1
|
Paditz E. Postnatal Development of the Circadian Rhythmicity of Human Pineal Melatonin Synthesis and Secretion (Systematic Review). CHILDREN (BASEL, SWITZERLAND) 2024; 11:1197. [PMID: 39457162 PMCID: PMC11506472 DOI: 10.3390/children11101197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024]
Abstract
Introduction: According to current knowledge, at birth, the pineal gland and melatonin receptors are already present and the suprachiasmatic nucleus is largely functional, and noradrenaline, the key pineal transmitter, can be detected in the early foetal period. It is still unclear why the pineal gland is not able to start its own pulsatile synthesis and secretion of melatonin in the first months of life, and as a result, infants during this time are dependent on an external supply of melatonin. Method: The causes and consequences of this physiological melatonin deficiency in human infancy are examined in a systematic review of the literature, in which 40 of 115 initially selected publications were evaluated in detail. The references of these studies were checked for relevant studies on this topic. References from previous reviews by the author were taken into account. Results: The development and differentiation of the pineal gland, the pinealocytes, as the site of melatonin synthesis, and the development and synaptic coupling of the associated predominantly noradrenergic neural pathways and vessels and the associated Lhx4 homebox only occurs during the first year of life. Discussion: The resulting physiological melatonin deficiency is associated with sleep disorders, infant colic, and increased crying in babies. Intervention studies indicate that this deficiency should be compensated for through breastfeeding, the administration of nonpooled donor milk, or through industrially produced chrononutrition made from nonpooled cow's milk with melatonin-poor day milk and melatonin-rich night milk.
Collapse
Affiliation(s)
- Ekkehart Paditz
- Center for Applied Prevention®, Blasewitzer Str. 41, D-01307 Dresden, Germany
| |
Collapse
|
2
|
Gou ZX, Zhou Y, Fan Y, Zhang F, Ning XM, Tang F, Lu LQ. Melatonin Improves Oxidative Stress Injury in Retinopathy of Prematurity by Targeting miR-23a-3p/Nrf2. Curr Eye Res 2024:1-13. [PMID: 39103986 DOI: 10.1080/02713683.2024.2380433] [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: 11/23/2023] [Revised: 06/13/2024] [Accepted: 07/10/2024] [Indexed: 08/07/2024]
Abstract
PURPOSE Melatonin has promising protective effects for retinopathy. However, its roles in retinopathy of prematurity (ROP) and the underlying mechanisms remain unknown. We aimed to explore its roles and mechanisms in a ROP model. METHODS Hematoxylin and eosin staining were used to observe the morphology of the retina. Immunofluorescence was used to detect positive (Nrf2+ and VEGF+) cells. Immunohistochemistry was used to detect the level of nuclear expression of PCNA in retinal tissue. Transmission electron microscope (TEM) was used to observe the morphology and structure of pigment cells. qRT-PCR was used to assay the expression of miR-23a-3p, Nrf2, and HO-1. Western blotting was used to detect the expression of Nrf2, HO-1, β-actin, and Lamin B1. RESULTS Melatonin or miR-23a-3p antagomir treatment could ameliorate the Oxygen-induced pathological changes, increased the expression of Nrf2 and HO-1, SOD, and GSH-Px, and decreased the expression of VEGF, miR-23a-3p, MDA and the apoptosis in the ROP model. Further target prediction and luciferase reporter assays confirmed the targeted binding relationship between miR-23a-3p and Nrf2. CONCLUSION Our study showed that melatonin could ameliorate H2O2-induced apoptosis and oxidative stress injury in RGC cells by mediating miR-23a-3p/Nrf2 signaling pathway, thereby improving retinal degeneration.
Collapse
Affiliation(s)
- Zhi-Xian Gou
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, P.R. China
- Department of Pediatrics, School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Yue Zhou
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, P.R. China
- Department of Pediatrics, School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Yang Fan
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, P.R. China
- Department of Pediatrics, School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Feng Zhang
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, P.R. China
- Department of Pediatrics, School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Xue-Mei Ning
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, P.R. China
- Department of Pediatrics, School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Fei Tang
- Clinic Medical College, Chengdu Medical College, Chengdu, P.R. China
| | - Li-Qun Lu
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, P.R. China
- Department of Pediatrics, School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| |
Collapse
|
3
|
Sánchez-Borja C, Cristóbal-Cañadas D, Rodríguez-Lucenilla MI, Muñoz-Hoyos A, Agil A, Vázquez-López MÁ, Parrón-Carreño T, Nievas-Soriano BJ, Bonillo-Perales A, Bonillo-Perales JC. Lower plasma melatonin levels in non-hypoxic premature newborns associated with neonatal pain. Eur J Pediatr 2024; 183:3607-3615. [PMID: 38842550 PMCID: PMC11263426 DOI: 10.1007/s00431-024-05632-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/27/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
We analyzed plasma melatonin levels in different groups of preterm newborns without hypoxia and their relationship with several perinatal variables like gestational age or neonatal pain. Prospective cohort study of preterm newborns (PTNB) without perinatal hypoxia, Apgar > 6 at 5 min, and oxygen needs on the third day of life. We compared melatonin levels at day 3 of life in different groups of non-hypoxic preterm infants (Student's t-tests, Mann-Whitney U, and chi2) and analyzed the relationship of melatonin with GA, birth weight, neonatal pain (Premature Infant Pain Profile (PIPP) scale), caffeine treatment, parenteral nutrition, or the development of free radical diseases (correlation study, linear regression) and factors associated with moderate/intense pain and free radical diseases (logistic regression analysis). Sixty-one preterm infants with gestational age (GA) of 30.7 ± 2.0 weeks with no oxygen requirements at day 3 of life were studied with plasma melatonin levels of 33.8 ± 12.01 pg/ml. Preterm infants weighing < 1250 g at birth had lower plasma melatonin levels (p = 0.05). Preterm infants with moderate or severe pain (PPIPP > 5) have lower melatonin levels (p = 0.01), and being preterm with PIPP > 5 is associated with lower plasma melatonin levels (p = 0.03). Being very preterm (GA < 32 GS), having low weight for gestational age (LWGA), receiving caffeine treatment, or requiring parenteral nutrition did not modify melatonin levels in non-hypoxic preterm infants (p = NS). Melatonin on day 3 of life in non-hypoxic preterm infants is not associated with later development of free radical diseases (BPD, sepsis, ROP, HIV, NEC). CONCLUSION We observed that preterm infants with moderate to severe pain have lower melatonin levels. These findings are relevant because they reinforce the findings of other authors that melatonin supplementation decreases pain and oxidative stress in painful procedures in premature infants. Further studies are needed to evaluate whether melatonin could be used as an analgesic in painful procedures in preterm infants. TRIAL REGISTRATION Trial registration was not required since this was an observational study. WHAT IS KNOWN • Melatonin is a potent antioxidant and free radical scavenger in newborns under stress conditions: hypoxia, acidosis, hypotension, painful procedures, or parenteral nutrition. • Pain stimulates the production of melatonin. • Various studies conclude that melatonin administration decreases pain during the neonatal period. WHAT IS NEW • Non-hypoxic preterm infants with moderate to severe pain (PIPP>5) have lower levels of melatonin. • Administration of caffeine and treatment with parenteral nutrition do not modify melatonin levels in non-hypoxic preterm infants.
Collapse
Affiliation(s)
| | | | | | | | - Ahmad Agil
- Department of Pharmacology, Institute Biohelath & Institute of Neuroscience, University of Granada, Granada, Spain
| | | | - Tesifón Parrón-Carreño
- Nursing, Physiotherapy, and Medicine Department, University of Almería, Ctra. de Sacramento, s/n, La Cañada, Almería, 01410, Spain
| | - Bruno José Nievas-Soriano
- Nursing, Physiotherapy, and Medicine Department, University of Almería, Ctra. de Sacramento, s/n, La Cañada, Almería, 01410, Spain.
| | | | | |
Collapse
|
4
|
Giri A, Mehan S, Khan Z, Das Gupta G, Narula AS, Kalfin R. Modulation of neural circuits by melatonin in neurodegenerative and neuropsychiatric disorders. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3867-3895. [PMID: 38225412 DOI: 10.1007/s00210-023-02939-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/30/2023] [Indexed: 01/17/2024]
Abstract
Neurodegenerative and neuropsychiatric disorders are two broad categories of neurological disorders characterized by progressive impairments in movement and cognitive functions within the central and peripheral nervous systems, and have emerged as a significant cause of mortality. Oxidative stress, neuroinflammation, and neurotransmitter imbalances are recognized as prominent pathogenic factors contributing to cognitive deficits and neurobehavioral anomalies. Consequently, preventing neurodegenerative and neuropsychiatric diseases has surfaced as a pivotal challenge in contemporary public health. This review explores the investigation of neurodegenerative and neuropsychiatric disorders using both synthetic and natural bioactive compounds. A central focus lies on melatonin, a neuroregulatory hormone secreted by the pineal gland in response to light-dark cycles. Melatonin, an amphiphilic molecule, assumes multifaceted roles, including scavenging free radicals, modulating energy metabolism, and synchronizing circadian rhythms. Noteworthy for its robust antioxidant and antiapoptotic properties, melatonin exhibits diverse neuroprotective effects. The inherent attributes of melatonin position it as a potential key player in the pathophysiology of neurological disorders. Preclinical and clinical studies have demonstrated melatonin's efficacy in alleviating neuropathological symptoms across neurodegenerative and neuropsychiatric conditions (depression, schizophrenia, bipolar disorder, and autism spectrum disorder). The documented neuroprotective prowess of melatonin introduces novel therapeutic avenues for addressing neurodegenerative and psychiatric disorders. This comprehensive review encompasses many of melatonin's applications in treating diverse brain disorders. Despite the strides made, realizing melatonin's full neuroprotective potential necessitates further rigorous clinical investigations. By unravelling the extended neuroprotective benefits of melatonin, future studies promise to deepen our understanding and augment the therapeutic implications against neurological deficits.
Collapse
Affiliation(s)
- Aditi Giri
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy Moga, Punjab, India.
- IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy Moga, Punjab, India
- IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | | | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, Sofia, 1113, Bulgaria
- Department of Healthcare, South-West University "NeofitRilski", Ivan Mihailov St. 66, Blagoevgrad, 2700, Bulgaria
| |
Collapse
|
5
|
Osipova NA, Panova AY, Efremov AM, Lozinskaya NA, Beznos OV, Katargina LA. Melatonin and its bioisosteres as potential therapeutic agents for the treatment of retinopathy of prematurity. Chem Biol Drug Des 2024; 103:e14504. [PMID: 38480485 DOI: 10.1111/cbdd.14504] [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: 11/14/2023] [Revised: 02/06/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
We conducted a study on the impact of intraperitoneal injections of melatonin and its three bioisosteres (compounds 1-3) on the development of oxygen-induced retinopathy in newborn rats during a 21-day experiment. It was demonstrated that melatonin and its analogues 1-3 effectively reduce the total protein concentration in the vitreous body of rat pups, decrease concentration of VEGF-A, and lower the level of oxidative stress (as indicated by normalization of antioxidant activity in the vitreous body). Melatonin and its analogues 1-3 equally normalize the level of VEGF-A. Analogues 1 and 2 even exceed melatonin in their ability to reduce protein influx into the vitreous body. However, analogue 2 had no effect on antioxidant activity, while analogues 1 and 3 caused a significant increase in this parameter, with analogue 3 even slightly exceeding melatonin. Thus, it can be concluded that analogues 1-3 are comparable to melatonin and can be utilized as potential therapeutic agents for the treatment of retinopathy of prematurity.
Collapse
Affiliation(s)
- N A Osipova
- Helmholtz National Medical Center of Eye Diseases, Moscow, Russia
| | - A Y Panova
- Helmholtz National Medical Center of Eye Diseases, Moscow, Russia
| | - A M Efremov
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - N A Lozinskaya
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - O V Beznos
- Helmholtz National Medical Center of Eye Diseases, Moscow, Russia
| | - L A Katargina
- Helmholtz National Medical Center of Eye Diseases, Moscow, Russia
| |
Collapse
|
6
|
Garofoli F, Franco V, Accorsi P, Albertini R, Angelini M, Asteggiano C, Aversa S, Ballante E, Borgatti R, Cabini RF, Caporali C, Chiapparini L, Cociglio S, Fazzi E, Longo S, Malerba L, Materia V, Mazzocchi L, Naboni C, Palmisani M, Pichiecchio A, Pinelli L, Pisoni C, Preda L, Riboli A, Risso FM, Rizzo V, Rognone E, Simoncelli AM, Villani P, Tzialla C, Ghirardello S, Orcesi S. Fate of melatonin orally administered in preterm newborns: Antioxidant performance and basis for neuroprotection. J Pineal Res 2024; 76:e12932. [PMID: 38111174 DOI: 10.1111/jpi.12932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/10/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
Preterm infants cannot counteract excessive reactive oxygen species (ROS) production due to preterm birth, leading to an excess of lipid peroxidation with malondialdehyde (MDA) production, capable of contributing to brain damage. Melatonin (ME), an endogenous brain hormone, and its metabolites, act as a free radical scavenger against ROS. Unfortunately, preterms have an impaired antioxidant system, resulting in the inability to produce and release ME. This prospective, multicenter, parallel groups, randomized, double-blind, placebo-controlled trial aimed to assess: (i) the endogenous production of ME in very preterm infants (gestational age ≤ 29 + 6 WE, 28 infants in the ME and 26 in the placebo group); (ii) the exogenous hormone availability and its metabolization to the main metabolite, 6-OH-ME after 15 days of ME oral treatment; (iii) difference of MDA plasma concentration, as peroxidation marker, after treatment. Blood was collected before the first administration (T1) and after 15 days of administration (T2). ME and 6-OH-ME were detected by liquid chromatography tandem mass spectrometry, MDA was measured by liquid chromatograph with fluorescence detection. ME and 6-OH-ME were not detectable in the placebo group at any study time-point. ME was absent in the active group at T1. In contrast, after oral administration, ME and 6-OH-ME resulted highly detectable and the difference between concentrations T2 versus T1 was statistically significant, as well as the difference between treated and placebo groups at T2. MDA levels seemed stable during the 15 days of treatment in both groups. Nevertheless, a trend in the percentage of neonates with reduced MDA concentration at T2/T1 was 48.1% in the ME group versus 38.5% in the placebo group. We demonstrated that very preterm infants are not able to produce endogenous detectable plasma levels of ME during their first days of life. Still, following ME oral administration, appreciable amounts of ME and 6-OH-ME were available. The trend of MDA reduction in the active group requires further clinical trials to fix the dosage, the length of ME therapy and to identify more appropriate indexes to demonstrate, at biological and clinical levels, the antioxidant activity and consequent neuroprotectant potential of ME in very preterm newborns.
Collapse
Affiliation(s)
- Francesca Garofoli
- 1Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Valentina Franco
- Department of Internal Medicine and Therapeutics, Clinical and Experimental Pharmacology Unit, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Pavia, Italy
| | - Patrizia Accorsi
- Unit of Child Neurology and Psychiatry, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Riccardo Albertini
- Laboratory of Clinical Chemistry, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Micol Angelini
- 1Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Carlo Asteggiano
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Department of Neuroradiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Salvatore Aversa
- Neonatal Intensive Care Unit, Children's Hospital, University Hospital "Spedali Civili" of Brescia, Brescia, Italy
| | - Elena Ballante
- Political and Social Sciences, University of Pavia, Pavia, Italy
- BioData Science Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Renato Borgatti
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Child Neurology and Psychiatry Unit IRCCS Mondino Foundation, Pavia, Italy
| | | | - Camilla Caporali
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Luisa Chiapparini
- Radiodiagnostic Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Sara Cociglio
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Elisa Fazzi
- Unit of Child Neurology and Psychiatry, ASST-Spedali Civili of Brescia, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefania Longo
- 1Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Laura Malerba
- Unit of Child Neurology and Psychiatry, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Valeria Materia
- Neonatal Intensive Care Unit, Children's Hospital, University Hospital "Spedali Civili" of Brescia, Brescia, Italy
| | - Laura Mazzocchi
- Department of Neuroradiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Cecilia Naboni
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Child Neurology and Psychiatry Unit IRCCS Mondino Foundation, Pavia, Italy
| | - Michela Palmisani
- Department of Internal Medicine and Therapeutics, Clinical and Experimental Pharmacology Unit, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Pavia, Italy
| | - Anna Pichiecchio
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Department of Neuroradiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Lorenzo Pinelli
- Neuroradiology Department, Pediatric Neuroradiology Section, Spedali Civili, Brescia, Italy
| | - Camilla Pisoni
- 1Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lorenzo Preda
- Radiodiagnostic Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostics and Pediatric sciences, University of Pavia, Italy
| | - Alice Riboli
- Hospital Pediatric Psychology, Unit of Psychology, Children's Hospital "Spedali Civili" of Brescia, Brescia, Italy
| | - Francesco M Risso
- Neonatal Intensive Care Unit, Children's Hospital, University Hospital "Spedali Civili" of Brescia, Brescia, Italy
| | - Vittoria Rizzo
- Laboratory of Clinical Chemistry, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Rognone
- Department of Neuroradiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Anna M Simoncelli
- Radiodiagnostic Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Paola Villani
- Laboratory of Clinical Chemistry, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Chryssoula Tzialla
- Neonatal and Pediatric Unit, Polo Ospedaliero Oltrepò, ASST Pavia, Pavia, Italy
| | - Stefano Ghirardello
- 1Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Simona Orcesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Child Neurology and Psychiatry Unit IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
7
|
Iavarone S, Massoud M, Di Felice G, Pulcinelli F, Rapini N, Luciani M. Antiplatelet Effect of Melatonin through Breastfeeding: A Pediatric Case Report. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1839. [PMID: 38136041 PMCID: PMC10741506 DOI: 10.3390/children10121839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/21/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023]
Abstract
We present a pediatric case of the antiplatelet effect of melatonin taken through breast milk in an 18-month-old child. The child was referred to our hematology outpatient clinic because of bleeding episodes that she presented since birth. Blood tests excluded the presence of blood coagulation diseases. The family history was negative for bleeding disorders. The child did not consume any drugs, food supplements, herbal teas or infusions. We performed an aggregation platelet test, which showed a reduced platelet aggregation. Shortly before, the baby had been breastfed. We speculated that breast milk could interfere with the result of the test; therefore, we decided to repeat the test in a fasting state. This time the test showed a normal platelet aggregation time. We learned that the child's mother was taking a mixture of valerian and melatonin. Thus, we decided to suspend maternal intake of melatonin and perform a new platelet aggregation test after three months. The test results were negative. After the suspension of melatonin, the patient did not present further bleeding events. In this case, melatonin, through the inhibition of platelet aggregation, had an important role on the hemostatic system of the child. Melatonin is considered as a dietary supplement and is mostly available as an alternative medicine without formal prescription and dosage regulation. It is important, especially during breastfeeding, to investigate personal and medication history, including also homeopathic remedies or dietary supplements.
Collapse
Affiliation(s)
- Sonia Iavarone
- Onco-Hematology, Cell and Gene Therapy and Bone Marrow Transplant Clinic Area, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.I.); (M.M.)
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Michela Massoud
- Onco-Hematology, Cell and Gene Therapy and Bone Marrow Transplant Clinic Area, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.I.); (M.M.)
| | - Giovina Di Felice
- Clinical Laboratory Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Fabio Pulcinelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Novella Rapini
- Unit of Endocrinology and Diabetes, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Matteo Luciani
- Onco-Hematology, Cell and Gene Therapy and Bone Marrow Transplant Clinic Area, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.I.); (M.M.)
| |
Collapse
|
8
|
Albertini MC, Vanzolini T, Perrone S, Weiss MD, Buonocore G, Dell'Orto V, Balduini W, Carloni S. MiR-126 and miR-146a as Melatonin-Responsive Biomarkers for Neonatal Brain Ischemia. J Mol Neurosci 2023; 73:763-772. [PMID: 37725287 PMCID: PMC10694110 DOI: 10.1007/s12031-023-02155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023]
Abstract
Despite advances in obstetric and neonatal care, challenges remain in early identification of neonates with encephalopathy due to hypoxia-ischemia who are undergoing therapeutic hypothermia. Therefore, there is a deep search for biomarkers that can identify brain injury. The aims of this study were to investigate the serum and brain expressions of two potential biomarkers, miR-126/miR-146a, in a preclinical model of hypoxia-ischemia (HI)-induced brain injury, and to explore their modulation during melatonin treatment. Seven-day-old rats were subjected to permanent ligation of the right carotid artery followed by 2.5 h hypoxia (HI). Melatonin (15 mg/kg) was administered 5 min after HI. Serum and brain samples were collected 1, 6 and 24 h after HI. Results show that HI caused a significant increase in the circulating levels of both miR-126 and miR-146a during the early phase of ischemic brain damage development (i.e. 1 h), with a parallel and opposite pattern in the ischemic cerebral cortex. These effects are not observed 24 h later. Treatment with melatonin restored the HI-induced effects on miR-126/miR-146a expressions, both in the cerebral cortex and in serum. We conclude that miR-126/miR-146a are promising biomarkers of HI injury and demonstrate an associated change in concentration following melatonin treatment.
Collapse
Affiliation(s)
- Maria Cristina Albertini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 PU, Urbino, Italy.
| | - Tania Vanzolini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 PU, Urbino, Italy
| | - Serafina Perrone
- Neonatology Unit, University Medical Center of Parma (AOUP) and University of Parma, Parma, Italy
| | - Michael D Weiss
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Valentina Dell'Orto
- Neonatology Unit, University Medical Center of Parma (AOUP) and University of Parma, Parma, Italy
| | - Walter Balduini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 PU, Urbino, Italy
| | - Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 PU, Urbino, Italy.
| |
Collapse
|
9
|
Häusler S, Robertson NJ, Golhen K, van den Anker J, Tucker K, Felder TK. Melatonin as a Therapy for Preterm Brain Injury: What Is the Evidence? Antioxidants (Basel) 2023; 12:1630. [PMID: 37627625 PMCID: PMC10451719 DOI: 10.3390/antiox12081630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/28/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Despite significant improvements in survival following preterm birth in recent years, the neurodevelopmental burden of prematurity, with its long-term cognitive and behavioral consequences, remains a significant challenge in neonatology. Neuroprotective treatment options to improve neurodevelopmental outcomes in preterm infants are therefore urgently needed. Alleviating inflammatory and oxidative stress (OS), melatonin might modify important triggers of preterm brain injury, a complex combination of destructive and developmental abnormalities termed encephalopathy of prematurity (EoP). Preliminary data also suggests that melatonin has a direct neurotrophic impact, emphasizing its therapeutic potential with a favorable safety profile in the preterm setting. The current review outlines the most important pathomechanisms underlying preterm brain injury and correlates them with melatonin's neuroprotective potential, while underlining significant pharmacokinetic/pharmacodynamic uncertainties that need to be addressed in future studies.
Collapse
Affiliation(s)
- Silke Häusler
- Division of Neonatology, Department of Pediatrics, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Nicola J. Robertson
- EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK; (N.J.R.); (K.T.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Klervi Golhen
- Pediatric Pharmacology and Pharmacometrics, University Children’s Hospital Basel (UKBB), University of Basel, 4001 Basel, Switzerland; (K.G.); (J.v.d.A.)
| | - John van den Anker
- Pediatric Pharmacology and Pharmacometrics, University Children’s Hospital Basel (UKBB), University of Basel, 4001 Basel, Switzerland; (K.G.); (J.v.d.A.)
- Division of Clinical Pharmacology, Children’s National Hospital, Washington, DC 20001, USA
| | - Katie Tucker
- EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK; (N.J.R.); (K.T.)
| | - Thomas K. Felder
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria;
| |
Collapse
|
10
|
刘 一, 夏 世. [Research research on the use of melatonin in combination with therapeutic hypothermia for the treatment of neonatal hypoxic-ischemic encephalopathy]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:864-869. [PMID: 37668036 PMCID: PMC10484075 DOI: 10.7499/j.issn.1008-8830.2302099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/21/2023] [Indexed: 09/06/2023]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) remains one of the leading causes of death and long-term neurodevelopmental disorders in full-term neonates, and there is currently no curative treatment. Therapeutic hypothermia is now a standard therapy for HIE in the neonatal intensive care unit, but its safety and efficacy in remote areas remains unclear. Melatonin is an indole endocrine hormone mainly produced by the pineal gland and it has the ability to easily penetrate the blood-brain barrier. Through receptor and non-receptor mechanisms, melatonin exerts anti-oxidative and anti-inflammatory effects and participates in the regulation of organelle function and the inhibition of cell death. Melatonin is considered one of the most promising drugs for the treatment of HIE based on its reliable safety profile and clinical/preclinical results. This article reviews the recent research on the use of melatonin in combination with therapeutic hypothermia for the treatment of neonatal HIE.
Collapse
|
11
|
Kitase Y, Madurai NK, Hamimi S, Hellinger RL, Odukoya OA, Ramachandra S, Muthukumar S, Vasan V, Sevensky R, Kirk SE, Gall A, Heck T, Ozen M, Orsburn BC, Robinson S, Jantzie LL. Chorioamnionitis disrupts erythropoietin and melatonin homeostasis through the placental-fetal-brain axis during critical developmental periods. Front Physiol 2023; 14:1201699. [PMID: 37546540 PMCID: PMC10398572 DOI: 10.3389/fphys.2023.1201699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/06/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction: Novel therapeutics are emerging to mitigate damage from perinatal brain injury (PBI). Few newborns with PBI suffer from a singular etiology. Most experience cumulative insults from prenatal inflammation, genetic and epigenetic vulnerability, toxins (opioids, other drug exposures, environmental exposure), hypoxia-ischemia, and postnatal stressors such as sepsis and seizures. Accordingly, tailoring of emerging therapeutic regimens with endogenous repair or neuro-immunomodulatory agents for individuals requires a more precise understanding of ligand, receptor-, and non-receptor-mediated regulation of essential developmental hormones. Given the recent clinical focus on neurorepair for PBI, we hypothesized that there would be injury-induced changes in erythropoietin (EPO), erythropoietin receptor (EPOR), melatonin receptor (MLTR), NAD-dependent deacetylase sirtuin-1 (SIRT1) signaling, and hypoxia inducible factors (HIF1α, HIF2α). Specifically, we predicted that EPO, EPOR, MLTR1, SIRT1, HIF1α and HIF2α alterations after chorioamnionitis (CHORIO) would reflect relative changes observed in human preterm infants. Similarly, we expected unique developmental regulation after injury that would reveal potential clues to mechanisms and timing of inflammatory and oxidative injury after CHORIO that could inform future therapeutic development to treat PBI. Methods: To induce CHORIO, a laparotomy was performed on embryonic day 18 (E18) in rats with transient uterine artery occlusion plus intra-amniotic injection of lipopolysaccharide (LPS). Placentae and fetal brains were collected at 24 h. Brains were also collected on postnatal day 2 (P2), P7, and P21. EPO, EPOR, MLTR1, SIRT1, HIF1α and HIF2α levels were quantified using a clinical electrochemiluminescent biomarker platform, qPCR, and/or RNAscope. MLT levels were quantified with liquid chromatography mass spectrometry. Results: Examination of EPO, EPOR, and MLTR1 at 24 h showed that while placental levels of EPO and MLTR1 mRNA were decreased acutely after CHORIO, cerebral levels of EPO, EPOR and MLTR1 mRNA were increased compared to control. Notably, CHORIO brains at P2 were SIRT1 mRNA deficient with increased HIF1α and HIF2α despite normalized levels of EPO, EPOR and MLTR1, and in the presence of elevated serum EPO levels. Uniquely, brain levels of EPO, EPOR and MLTR1 shifted at P7 and P21, with prominent CHORIO-induced changes in mRNA expression. Reductions at P21 were concomitant with increased serum EPO levels in CHORIO rats compared to controls and variable MLT levels. Discussion: These data reveal that commensurate with robust inflammation through the maternal placental-fetal axis, CHORIO impacts EPO, MLT, SIRT1, and HIF signal transduction defined by dynamic changes in EPO, EPOR, MLTR1, SIRT1, HIF1α and HIF2α mRNA, and EPO protein. Notably, ligand-receptor mismatch, tissue compartment differential regulation, and non-receptor-mediated signaling highlight the importance, complexity and nuance of neural and immune cell development and provide essential clues to mechanisms of injury in PBI. As the placenta, immune cells, and neural cells share many common, developmentally regulated signal transduction pathways, further studies are needed to clarify the perinatal dynamics of EPO and MLT signaling and to capitalize on therapies that target endogenous neurorepair mechanisms.
Collapse
Affiliation(s)
- Yuma Kitase
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nethra K. Madurai
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah Hamimi
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ryan L. Hellinger
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - O. Angel Odukoya
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sindhu Ramachandra
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sankar Muthukumar
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Vikram Vasan
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Riley Sevensky
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shannon E. Kirk
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alexander Gall
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Timothy Heck
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Maide Ozen
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Benjamin C. Orsburn
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shenandoah Robinson
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lauren L. Jantzie
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Kennedy Krieger Institute, Baltimore, MD, United States
| |
Collapse
|
12
|
Bell A, Hewins B, Bishop C, Fortin A, Wang J, Creamer JL, Collen J, Werner JK. Traumatic Brain Injury, Sleep, and Melatonin-Intrinsic Changes with Therapeutic Potential. Clocks Sleep 2023; 5:177-203. [PMID: 37092428 PMCID: PMC10123665 DOI: 10.3390/clockssleep5020016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the most prevalent causes of morbidity in the United States and is associated with numerous chronic sequelae long after the point of injury. One of the most common long-term complaints in patients with TBI is sleep dysfunction. It is reported that alterations in melatonin follow TBI and may be linked with various sleep and circadian disorders directly (via cellular signaling) or indirectly (via free radicals and inflammatory signaling). Work over the past two decades has contributed to our understanding of the role of melatonin as a sleep regulator and neuroprotective anti-inflammatory agent. Although there is increasing interest in the treatment of insomnia following TBI, a lack of standardization and rigor in melatonin research has left behind a trail of non-generalizable data and ambiguous treatment recommendations. This narrative review describes the underlying biochemical properties of melatonin as they are relevant to TBI. We also discuss potential benefits and a path forward regarding the therapeutic management of TBI with melatonin treatment, including its role as a neuroprotectant, a somnogen, and a modulator of the circadian rhythm.
Collapse
Affiliation(s)
- Allen Bell
- Walter Reed National Military Medical Center, Bethesda, MD 20814, USA
| | - Bryson Hewins
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (B.H.)
| | - Courtney Bishop
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (B.H.)
| | - Amanda Fortin
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (B.H.)
| | - Jonathan Wang
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (B.H.)
| | | | - Jacob Collen
- Walter Reed National Military Medical Center, Bethesda, MD 20814, USA
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (B.H.)
| | - J. Kent Werner
- Walter Reed National Military Medical Center, Bethesda, MD 20814, USA
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (B.H.)
| |
Collapse
|
13
|
The Role of Melatonin in Pregnancy and the Health Benefits for the Newborn. Biomedicines 2022; 10:biomedicines10123252. [PMID: 36552008 PMCID: PMC9775355 DOI: 10.3390/biomedicines10123252] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/08/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
In the last few years, there have been significant evolutions in the understanding of the hormone melatonin in terms of its physiology, regulatory role, and potential utility in various domains of clinical medicine. Melatonin's properties include, among others, the regulation of mitochondrial function, anti-inflammatory, anti-oxidative and neuro-protective effects, sleep promotion and immune enhancement. As it is also bioavailable and has little or no toxicity, it has been proposed as safe and effective for the treatment of numerous diseases and to preserve human health. In this manuscript, we tried to evaluate the role of melatonin at the beginning of human life, in pregnancy, in the fetus and in newborns through newly published literature studies.
Collapse
|
14
|
Liu X, Cui Y, Zhang Y, Xiang G, Yu M, Wang X, Qiu B, Li XG, Liu W, Zhang D. Rescue of social deficits by early-life melatonin supplementation through modulation of gut microbiota in a murine model of autism. Biomed Pharmacother 2022; 156:113949. [DOI: 10.1016/j.biopha.2022.113949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
|
15
|
Pedroza-García KA, Calderón-Vallejo D, Quintanar JL. Neonatal Hypoxic-Ischemic Encephalopathy: Perspectives of Neuroprotective and Neuroregenerative Treatments. Neuropediatrics 2022; 53:402-417. [PMID: 36030792 DOI: 10.1055/s-0042-1755235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a serious condition that could have deleterious neurological outcomes, such as cerebral palsy, neuromotor disability, developmental disability, epilepsy, and sensitive or cognitive problems, and increase the risk of death in severe cases. Once HIE occurs, molecular cascades are triggered favoring the oxidative stress, excitotoxicity, and inflammation damage that promote cell death via apoptosis or necrosis. Currently, the therapeutic hypothermia is the standard of care in HIE; however, it has a small window of action and only can be used in children of more than 36 gestational weeks; for this reason, it is very important to develop new therapies to prevent the progression of the hypoxic-ischemic injury or to develop neuroregenerative therapies in severe HIE cases. The objective of this revision is to describe the emerging treatments for HIE, either preventing cell death for oxidative stress, excitotoxicity, or exacerbated inflammation, as well as describing a new therapeutic approach for neuroregeneration, such as mesenchymal stem cells, brain-derived neurotrophic factor, and gonadotropin realizing hormone agonists.
Collapse
Affiliation(s)
- Karina A Pedroza-García
- Departamento de Fisiología y Farmacología, Laboratorio de Neurofisiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
| | - Denisse Calderón-Vallejo
- Departamento de Fisiología y Farmacología, Laboratorio de Neurofisiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México.,Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
| | - J Luis Quintanar
- Departamento de Fisiología y Farmacología, Laboratorio de Neurofisiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
| |
Collapse
|
16
|
D'angelo G, Cannavò L, Reiter RJ, Gitto E. Melatonin Administration from 2000 to 2020 to Human Newborns with Hypoxic-Ischemic Encephalopathy. Am J Perinatol 2022; 39:824-829. [PMID: 33129208 DOI: 10.1055/s-0040-1719151] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is the main cause of long-term neurodevelopmental morbidity in term born infants worldwide. Melatonin is a hormone with antioxidant and anti-inflammatory effects that make it a promising molecule for the treatment of perinatal asphyxia. Probably, the synergistic use of hypothermia associated with melatonin treatment may improve the neurological outcome in infants with HIE. In the past 20 years, the efficacy of melatonin in reducing oxidative stress has been demonstrated in animals; however, clinical trials with sufficient sample size of newborns are lacking to date. Since in 2000 we were among the first to study the neuroprotective properties of melatonin on infants, in this review, we want to summarize the advantages and limitations of the investigations conducted to date. KEY POINTS: · HIE is the main cause of morbidity in term born infants worldwide.. · Melatonin is a promising molecule for the treatment of perinatal asphyxia.. · This review summarizes advantages and limitations of the investigations conducted on melatonin..
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, Messina, Italy
| | - Laura Cannavò
- Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi," University of Messina, Messina, Italy
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Eloisa Gitto
- Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi," University of Messina, Messina, Italy
| |
Collapse
|
17
|
Bishop-Freeman SC, Young KA, Labay LM, Beuhler MC, Hudson JS. Melatonin Supplementation in Undetermined Pediatric Deaths. J Anal Toxicol 2022; 46:808-816. [PMID: 35639879 DOI: 10.1093/jat/bkac033] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/12/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Since 2015, the North Carolina Office of the Chief Medical Examiner has investigated seven deaths of infants and toddlers, ages 2 months to 3 years, with exogenous melatonin detected upon toxicological analysis. Melatonin concentrations ranged from 3-1400 ng/mL in postmortem whole blood. While the cause and the manner of all seven deaths were classified as undetermined, the analytical findings are noteworthy. Melatonin is generally considered a safe, natural product appearing in many over-the-counter supplements geared towards young children to facilitate calmness and improve sleep. Melatonin is a neurohormone, which regulates not only circadian rhythms and natural sleep, but other physiological functions. Endogenous melatonin production, derived from essential amino acid metabolism, does not begin until pineal gland maturation at around three months of age with concentrations in plasma peaking during periods of darkness at approximately 0.2 ng/mL. Administering commercially available melatonin supplements to infants results in levels orders of magnitude greater than endogenous sources which should not be assumed to be safe just because of its endogenous nature. The finding of exogenous concentrations in some postmortem pediatric cases warrants attention. Several topics of interest surrounding these postmortem melatonin findings will be considered, such as minimal regulatory control over commercial products as well as the potential impact on hazardous sleeping conditions. This manuscript will outline the physiological effects of melatonin and detail the case studies from the NC medical examiner system. Forensic toxicology laboratories should consider including melatonin at exogenous concentrations in their testing schemes for appropriate postmortem infant and toddler cases.
Collapse
Affiliation(s)
- Sandra C Bishop-Freeman
- North Carolina Office of Chief Medical Examiner, 4312 District Dr. Raleigh, NC 27607, USA.,University of North Carolina, Department of Pathology and Laboratory Medicine, Chapel Hill, NC 27514, USA
| | - Kerry A Young
- North Carolina Office of Chief Medical Examiner, 4312 District Dr. Raleigh, NC 27607, USA
| | | | - Michael C Beuhler
- Carolinas Poisons Control, 5000 Airport Center Pkwy Suite B Charlotte, NC 28208, USA
| | - Jason S Hudson
- North Carolina Office of Chief Medical Examiner, 4312 District Dr. Raleigh, NC 27607, USA.,University of North Carolina, Department of Pathology and Laboratory Medicine, Chapel Hill, NC 27514, USA
| |
Collapse
|
18
|
Siahanidou T, Spiliopoulou C. Pharmacological Neuroprotection of the Preterm Brain: Current Evidence and Perspectives. Am J Perinatol 2022; 39:479-491. [PMID: 32961562 DOI: 10.1055/s-0040-1716710] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite improvements in viability, the long-term neurodevelopmental outcomes of preterm babies remain serious concern as a significant percentage of these infants develop neurological and/or intellectual impairment, and they are also at increased risk of psychiatric illnesses later in life. The current challenge is to develop neuroprotective approaches to improve adverse outcomes in preterm survivors. The purpose of this review was to provide an overview of the current evidence on pharmacological agents targeting the neuroprotection of the preterm brain. Among them, magnesium sulfate, given antenatally to pregnant women with imminent preterm birth before 30 to 34 weeks of gestation, as well as caffeine administered to preterm infants after birth, exhibited neuroprotective effects for human preterm brain. Erythropoietin treatment of preterm infants did not result in neuroprotection at 2 years of age in two out of three published large randomized controlled trials; however, long-term follow-up of these infants is needed to come to definite conclusions. Further studies are also required to assess whether melatonin, neurosteroids, inhaled nitric oxide, allopurinol, or dietary supplements (omega-3 fatty acids, choline, curcumin, etc.) could be implemented as neuroprotectants in clinical practice. Furthermore, other pharmacological agents showing promising signs of neuroprotective efficacy in preclinical studies (growth factors, hyaluronidase inhibitors or treatment, antidiabetic drugs, cannabidiol, histamine-H3 receptor antagonists, etc.), as well as stem cell- or exosomal-based therapies and nanomedicine, may prove useful in the future as potential neuroprotective approaches for human preterm brain. KEY POINTS: · Magnesium and caffeine have neuroprotective effects for the preterm brain.. · Follow-up of infants treated with erythropoietin is needed.. · Neuroprotective efficacy of several drugs in animals needs to be shown in humans..
Collapse
Affiliation(s)
- Tania Siahanidou
- Neonatal Unit of the First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | |
Collapse
|
19
|
Abiramalatha T, Ramaswamy VV, Ponnala AK, Kallem VR, Murkunde YV, Punnoose AM, Vivekanandhan A, Pullattayil AK, Amboiram P. Emerging neuroprotective interventions in periventricular leukomalacia: A systematic review of preclinical studies. Expert Opin Investig Drugs 2022; 31:305-330. [PMID: 35143732 DOI: 10.1080/13543784.2022.2040479] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Periventricular leukomalacia (PVL) is a result of various antenatal, intrapartum, or postnatal insults to the developing brain and is an important harbinger of cerebral palsy in preterm neonates. There is no proven therapy for PVL. This calls for appraisal of targeted therapies that have been investigated in animal models to evaluate their relevance in clinical research context. AREAS COVERED This systematic review identifies interventions that were evaluated in preclinical studies for neuroprotective efficacy against PVL. We identified 142 studies evaluating various interventions in PVL animal models. (Search method is detailed in section 2). EXPERT OPINION Interventions that have yielded significant results in preclinical research, and that have been evaluated in a limited number of clinical trials include stem cells, erythropoietin, and melatonin. Many other therapeutic modalities evaluated in preclinical studies have been identified, but more data on their neuroprotective potential in PVL must be garnered before they can be considered for clinical trials. Because most of the tested interventions had only a partial efficacy, a combination of interventions that could be synergistic should be investigated in future preclinical studies. Furthermore, since the nature and pattern of perinatal insults to preterm brain predisposing it to PVL are substantially variable, individualised approaches for the choice of appropriate neuroprotective interventions tailored to different sub-groups of preterm neonates should be explored.
Collapse
Affiliation(s)
- Thangaraj Abiramalatha
- Consultant Neonatologist, Kovai Medical Center and Hospital (KMCH).,Department of Pediatrics and Neonatology, KMCH Institute of Health Sciences and Research, Coimbatore, India
| | | | - Andelsivj Kumar Ponnala
- Centre for Toxicology and Developmental Research (CEFTE), Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | | | - Yogeshkumar V Murkunde
- Centre for Toxicology and Developmental Research (CEFTE), Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Alan Mathew Punnoose
- Department of Stem Cell Research and Regenerative Medicine, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | | | | | - Prakash Amboiram
- Department of Neonatology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
20
|
Antioxidant Effect of Melatonin in Preterm Newborns. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6308255. [PMID: 34840669 PMCID: PMC8626170 DOI: 10.1155/2021/6308255] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/08/2021] [Accepted: 10/29/2021] [Indexed: 01/13/2023]
Abstract
Introduction Preterm infants are at risk of free radical-mediated diseases from oxidative stress (OS) injury. Increased free radical generation has been demonstrated in preterm infants during the first seven days of life. Melatonin (MEL) is a powerful antioxidant and scavenger of free radicals. In preterm neonates, melatonin deficiency has been reported. Exogenous melatonin administration appears a promising strategy in the treatment of neonatal morbidities in which OS has a leading role. Objective The aim was to evaluate plasma MEL concentrations and OS biomarkers in preterm newborns after early administration of melatonin. Methods A prospective, randomized double-blind placebo-controlled pilot study was conducted from January 2019 to September 2020. Thirty-six preterm newborns were enrolled. Starting from the first day of life, 21 received a single dose of oral melatonin 0.5 mg/kg once a day, in the morning (MEL group); 15 newborns received an equivalent dose of placebo (placebo group). Samples of 0.2 mL of plasma were collected at 24 and 48 hours after MEL administration. Plasma concentrations of melatonin, non-protein-bound iron (NPBI), advanced oxidation protein products (AOPP), and F2-isoprostanes (F2-Isopr) were measured. Babies were clinically followed until discharge. Results At 24 and 48 hours after MEL administration, the MEL concentrations were significantly higher in the MEL group than in the placebo group (52759.30 ± 63529.09 vs. 28.57 ± 46.24 pg/mL and 279397.6 ± 516344.2 vs. 38.50 ± 44.01 pg/mL, respectively). NPBI and AOPP did not show any statistically significant differences between the groups both at 24 and 48 hours. At 48 hours, the mean blood concentrations of F2-Isopr were significantly lower in the MEL group than in the placebo group (36.48 ± 33.85 pg/mL vs.89.97 ± 52.01 pg/mL). Conclusions Early melatonin administration in preterm newborns reduces lipid peroxidation in the first days of life showing a potential role to protect high-risk newborns. Trial Registration. This trial is registered with NCT04785183, Early Supplementation of Melatonin in Preterm Newborns: the Effects on Oxidative Stress.
Collapse
|
21
|
Vine T, Brown GM, Frey BN. Melatonin use during pregnancy and lactation: A scoping review of human studies. BRAZILIAN JOURNAL OF PSYCHIATRY 2021; 44:342-348. [PMID: 34730672 PMCID: PMC9169489 DOI: 10.1590/1516-4446-2021-2156] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/01/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The prevalence of sleep disorders during the perinatal period is high and large health administrative database surveys have shown that the use of exogenous melatonin in pregnant populations is quite common, about 4%. Much of the concern about using melatonin during pregnancy and breastfeeding stems from animal research. Thus, the objective of this article is to provide a critical review of human studies related to exogenous melatonin use during pregnancy and breastfeeding. METHODS The electronic databases Ovid, MEDLINE, Embase, and the Cochrane Library were searched using terms and keywords related to melatonin, pregnancy, and breastfeeding. RESULTS Fifteen studies were included in this review. Eight focused on melatonin use during pregnancy and seven focused on melatonin use during breastfeeding. There was a variety of study designs, including case reports, cohort studies, and clinical trials. There is a lack of randomized, controlled trials examining the efficacy and safety of melatonin as a treatment for sleep disorders during pregnancy or breastfeeding and, notably, insomnia was not the primary outcome measure in any of the studies included in this review. Clinical trials that used exogenous melatonin during pregnancy and breastfeeding for other clinical conditions have not suggested major safety concerns or adverse events. CONCLUSION Contrary to what animal studies have suggested, evidence from clinical studies to date suggests that melatonin use during pregnancy and breastfeeding is probably safe in humans. This review further emphasizes the need for clinical studies on sleep disorders, including exogenous melatonin, during pregnancy and lactation.
Collapse
Affiliation(s)
- Tya Vine
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - Gregory M Brown
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Molecular Brain Science Department, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Benicio N Frey
- Women's Health Concerns Clinic and Mood Disorder Program, St Joseph's Healthcare Hamilton, Hamilton, ON, Canada.,Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
22
|
Neuroprotective Agents for Neonates with Hypoxic-Ischemic Encephalopathy. Neonatal Netw 2021; 40:406-413. [PMID: 34845092 DOI: 10.1891/11-t-755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 11/25/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE) remains a significant source of long-term neurodevelopmental impairment despite overall improvements in survival without disability in neonates who undergo therapeutic hypothermia. Each phase in the evolution of hypoxic-ischemic injury presents potential pharmacologic targets for neuroprotective agents. Melatonin is a promising emerging therapy for early phases of ischemic injury, but utility is currently limited by the lack of pharmaceutical-grade products. Magnesium has been extensively studied for its neuroprotective effects in the preterm population. Studies in neonates with HIE have produced mixed outcomes. Erythropoietin use in HIE with or without therapeutic hypothermia appears to be safe and may provide additional benefit. Dexmedetomidine, N-acetylcysteine, xenon, and topiramate all have promising animal data, but need additional human trials to elucidate what role they may play in HIE. Frequent review of existing literature is required to ensure provision of evidence-based pharmacologic agents for neuroprotection following HIE.
Collapse
|
23
|
O'Connor KM, Ashoori M, Dias ML, Dempsey EM, O'Halloran KD, McDonald FB. Influence of innate immune activation on endocrine and metabolic pathways in infancy. Am J Physiol Endocrinol Metab 2021; 321:E24-E46. [PMID: 33900849 DOI: 10.1152/ajpendo.00542.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prematurity is the leading cause of neonatal morbidity and mortality worldwide. Premature infants often require extended hospital stays, with increased risk of developing infection compared with term infants. A picture is emerging of wide-ranging deleterious consequences resulting from innate immune system activation in the newborn infant. Those who survive infection have been exposed to a stimulus that can impose long-lasting alterations into later life. In this review, we discuss sepsis-driven alterations in integrated neuroendocrine and metabolic pathways and highlight current knowledge gaps in respect of neonatal sepsis. We review established biomarkers for sepsis and extend the discussion to examine emerging findings from human and animal models of neonatal sepsis that propose novel biomarkers for early identification of sepsis. Future research in this area is required to establish a greater understanding of the distinct neonatal signature of early and late-stage infection, to improve diagnosis, curtail inappropriate antibiotic use, and promote precision medicine through a biomarker-guided empirical and adjunctive treatment approach for neonatal sepsis. There is an unmet clinical need to decrease sepsis-induced morbidity in neonates, to limit and prevent adverse consequences in later life and decrease mortality.
Collapse
Affiliation(s)
- K M O'Connor
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - M Ashoori
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| | - M L Dias
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - E M Dempsey
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, School of Medicine, College of Medicine and Health, Cork University Hospital, Wilton, Cork, Ireland
| | - K D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| | - F B McDonald
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
- Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| |
Collapse
|
24
|
Pang R, Advic-Belltheus A, Meehan C, Fullen DJ, Golay X, Robertson NJ. Melatonin for Neonatal Encephalopathy: From Bench to Bedside. Int J Mol Sci 2021; 22:5481. [PMID: 34067448 PMCID: PMC8196955 DOI: 10.3390/ijms22115481] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/21/2022] Open
Abstract
Neonatal encephalopathy is a leading cause of morbidity and mortality worldwide. Although therapeutic hypothermia (HT) is now standard practice in most neonatal intensive care units in high resource settings, some infants still develop long-term adverse neurological sequelae. In low resource settings, HT may not be safe or efficacious. Therefore, additional neuroprotective interventions are urgently needed. Melatonin's diverse neuroprotective properties include antioxidant, anti-inflammatory, and anti-apoptotic effects. Its strong safety profile and compelling preclinical data suggests that melatonin is a promising agent to improve the outcomes of infants with NE. Over the past decade, the safety and efficacy of melatonin to augment HT has been studied in the neonatal piglet model of perinatal asphyxia. From this model, we have observed that the neuroprotective effects of melatonin are time-critical and dose dependent. Therapeutic melatonin levels are likely to be 15-30 mg/L and for optimal effect, these need to be achieved within the first 2-3 h after birth. This review summarises the neuroprotective properties of melatonin, the key findings from the piglet and other animal studies to date, and the challenges we face to translate melatonin from bench to bedside.
Collapse
Affiliation(s)
- Raymand Pang
- Institute for Women’s Health, University College London, London WC1E 6HU, UK; (R.P.); (A.A.-B.); (C.M.)
| | - Adnan Advic-Belltheus
- Institute for Women’s Health, University College London, London WC1E 6HU, UK; (R.P.); (A.A.-B.); (C.M.)
| | - Christopher Meehan
- Institute for Women’s Health, University College London, London WC1E 6HU, UK; (R.P.); (A.A.-B.); (C.M.)
| | - Daniel J. Fullen
- Translational Research Office, University College London, London W1T 7NF, UK;
| | - Xavier Golay
- Department of Brain Repair and Rehabilitation, Institute of Neurology, University College London, London WC1N 3BG, UK;
| | - Nicola J. Robertson
- Institute for Women’s Health, University College London, London WC1E 6HU, UK; (R.P.); (A.A.-B.); (C.M.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| |
Collapse
|
25
|
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: 2.3] [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
|
26
|
Abstract
Oxidative stress (OS) plays a key role in the pathophysiology of preterm infants. Accurate assessment of OS remains an analytical challenge that has been partially addressed during the last few decades. A plethora of approaches have been developed to assess preterm biofluids to demonstrate a link postnatally with preterm OS, giving rise to a set of widely employed biomarkers. However, the vast number of different analytic methods and lack of standardization hampers reliable comparison of OS-related biomarkers. In this chapter, we discuss approaches for the study of OS in prematurity with respect to methodologic considerations, the metabolic source of different biomarkers and their role in clinical studies.
Collapse
|
27
|
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: 6] [Impact Index Per Article: 1.5] [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
|
28
|
Wu ZY, Huang SD, Zou JJ, Wang QX, Naveed M, Bao HN, Wang W, Fukunaga K, Han F. Autism spectrum disorder (ASD): Disturbance of the melatonin system and its implications. Biomed Pharmacother 2020; 130:110496. [DOI: 10.1016/j.biopha.2020.110496] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
|
29
|
Procaccini DE, Lobner K, Anton B, Kudchadkar SR. Melatonin Use in Hospitalized Children for Non-Anesthetic Indications: A Systematic Review. Pharmacotherapy 2020; 40:692-703. [PMID: 32342515 DOI: 10.1002/phar.2408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Melatonin, a potent free radical scavenger, plays an important role in homeostasis of cell and organ physiology. The increased demand and synthesis from the pineal gland during times of oxidative stress suggests a potential benefit of melatonin supplementation during hospitalization for acute illness. Yet, the paucity of clinical studies for non-anesthetic-associated indications in pediatric populations hampers the safe, effective, and consistent use of melatonin. The objective of this study was to systematically review published studies of melatonin use for non-sedative and non-analgesic indications in hospitalized pediatric patients. We conducted a search of PubMed, EMBASE, Cumulative Index to Nursing and Allied Health Literature (CINAHL), Web of Science, and Scopus databases for articles on the use of melatonin for pediatric patients in a hospital setting. Thirteen eligible studies, all in neonates, were identified. Data elements extracted included study design, number of study subjects, indication for melatonin therapy, and melatonin regimen (formulation, dosage, and duration). Because study methodologies were very heterogeneous, a quantitative synthesis of the published findings was not possible. The identified studies were therefore categorized by the indication of melatonin (adjuvant-antioxidant or anti-inflammatory therapy) in the following specific disease states: (i) acute infections, (ii) respiratory distress syndrome, (iii) neurologic injury, and (iv) jaundice. The current data suggest that melatonin is safe for use in hospitalized neonates. Melatonin may be beneficial for reducing inflammatory markers in neonatal patients with disease states and clinical sequelae that are associated with increased inflammation and oxidative stress. Melatonin, in conjunction with phototherapy, is not superior to use of vitamin D with phototherapy for treatment of neonatal jaundice. However, studies in other pediatric populations are needed given widespread use across clinical inpatient settings.
Collapse
Affiliation(s)
- David E Procaccini
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Katie Lobner
- Welch Medical Library, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Blair Anton
- Welch Medical Library, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sapna R Kudchadkar
- Departments of Anesthesiology & Critical Care Medicine, Pediatrics, and Physical Medicine & Rehabilitation, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| |
Collapse
|
30
|
Pourhanifeh MH, Hosseinzadeh A, Dehdashtian E, Hemati K, Mehrzadi S. Melatonin: new insights on its therapeutic properties in diabetic complications. Diabetol Metab Syndr 2020; 12:30. [PMID: 32280378 PMCID: PMC7140344 DOI: 10.1186/s13098-020-00537-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetes and diabetic complications are considered as leading causes of both morbidity and mortality in the world. Unfortunately, routine medical treatments used for affected patients possess undesirable side effects, including kidney and liver damages as well as gastrointestinal adverse reactions. Therefore, exploring the novel therapeutic strategies for diabetic patients is a crucial issue. It has been recently shown that melatonin, as main product of the pineal gland, despite its various pharmacological features including anticancer, anti-aging, antioxidant and anti-inflammatory effects, exerts anti-diabetic properties through regulating various cellular mechanisms. The aim of the present review is to describe potential roles of melatonin in the treatment of diabetes and its complications.
Collapse
Affiliation(s)
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Dehdashtian
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Karim Hemati
- Department of Anesthesiology, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Cardinali DP. An Assessment of Melatonin's Therapeutic Value in the Hypoxic-Ischemic Encephalopathy of the Newborn. Front Synaptic Neurosci 2019; 11:34. [PMID: 31920617 PMCID: PMC6914689 DOI: 10.3389/fnsyn.2019.00034] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/26/2019] [Indexed: 12/17/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is one of the most frequent causes of brain injury in the newborn. From a pathophysiological standpoint, a complex process takes place at the cellular and tissue level during the development of newborn brain damage in the absence of oxygen. Initially, the lesion is triggered by a deficit in the supply of oxygen to cells and tissues, causing a primary energy insufficiency. Subsequently, high energy phosphate levels recover transiently (the latent phase) that is followed by a secondary phase, in which many of the pathophysiological mechanisms involved in the development of neonatal brain damage ensue (i.e., excitotoxicity, massive influx of Ca2+, oxidative and nitrosative stress, inflammation). This leads to cell death by necrosis or apoptosis. Eventually, a tertiary phase occurs, characterized by the persistence of brain damage for months and even years after the HI insult. Hypothermia is the only therapeutic strategy against HIE that has been incorporated into neonatal intensive care units with limited success. Thus, there is an urgent need for agents with the capacity to curtail acute and chronic damage in HIE. Melatonin, a molecule of unusual phylogenetic conservation present in all known aerobic organisms, has a potential role as a neuroprotective agent both acutely and chronically in HIE. Melatonin displays a remarkable antioxidant and anti-inflammatory activity and is capable to cross the blood-brain barrier readily. Moreover, in many animal models of brain degeneration, melatonin was effective to impair chronic mechanisms of neuronal death. In animal models, and in a limited number of clinical studies, melatonin increased the level of protection developed by hypothermia in newborn asphyxia. This review article summarizes briefly the available therapeutic strategies in HIE and assesses the role of melatonin as a potentially relevant therapeutic tool to cover the hypoxia-ischemia phase and the secondary and tertiary phases following a hypoxic-ischemic insult.
Collapse
Affiliation(s)
- Daniel P. Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| |
Collapse
|
32
|
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
|
33
|
Variations in melatonin levels in preterm and term human breast milk during the first month after delivery. Sci Rep 2019; 9:17984. [PMID: 31784629 PMCID: PMC6884443 DOI: 10.1038/s41598-019-54530-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/11/2019] [Indexed: 12/20/2022] Open
Abstract
The objectives of the present study were to examine the dynamic changes in breast milk melatonin throughout the course of lactation and to explore factors associated with changes in melatonin concentrations and rhythms in both preterm and term breast milk. Breast milk was collected sequentially at 03:00, 09:00, 15:00, and 21:00 in one day. Melatonin was analyzed in 392 breast milk samples from 98 healthy nursing mothers at 0 to 30 days postpartum. In both preterm and term breast milk, the melatonin concentration presented a circadian rhythm with the acrophase at around 03:00. Subgroup analysis showed the peak melatonin concentrations differed significantly across lactation stages, with the highest concentration in the colostrum, followed by transitional and mature breast milk. At 03:00, preterm breast milk had a higher concentration of melatonin than term breast milk in the colostrum (28.67 pg/mL vs. 25.31 pg/mL, p < 0.022), transitional breast milk (24.70 pg/mL vs. 22.55 pg/mL), and mature breast milk (22.37 pg/mL vs. 20.12 pg /mL). Further studies are warranted for their roles and significance on melatonin in breast milk in nutrition and metabolism of neonates.
Collapse
|
34
|
Romeo DM, Leo G, Lapenta L, Leone D, Turrini I, Brogna C, Gallini F, Cota F, Vento G, Mercuri E. Sleep disorders in low-risk preschool very preterm children. Sleep Med 2019; 63:137-141. [DOI: 10.1016/j.sleep.2019.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 04/17/2019] [Accepted: 04/19/2019] [Indexed: 01/09/2023]
|
35
|
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: 1.2] [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
|
36
|
Balduini W, Weiss MD, Carloni S, Rocchi M, Sura L, Rossignol C, Longini M, Bazzini F, Perrone S, Ott D, Wadhawan R, Buonocore G. Melatonin pharmacokinetics and dose extrapolation after enteral infusion in neonates subjected to hypothermia. J Pineal Res 2019; 66:e12565. [PMID: 30734962 DOI: 10.1111/jpi.12565] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/01/2019] [Accepted: 02/01/2019] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Neonates with hypoxic-ischemic encephalopathy (HIE) undergoing hypothermia may benefit from adjunctive therapy with melatonin. However, melatonin safety, pharmacokinetics (PK), and dosage in this sensitive population are still unknown. METHODS AND RESULTS This study assessed the PK and safety of melatonin enteral administration to neonates with HIE undergoing hypothermia. Melatonin was infused at 0.5 mg/kg in five neonates with HIE undergoing hypothermia. Infusion started 1 hour after the neonates reached the target temperature of 33.5°C. Blood samples were collected before and at selective times after melatonin infusion. Abdominal complications or clinically significant changes in patients' vital signs were not found during or after melatonin. The peak plasma concentration reached 0.25 µg/mL. The area under the curve in 24 hours was 4.35 µg/mL*h. DISCUSSION Melatonin half-life and clearance were prolonged, and the distribution volume decreased compared to adults. In silico simulation estimated that the steady state can be reached after four infusions. Hypothermia does not affect melatonin PK. In humans high blood concentrations with lower doses can be achieved compared to animal experimentation, although intravenous administration is advised in the neonate population. Our study is a preparatory step for future clinical studies aimed at assessing melatonin efficacy in HIE.
Collapse
Affiliation(s)
- Walter Balduini
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Michael D Weiss
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Marco Rocchi
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Livia Sura
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Candace Rossignol
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Mariangela Longini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Francesco Bazzini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Serafina Perrone
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Deborah Ott
- Department of Pediatrics, Florida Hospital, Orlando, Florida
| | - Rajan Wadhawan
- Department of Pediatrics, Florida Hospital, Orlando, Florida
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
37
|
Biran V, Decobert F, Bednarek N, Boizeau P, Benoist JF, Claustrat B, Barré J, Colella M, Frérot A, Garnotel R, Graesslin O, Haddad B, Launay JM, Schmitz T, Schroedt J, Virlouvet AL, Guilmin-Crépon S, Yacoubi A, Jacqz-Aigrain E, Gressens P, Alberti C, Baud O. Melatonin Levels in Preterm and Term Infants and Their Mothers. Int J Mol Sci 2019; 20:ijms20092077. [PMID: 31035572 PMCID: PMC6540351 DOI: 10.3390/ijms20092077] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 04/23/2019] [Indexed: 11/16/2022] Open
Abstract
The prevention of perinatal brain damage following preterm birth remains a public health priority. Melatonin has been shown to be a promising neuroprotectant in neonatal preclinical models of brain damage, but few studies have investigated melatonin secretion in newborns. We hypothesized that melatonin circulating levels would be lower in preterm compared to term infants. We conducted a prospective, longitudinal, multicenter study to assess melatonin, and 6-sulfatoxy-melatonin (aMT6s) concentrations, measured by radioimmunoassay. Among 209 neonates recruited, 110 were born before 34 gestational weeks (GW) and 99 born after 34 GW. Plasma melatonin concentrations, measured at birth and on Day 3 were below detectable levels (≤7 pg/mL) in 78% and 81%, respectively, of infants born before 34 GW compared to 57% and 34%, respectively, of infants born after 34 GW. The distribution of plasma melatonin concentrations was found to be correlated with gestational age at both time-points (p < 0.001). Median urine aMT6s concentrations were significantly lower in infants born before 34 GW, both on Day 1 (230 ng/L vs. 533 ng/L, p < 0.0001) and on Day 3 (197 ng/L vs. 359 ng/L, p < 0.0001). In conclusion, melatonin secretion appears very low in preterm infants, providing the rationale for testing supplemental melatonin as a neuroprotectant in clinical trials.
Collapse
Affiliation(s)
- Valérie Biran
- Neonatal Intensive Care Unit, Assistance Publique-Hôpitaux de Paris, Robert Debré Children's Hospital, University Paris Diderot, Sorbonne Paris-Cité, 75019 Paris, France.
- PROTECT, Inserm 1141, Université Paris Diderot, Sorbonne Paris Cité, 75019 Paris, France.
- PremUP Foundation, 75014 Paris, France.
| | - Fabrice Decobert
- PremUP Foundation, 75014 Paris, France.
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal, 94010 Créteil, France.
| | - Nathalie Bednarek
- Neonatal Intensive Care Unit, American Memorial Hospital, 51100 Reims, France.
| | - Priscilla Boizeau
- Unit of Clinical Epidemiology, Assistance Publique-Hôpitaux de Paris, Robert Debré Children's Hospital, University Paris Diderot, Sorbonne Paris-Cité, Inserm U1123 and CIC-EC 1426, 75019 Paris, France.
| | - Jean-François Benoist
- Biochemistry Department, Assistance Publique-Hôpitaux de Paris, Robert Debré Children's Hospital, 75019 Paris, France.
| | - Bruno Claustrat
- Hormonology Department, Groupement hospitalier Est-Hospices Civils de Lyon, 69500 Bron, France.
| | - Jérôme Barré
- Centre de Ressources Biologiques, Centre Hospitalier Intercommunal Créteil, 94010 Créteil, France.
| | - Marina Colella
- Neonatal Intensive Care Unit, Robert Debré Hospital, 75019 Paris, France.
| | - Alice Frérot
- Neonatal Intensive Care Unit, Robert Debré Hospital, 75019 Paris, France.
| | - Roselyne Garnotel
- Biochemistry Laboratory, American Memorial Hospital Reims, 51100 Reims, France.
| | - Olivier Graesslin
- Department of Gynecology and Obstetrics, American Memorial Hospital Reims, 51100 Reims, France.
| | - Bassam Haddad
- Department of Gynecology and Obstetrics, Centre Hospitalier Intercommunal Créteil, 94010 Créteil, France.
| | - Jean-Marie Launay
- Biochemistry and Molecular Laboratory, Lariboisière Hospital, 75019 Paris, France.
| | - Thomas Schmitz
- Department of Gynecology and Obstetrics, Robert Debré Hospital, 75019 Paris, France.
| | | | | | | | - Adyla Yacoubi
- UEC CIC 1426, Robert Debré Hospital, 75019 Paris, France.
| | - Evelyne Jacqz-Aigrain
- Department of Pharmacology and Pharmacogenetics, Assistance Publique-Hôpitaux de Paris, Robert Debré Children's Hospital, 75019 Paris, France.
| | - Pierre Gressens
- PROTECT, Inserm 1141, Université Paris Diderot, Sorbonne Paris Cité, 75019 Paris, France.
- PremUP Foundation, 75014 Paris, France.
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, UK.
| | - Corinne Alberti
- Unit of Clinical Epidemiology, Assistance Publique-Hôpitaux de Paris, Robert Debré Children's Hospital, University Paris Diderot, Sorbonne Paris-Cité, Inserm U1123 and CIC-EC 1426, 75019 Paris, France.
| | - Olivier Baud
- Neonatal Intensive Care Unit, Assistance Publique-Hôpitaux de Paris, Robert Debré Children's Hospital, University Paris Diderot, Sorbonne Paris-Cité, 75019 Paris, France.
- PROTECT, Inserm 1141, Université Paris Diderot, Sorbonne Paris Cité, 75019 Paris, France.
- PremUP Foundation, 75014 Paris, France.
- Division of Neonatology and Pediatric Intensive Care, Children's University Hospital and University of Geneva, 1205 Geneva, Switzerland.
| |
Collapse
|
38
|
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: 167] [Impact Index Per Article: 33.4] [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
|
39
|
Finch-Edmondson M, Morgan C, Hunt RW, Novak I. Emergent Prophylactic, Reparative and Restorative Brain Interventions for Infants Born Preterm With Cerebral Palsy. Front Physiol 2019; 10:15. [PMID: 30745876 PMCID: PMC6360173 DOI: 10.3389/fphys.2019.00015] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/08/2019] [Indexed: 12/13/2022] Open
Abstract
Worldwide, an estimated 15 million babies are born preterm (<37 weeks' gestation) every year. Despite significant improvements in survival rates, preterm infants often face a lifetime of neurodevelopmental disability including cognitive, behavioral, and motor impairments. Indeed, prematurity remains the largest risk factor for the development of cerebral palsy. The developing brain of the preterm infant is particularly fragile; preterm babies exhibit varying severities of cerebral palsy arising from reductions in both cerebral white and gray matter volumes, as well as altered brain microstructure and connectivity. Current intensive care therapies aim to optimize cardiovascular and respiratory function to protect the brain from injury by preserving oxygenation and blood flow. If a brain injury does occur, definitive diagnosis of cerebral palsy in the first few hours and weeks of life is difficult, especially when the lesions are subtle and not apparent on cranial ultrasound. However, early diagnosis of mildly affected infants is critical, because these are the patients most likely to respond to emergent treatments inducing neuroplasticity via high-intensity motor training programs and regenerative therapies involving stem cells. A current controversy is whether to test universal treatment in all infants at risk of brain injury, accepting that some patients never required treatment, because the perceived potential benefits outweigh the risk of harm. Versus, waiting for a diagnosis before commencing targeted treatment for infants with a brain injury, and potentially missing the therapeutic window. In this review, we discuss the emerging prophylactic, reparative, and restorative brain interventions for infants born preterm, who are at high risk of developing cerebral palsy. We examine the current evidence, considering the timing of the intervention with relation to the proposed mechanism/s of action. Finally, we consider the development of novel markers of preterm brain injury, which will undoubtedly lead to improved diagnostic and prognostic capability, and more accurate instruments to assess the efficacy of emerging interventions for this most vulnerable group of infants.
Collapse
Affiliation(s)
- Megan Finch-Edmondson
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Catherine Morgan
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Rod W. Hunt
- Department of Neonatal Medicine, The Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
- Neonatal Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Iona Novak
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
40
|
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: 4.4] [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
|
41
|
Abstract
Despite notable advances in the care and survival of preterm infants, a significant proportion of preterm neonates will have life-long cognitive, behavioral, and motor deficits, and robustly effective neuroprotective strategies are still missing. These therapies must target the pathophysiologic mechanisms observed in contemporaneous infants and rely on modern epidemiology, imaging, and experimental models and assessment techniques. Two drugs, magnesium sulfate and caffeine, are already in use in several units, and although their targets are apnea of prematurity and myometrial contractility (respectively), they do offer improved odds of positive outcomes. Nevertheless, these drugs have limited efficacy, and NICU-to-NICU administration varies greatly. As such, there is an obvious need for additional specific neurotherapeutic strategies to further enhance the outcome of this very fragile population of neonates. The chapter reviews these issues, highlights bottlenecks that need to be solved for meaningful progress in the field, and proposes future innovative avenues for intervention, including delayed interventions.
Collapse
Affiliation(s)
- Bobbi Fleiss
- NeuroDiderot, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, London, United Kingdom
| | - Pierre Gressens
- NeuroDiderot, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, London, United Kingdom.
| |
Collapse
|
42
|
Braam W, Ehrhart F, Maas APHM, Smits MG, Curfs L. Low maternal melatonin level increases autism spectrum disorder risk in children. RESEARCH IN DEVELOPMENTAL DISABILITIES 2018; 82:79-89. [PMID: 29501372 DOI: 10.1016/j.ridd.2018.02.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 02/05/2018] [Accepted: 02/17/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND It is assumed that autism spectrum disorder (ASD) is caused by a combination of de novo inherited variation and common variation as well as environmental factors. It often co-occurs with intellectual disability (ID). Almost eight hundred potential causative genetic variations have been found in ASD patients. However, not one of them is responsible for more than 1% of ASD cases. Low melatonin levels are a frequent finding in ASD patients. Melatonin levels are negatively correlated with severity of autistic impairments, it is important for normal neurodevelopment and is highly effective in protecting DNA from oxidative damage. Melatonin deficiency could be a major factor, and well a common heritable variation, that increases the susceptibility to environmental risk factors for ASD. ASD is already present at birth. As the fetus does not produce melatonin, low maternal melatonin levels may be involved. METHODS We measured 6-sulfatoxymelatonin in urine of 60 mothers of a child with ASD and controls. RESULTS 6-sulfatoxymelatonin levels were significantly lower in mothers with an ASD child than in controls (p = 0.012). CONCLUSIONS Low parental melatonin levels could be one of the contributors to ASD and possibly ID etiology. Our findings need to be duplicated on a larger scale. If our hypothesis is correct, this could lead to policies to detect future parents who are at risk and to treatment strategies to ASD and intellectual disability risk.
Collapse
Affiliation(s)
- Wiebe Braam
- 's Heeren Loo, Department Advisium, Wekerom, The Netherlands; Governor Kremers Centre, Maastricht University Medical Centre, The Netherlands.
| | - Friederike Ehrhart
- Governor Kremers Centre, Maastricht University Medical Centre, The Netherlands; Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, The Netherlands
| | - Anneke P H M Maas
- Governor Kremers Centre, Maastricht University Medical Centre, The Netherlands; Department of Special Education, Radboud University, Nijmegen, The Netherlands
| | - Marcel G Smits
- Governor Kremers Centre, Maastricht University Medical Centre, The Netherlands; Multidisciplinary expert centre for sleep-wake disturbances and chronobiology, Gelderse Vallei Hospital, Ede, The Netherlands
| | - Leopold Curfs
- Governor Kremers Centre, Maastricht University Medical Centre, The Netherlands
| |
Collapse
|
43
|
Dhillon SK, Lear CA, Galinsky R, Wassink G, Davidson JO, Juul S, Robertson NJ, Gunn AJ, Bennet L. The fetus at the tipping point: modifying the outcome of fetal asphyxia. J Physiol 2018; 596:5571-5592. [PMID: 29774532 DOI: 10.1113/jp274949] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022] Open
Abstract
Brain injury around birth is associated with nearly half of all cases of cerebral palsy. Although brain injury is multifactorial, particularly after preterm birth, acute hypoxia-ischaemia is a major contributor to injury. It is now well established that the severity of injury after hypoxia-ischaemia is determined by a dynamic balance between injurious and protective processes. In addition, mothers who are at risk of premature delivery have high rates of diabetes and antepartum infection/inflammation and are almost universally given treatments such as antenatal glucocorticoids and magnesium sulphate to reduce the risk of death and complications after preterm birth. We review evidence that these common factors affect responses to fetal asphyxia, often in unexpected ways. For example, glucocorticoid exposure dramatically increases delayed cell loss after acute hypoxia-ischaemia, largely through secondary hyperglycaemia. This critical new information is important to understand the effects of clinical treatments of women whose fetuses are at risk of perinatal asphyxia.
Collapse
Affiliation(s)
| | - Christopher A Lear
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Robert Galinsky
- The Department of Physiology, University of Auckland, Auckland, New Zealand.,The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
| | - Guido Wassink
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Joanne O Davidson
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Sandra Juul
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Alistair J Gunn
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- The Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
44
|
Aridas JDS, Yawno T, Sutherland AE, Nitsos I, Ditchfield M, Wong FY, Hunt RW, Fahey MC, Malhotra A, Wallace EM, Jenkin G, Miller SL. Systemic and transdermal melatonin administration prevents neuropathology in response to perinatal asphyxia in newborn lambs. J Pineal Res 2018; 64:e12479. [PMID: 29464766 PMCID: PMC5947141 DOI: 10.1111/jpi.12479] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/06/2018] [Indexed: 01/19/2023]
Abstract
Perinatal asphyxia remains a principal cause of infant mortality and long-term neurological morbidity, particularly in low-resource countries. No neuroprotective interventions are currently available. Melatonin (MLT), a potent antioxidant, anti-inflammatory and antiapoptotic agent, offers promise as an intravenous (IV) or transdermal therapy to protect the brain. We aimed to determine the effect of melatonin (IV or transdermal patch) on neuropathology in a lamb model of perinatal asphyxia. Asphyxia was induced in newborn lambs via umbilical cord occlusion at birth. Animals were randomly allocated to melatonin commencing 30 minutes after birth (60 mg in 24 hours; IV or transdermal patch). Brain magnetic resonance spectroscopy (MRS) was undertaken at 12 and 72 hours. Animals (control n = 9; control+MLT n = 6; asphyxia n = 16; asphyxia+MLT [IV n = 14; patch n = 4]) were euthanised at 72 hours, and cerebrospinal fluid (CSF) and brains were collected for analysis. Asphyxia resulted in severe acidosis (pH 6.9 ± 0.0; lactate 9 ± 2 mmol/L) and altered determinants of encephalopathy. MRS lactate:N-acetyl aspartate ratio was 2.5-fold higher in asphyxia lambs compared with controls at 12 hours and 3-fold higher at 72 hours (P < .05). Melatonin prevented this rise (3.5-fold reduced vs asphyxia; P = .02). Asphyxia significantly increased brain white and grey matter apoptotic cell death (activated caspase-3), lipid peroxidation (4HNE) and neuroinflammation (IBA-1). These changes were significantly mitigated by both IV and patch melatonin. Systemic or transdermal neonatal melatonin administration significantly reduces the neuropathology and encephalopathy signs associated with perinatal asphyxia. A simple melatonin patch, administered soon after birth, may improve outcome in infants affected by asphyxia, especially in low-resource settings.
Collapse
Affiliation(s)
- James D. S. Aridas
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
| | - Tamara Yawno
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVic.Australia
| | - Amy E. Sutherland
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
| | - Ilias Nitsos
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVic.Australia
| | | | - Flora Y. Wong
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Monash Children's HospitalMonash HealthClaytonVic.Australia
| | - Rod W. Hunt
- Murdoch Children's Research InstituteMelbourneVic.Australia
| | - Michael C. Fahey
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Monash Children's HospitalMonash HealthClaytonVic.Australia
| | - Atul Malhotra
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Monash Children's HospitalMonash HealthClaytonVic.Australia
| | - Euan M. Wallace
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVic.Australia
| | - Graham Jenkin
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVic.Australia
| | - Suzanne L. Miller
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVic.Australia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVic.Australia
| |
Collapse
|
45
|
Carloni S, Proietti F, Rocchi M, Longini M, Marseglia L, D'Angelo G, Balduini W, Gitto E, Buonocore G. Melatonin Pharmacokinetics Following Oral Administration in Preterm Neonates. Molecules 2017; 22:molecules22122115. [PMID: 29194416 PMCID: PMC6149762 DOI: 10.3390/molecules22122115] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 11/29/2017] [Accepted: 11/29/2017] [Indexed: 12/11/2022] Open
Abstract
Melatonin possesses potential efficacy in perinatal brain injuries, and has been proposed as adjunctive pharmacological therapy in combination with hypothermia in the clinical setting. However, the pharmacokinetics of melatonin in preterm and term newborns is still unknown. The aim of this study was to analyze the pharmacokinetics of melatonin after intragastric administration in preterm infants. Preterm newborns were enrolled 24–72 h after birth, and randomly assigned to three groups receiving a single bolus of 0.5 mg·kg−1 melatonin, or 3 boluses of 1 or 5 mg·kg−1 of melatonin at 24-h intervals. Blood samples were collected before and at selective times after melatonin administration. The half-life of melatonin in plasma ranged from 7.98 to 10.94 h, and the area under the curve (AUC) from 10.48 to 118.17 µg·mL−1·h−1. Our results indicate a different pharmacokinetic profile in premature newborns, compared to adults and experimental animals. The high peak plasma concentrations and the long half-life indicate that in the neonatal clinical setting, it is possible to obtain and maintain high serum concentrations using a single administration of melatonin repeated every 12/24 h.
Collapse
Affiliation(s)
- Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy.
| | - Fabrizio Proietti
- Department of Molecular and Developmental Medicine, University of Siena, Viale Bracci, 53100 Siena, Italy.
| | - Marco Rocchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy.
| | - Mariangela Longini
- Department of Molecular and Developmental Medicine, University of Siena, Viale Bracci, 53100 Siena, Italy.
| | - Lucia Marseglia
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi"-Neonatal Intensive Care Unit, University of Messina, Via Consolare Valeria 1, 98125 Gazzi Messina, Italy.
| | - Gabriella D'Angelo
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi"-Neonatal Intensive Care Unit, University of Messina, Via Consolare Valeria 1, 98125 Gazzi Messina, Italy.
| | - Walter Balduini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy.
| | - Eloisa Gitto
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi"-Neonatal Intensive Care Unit, University of Messina, Via Consolare Valeria 1, 98125 Gazzi Messina, Italy.
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, Viale Bracci, 53100 Siena, Italy.
| |
Collapse
|
46
|
Yawno T, Mahen M, Li J, Fahey MC, Jenkin G, Miller SL. The Beneficial Effects of Melatonin Administration Following Hypoxia-Ischemia in Preterm Fetal Sheep. Front Cell Neurosci 2017; 11:296. [PMID: 29018332 PMCID: PMC5615225 DOI: 10.3389/fncel.2017.00296] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/06/2017] [Indexed: 11/24/2022] Open
Abstract
Melatonin (MLT) is an endogenous hormone that controls circadian cycle. MLT has additional important properties that make it appealing as a neuroprotective agent—it is a potent anti-oxidant, with anti-apoptotic and anti-inflammatory properties. MLT is safe for administration during pregnancy or to the newborn after birth, and can reduce white matter brain injury under conditions of chronic fetal hypoxia. Accordingly, in the current study, we examined whether an intermediate dose of MLT could restore white matter brain development when administered after an acute hypoxic ischemic (HI) insult in preterm fetal sheep. Fifteen fetal sheep at 95–98 days gestation were instrumented with femoral artery and vein catheters, and a silastic cuff placed around the umbilical cord. At 102 days gestation, the cuff was inflated, causing complete umbilical cord occlusion for 25 min in 10 fetuses, to induce acute severe HI. Five HI fetuses received intravenous MLT for 24 h beginning at 2 h after HI. The remaining five fetuses were administered saline alone. Ten days after HI, the fetal brain was collected from each animal and white and gray matter neuropathology assessed. HI caused a significant increase in apoptotic cell death (TUNEL+), activated microglia (Iba-1+), and oxidative stress (8-OHdG+) within the subventricular and subcortical white matter. HI reduced the total number of oligodendrocytes and CNPase+ myelin density. MLT administration following HI decreased apoptosis, inflammation and oxidative stress within the white matter. MLT had intermediate benefits for the developing white matter: it increased oligodendrocyte cell number within the periventricular white matter only, and improved CNPase+ myelin density within the subcortical but not the striatal white matter. MLT administration following HI was also associated with improved neuronal survival within the cortex. Neuropathology in preterm infants is complex and mediated by multiple mechanisms, including inflammation, oxidative stress and apoptotic pathways. Treatment with MLT presents a safe approach to neuroprotective therapy in preterm infants but appears to have brain region-specific benefits within the white matter.
Collapse
Affiliation(s)
- Tamara Yawno
- The Ritchie Centre, Hudson Institute of Medical Research, ClaytonVIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, ClaytonVIC, Australia
| | - Mawin Mahen
- The Ritchie Centre, Hudson Institute of Medical Research, ClaytonVIC, Australia
| | - Jingang Li
- The Ritchie Centre, Hudson Institute of Medical Research, ClaytonVIC, Australia
| | - Michael C Fahey
- The Ritchie Centre, Hudson Institute of Medical Research, ClaytonVIC, Australia.,Department of Paediatrics, Monash Medical Centre, ClaytonVIC, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, ClaytonVIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, ClaytonVIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, ClaytonVIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, ClaytonVIC, Australia
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Hypoxic-ischemic brain injury is a leading cause of mortality and morbidity in neonates. Treating such injury by interrupting the excitotoxic-oxidative cascade is of immense importance. This review will focus on novel techniques of neuroprotection and describe the latest advances in established therapeutic methods. KEY FINDINGS Although the primacy of therapeutic hypothermia in treating hypoxic-ischemic encephalopathy is well established, recent research establishes that the arbitrarily chosen regimen of cooling to 33°C for 72 h may indeed be the most appropriate method. The optimal duration of antenatal magnesium therapy for neuroprotection remains unsettled, though it is reassuring that even 12 h or less of magnesium therapy results in comparable neurological outcomes. Combining adjuvant therapies such as melatonin or erythropoietin with therapeutic hypothermia results in favorable neurological outcomes compared with hypothermia alone. Finally, stem cell-based therapies show considerable potential in preclinical studies. SUMMARY Significant advances have occurred in the management of neonatal brain injury. With establishment of the optimal temperature and duration of hypothermia, combinatory therapies using adjuncts hold the greatest promise. Promising preclinical approaches such as stem cell-based therapy and use of noble gases need to be confirmed with clinical trials.
Collapse
|
48
|
Andersen LPH, Gögenur I, Rosenberg J, Reiter RJ. The Safety of Melatonin in Humans. Clin Drug Investig 2015; 36:169-75. [DOI: 10.1007/s40261-015-0368-5] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
49
|
Turner MA. Clinical trials of medicines in neonates: the influence of ethical and practical issues on design and conduct. Br J Clin Pharmacol 2015; 79:370-8. [PMID: 25041601 DOI: 10.1111/bcp.12467] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 06/24/2014] [Indexed: 01/03/2023] Open
Abstract
In the past, there has been a perception that ethical and practical problems limit the opportunities for research in neonates. This perception is no longer appropriate. It is now clear that research about the medicines used in neonates is an ethical requirement. It is possible to conduct high quality research in neonates if the research team adapt to the characteristics of this population. Good practice involves respecting the specific needs of newborn babies and their families by adopting relevant approaches to study design, recruitment, pharmacokinetic studies and safety assessment. Neonatal units have a unique culture that requires careful development in a research setting. Clinical investigators need to recognize the clinical and ethical imperative to conduct rigorous research. Industry needs to engage with neonatal networks early in the process of drug development, preferably before contacting regulatory agencies. Follow-up over 3-5 years is essential for the evaluation of medicines in neonates and explicit funding for this is required for the assessment of the benefit and risk of treatments given to sick newborn babies. The views of parents must be central to the development of studies and the research agenda. Ethical and practical problems are no longer barriers to research in neonates. The current challenges are to disseminate good practice and maximize capacity in order to meet the need for research among newborn babies.
Collapse
Affiliation(s)
- Mark A Turner
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
50
|
Berger HR, Morken TS, Vettukattil R, Brubakk AM, Sonnewald U, Widerøe M. No improvement of neuronal metabolism in the reperfusion phase with melatonin treatment after hypoxic-ischemic brain injury in the neonatal rat. J Neurochem 2015; 136:339-50. [PMID: 26526584 DOI: 10.1111/jnc.13420] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/22/2015] [Accepted: 10/23/2015] [Indexed: 01/13/2023]
Abstract
Mitochondrial impairment is a key feature underlying neonatal hypoxic-ischemic (HI) brain injury and melatonin is potentially neuroprotective through its effects on mitochondria. In this study, we have used (1) H and (13) C NMR spectroscopy after injection of [1-(13) C]glucose and [1,2-(13) C]acetate to examine neuronal and astrocytic metabolism in the early reperfusion phase after unilateral HI brain injury in 7-day-old rat pups, exploring the effects of HI on mitochondrial function and the potential protective effects of melatonin on brain metabolism. One hour after hypoxia-ischemia, astrocytic metabolism was recovered and glycolysis was normalized, whereas mitochondrial metabolism in neurons was clearly impaired. Pyruvate carboxylation was also lower in both hemispheres after HI. The transfer of glutamate from neurons to astrocytes was higher whereas the transfer of glutamine from astrocytes to neurons was lower 1 h after HI in the contralateral hemisphere. Neuronal metabolism was equally affected in pups treated with melatonin (10 mg/kg) immediately after HI as in vehicle treated pups indicating that the given dose of melatonin was not capable of protecting the neuronal mitochondria in this early phase after HI brain injury. However, any beneficial effects of melatonin might have been masked by modulatory effects of the solvent dimethyl sulfoxide on cerebral metabolism. Neuronal and astrocytic metabolism was examined by (13) C and (1) H NMR spectroscopy in the early reperfusion phase after unilateral hypoxic-ischemic brain injury and melatonin treatment in neonatal rats. One hour after hypoxia-ischemia astrocytic mitochondrial metabolism had recovered and glycolysis was normalized, whereas mitochondrial metabolism in neurons was impaired. Melatonin treatment did not show a protective effect on neuronal metabolism.
Collapse
Affiliation(s)
- Hester R Berger
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tora Sund Morken
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Ophthalmology, Trondheim University Hospital, Trondheim, Norway
| | - Riyas Vettukattil
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ann-Mari Brubakk
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ursula Sonnewald
- Department of Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marius Widerøe
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|