory properties against breast cancer (BC) cell lines. All the compounds except for ferulic acid, boropinic acid, and umbelliferone had binding affinities to melatonin receptors in the nM to μM range, and both auraptene and umbellinprenin reduced BC cell proliferation and migration in phenotypically diverse BC including triple negative.
Collapse
Affiliation(s)
- Mahmud Hasan
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University , Pittsburgh, Pennsylvania 15282, United States
| | - Salvatore Genovese
- Department of Pharmacy, University "G. d'Annunzio" of Chieti e Pescara , Chieti 66100, Italy
| | - Serena Fiorito
- Department of Pharmacy, University "G. d'Annunzio" of Chieti e Pescara , Chieti 66100, Italy
| | - Francesco Epifano
- Department of Pharmacy, University "G. d'Annunzio" of Chieti e Pescara , Chieti 66100, Italy
| | - Paula A Witt-Enderby
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University , Pittsburgh, Pennsylvania 15282, United States
| |
Collapse
|
50
|
Checa-Ros A, Muñoz-Hoyos A, Molina-Carballo A, Muñoz-Gallego A, Narbona-Galdó S, Jerez-Calero A, Augustín-Morales MDC. Analysis of Different Melatonin Secretion Patterns in Children With Sleep Disorders: Melatonin Secretion Patterns in Children. J Child Neurol 2017; 32:1000-1008. [PMID: 28911277 DOI: 10.1177/0883073817726680] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The objective of this study was to analyze circadian patterns of urinary 6-sulphatoxymelatonin (aMT6s) excretion in children with primary sleep disorders in comparison with healthy controls. A total of 124 control children and 124 patients (aged 4-14 years) diagnosed with diverse primary sleep disorders were recruited. aMT6s concentrations were measured in diurnal and nocturnal urine, as well as in 24-hour urine. aMT6s levels were significantly higher and showed significantly more evident circadian variations in the control group ( P < .001). Four different melatonin (aMT) production and excretion patterns were distinguished in the group with sleep disorders: (1) standard aMT production pattern, (2) low aMT production pattern, (3) aMT production pattern with absence of circadian variation, and (4) aMT hyperproduction pattern. This study highlights the importance of analyzing specific alterations of aMT secretion in each sleep disorder and provides evidences to explain why not all children with sleep disturbances do respond to aMT treatment.
Collapse
Affiliation(s)
- Ana Checa-Ros
- 1 San Cecilio University Hospital, Department of Pediatrics, School of Medicine, University of Granada, Spain
| | - Antonio Muñoz-Hoyos
- 1 San Cecilio University Hospital, Department of Pediatrics, School of Medicine, University of Granada, Spain
| | - Antonio Molina-Carballo
- 1 San Cecilio University Hospital, Department of Pediatrics, School of Medicine, University of Granada, Spain
| | | | - Susana Narbona-Galdó
- 1 San Cecilio University Hospital, Department of Pediatrics, School of Medicine, University of Granada, Spain
| | - Antonio Jerez-Calero
- 1 San Cecilio University Hospital, Department of Pediatrics, School of Medicine, University of Granada, Spain
| | | |
Collapse
|