1
|
Quach D, Mol BW, Springer J, Tully E, Higgins C, Jones M, Hennes D, Pham Y, Swarnamani K, Palmer K, Davies‐Tuck M. A double-blind, randomized, placebo-controlled trial of melatonin as an adjuvant agent for induction of labor: The MILO trial. Acta Obstet Gynecol Scand 2024; 103:2442-2454. [PMID: 39446608 PMCID: PMC11609988 DOI: 10.1111/aogs.14951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/23/2024] [Accepted: 08/07/2024] [Indexed: 10/26/2024]
Abstract
INTRODUCTION Melatonin has been suggested to have a biological role in the onset and progress of labor. We tested the hypothesis that the addition of melatonin during an induction of labor will reduce the need for a cesarean birth. MATERIAL AND METHODS This trial underwent protocol amendments that are detailed in the main text of the article. This trial is registered with the Australian and New Zealand Clinical Trials Registry (ACTRN12616000311459). At a multi-center health service including secondary and tertiary obstetric hospitals, we performed a randomized, double-blind, placebo-controlled trial in women with a singleton cephalic pregnancy, free of significant maternal or perinatal complications who were undergoing induction of labor (with or without cervical ripening). Women were randomized to 10 mg melatonin vs placebo, with cervical ripening as required, and then 6-h during their induction of labor to a maximum of four doses or until birth. The primary outcome was cesarean birth. Secondary outcomes included labor, maternal, and neonatal outcomes. Data were analyzed using intention to treat. Sub-group analyses based on mode of ripening and parity were also performed. RESULTS Between 2019 and 2021 we randomized 189 women (103 to melatonin and 86 to placebo). The study was prematurely terminated due to logistical complications resulting from the COVID-19 pandemic. Cesarean rates were 28/103 (27.2%) in the melatonin group vs 20/84 (23.3%) in the placebo group (RR 1.17 95% CI 0.71-1.92). There were no significant differences in rate of cesarean birth between the melatonin and placebo groups for failure to progress (13.4% and 9.3%, respectively, RR 1.46; 95% CI 0.64-3.32) or suspected fetal distress (10.7% and 10.5%, respectively, RR 1.02; 95% CI 0.44-2.34). The melatonin group had significantly lower rates of spontaneous vaginal birth within 24 h (35.0% vs. 50.0%; RR 0.70 95% CI 0.50-0.98). The rates of secondary outcomes such as total length of labor, rate of postpartum hemorrhage, and instrumental birth were comparable. Babies born in the melatonin group were more likely to need admission to the special care nursery, namely for hypoglycemic monitoring (18.5% vs. 8.1% RR 2.26; 95% CI 1.00-5.10). CONCLUSIONS In women undergoing induction of labor, melatonin does not reduce the cesarean section rate. Melatonin use intrapartum may also be associated with neonatal hypoglycemia.
Collapse
Affiliation(s)
- Diane Quach
- Department of Obstetrics and GynecologyMonash Women's, Monash HealthClaytonVictoriaAustralia
- Department of Obstetrics and GynecologyMonash UniversityClaytonVictoriaAustralia
| | - Ben W. Mol
- Department of Obstetrics and GynecologyMonash UniversityClaytonVictoriaAustralia
- Aberdeen Centre for Women's Health Research, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenAberdeenUK
| | - Jamie Springer
- Department of Obstetrics and GynecologyMonash Women's, Monash HealthClaytonVictoriaAustralia
- Department of Obstetrics and GynecologyMonash UniversityClaytonVictoriaAustralia
| | - Erin Tully
- Department of Obstetrics and GynecologyMonash Women's, Monash HealthClaytonVictoriaAustralia
- Department of Obstetrics and GynecologyMonash UniversityClaytonVictoriaAustralia
| | - Chloe Higgins
- Department of Obstetrics and GynecologyMonash Women's, Monash HealthClaytonVictoriaAustralia
| | - Madeleine Jones
- Department of Obstetrics and GynecologyMonash Women's, Monash HealthClaytonVictoriaAustralia
- Department of Obstetrics and GynecologyMonash UniversityClaytonVictoriaAustralia
| | - David Hennes
- Department of Obstetrics and GynecologyMonash Women's, Monash HealthClaytonVictoriaAustralia
- Department of Obstetrics and GynecologyMonash UniversityClaytonVictoriaAustralia
| | - Yen Pham
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
| | - Kamala Swarnamani
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
| | - Kirsten Palmer
- Department of Obstetrics and GynecologyMonash Women's, Monash HealthClaytonVictoriaAustralia
- Department of Obstetrics and GynecologyMonash UniversityClaytonVictoriaAustralia
| | - Miranda Davies‐Tuck
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
| |
Collapse
|
2
|
Li Y, Cheng Z, Ma W, Qiu Y, Liu T, Nan B, Li M, Sun L, Liu W, Yin H, Wang C, Li X, Zang C. Effect of Exogenous Melatonin on Performance and Mastitis in Dairy Cows. Vet Sci 2024; 11:431. [PMID: 39330810 PMCID: PMC11435509 DOI: 10.3390/vetsci11090431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Mastitis is an important factor affecting the health of cows that leads to elevated somatic cell counts in milk, which can seriously affect milk quality and result in huge economic losses for the livestock industry. Therefore, the aim of this trial was to investigate the effect of melatonin on performance and mastitis in dairy cows. Forty-eight Holstein cows with a similar body weight (470 ± 10 kg), parity (2.75 ± 1.23), number of lactation days (143 ± 43 days), BCS (3.0-3.5), milk yield (36.80 ± 4.18 kg), and somatic cell count (300,000-500,000 cells/mL) were selected and randomly divided into four groups: control (CON group), trial Ⅰ (T80 group), trial Ⅱ (T120 group), and trial Ⅲ (T160 group). Twelve cows in trial groups I, II, and III were pre-dispensed 80, 120, and 160 mg of melatonin in edible glutinous rice capsules along with the basal ration, respectively, while the control group was fed an empty glutinous rice capsule along with the ration. The trial period was 37 days, which included a 7-day adaptive phase followed by a 30-day experimental period. At the end of the trial period, feeding was ended and the cows were observed for 7 days. Milk samples were collected on days 0, 7, 14, 21, 28, and 37 to determine the somatic cell number and milk composition. Blood samples were collected on days 0, 15, 30, and 37 of the trial to determine the serum biochemical indicators, antioxidant and immune indicators, and the amount of melatonin in the blood. The results showed that the somatic cell counts of lactating cows in the CON group were lower than those in the T120 group on days 14 (p < 0.05) and 28 (p < 0.01) at 1 week after melatonin cessation. The milk protein percentage and milk fat percentage of cows in the T120 group were higher than those in the CON group (p < 0.01). The total protein and globulin content in the T120 group were higher than those in the CON group (p < 0.01). In terms of antioxidant capacity and immunity, the cows 1 week after melatonin cessation showed higher superoxide dismutase activity and interleukin-10 contents (p < 0.01) compared with the CON group and lower malondialdehyde and tumor necrosis factor-alpha contents (p < 0.01) compared with the T120 group. The melatonin content in the T120 group was increased relative to that in the other groups. In conclusion, exogenous melatonin can increase the content of milk components, reduce the somatic cell count, and improve the antioxidant capacity and immune responses to a certain extent. Under the experimental conditions, 120 mg/day melatonin is recommended for mid- to late-lactation cows.
Collapse
Affiliation(s)
- Yunmeng Li
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| | - Zhiqiang Cheng
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| | - Wenting Ma
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| | - Yaqi Qiu
- Karamay Green City Agricultural Development Co., Karamay 834000, China;
| | - Tuo Liu
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| | - Bingyu Nan
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| | - Mengfei Li
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| | - Long Sun
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| | - Wentao Liu
- Xinjiang Urumqi Rural Revitalization Guidance Service Center, Urumqi 830000, China;
| | - Haina Yin
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| | - Caidie Wang
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| | - Xiaobin Li
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| | - Changjiang Zang
- Xinjiang Laboratory of Herbivore Nutrition for Meat and Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (Y.L.); (Z.C.); (W.M.); (T.L.); (B.N.); (M.L.); (L.S.); (H.Y.); (C.W.); (X.L.)
| |
Collapse
|
3
|
Wāng Y, Han Y, Xu DX. Developmental impacts and toxicological hallmarks of silver nanoparticles across diverse biological models. ENVIRONMENTAL SCIENCE AND ECOTECHNOLOGY 2024; 19:100325. [PMID: 38046179 PMCID: PMC10692670 DOI: 10.1016/j.ese.2023.100325] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 12/05/2023]
Abstract
Silver nanoparticles (AgNPs), revered for their antimicrobial prowess, have become ubiquitous in a range of products, from biomedical equipment to food packaging. However, amidst their rising popularity, concerns loom over their possible detrimental effects on fetal development and subsequent adult life. This review delves into the developmental toxicity of AgNPs across diverse models, from aquatic species like zebrafish and catfish to mammalian rodents and in vitro embryonic stem cells. Our focus encompasses the fate of AgNPs in different contexts, elucidating associated hazardous results such as embryotoxicity and adverse pregnancy outcomes. Furthermore, we scrutinize the enduring adverse impacts on offspring, spanning impaired neurobehavior function, reproductive disorders, cardiopulmonary lesions, and hepatotoxicity. Key hallmarks of developmental harm are identified, encompassing redox imbalances, inflammatory cascades, DNA damage, and mitochondrial stress. Notably, we explore potential explanations, linking immunoregulatory dysfunction and disrupted epigenetic modifications to AgNPs-induced developmental failures. Despite substantial progress, our understanding of the developmental risks posed by AgNPs remains incomplete, underscoring the urgency of further research in this critical area.
Collapse
Affiliation(s)
- Yán Wāng
- Department of Toxicology, School of Public Health & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Yapeng Han
- Department of Toxicology, School of Public Health & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
4
|
Marques KL, Rodrigues V, Balduci CTN, Montes GC, Barradas PC, Cunha-Rodrigues MC. Emerging therapeutic strategies in hypoxic-ischemic encephalopathy: a focus on cognitive outcomes. Front Pharmacol 2024; 15:1347529. [PMID: 38469401 PMCID: PMC10925695 DOI: 10.3389/fphar.2024.1347529] [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: 11/30/2023] [Accepted: 01/30/2024] [Indexed: 03/13/2024] Open
Abstract
Perinatal hypoxia-ischemia represents a significant risk to CNS development, leading to high mortality rates, diverse damages, and persistent neurological deficits. Despite advances in neonatal medicine in recent decades, the incidence of HIE remains substantial. Motor deficits can manifest early, while cognitive impairments may be diagnosed later, emphasizing the need for extended follow-up. This review aims to explore potential candidates for therapeutic interventions for hypoxic-ischemic encephalopathy (HIE), with a focus on cognitive deficits. We searched randomized clinical trials (RCT) that tested drug treatments for HIE and evaluated cognitive outcomes. The results included studies on erythropoietin, melatonin, magnesium sulfate, topiramate, and a combination of vitamin C and ibuprofen. Although there are several indications of the efficacy of these drugs among animal models, considering neuroprotective properties, the RCTs failed to provide complete effectiveness in the context of cognitive impairments derived from HIE. More robust RCTs are still needed to advance our knowledge and to establish standardized treatments for HIE.
Collapse
Affiliation(s)
- Kethely L. Marques
- Laboratory of Neurobiology, Pharmacology and Psychobiology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor Rodrigues
- Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cassiana T. N. Balduci
- Laboratory of Neurobiology, Pharmacology and Psychobiology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- Rare Diseases Sales Force, Daiichi Sankyo Brazil, São Paulo, Brazil
| | - Guilherme C. Montes
- Laboratory of Neurobiology, Pharmacology and Psychobiology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Penha C. Barradas
- Laboratory of Neurobiology, Pharmacology and Psychobiology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marta C. Cunha-Rodrigues
- Laboratory of Neurobiology, Pharmacology and Psychobiology Department, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Nilofar N, Zengin G, Acar M, Bouyayha A, Youssra A, Eldahshan O, Fayez S, Fahmy N. Assessing the Chemical Composition, Antioxidant and enzyme Inhibitory Effects of Pentapleura subulifera and Cyclotrichium glabrescens Extracts. Chem Biodivers 2024; 21:e202301651. [PMID: 38016080 DOI: 10.1002/cbdv.202301651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 11/30/2023]
Abstract
The Lamiaceae family, encompassing diverse plant species, holds significant value in food, medicine, and cosmetics. Within this family, Pentapleura subulifera and Cyclotrichium glabrescens, relatively unexplored species, were investigated for their chemical composition, antioxidant capacity, and enzyme-inhibiting effects. The chemical composition of hexane, methanolic, and aqueous extracts from P. subulifera and C. glabrescens were analyzed using LC-ESI-MS/MS and the non-polar hexane fraction was investigated via GC-MS. The antioxidant potential of the extracts was determined through radical scavenging, reducing power and metal chelating assays. Additionally, inhibitory activity against six enzymes - acetylcholinesterase (AChE), butyrylcholinesterase (BChE), tyrosinase, amylase, and glucosidase - was examined. The aqueous extract of P. subulifera and the methanolic extract of C. glabrescens exhibited elevated phenolic content at 129.47 mg gallic acid equivalent (GAE)/g and 55.97 mg GAE/g, respectively. Chemical profiling of the constituents of the two plant species resulted in the identification of a total of twenty compounds. The majority of which belonged to flavonoids and quinic acid derivatives, primarily concentrated in the methanol and aqueous extracts. Among all antioxidant assays, the aqueous extracts of P. subulifera demonstrated superior antioxidant activity, with the highest recorded activity of 404.93 mg trolox equivalent (TE)/g in the cupric reducing antioxidant capacity (CUPRAC) test. Meanwhile, the hexane extract of C. glabrescens exhibited the highest AChE inhibitory activity at 2.71 mg galanthamine equivalent (GALAE)/g, followed by the methanol extract of P. subulifera at 2.41 mg GALAE/g. These findings unequivocally establish the notable antioxidant and enzyme inhibitory activity of P. subulifera and C. glabrescens extracts, underscoring their potential as a source of valuable natural antioxidants.
Collapse
Affiliation(s)
- Nilofar Nilofar
- Physiology and Biochemistry Laboratory, Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
- Department of Pharmacy, Botanic Garden "Giardino dei Semplici", Università degli Studi "Gabriele d'Annunzio", via dei Vestini 31, 66100, Chieti, Italy
| | - Gokhan Zengin
- Physiology and Biochemistry Laboratory, Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
| | - Mikail Acar
- Munzur University, Department of Plant and Animal Production, Tunceli Vocational School of Higher Education, Tunceli, 62000, Turkey
| | - Abdelhakim Bouyayha
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco
| | - Aalilou Youssra
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco
| | - Omayma Eldahshan
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
- Center of Drug Discovery Research and Development, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Shaimaa Fayez
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| | - Nouran Fahmy
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| |
Collapse
|
6
|
Kose C, Ibanoglu MC, Erdogan K, Arslan B, Uzlu SE, Akpinar F, Karadeniz RS, Engin-Ustun Y. The effect of fetal hypoxia on myeloperoxidase levels in cord blood: a prospective study. Minerva Obstet Gynecol 2024; 76:1-6. [PMID: 35420291 DOI: 10.23736/s2724-606x.22.05090-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND We aimed to compare myeloperoxidase (MPO) levels in cord blood samples of mothers with and without perinatal hypoxia, since fetal hypoxia results in decreased pH, base excess, and an increase in pCO2 and lactate levels. METHODS We enrolled 42 pregnant women to this cross-sectional analytic study if they had met following criteria: uneventful gestational follow-ups, no known chronic or pregnancy-associated diseases, a BMI of <29.9, a singleton pregnancy, those with pregnancy over 34 weeks. The exclusion criteria for the study and control groups were as follows: presence of multiple pregnancies, fetal abnormality, any disease diagnosed before or during antenatal follow-up e.g., diabetes, hypertension, thyroid dysfunction, uncontrolled endocrine disease or abnormal kidney function, autoimmune disease, chronic inflammatory diseases, IUGR, preeclampsia), maternal age below 18 or above 35, intrauterine exitus, pregnancy with assisted reproductive technique, alcohol or smoking addiction, and any chronic drug use. The subjects were 1:1 randomized to either hypoxic newborns (N.=21) and those in the control group (N.=21) and their myeloperoxidase levels were measured from cord blood samples. Results were expressed as U/L. Patient data regarding age, gestation, parity, birth weight, birth length, APGAR scores, and neonatal complications were collected. All the women signed written informed consent forms and accepted verbal consent before being included in the study. RESULTS The mean age of the study population was 26.9±5.3 years. The mean BMI was 28.3±3.5 kg/m2. For the hypoxic group, 21 newborns with cord blood below 7.25 were included in the study group. The bloods with pH above 7.25 formed the control group. Mean pH and five (5) minute APGAR scores were found to be significantly lower in the study group, while base excess (BE) was found to be significantly higher. In this study, we compared the MPO levels of hypoxic newborns and those in the control group, and we did not find a significant difference between the two groups (P=0.147). Pearson Correlation Analysis is at -0.566 with P value (0.008) showing significant negative correlation between MPO and pH in the study group. CONCLUSIONS We found that MPO values are negatively correlated with cord blood pH among newborns diagnosed with fetal hypoxia.
Collapse
Affiliation(s)
- Caner Kose
- Department of Obstetrics and Gynecology, Etlik Zubeyde Hanım Women's Health Training and Research Hospital, Ankara, Türkiye
| | - Mujde C Ibanoglu
- Department of Obstetrics and Gynecology, Etlik Zubeyde Hanım Women's Health Training and Research Hospital, Ankara, Türkiye -
| | - Kadriye Erdogan
- Department of Obstetrics and Gynecology, Etlik Zubeyde Hanım Women's Health Training and Research Hospital, Ankara, Türkiye
| | - Burak Arslan
- Department of Biochemistry, Ankara Etlik Zubeyde Hanım Women's Health Training and Research Hospital, Ankara, Türkiye
| | - Safiye E Uzlu
- Department of Neonatology, Ankara Etlik Zubeyde Hanım Women's Health Training and Research Hospital, Ankara, Türkiye
| | - Funda Akpinar
- Department of Obstetrics and Gynecology, Etlik Zubeyde Hanım Women's Health Training and Research Hospital, Ankara, Türkiye
| | - R Sinan Karadeniz
- Department of Obstetrics and Gynecology, Etlik Zubeyde Hanım Women's Health Training and Research Hospital, Ankara, Türkiye
| | - Yaprak Engin-Ustun
- Department of Obstetrics and Gynecology, Etlik Zubeyde Hanım Women's Health Training and Research Hospital, Ankara, Türkiye
| |
Collapse
|
7
|
Liang Y, Wang Y, Zhang X, Jin S, Guo Y, Yu Z, Xu X, Shuai Q, Feng Z, Chen B, Liang T, Ao R, Li J, Zhang J, Cao R, Zhao H, Chen Z, Liu Z, Xie J. Melatonin alleviates valproic acid-induced neural tube defects by modulating Src/PI3K/ERK signaling and oxidative stress. Acta Biochim Biophys Sin (Shanghai) 2024; 56:23-33. [PMID: 38062774 PMCID: PMC10875364 DOI: 10.3724/abbs.2023234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/27/2023] [Indexed: 01/26/2024] Open
Abstract
Neural tube defects (NTDs) represent a developmental disorder of the nervous system that can lead to significant disability in children and impose substantial social burdens. Valproic acid (VPA), a widely prescribed first-line antiepileptic drug for epilepsy and various neurological conditions, has been associated with a 4-fold increase in the risk of NTDs when used during pregnancy. Consequently, urgent efforts are required to identify innovative prevention and treatment approaches for VPA-induced NTDs. Studies have demonstrated that the disruption in the delicate balance between cell proliferation and apoptosis is a crucial factor contributing to NTDs induced by VPA. Encouragingly, our current data reveal that melatonin (MT) significantly inhibits apoptosis while promoting the restoration of neuroepithelial cell proliferation impaired by VPA. Moreover, further investigations demonstrate that MT substantially reduces the incidence of neural tube malformations resulted from VPA exposure, primarily by suppressing apoptosis through the modulation of intracellular reactive oxygen species levels. In addition, the Src/PI3K/ERK signaling pathway appears to play a pivotal role in VPA-induced NTDs, with significant inhibition observed in the affected samples. Notably, MT treatment successfully reinstates Src/PI3K/ERK signaling, thereby offering a potential underlying mechanism for the protective effects of MT against VPA-induced NTDs. In summary, our current study substantiates the considerable protective potential of MT in mitigating VPA-triggered NTDs, thereby offering valuable strategies for the clinical management of VPA-related birth defects.
Collapse
Affiliation(s)
- Yuxiang Liang
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
- Experimental Animal Center of Shanxi Medical UniversityShanxi Key Laboratory of Human Disease and Animal ModelsTaiyuan030001China
| | - Ying Wang
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Xiao Zhang
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
- School of PharmacyShanxi Medical UniversityTaiyuan030001China
| | - Shanshan Jin
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Yuqian Guo
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Zhaowei Yu
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
- School of PharmacyShanxi Medical UniversityTaiyuan030001China
| | - Xinrui Xu
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Qizhi Shuai
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Zihan Feng
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Binghong Chen
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Ting Liang
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Ruifang Ao
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Jianting Li
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Juan Zhang
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Rui Cao
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Hong Zhao
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Zhaoyang Chen
- Experimental Animal Center of Shanxi Medical UniversityShanxi Key Laboratory of Human Disease and Animal ModelsTaiyuan030001China
| | - Zhizhen Liu
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Jun Xie
- Department of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| |
Collapse
|
8
|
Grzeszczak K, Łanocha-Arendarczyk N, Malinowski W, Ziętek P, Kosik-Bogacka D. Oxidative Stress in Pregnancy. Biomolecules 2023; 13:1768. [PMID: 38136639 PMCID: PMC10741771 DOI: 10.3390/biom13121768] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Recent years have seen an increased interest in the role of oxidative stress (OS) in pregnancy. Pregnancy inherently heightens susceptibility to OS, a condition fueled by a systemic inflammatory response that culminates in an elevated presence of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in the circulatory system. The amplified OS in pregnancy can trigger a series of detrimental outcomes such as underdevelopment, abnormal placental function, and a host of pregnancy complications, including pre-eclampsia, embryonic resorption, recurrent pregnancy loss, fetal developmental anomalies, intrauterine growth restriction, and, in extreme instances, fetal death. The body's response to mitigate the uncontrolled increase in RNS/ROS levels requires trace elements that take part in non-enzymatic and enzymatic defense processes, namely, copper (Cu), zinc (Zn), manganese (Mn), and selenium (Se). Determination of ROS concentrations poses a challenge due to their short half-lives, prompting the use of marker proteins, including malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT), and glutathione (GSH). These markers, indicative of oxidative stress intensity, can offer indirect assessments of pregnancy complications. Given the limitations of conducting experimental studies on pregnant women, animal models serve as valuable substitutes for in-depth research. This review of such models delves into the mechanism of OS in pregnancy and underscores the pivotal role of OS markers in their evaluation.
Collapse
Affiliation(s)
- Konrad Grzeszczak
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland; (K.G.); (N.Ł.-A.)
- Department of Laboratory Diagnostics, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Natalia Łanocha-Arendarczyk
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland; (K.G.); (N.Ł.-A.)
| | - Witold Malinowski
- Faculty of Health Sciences, The Masovian. Public University in Płock, Plac Dąbrowskiego 2, 09-402 Płock, Poland;
| | - Paweł Ziętek
- Department of Orthopaedics, Traumatology and Orthopaedic Oncology, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Danuta Kosik-Bogacka
- Independent Laboratory of Pharmaceutical Botany, Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland
| |
Collapse
|
9
|
Alrouji M, Al‐kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, Jabir MS, Saad HM, Batiha GE. NF-κB/NLRP3 inflammasome axis and risk of Parkinson's disease in Type 2 diabetes mellitus: A narrative review and new perspective. J Cell Mol Med 2023; 27:1775-1789. [PMID: 37210624 PMCID: PMC10315781 DOI: 10.1111/jcmm.17784] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/27/2023] [Accepted: 05/10/2023] [Indexed: 05/22/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease (AD). Genetic predisposition and immune dysfunction are involved in the pathogenesis of PD. Notably, peripheral inflammatory disorders and neuroinflammation are associated with PD neuropathology. Type 2 diabetes mellitus (T2DM) is associated with inflammatory disorders due to hyperglycaemia-induced oxidative stress and the release of pro-inflammatory cytokines. Particularly, insulin resistance (IR) in T2DM promotes the degeneration of dopaminergic neurons in the substantia nigra (SN). Thus, T2DM-induced inflammatory disorders predispose to the development and progression of PD, and their targeting may reduce PD risk in T2DM. Therefore, this narrative review aims to find the potential link between T2DM and PD by investigating the role of inflammatory signalling pathways, mainly the nuclear factor kappa B (NF-κB) and the nod-like receptor pyrin 3 (NLRP3) inflammasome. NF-κB is implicated in the pathogenesis of T2DM, and activation of NF-κB with induction of neuronal apoptosis was also confirmed in PD patients. Systemic activation of NLRP3 inflammasome promotes the accumulation of α-synuclein and degeneration of dopaminergic neurons in the SN. Increasing α-synuclein in PD patients enhances NLRP3 inflammasome activation and the release of interleukin (IL)-1β followed by the development of systemic inflammation and neuroinflammation. In conclusion, activation of the NF-κB/NLRP3 inflammasome axis in T2DM patients could be the causal pathway in the development of PD. The inflammatory mechanisms triggered by activated NLRP3 inflammasome lead to pancreatic β-cell dysfunction and the development of T2DM. Therefore, attenuation of inflammatory changes by inhibiting the NF-κB/NLRP3 inflammasome axis in the early T2DM may reduce future PD risk.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesShaqra UniversityShaqraSaudi Arabia
| | - Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐HerdeckeWuppertalGermany
| | - Majid S. Jabir
- Applied Science DepartmentUniversity of TechnologyBaghdadIraq
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMatrouhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
10
|
Olugbodi JO, Lawal B, Bako G, Onikanni AS, Abolenin SM, Mohammud SS, Ataya FS, Batiha GES. Effect of sub-dermal exposure of silver nanoparticles on hepatic, renal and cardiac functions accompanying oxidative damage in male Wistar rats. Sci Rep 2023; 13:10539. [PMID: 37386048 PMCID: PMC10310751 DOI: 10.1038/s41598-023-37178-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 06/17/2023] [Indexed: 07/01/2023] Open
Abstract
Silver nanoparticles (AgNPs) have been generally used due to their strong antibacterial, antiviral and antifungal and antimicrobial properties. However, their toxicity is a subject of sustained debate, thus requiring further studies. Hence, this study examines the adverse effects of the sub-dermal administered dose of AgNPs (200 nm) on the liver, kidney and heart of male Wistar rats. Thirty male rats were randomly distributed into six groups of five animals per group. Group A and D served as the control and received distilled water for 14 and 28 days respectively. Groups B and C were sub-dermally exposed to AgNPs at 10 and 50 mg/kg daily for 14 days while E and F were sub-dermally exposed to AgNPs at 10 and 50 mg/kg daily for 28 days. The liver, kidney and heart of the animals were collected, processed and used for biochemical and histological analysis. Our results revealed that the subdermal administration of AgNPs induced significant increased (p < 0.05) activities of aspartate aminotransferase (AST), alanine transferase (ALT), alkaline phosphatase (ALP), urea, creatinine, and malondialdehyde (MDA) while decreasing the levels of glutathione (GSH), catalase (CAT), superoxide dismutase (SOD), and total thiol groups in the rat tissues. Our findings suggest that the subdermal administration of AgNPs induced oxidative stress and impaired the hepatic, renal and cardiac functions of male Wistar rats.
Collapse
Affiliation(s)
- Janet Olayemi Olugbodi
- Department of Biochemistry, Bingham University, Abuja-Keffi Expressway Road, P.M.B 005, Karu, Nigeria.
| | - Bashir Lawal
- Department of Pathology, University of Pittsburgh, Pittsburgh, United States
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, United States
| | - Godiya Bako
- Department of Biochemistry, Bingham University, Abuja-Keffi Expressway Road, P.M.B 005, Karu, Nigeria
| | - Amos Sunday Onikanni
- Biochemistry Unit, Department of Chemical Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
- College of Medicine, Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Sulama M Abolenin
- Biology Department, Thurobah University College, Thurobah, Republic of Congo
| | - Soliman S Mohammud
- Biology Department, Thurobah University College, Thurobah, Republic of Congo
| | - Farid S Ataya
- Department of Biochemistry, College of Science, King Saud University, P. O. Box 2455, 11451, Riyadh, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
11
|
Di W, Jin Z, Lei W, Liu Q, Yang W, Zhang S, Lu C, Xu X, Yang Y, Zhao H. Protection of melatonin treatment and combination with traditional antibiotics against septic myocardial injury. Cell Mol Biol Lett 2023; 28:35. [PMID: 37101253 PMCID: PMC10134561 DOI: 10.1186/s11658-022-00415-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/23/2022] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Heart failure is a common complication of sepsis with a high mortality rate. It has been reported that melatonin can attenuate septic injury due to various properties. On the basis of previous reports, this study will further explore the effects and mechanisms of melatonin pretreatment, posttreatment, and combination with antibiotics in the treatment of sepsis and septic myocardial injury. METHODS AND RESULTS Our results showed that melatonin pretreatment showed an obvious protective effect on sepsis and septic myocardial injury, which was related to the attenuation of inflammation and oxidative stress, the improvement of mitochondrial function, the regulation of endoplasmic reticulum stress (ERS), and the activation of the AMPK signaling pathway. In particular, AMPK serves as a key effector for melatonin-initiated myocardial benefits. In addition, melatonin posttreatment also had a certain degree of protection, while its effect was not as remarkable as that of pretreatment. The combination of melatonin and classical antibiotics had a slight but limited effect. RNA-seq detection clarified the cardioprotective mechanism of melatonin. CONCLUSION Altogether, this study provides a theoretical basis for the application strategy and combination of melatonin in septic myocardial injury.
Collapse
Affiliation(s)
- Wencheng Di
- Department of Cardiovascular Medicine, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, 29 Bulan Road, Shenzhen, Guangdong Province, China
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Airforce Military Medical University, 127 Changle West Road, Xi'an, China
| | - Wangrui Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Qiong Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Wenwen Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Shaofei Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Chenxi Lu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Xiaoling Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China.
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China.
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, The Airforce Military Medical University, 1 Xinsi Road, Xi'an, China.
| |
Collapse
|
12
|
Vendrame SA, Stein CDS, Naidon ÂM, Gonçalves dos Santos I, Moresco RN, Gonçalves TDL. Vaginal delivery is associated with lower levels of thiol groups, vitamin C and ferric reducing ability in colostrum compared with caesarean section. J OBSTET GYNAECOL 2022; 42:3509-3513. [PMID: 36457295 DOI: 10.1080/01443615.2022.2151346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
This study aimed to investigate the effects of delivery type (normal or caesarean) on the antioxidant and oxidative capacity of colostrum collected shortly after delivery. A total of 61 parturients were included in the study and divided into two groups: those who underwent vaginal delivery (n = 36) and those who underwent elective caesarean section (n = 25). Colostrum samples were collected by manual milking up to 48 h post parturition and analysed for thiol groups (-SH), vitamin C, ferric reducing ability (FRAP), nitrate/nitrite oxides (NOx), and advanced oxidation protein products (AOPP). Colostrum levels of -SH (p = 0.0042), vitamin C (p = 0.0455), and FRAP (p = 0.0374) were significantly lower in the vaginal delivery group. The results suggest that vaginal delivery, compared to caesarean section, is associated with lower levels of antioxidants in colostrum and the mode of delivery plays an important role in the composition of antioxidants in maternal colostrum that help protect newborns from oxidative damage.IMPACT STATEMENTWhat is already known on this subject? Colostrum is the first biological fluid produced by the mother after delivery and is responsible for a child's growth, cognitive development and health. It is known that childbirth can cause oxidative imbalance, and its effects have already been evaluated in maternal and foetal blood, however, there are few studies evaluating the effects of childbirth on colostrum composition.What do the results of this study add? Previously, a study showed that caesarean section caused greater oxidation of colostrum compared to vaginal delivery. Thus, we sought to evaluate other markers (thiol groups, vitamin C, ferric reducing ability, nitrate/nitrite oxides, and advanced oxidation protein products), in a short period of time after delivery, in order to elucidate this still little discussed issue. Unlike the previous one, our study suggests that vaginal delivery, compared to caesarean section, is associated with lower levels of antioxidants in colostrum, which may make it difficult to protect newborns from oxidative damage.What are the implications of these findings for clinical practice and/or further research? Our study suggests that normal delivery can influence the antioxidant composition of maternal colostrum, and it is debateable for future clinical practice to improve eating habits during pregnancy and lactation, in order to strengthen the antioxidant capacity of colostrum and reduce oxidative damage to newborns.
Collapse
Affiliation(s)
- Silmara Ana Vendrame
- Departamento de Análises Clínicas e toxicológicas, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Carolina dos Santos Stein
- Departamento de Análises Clínicas e toxicológicas, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | | | | | - Rafael Noal Moresco
- Departamento de Análises Clínicas e toxicológicas, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Thissiane de Lima Gonçalves
- Departamento de Análises Clínicas e toxicológicas, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| |
Collapse
|
13
|
Kimble A, Robbins ME, Perez M. Pathogenesis of Bronchopulmonary Dysplasia: Role of Oxidative Stress from 'Omics' Studies. Antioxidants (Basel) 2022; 11:2380. [PMID: 36552588 PMCID: PMC9774798 DOI: 10.3390/antiox11122380] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/24/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) remains the most common respiratory complication of prematurity as younger and smaller infants are surviving beyond the immediate neonatal period. The recognition that oxidative stress (OS) plays a key role in BPD pathogenesis has been widely accepted since at least the 1980s. In this article, we examine the interplay between OS and genetic regulation and review 'omics' data related to OS in BPD. Data from animal models (largely models of hyperoxic lung injury) and from human studies are presented. Epigenetic and transcriptomic analyses have demonstrated several genes related to OS to be differentially expressed in murine models that mimic BPD as well as in premature infants at risk of BPD development and infants with established lung disease. Alterations in the genetic regulation of antioxidant enzymes is a common theme in these studies. Data from metabolomics and proteomics have also demonstrated the potential involvement of OS-related pathways in BPD. A limitation of many studies includes the difficulty of obtaining timely and appropriate samples from human patients. Additional 'omics' studies could further our understanding of the role of OS in BPD pathogenesis, which may prove beneficial for prevention and timely diagnosis, and aid in the development of targeted therapies.
Collapse
Affiliation(s)
- Ashley Kimble
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Ann and Robert H Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Mary E. Robbins
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Ann and Robert H Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Stanley Manne Children’s Research Institute of Chicago, Chicago, IL 60611, USA
| | - Marta Perez
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Ann and Robert H Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Stanley Manne Children’s Research Institute of Chicago, Chicago, IL 60611, USA
| |
Collapse
|
14
|
Tomruk A, Ozgur-Buyukatalay E, Ozturk GG, Ulusu NN. Short-term exposure to radiofrequency radiation and metabolic enzymes' activities during pregnancy and prenatal development. Electromagn Biol Med 2022; 41:370-378. [PMID: 35904122 DOI: 10.1080/15368378.2022.2104309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiofrequency radiation (RFR) as an environmental and physical pollutant may induce vulnerability to toxicity and disturb fetal development. Therefore, the potential health effects of short-term mobile phone like RFR exposure (GSM 1800 MHz; 14 V/m, 2 mW/kg specific absorption rate (SAR) during 15 min/day for a week) during pregnancy and also the development of fetuses were investigated. Hepatic glucose regulation and glutathione-dependent enzymes' capacities were biochemically analyzed in adult (female) and pregnant New Zealand White rabbits. Pregnant rabbits' two-day-old offspring were included to understand their developmental stages under short-term maternal RFR exposure. We analyzed two regulatory enzymes in the oxidative phase of phosphogluconate pathways to interpret the cytosolic NADPH's biosynthesis for maintaining mitochondrial energy metabolism. Moreover, the efficiencies of maternal glutathione-dependent enzymes on both the removal of metabolic disturbances during pregnancy and fetus development were examined. Whole-body RFR exposures were applied to pregnant animals from the 15th to the 22nd day of their gestations, i.e., the maturation periods of tissues and organs for rabbit fetuses. There were significant differences in hepatic glucose regulation and GSH-dependent enzymes' capacities with pregnancy and short-term RFR exposure. Consequently, we observed that intrauterine exposure to RFR might lead to cellular ROS- dependent disturbances in metabolic activity and any deficiency in the intracellular antioxidant (ROS-scavenging) system. This study might be a novel insight into further studies on the possible effects of short-term RF exposure and prenatal development.
Collapse
Affiliation(s)
- Arın Tomruk
- Department of Biophysics, Faculty of Medicine, Gazi University, Ankara, Turkey
| | | | - Goknur Guler Ozturk
- Department of Biophysics, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - N Nuray Ulusu
- Department of Biochemistry, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
15
|
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: 2.0] [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
|
16
|
Ates G, Tamer S, Yorulmaz H, Mutlu S, Olgac V, Aksu A, Caglar NB, Özkök E. Melatonin pretreatment modulates anti-inflammatory, antioxidant, YKL-40, and matrix metalloproteinases in endotoxemic rat lung tissue. Exp Biol Med (Maywood) 2022; 247:1080-1089. [PMID: 35369768 PMCID: PMC9265525 DOI: 10.1177/15353702221084933] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 02/14/2022] [Indexed: 08/30/2023] Open
Abstract
We aimed to investigate the effects of melatonin administered before and during endotoxemia on the lung tissue of rats, cytokine, YKL-40, matrix metalloproteinase (MMP) and inhibitor levels, oxidative stress parameters, and energy balance. Sepsis was induced with lipopolysaccharide (LPS), the cell wall molecule of gram negative bacteria. Rats were divided into four groups, Control, LPS (Escherichia coli O127:B8, 20 mg/kg), melatonin (10 mg/kg), and melatonin+LPS (M+LPS). After injections, lung tissues samples were taken for experimental analyses. YKL-40, thiobarbituric acid reactive substances (TBARS), glutathione reductase (GR), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD) enzymes levels were measured, high-energy components were analyzed; tumor necrosis factor-alpha (TNF-α), MMP-2, YKL-40, MMP-9, myeloperoxidase (MPO), tissue inhibitors of matrix metalloproteinase (TIMP)-1, and interleukin (IL)-10 immunoreactivities were investigated. In LPS group, YKL-40, creatine phosphate (both, p < 0.05), SOD, GR, adenosine mono-phophate (AMP), adenosine tri-phosphate (ATP) (for all, p < 0.01) were significantly decreased, while TBARS and adenosine di-phosphate (ADP) levels were increased (p < 0.01, p < 0.05; respectively) compared to other groups. MMP-2 and -9, TIMP-1, TNF-α, IL-10, and MPO immunoreactivity were investigated in LPS group. On the contrary, in M+LPS group, MMP-9, TIMP-1 immunoreactivities were not found and IL-10 and MMP-2 immunoreactivities were found with little involvement. In M+LPS group, YKL-40, GR, AMP, ATP, creatine phosphate (for all, p < 0.05), and SOD (p < 0.01) levels were significantly increased and TBARS levels were decreased (p < 0.05). In our study, we suggest that melatonin exerts a protective and curative effect by reducing the matrix metalloproteinase levels responsible for tissue damage balance, stimulating the release of antioxidant enzymes, regulating cytokines and energy balance during endotoxemia.
Collapse
Affiliation(s)
- Gulten Ates
- Department of Physiology, Faculty of Medicine, Istanbul Yeni Yuzyil University, Istanbul 34010, Turkey
| | - Sule Tamer
- Department of Physiology, Istanbul Medical Faculty, Istanbul University, Istanbul 34093, Turkey
| | - Hatice Yorulmaz
- Faculty of Health Sciences, Halic University, Istanbul 34445, Turkey
| | - Sevcihan Mutlu
- Department of Clinical Oncological, Institute of Oncology, Istanbul University, Istanbul 34093, Turkey
| | - Vakur Olgac
- Department of Pathology, Institute of Oncology, Istanbul University, Istanbul 34093, Turkey
| | - Abdullah Aksu
- Department of Chemical Oceanography, Institute of Marine Sciences and Management, Istanbul University, Istanbul 34134, Turkey
| | - Nuray Balkis Caglar
- Department of Chemical Oceanography, Institute of Marine Sciences and Management, Istanbul University, Istanbul 34134, Turkey
| | - Elif Özkök
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul 34093, Turkey
| |
Collapse
|
17
|
Tran NT, Kowalski GM, Muccini AM, Nitsos I, Hale N, Snow RJ, Walker DW, Ellery SJ. Creatine supplementation reduces the cerebral oxidative and metabolic stress responses to acute in utero hypoxia in the late-gestation fetal sheep. J Physiol 2022; 600:3193-3210. [PMID: 35587817 PMCID: PMC9542404 DOI: 10.1113/jp282840] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/22/2022] [Indexed: 11/24/2022] Open
Abstract
Abstract Prophylactic creatine treatment may reduce hypoxic brain injury due to its ability to sustain intracellular ATP levels thereby reducing oxidative and metabolic stress responses during oxygen deprivation. Using microdialysis, we investigated the real‐time in vivo effects of fetal creatine supplementation on cerebral metabolism following acute in utero hypoxia caused by umbilical cord occlusion (UCO). Fetal sheep (118 days’ gestational age (dGA)) were implanted with an inflatable Silastic cuff around the umbilical cord and a microdialysis probe inserted into the right cerebral hemisphere for interstitial fluid sampling. Creatine (6 mg kg−1 h−1) or saline was continuously infused intravenously from 122 dGA. At 131 dGA, a 10 min UCO was induced. Hourly microdialysis samples were obtained from −24 to 72 h post‐UCO and analysed for percentage change of hydroxyl radicals (•OH) and interstitial metabolites (lactate, pyruvate, glutamate, glycerol, glycine). Histochemical markers of protein and lipid oxidation were assessed at post‐mortem 72 h post‐UCO. Prior to UCO, creatine treatment reduced pyruvate and glycerol concentrations in the microdialysate outflow. Creatine treatment reduced interstitial cerebral •OH outflow 0 to 24 h post‐UCO. Fetuses with higher arterial creatine concentrations before UCO presented with reduced levels of hypoxaemia (PO2 and SO2) during UCO which associated with reduced interstitial cerebral pyruvate, lactate and •OH accumulation. No effects of creatine treatment on immunohistochemical markers of oxidative stress were found. In conclusion, fetal creatine treatment decreased cerebral outflow of •OH and was associated with an improvement in cerebral bioenergetics following acute hypoxia.
![]() Key points Fetal hypoxia can cause persistent metabolic and oxidative stress responses that disturb energy homeostasis in the brain. Creatine in its phosphorylated form is an endogenous phosphagen; therefore, supplementation is a proposed prophylactic treatment for fetal hypoxia. Fetal sheep instrumented with a cerebral microdialysis probe were continuously infused with or without creatine‐monohydrate for 10 days before induction of 10 min umbilical cord occlusion (UCO; 131 days’ gestation). Cerebral interstitial fluid was collected up to 72 h following UCO. Prior to UCO, fetal creatine supplementation reduced interstitial cerebral pyruvate and glycerol concentrations. Fetal creatine supplementation reduced cerebral hydroxyl radical efflux up to 24 h post‐UCO. Fetuses with higher arterial creatine concentrations before UCO and reduced levels of systemic hypoxaemia during UCO were associated with reduced cerebral interstitial pyruvate, lactate and •OH following UCO. Creatine supplementation leads to some improvements in cerebral bioenergetics following in utero acute hypoxia.
Collapse
Affiliation(s)
- Nhi Thao Tran
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Monash Medical Centre, Clayton, Melbourne, Victoria, Australia
| | - Greg M Kowalski
- Institute for Physical Activity & Nutrition, Deakin University, Burwood, School of Exercise & Nutrition Sciences, Deakin University, Geelong Melbourne, Victoria, Australia.,Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Anna M Muccini
- The Ritchie Centre, Hudson Institute of Medical Research, Monash Medical Centre, Clayton, Melbourne, Victoria, Australia.,Genetic Research Services, University of Queensland, Queensland, Australia
| | - Ilias Nitsos
- The Ritchie Centre, Hudson Institute of Medical Research, Monash Medical Centre, Clayton, Melbourne, Victoria, Australia.,Department of Obstetrics & Gynecology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Nadia Hale
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Monash Medical Centre, Clayton, Melbourne, Victoria, Australia
| | - Rod J Snow
- Institute for Physical Activity & Nutrition, Deakin University, Burwood, School of Exercise & Nutrition Sciences, Deakin University, Geelong Melbourne, Victoria, Australia
| | - David W Walker
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria, Australia
| | - Stacey J Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, Monash Medical Centre, Clayton, Melbourne, Victoria, Australia.,Department of Obstetrics & Gynecology, Monash University, Clayton, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Ibanoglu MC, Köse C, Yilmaz-Ergani S, Arslan B, Akpınar F, Engin-Ustun Y. A prospective study of myeloperoxidase levels in water immersion. Placenta 2022; 123:1-4. [DOI: 10.1016/j.placenta.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/18/2022] [Accepted: 04/19/2022] [Indexed: 11/16/2022]
|
19
|
Shcherbitskaia AD, Vasilev DS, Milyutina YP, Tumanova NL, Mikhel AV, Zalozniaia IV, Arutjunyan AV. Prenatal Hyperhomocysteinemia Induces Glial Activation and Alters Neuroinflammatory Marker Expression in Infant Rat Hippocampus. Cells 2021; 10:cells10061536. [PMID: 34207057 PMCID: PMC8234222 DOI: 10.3390/cells10061536] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/15/2022] Open
Abstract
Maternal hyperhomocysteinemia is one of the common complications of pregnancy that causes offspring cognitive deficits during postnatal development. In this study, we investigated the effect of prenatal hyperhomocysteinemia (PHHC) on inflammatory, glial activation, and neuronal cell death markers in the hippocampus of infant rats. Female Wistar rats received L-methionine (0.6 g/kg b.w.) by oral administration during pregnancy. On postnatal days 5 and 20, the offspring’s hippocampus was removed to perform histological and biochemical studies. After PHHC, the offspring exhibited increased brain interleukin-1β and interleukin-6 levels and glial activation, as well as reduced anti-inflammatory interleukin-10 level in the hippocampus. Additionally, the activity of acetylcholinesterase was increased in the hippocampus of the pups. Exposure to PHHC also resulted in the reduced number of neurons and disrupted neuronal ultrastructure. At the same time, no changes in the content and activity of caspase-3 were found in the hippocampus of the pups. In conclusion, our findings support the hypothesis that neuroinflammation and glial activation could be involved in altering the hippocampus cellular composition following PHHC, and these alterations could be associated with cognitive disorders later in life.
Collapse
Affiliation(s)
- Anastasiia D. Shcherbitskaia
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Y.P.M.); (A.V.M.); (I.V.Z.); (A.V.A.)
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 St. Petersburg, Russia; (D.S.V.); (N.L.T.)
- Correspondence:
| | - Dmitrii S. Vasilev
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 St. Petersburg, Russia; (D.S.V.); (N.L.T.)
| | - Yulia P. Milyutina
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Y.P.M.); (A.V.M.); (I.V.Z.); (A.V.A.)
| | - Natalia L. Tumanova
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 St. Petersburg, Russia; (D.S.V.); (N.L.T.)
| | - Anastasiia V. Mikhel
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Y.P.M.); (A.V.M.); (I.V.Z.); (A.V.A.)
| | - Irina V. Zalozniaia
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Y.P.M.); (A.V.M.); (I.V.Z.); (A.V.A.)
| | - Alexander V. Arutjunyan
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Y.P.M.); (A.V.M.); (I.V.Z.); (A.V.A.)
| |
Collapse
|
20
|
Peila C, Bertino E, Cresi F, Coscia A. Interactions between preeclampsia and composition of the human milk: what do we know? J Matern Fetal Neonatal Med 2021; 35:6219-6225. [PMID: 34121581 DOI: 10.1080/14767058.2021.1910655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Purpose: Preeclampsia (PE) is a gestational hypertensive syndrome characterized by a complex disease with variable clinical manifestation. An aspect of increased interest is the possible effect of PE on the mammary gland (and therefore on breast milk composition) considering that a mother's own milk is always considered the first choice for the nutrition of all neonates. Human milk (HM) is a unique food believed to contain biological factors involved in both short and long-term benefits. This article aims to review the published papers on this topic and to offer additional insights on the role of this gestational pathology on the composition of HM.Materials and methods: This review was performed by searching the MEDLINE, EMBASE, CINHAL, and Cochrane Library databases.Results: A total of 15 articles were selected.Conclusions: Overall, the findings from the literature suggest that PE can alter the composition of HM milk. Nevertheless, the number of studies in this field are scarce, and the related protocols present some limitation, for example, evaluating the variability of just a few specific milk biochemical markers in association with this syndrome.
Collapse
Affiliation(s)
- Chiara Peila
- Complex Structure Neonatology Unit, Department of Public Health and Paediatric, University of Turin, Torino, Italy
| | - Enrico Bertino
- Complex Structure Neonatology Unit, Department of Public Health and Paediatric, University of Turin, Torino, Italy
| | - Francesco Cresi
- Complex Structure Neonatology Unit, Department of Public Health and Paediatric, University of Turin, Torino, Italy
| | - Alessandra Coscia
- Complex Structure Neonatology Unit, Department of Public Health and Paediatric, University of Turin, Torino, Italy
| |
Collapse
|
21
|
Ahmed J, Pullattayil S AK, Robertson NJ, More K. Melatonin for neuroprotection in neonatal encephalopathy: A systematic review & meta-analysis of clinical trials. Eur J Paediatr Neurol 2021; 31:38-45. [PMID: 33601197 DOI: 10.1016/j.ejpn.2021.02.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/11/2021] [Accepted: 02/04/2021] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Melatonin has shown neuroprotective properties in pre-clinical studies of perinatal asphyxia through antioxidant, anti-apoptotic and anti-inflammatory actions. Studies have also demonstrated its safety and efficacy in neonatal encephalopathy (NE). However, its role in the current era of therapeutic hypothermia (HT) is unclear. The review aims to describe the currently available clinical evidence for Melatonin as a potential therapy for NE. METHODS Data Sources: We searched Medline, EMBASE, CINAHL, LILACS, and Cochrane central databases, published journals, and conference proceedings from inception to May 31, 2020. STUDY SELECTION Randomized controlled trials (RCTs) of Melatonin for NE in term or late preterm infants reporting neurodevelopmental outcomes, death, or both. The evidence quality was evaluated using the GRADE system, while the recommendations were taken according to the quality. RESULTS We included five RCTs involving 215 neonates. Long-term development outcome data is lacking in all except in one small study, reporting significantly higher composite cognition scores at 18 months. One study reported intermediate 6-month favorable development on follow-up. Meta-analysis of mortality in combined HT + Melatonin group vs HT alone (Studies = 2, participants = 54) demonstrated no significant reduction with relative risk (RR) 0.42; 95%CI, 0.99-1.12). The overall GRADE evidence quality was very low for a very small sample size. We did not meta-analyze the data for Melatonin alone therapy without HT, as the included studies were of very low quality. CONCLUSIONS Despite strong experimental data supporting the role of Melatonin as a neuroprotective agent in NE (both alone and as an adjunct with therapeutic hypothermia), the clinical data supporting the neuroprotective effects in neonates is limited. Larger well designed, adequately powered multicentre clinical trials are urgently needed to define the neuroprotective role of Melatonin in optimizing outcomes of NE.
Collapse
Affiliation(s)
- Javed Ahmed
- Division of Neonatology, Women's Wellness and Research Centre, Hamad Medical Corporation, Doha, Qatar.
| | | | - Nicola J Robertson
- Institute for Women's Health, University College London, London, WC1E 6HX, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, BioQuarter, 49 Little France Crescent, Edinburgh, EH16 4SB, UK; The Roslin Institute, University of Edinburgh, Easter Bush Campus, EH25 9RG, UK.
| | - Kiran More
- Division of Neonatology, Sidra Medicine, Doha, Qatar; Weill Cornell Medicine, Doha, Qatar.
| |
Collapse
|
22
|
Yates N, Gunn AJ, Bennet L, Dhillon SK, Davidson JO. Preventing Brain Injury in the Preterm Infant-Current Controversies and Potential Therapies. Int J Mol Sci 2021; 22:1671. [PMID: 33562339 PMCID: PMC7915709 DOI: 10.3390/ijms22041671] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/12/2022] Open
Abstract
Preterm birth is associated with a high risk of morbidity and mortality including brain damage and cerebral palsy. The development of brain injury in the preterm infant may be influenced by many factors including perinatal asphyxia, infection/inflammation, chronic hypoxia and exposure to treatments such as mechanical ventilation and corticosteroids. There are currently very limited treatment options available. In clinical trials, magnesium sulfate has been associated with a small, significant reduction in the risk of cerebral palsy and gross motor dysfunction in early childhood but no effect on the combined outcome of death or disability, and longer-term follow up to date has not shown improved neurological outcomes in school-age children. Recombinant erythropoietin has shown neuroprotective potential in preclinical studies but two large randomized trials, in extremely preterm infants, of treatment started within 24 or 48 h of birth showed no effect on the risk of severe neurodevelopmental impairment or death at 2 years of age. Preclinical studies have highlighted a number of promising neuroprotective treatments, such as therapeutic hypothermia, melatonin, human amnion epithelial cells, umbilical cord blood and vitamin D supplementation, which may be useful at reducing brain damage in preterm infants. Moreover, refinements of clinical care of preterm infants have the potential to influence later neurological outcomes, including the administration of antenatal and postnatal corticosteroids and more accurate identification and targeted treatment of seizures.
Collapse
Affiliation(s)
- Nathanael Yates
- The Queensland Brain Institute, University of Queensland, St Lucia, QLD 4072, Australia;
- School of Human Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Alistair J. Gunn
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| | - Laura Bennet
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| | - Simerdeep K. Dhillon
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| | - Joanne O. Davidson
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| |
Collapse
|
23
|
Flinn T, Kleemann DO, Swinbourne AM, Kelly JM, Weaver AC, Walker SK, Gatford KL, Kind KL, van Wettere WHEJ. Neonatal lamb mortality: major risk factors and the potential ameliorative role of melatonin. J Anim Sci Biotechnol 2020; 11:107. [PMID: 33292527 PMCID: PMC7643391 DOI: 10.1186/s40104-020-00510-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
High incidences of pre-weaning mortality continue to limit global sheep production, constituting a major economic and welfare concern. Despite significant advances in genetics, nutrition, and management, the proportion of lamb deaths has remained stable at 15–20% over the past four decades. There is mounting evidence that melatonin can improve outcomes in compromised ovine pregnancies via enhanced uterine bloodflow and neonatal neuroprotection. This review provides an overview of the major risk factors and underlying mechanisms involved in perinatal lamb mortality and discusses the potential of melatonin treatment as a remedial strategy. Supplementing pregnant ewes with melatonin enhances uterine bloodflow and fetal oxygenation, and potentially birthweight and neonatal thermogenic capacity. Melatonin freely crosses the ovine placenta and blood-brain barrier and provides neuroprotection to the fetal lamb during periods of chronic and acute hypoxia throughout gestation, with improved behavioural outcomes in hypoxic neonates. The current literature provides strong evidence that maternal melatonin treatment improves outcomes for lambs which experience compromised in utero development or prolonged parturition, though to date this has not been investigated in livestock production systems. As such there is a clear basis for continued research into the effects of maternal melatonin supplementation during gestation on pre-weaning survival under extensive production conditions.
Collapse
Affiliation(s)
- Tom Flinn
- Davies Livestock Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia.
| | - David O Kleemann
- Turretfield Research Centre, South Australian Research and Development Institute, Rosedale, SA, Australia
| | - Alyce M Swinbourne
- Davies Livestock Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Jennifer M Kelly
- Turretfield Research Centre, South Australian Research and Development Institute, Rosedale, SA, Australia
| | - Alice C Weaver
- Turretfield Research Centre, South Australian Research and Development Institute, Rosedale, SA, Australia
| | - Simon K Walker
- Turretfield Research Centre, South Australian Research and Development Institute, Rosedale, SA, Australia
| | - Kathryn L Gatford
- Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Karen L Kind
- Davies Livestock Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - William H E J van Wettere
- Davies Livestock Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| |
Collapse
|
24
|
Hurley T, O'Dea M, Aslam S, Aly H, Robertson N, Molloy E. Melatonin treatment for newborns with hypoxic ischaemic encephalopathy. Hippokratia 2020. [DOI: 10.1002/14651858.cd013754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Tim Hurley
- Department of Paediatrics; Trinity College Dublin; Dublin Ireland
| | - Mary O'Dea
- Paediatric and Child Health; Trinity College Dublin; Dublin Ireland
| | - Saima Aslam
- Neonatology; National Maternity Hospital; Dublin Ireland
| | - Hany Aly
- Neonatology; Cleveland Clinic Children’s Hospital; Cleveland OH USA
| | - Nikki Robertson
- Obstetrics and Gynaecology; University College London; London UK
| | - Eleanor Molloy
- Paediatric and Child Health; Trinity College Dublin; Dublin Ireland
- Department of Paediatrics; The National Children’s Hospital, Tallaght; Dublin Ireland
| |
Collapse
|
25
|
Chitimus DM, Popescu MR, Voiculescu SE, Panaitescu AM, Pavel B, Zagrean L, Zagrean AM. Melatonin's Impact on Antioxidative and Anti-Inflammatory Reprogramming in Homeostasis and Disease. Biomolecules 2020; 10:biom10091211. [PMID: 32825327 PMCID: PMC7563541 DOI: 10.3390/biom10091211] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/30/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022] Open
Abstract
There is a growing consensus that the antioxidant and anti-inflammatory properties of melatonin are of great importance in preserving the body functions and homeostasis, with great impact in the peripartum period and adult life. Melatonin promotes adaptation through allostasis and stands out as an endogenous, dietary, and therapeutic molecule with important health benefits. The anti-inflammatory and antioxidant effects of melatonin are intertwined and are exerted throughout pregnancy and later during development and aging. Melatonin supplementation during pregnancy can reduce ischemia-induced oxidative damage in the fetal brain, increase offspring survival in inflammatory states, and reduce blood pressure in the adult offspring. In adulthood, disturbances in melatonin production negatively impact the progression of cardiovascular risk factors and promote cardiovascular and neurodegenerative diseases. The most studied cardiovascular effects of melatonin are linked to hypertension and myocardial ischemia/reperfusion injury, while the most promising ones are linked to regaining control of metabolic syndrome components. In addition, there might be an emerging role for melatonin as an adjuvant in treating coronavirus disease 2019 (COVID 19). The present review summarizes and comments on important data regarding the roles exerted by melatonin in homeostasis and oxidative stress and inflammation related pathologies.
Collapse
Affiliation(s)
- Diana Maria Chitimus
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Mihaela Roxana Popescu
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, Elias University Hospital, 010164 Bucharest, Romania;
| | - Suzana Elena Voiculescu
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Anca Maria Panaitescu
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, Filantropia Clinical Hospital, 010164 Bucharest, Romania;
| | - Bogdan Pavel
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Leon Zagrean
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Ana-Maria Zagrean
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
- Correspondence:
| |
Collapse
|
26
|
Duan L, Li S, Wang L, Jing Y, Li G, Sun Y, Sun W, Li Y, Zhao L, Xin S. Melatonin Plays a Critical Protective Role in Nicotine-Related Abdominal Aortic Aneurysm. Front Physiol 2020; 11:866. [PMID: 32765304 PMCID: PMC7379742 DOI: 10.3389/fphys.2020.00866] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 06/26/2020] [Indexed: 01/10/2023] Open
Abstract
Aim: Smoking is a major risk factor for abdominal aortic aneurysm (AAA). Among the components of smoke, nicotine is known to exert pro-atherosclerotic, prothrombotic, and proangiogenic effects on vascular smooth muscle cells (VSMCs). The current study was designed to investigate the mechanisms through which nicotine induces vascular wall dysfunction and to examine whether melatonin protects against nicotine-related AAA. Methods: In this study, an enzyme-linked immunosorbent assay (ELISA) was used to measure melatonin and TNF-α levels, as well as total antioxidant status (TAS), in patients with AAA. We established a nicotine-related AAA model and explored the mechanisms underlying the therapeutic effects of melatonin. Tissue histopathology was used to assess vascular function, while western blotting (WB) and immunofluorescence staining were performed to detect protein expression. Results: We observed melatonin insufficiency in the serum from patients with AAA, particularly smokers. Moreover, melatonin level was positively correlated with antioxidant capacity. In the in vivo model, nicotine accelerated AAA expansion and destroyed vascular structure. Furthermore, OPN, LC3II, p62, matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9), NF-κB p65, TNF-α, phosphorylated AKT, and phosphorylated mTOR levels were increased, in vivo, following nicotine treatment, while SM22α and α-SMA levels were reduced. Additionally, melatonin attenuated the effects of nicotine on AAA and reversed changes in protein expression. Moreover, melatonin lost its protective effects following bafilomycin A1-mediated inhibition of autophagy. Conclusion: Based on our data, melatonin exerts a beneficial effect on rats with nicotine-related AAA by downregulating the AKT-mTOR signaling pathway, improving autophagy dysfunction, and restoring the VSMC phenotype.
Collapse
Affiliation(s)
- Liren Duan
- Department of Vascular Surgery, The First Hospital of China Medical University, Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, China
| | - Shenli Li
- Department of Anesthesiology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Lei Wang
- Department of Vascular Surgery, The First Hospital of China Medical University, Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, China
| | - Yuchen Jing
- Department of Vascular Surgery, The First Hospital of China Medical University, Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, China
| | - Guangxin Li
- Department of Surgery, Yale University School of Medicine, New Haven, CT, United States
| | - Yaodong Sun
- Department of Vascular Surgery, The First Hospital of China Medical University, Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, China
| | - Weifeng Sun
- Department of Vascular Surgery, The First Hospital of China Medical University, Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, China
| | - Yalun Li
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, China
| | - Shijie Xin
- Department of Vascular Surgery, The First Hospital of China Medical University, Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, China
| |
Collapse
|
27
|
Montaldo P, Cunnington A, Oliveira V, Swamy R, Bandya P, Pant S, Lally PJ, Ivain P, Mendoza J, Atreja G, Padmesh V, Baburaj M, Sebastian M, Yasashwi I, Kamalarathnam C, Chandramohan R, Mangalabharathi S, Kumaraswami K, Kumar S, Benakappa N, Manerkar S, Mondhkar J, Prakash V, Sajjid M, Seeralar A, Jahan I, Moni SC, Shahidullah M, Sujatha R, Chandrasekaran M, Ramji S, Shankaran S, Kaforou M, Herberg J, Thayyil S. Transcriptomic profile of adverse neurodevelopmental outcomes after neonatal encephalopathy. Sci Rep 2020; 10:13100. [PMID: 32753750 PMCID: PMC7403382 DOI: 10.1038/s41598-020-70131-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 06/16/2020] [Indexed: 12/20/2022] Open
Abstract
A rapid and early diagnostic test to identify the encephalopathic babies at risk of adverse outcome may accelerate the development of neuroprotectants. We examined if a whole blood transcriptomic signature measured soon after birth, predicts adverse neurodevelopmental outcome eighteen months after neonatal encephalopathy. We performed next generation sequencing on whole blood ribonucleic acid obtained within six hours of birth from the first 47 encephalopathic babies recruited to the Hypothermia for Encephalopathy in Low and middle-income countries (HELIX) trial. Two infants with blood culture positive sepsis were excluded, and the data from remaining 45 were analysed. A total of 855 genes were significantly differentially expressed between the good and adverse outcome groups, of which RGS1 and SMC4 were the most significant. Biological pathway analysis adjusted for gender, trial randomisation allocation (cooling therapy versus usual care) and estimated blood leukocyte proportions revealed over-representation of genes from pathways related to melatonin and polo-like kinase in babies with adverse outcome. These preliminary data suggest that transcriptomic profiling may be a promising tool for rapid risk stratification in neonatal encephalopathy. It may provide insights into biological mechanisms and identify novel therapeutic targets for neuroprotection.
Collapse
Affiliation(s)
- Paolo Montaldo
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK. .,Neonatal Unit, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| | - Aubrey Cunnington
- Paediatric Infectious Diseases, Department of Infectious Diseases, Imperial College London, London, UK
| | - Vania Oliveira
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Ravi Swamy
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Prathik Bandya
- Neonatal Medicine, Indira Gandhi Institute of Child Health, Bangalore, Karnataka, India
| | - Stuti Pant
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Peter J Lally
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Phoebe Ivain
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Josephine Mendoza
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Gaurav Atreja
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Vadakepat Padmesh
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Mythili Baburaj
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Monica Sebastian
- Neonatal Medicine, Institute of Child Health, Madras Medical College, Tamil Nadu, Chennai, India
| | - Indiramma Yasashwi
- Neonatal Medicine, Indira Gandhi Institute of Child Health, Bangalore, Karnataka, India
| | - Chinnathambi Kamalarathnam
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Rema Chandramohan
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Sundaram Mangalabharathi
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Kumutha Kumaraswami
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Shobha Kumar
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Naveen Benakappa
- Neonatal Medicine, Indira Gandhi Institute of Child Health, Bangalore, Karnataka, India
| | | | | | - Vinayagam Prakash
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Mohammed Sajjid
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Arasar Seeralar
- Neonatal Medicine, Institute of Child Health, Madras Medical College, Tamil Nadu, Chennai, India
| | - Ismat Jahan
- Neonatal Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | | | - Mohammod Shahidullah
- Neonatal Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Radhika Sujatha
- Neonatal Medicine, Government Medical College, Thiruvananthapuram, Kerala, India
| | - Manigandan Chandrasekaran
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Siddarth Ramji
- Neonatal Medicine, Maulana Azad Medical College, New Delhi, Delhi, India
| | - Seetha Shankaran
- Neonatal-Perinatal Medicine, Wayne State University, Detroit, MI, USA
| | - Myrsini Kaforou
- Paediatric Infectious Diseases, Department of Infectious Diseases, Imperial College London, London, UK
| | - Jethro Herberg
- Paediatric Infectious Diseases, Department of Infectious Diseases, Imperial College London, London, UK
| | - Sudhin Thayyil
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| |
Collapse
|
28
|
Abstract
OBJECTIVES To investigate the effect of adding melatonin to hypothermia treatment on neurodevelopmental outcomes in asphyctic newborns. DESIGN Pilot multicenter, randomized, controlled, double-blind clinical trial. Statistical comparison of results obtained in two intervention arms: hypothermia plus placebo and hypothermia plus melatonin. SETTING Level 3 neonatal ICU. PATIENTS Twenty-five newborns were recruited. INTERVENTIONS The hypothermia plus melatonin patients received a daily dose of IV melatonin, 5 mg per kg body weight, for 3 days. General laboratory variables were measured both at neonatal ICU admission and after intervention. All infants were studied with amplitude-integrated electroencephalography and brain MRI within the first week of life. The neurodevelopmental Bayley III test, the Gross Motor Function Classification System, and the Tardieu scale were applied at the ages of 6 and 18 months. MEASUREMENTS AND MAIN RESULTS Clinical characteristics, laboratory evaluations, MRI findings, and amplitude-integrated electroencephalography background did not differ between the treatment groups. The newborns in the hypothermia plus melatonin group achieved a significantly higher composite score for the cognitive section of the Bayley III test at 18 months old, with respect to the hypothermia plus placebo group (p = 0.05). There were no differences between the groups according to the Gross Motor Function Classification System and Tardieu motor assessment scales. CONCLUSIONS The early addition of IV melatonin to asphyctic neonates is feasible and may improve long-term neurodevelopment. To our knowledge, this is the first clinical trial to analyze the administration of IV melatonin as an adjuvant therapy to therapeutic hypothermia.
Collapse
|
29
|
Motallebzadeh E, Tameh AA, Zavareh SAT, Farhood B, Aliasgharzedeh A, Mohseni M. Neuroprotective effect of melatonin on radiation-induced oxidative stress and apoptosis in the brainstem of rats. J Cell Physiol 2020; 235:8791-8798. [PMID: 32324264 DOI: 10.1002/jcp.29722] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023]
Abstract
This study aimed to determine the effects of melatonin on irradiation-induced apoptosis and oxidative stress in the brainstem region of Wistar rats. Therefore, the animals underwent whole-brain X-radiation with a single dose of 25 Gy in the presence or absence of melatonin pretreatment at a concentration of 100 mg/kg BW. The rats were allocated into four groups (10 rats in each group): namely, vehicle control (VC), 100 mg/kg of melatonin alone (MLT), irradiation-only (RAD), and irradiation plus 100 mg/kg of melatonin (RAM). An hour before irradiation, the animals received intraperitoneal (IP) melatonin and then were killed after 6 hr, followed by measurement of nitric oxide (NO), malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase (CAT), and total antioxidant capacity (TAC) in the brainstem region. Furthermore, the western blot analysis technique was performed to assess the caspase-3 expression level. Results showed significantly higher MDA and NO levels in the brainstem tissues for the RAD group when compared with the VC group (p < .001). Moreover, the irradiated rats exhibited a significant decrease in the levels of CAT, SOD, GPx, and TAC (p < .01, p < .001, p < .001, and p < .001, respectively) in comparison to the VC group. The results of apoptosis assessment revealed that the expression level of caspase-3 significantly rose in the RAD group in comparison with the VC group (p < .001). Pretreatment with melatonin ameliorated the radiation-induced adverse effects by decreasing the MDA and NO levels (p < .001) and increasing the antioxidant enzyme activities (p < .001). Consequently, the caspase-3 protein expression level in the RAM group showed a significant reduction in comparison with the RAD group (p < .001). In conclusion, melatonin approximately showed a capacity for neuroprotective activity in managing irradiation-induced oxidative stress and apoptosis in the brainstem of rats; however, the use of melatonin as a neuroprotective agent in humans requires further study, particularly clinical trials.
Collapse
Affiliation(s)
- Elham Motallebzadeh
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.,Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Akbar Aliasgharzedeh
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mehran Mohseni
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
30
|
Arutjunyan AV, Milyutina YP, Shcherbitskaia AD, Kerkeshko GO, Zalozniaia IV, Mikhel AV. Neurotrophins of the Fetal Brain and Placenta in Prenatal Hyperhomocysteinemia. BIOCHEMISTRY (MOSCOW) 2020; 85:213-223. [PMID: 32093597 DOI: 10.1134/s000629792002008x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prenatal hyperhomocysteinemia (PHHC) in pregnant rats was induced by chronic L-methionine loading, resulting in a significant increase in the L-homocysteine content both in the mothers' blood and blood and brain of fetuses. Significant decrease in the weight of the placenta, fetus, and fetal brain was detected by the morphometric studies on day 20 of pregnancy. PHHC also activated maternal immune system due to the increase in the content of proinflammatory interleukin-1β in the rat blood and fetal part of the placenta. PHHC elevated the levels of the brain-derived neurotrophic factor (BDNF, 29 kDa) and nerve growth factor (NGF, 31 kDa) precursors in the placenta and the content of the BDNF isoform (29 kDa) in the fetal brain. The content of neuregulin 1 (NRG1) decreased in the placenta and increased in the fetal brain on day 20 of embryonic development. An increase in the caspase-3 activity was detected in the brains of fetuses subjected to PHHC. It was suggested that changes in the processing of neurotrophins induced by PPHC, oxidative stress, and inflammatory processes initiated by it, as well as apoptosis, play an important role in the development of brain disorders in the offspring.
Collapse
Affiliation(s)
- A V Arutjunyan
- Ott Institute of Obstetrics, Gynecology, and Reproductology, St. Petersburg, 199034, Russia.
| | - Yu P Milyutina
- Ott Institute of Obstetrics, Gynecology, and Reproductology, St. Petersburg, 199034, Russia
| | - A D Shcherbitskaia
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, 194223, Russia
| | - G O Kerkeshko
- Ott Institute of Obstetrics, Gynecology, and Reproductology, St. Petersburg, 199034, Russia
| | - I V Zalozniaia
- Ott Institute of Obstetrics, Gynecology, and Reproductology, St. Petersburg, 199034, Russia
| | - A V Mikhel
- Ott Institute of Obstetrics, Gynecology, and Reproductology, St. Petersburg, 199034, Russia
| |
Collapse
|
31
|
Stefanov G, Briyal S, Pais G, Puppala B, Gulati A. Relationship Between Oxidative Stress Markers and Endothelin-1 Levels in Newborns of Different Gestational Ages. Front Pediatr 2020; 8:279. [PMID: 32582590 PMCID: PMC7280445 DOI: 10.3389/fped.2020.00279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/04/2020] [Indexed: 11/13/2022] Open
Abstract
Oxidative stress results from excessive reactive oxygen species formation and/or inadequate antioxidant defense. Premature and critically ill infants are especially susceptible due to an immature intrinsic antioxidant system that cannot fully compensate for a free radical load. Oxidative stress is also associated with endothelial dysfunction and alterations in Endothelin-1 (ET-1) signaling pathways. However, the effects of the complex interaction between oxidative stress and ET-1 in newborns are not well-understood. The objective of this pilot study was to determine the relationship between levels of common oxidative stress biomarkers [glutathione (GSH), malondialdehyde (MDA)] and ET-1 in newborns of different gestational ages. In a level IV NICU, 63 neonates were prospectively enrolled and divided into groups based on gestational age at birth: Early Preterm (24 0/7-30 6/7 weeks), Late Preterm (31 0/7-36 6/7 weeks), and Term (37 0/7-42 weeks). Umbilical cord (1.5 mL) and 24(±4) h of life (24 h) (1 mL) blood samples were collected for GSH, MDA, and ET-1 analyses. GSH, MDA, and ET-1 were determined using established methodology. Mean cord MDA levels for all age groups, Early Preterm (2.93 ± 0.08 pg/ml), Late Preterm (2.73 ± 0.15 pg/ml), and Term (2.92 ± 0.13 pg/ml), were significantly higher than those at 24 h of life (p < 0.001). Mean cord ET-1 levels were significantly higher than 24 h samples in both Early and Late Preterm groups (p < 0.05). Cord and 24 h ET-1 levels did not correlate with MDA and GSH levels at birth (r2 = 0.03, p > 0.05 and r2 = 0.001, p > 0.05, respectively) or 24 h of life (r2 = 0.001, p > 0.05 and r2 = 0.03, p > 0.05, respectively). Preterm neonates exposed to prenatal corticosteroids (1.87 ± 0.31 pg/ml) had lower cord MDA levels than non-exposed neonates (2.85 ± 0.12 pg/ml) (p < 0.05). Both cord and 24 h OS markers were significantly higher in neonates treated with oxygen therapy (p < 0.005 and p < 0.05, respectively) than those who did not receive supplemental oxygen. Oxidative stress markers (MDA and GSH) and ET-1 levels act independently. MDA is higher in cord blood than at 24 h of life regardless of gestational age. In preterm neonates, ET-1 levels are higher in umbilical cord blood compared to 24 h of life.
Collapse
Affiliation(s)
- Gospodin Stefanov
- Division of Neonatology, Advocate Children's Hospital, Park Ridge, IL, United States
| | - Seema Briyal
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Gwendolyn Pais
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Bhagya Puppala
- Division of Neonatology, Advocate Children's Hospital, Park Ridge, IL, United States
| | - Anil Gulati
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| |
Collapse
|
32
|
Scarpato R, Testi S, Colosimo V, Garcia Crespo C, Micheli C, Azzarà A, Tozzi MG, Ghirri P. Role of oxidative stress, genome damage and DNA methylation as determinants of pathological conditions in the newborn: an overview from conception to early neonatal stage. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 783:108295. [DOI: 10.1016/j.mrrev.2019.108295] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 11/25/2019] [Accepted: 12/24/2019] [Indexed: 12/15/2022]
|
33
|
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: 34] [Impact Index Per Article: 5.7] [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
|
34
|
Gao L, Wang G, Yuan H, Xu E, Liu G, Liu J. Serum metabolomics in mice after paraquat posioning. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0049-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
35
|
Arutjunyan AV, Evsyukova II, Polyakova VO. The Role of Melatonin in Morphofunctional Development of the Brain in Early Ontogeny. NEUROCHEM J+ 2019. [DOI: 10.1134/s1819712419030036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Hamid ERA, Ali WH, Azmy A, Ahmed HH, Sherif LS, Saleh MT. Oxidative Stress and Anti-Oxidant Markers in Premature Infants with Respiratory Distress Syndrome. Open Access Maced J Med Sci 2019; 7:2858-2863. [PMID: 31844449 PMCID: PMC6901850 DOI: 10.3889/oamjms.2019.534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Neonatal respiratory distress syndrome (RDS) caused by decreased surfactant and structural lung immaturity. The imbalance between oxidative status and antioxidant defence system was suggested to be an important trigger for lung affection with RDS. AIM The goal of the current research was to elucidate the significance of the oxidant/ antioxidant status in the pathogenesis of RDS in preterm infants. PATIENTS AND METHODS This controlled study included 31 preterm neonates with RDS and 36 healthy preterm neonates. Quantification level of oxidative stress biomarkers; malondialdehyde (MDA) & hydrogen peroxide (H2O2) along with antioxidant enzymes activity; catalase (CAT) & superoxide dismutase (SOD) in plasma of healthy premature neonates compared with those with RDS. RESULTS status of oxidative stress markers (MDA & H2O2) showed a significant increase with decreased levels of antioxidant enzymes activity (CAT & SOD) in neonates with RDS when compared to healthy prematures. CONCLUSION The results obtained in this study indicate that the increased oxidative stress accompanied by reduced antioxidant defences may play a significant role in the pathogenesis of respiratory distress in preterm newborns.
Collapse
Affiliation(s)
| | - Walaa H Ali
- Child Health Department, National Research Centre, Dokki, Giza, Egypt
| | - Ashraf Azmy
- Child Health Department, National Research Centre, Dokki, Giza, Egypt
| | - Hanaa H Ahmed
- Hormones Department, National Research Centre, Dokki, Giza, Egypt
| | - Lobna S Sherif
- Child Health Department, National Research Centre, Dokki, Giza, Egypt
| | - Maysa T Saleh
- Child Health Department, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
37
|
Pramanik SK, Pal U, Choudhary P, Singh H, Reiter RJ, Ethirajan A, Swarnakar S, Das A. Stimuli-Responsive Nanocapsules for the Spatiotemporal Release of Melatonin: Protection against Gastric Inflammation. ACS APPLIED BIO MATERIALS 2019; 2:5218-5226. [DOI: 10.1021/acsabm.9b00236] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Sumit Kumar Pramanik
- CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, Gujarat 364 002, India
| | - Uttam Pal
- Chemical Sciences Division, Saha Institute of Nuclear Physics, Kolkata, West Bengal 700 064, India
| | - Preety Choudhary
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700 032, India
| | - Harwinder Singh
- CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, Gujarat 364 002, India
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas 78229, United States
| | - Anitha Ethirajan
- Institute for Materials Research (IMO), Hasselt University, Wetenschapspark 1, Diepenbeek 3590, Belgium
| | - Snehasikta Swarnakar
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700 032, India
| | - Amitava Das
- CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, Gujarat 364 002, India
| |
Collapse
|
38
|
Paprocka J, Kijonka M, Rzepka B, Sokół M. Melatonin in Hypoxic-Ischemic Brain Injury in Term and Preterm Babies. Int J Endocrinol 2019; 2019:9626715. [PMID: 30915118 PMCID: PMC6402213 DOI: 10.1155/2019/9626715] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/23/2019] [Accepted: 01/30/2019] [Indexed: 12/05/2022] Open
Abstract
Melatonin may serve as a potential therapeutic free radical scavenger and broad-spectrum antioxidant. It shows neuroprotective properties against hypoxic-ischemic brain injury in animal models. The authors review the studies focusing on the neuroprotective potential of melatonin and its possibility of treatment after perinatal asphyxia. Melatonin efficacy, low toxicity, and ability to readily cross through the blood-brain barrier make it a promising molecule. A very interesting thing is the difference between the half-life of melatonin in preterm neonates (15 hours) and adults (45-60 minutes). Probably, the use of synergic strategies-hypothermia coupled with melatonin treatment-may be promising in improving antioxidant action. The authors discuss and try to summarize the evidence surrounding the use of melatonin in hypoxic-ischemic events in term and preterm babies.
Collapse
Affiliation(s)
- Justyna Paprocka
- Department of Pediatric Neurology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marek Kijonka
- Department of Medical Physics, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Poland
| | - Beata Rzepka
- Students' Scientific Society, Department Pediatric Neurology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Maria Sokół
- Department of Medical Physics, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology Gliwice Branch, Poland
| |
Collapse
|
39
|
Hoffmann A, Spengler D. The Mitochondrion as Potential Interface in Early-Life Stress Brain Programming. Front Behav Neurosci 2018; 12:306. [PMID: 30574076 PMCID: PMC6291450 DOI: 10.3389/fnbeh.2018.00306] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/26/2018] [Indexed: 12/23/2022] Open
Abstract
Mitochondria play a central role in cellular energy-generating processes and are master regulators of cell life. They provide the energy necessary to reinstate and sustain homeostasis in response to stress, and to launch energy intensive adaptation programs to ensure an organism’s survival and future well-being. By this means, mitochondria are particularly apt to mediate brain programming by early-life stress (ELS) and to serve at the same time as subcellular substrate in the programming process. With a focus on mitochondria’s integrated role in metabolism, steroidogenesis and oxidative stress, we review current findings on altered mitochondrial function in the brain, the placenta and peripheral blood cells following ELS-dependent programming in rodents and recent insights from humans exposed to early life adversity (ELA). Concluding, we propose a role of the mitochondrion as subcellular intersection point connecting ELS, brain programming and mental well-being, and a role as a potential site for therapeutic interventions in individuals exposed to severe ELS.
Collapse
Affiliation(s)
- Anke Hoffmann
- Epigenomics of Early Life, Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Dietmar Spengler
- Epigenomics of Early Life, Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
40
|
Bebars GM, Kamel BA, Allam E. Comparison between preterm and full term neonatal cord selenium in correlation to maternal serum selenium levels. EGYPTIAN PEDIATRIC ASSOCIATION GAZETTE 2018. [DOI: 10.1016/j.epag.2018.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
41
|
Ahmad R, Sportelli V, Ziller M, Spengler D, Hoffmann A. Tracing Early Neurodevelopment in Schizophrenia with Induced Pluripotent Stem Cells. Cells 2018; 7:E140. [PMID: 30227641 PMCID: PMC6162757 DOI: 10.3390/cells7090140] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/29/2022] Open
Abstract
Schizophrenia (SCZ) is a devastating mental disorder that is characterized by distortions in thinking, perception, emotion, language, sense of self, and behavior. Epidemiological evidence suggests that subtle perturbations in early neurodevelopment increase later susceptibility for disease, which typically manifests in adolescence to early adulthood. Early perturbations are thought to be significantly mediated through incompletely understood genetic risk factors. The advent of induced pluripotent stem cell (iPSC) technology allows for the in vitro analysis of disease-relevant neuronal cell types from the early stages of human brain development. Since iPSCs capture each donor's genotype, comparison between neuronal cells derived from healthy and diseased individuals can provide important insights into the molecular and cellular basis of SCZ. In this review, we discuss results from an increasing number of iPSC-based SCZ/control studies that highlight alterations in neuronal differentiation, maturation, and neurotransmission in addition to perturbed mitochondrial function and micro-RNA expression. In light of this remarkable progress, we consider also ongoing challenges from the field of iPSC-based disease modeling that call for further improvements on the generation and design of patient-specific iPSC studies to ultimately progress from basic studies on SCZ to tailored treatments.
Collapse
Affiliation(s)
- Ruhel Ahmad
- Max Planck Institute of Psychiatry, Translational Psychiatry, 80804 Munich, Germany.
| | - Vincenza Sportelli
- Max Planck Institute of Psychiatry, Translational Psychiatry, 80804 Munich, Germany.
| | - Michael Ziller
- Max Planck Institute of Psychiatry, Translational Psychiatry, 80804 Munich, Germany.
| | - Dietmar Spengler
- Max Planck Institute of Psychiatry, Translational Psychiatry, 80804 Munich, Germany.
| | - Anke Hoffmann
- Max Planck Institute of Psychiatry, Translational Psychiatry, 80804 Munich, Germany.
| |
Collapse
|
42
|
Wesselink E, Koekkoek WAC, Grefte S, Witkamp RF, van Zanten ARH. Feeding mitochondria: Potential role of nutritional components to improve critical illness convalescence. Clin Nutr 2018; 38:982-995. [PMID: 30201141 DOI: 10.1016/j.clnu.2018.08.032] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 08/03/2018] [Accepted: 08/25/2018] [Indexed: 12/30/2022]
Abstract
Persistent physical impairment is frequently encountered after critical illness. Recent data point towards mitochondrial dysfunction as an important determinant of this phenomenon. This narrative review provides a comprehensive overview of the present knowledge of mitochondrial function during and after critical illness and the role and potential therapeutic applications of specific micronutrients to restore mitochondrial function. Increased lactate levels and decreased mitochondrial ATP-production are common findings during critical illness and considered to be associated with decreased activity of muscle mitochondrial complexes in the electron transfer system. Adequate nutrient levels are essential for mitochondrial function as several specific micronutrients play crucial roles in energy metabolism and ATP-production. We have addressed the role of B vitamins, ascorbic acid, α-tocopherol, selenium, zinc, coenzyme Q10, caffeine, melatonin, carnitine, nitrate, lipoic acid and taurine in mitochondrial function. B vitamins and lipoic acid are essential in the tricarboxylic acid cycle, while selenium, α-tocopherol, Coenzyme Q10, caffeine, and melatonin are suggested to boost the electron transfer system function. Carnitine is essential for fatty acid beta-oxidation. Selenium is involved in mitochondrial biogenesis. Notwithstanding the documented importance of several nutritional components for optimal mitochondrial function, at present, there are no studies providing directions for optimal requirements during or after critical illness although deficiencies of these specific micronutrients involved in mitochondrial metabolism are common. Considering the interplay between these specific micronutrients, future research should pay more attention to their combined supply to provide guidance for use in clinical practise. REVISION NUMBER: YCLNU-D-17-01092R2.
Collapse
Affiliation(s)
- E Wesselink
- Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| | - W A C Koekkoek
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Willy Brandtlaan 10, 6716, Ede, The Netherlands.
| | - S Grefte
- Human and Animal Physiology, Wageningen University, De Elst 1, 6708 DW, Wageningen, The Netherlands.
| | - R F Witkamp
- Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| | - A R H van Zanten
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Willy Brandtlaan 10, 6716, Ede, The Netherlands.
| |
Collapse
|
43
|
Galano A, Reiter RJ. Melatonin and its metabolites vs oxidative stress: From individual actions to collective protection. J Pineal Res 2018; 65:e12514. [PMID: 29888508 DOI: 10.1111/jpi.12514] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/05/2018] [Indexed: 12/11/2022]
Abstract
Oxidative stress (OS) represents a threat to the chemical integrity of biomolecules including lipids, proteins, and DNA. The associated molecular damage frequently results in serious health issues, which justifies our concern about this phenomenon. In addition to enzymatic defense mechanisms, there are compounds (usually referred to as antioxidants) that offer chemical protection against oxidative events. Among them, melatonin and its metabolites constitute a particularly efficient chemical family. They offer protection against OS as individual chemical entities through a wide variety of mechanisms including electron transfer, hydrogen transfer, radical adduct formation, and metal chelation, and by repairing biological targets. In fact, many of them including melatonin can be classified as multipurpose antioxidants. However, what seems to be unique to the melatonin's family is their collective effects. Because the members of this family are metabolically related, most of them are expected to be present in living organisms wherever melatonin is produced. Therefore, the protection exerted by melatonin against OS may be viewed as a result of the combined antioxidant effects of the parent molecule and its metabolites. Melatonin's family is rather exceptional in this regard, offering versatile and collective antioxidant protection against OS. It certainly seems that melatonin is one of the best nature's defenses against oxidative damage.
Collapse
Affiliation(s)
- Annia Galano
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, México City, México
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, TX, USA
| |
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: 41] [Impact Index Per Article: 5.9] [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
|
Ali T, Rehman SU, Shah FA, Kim MO. Acute dose of melatonin via Nrf2 dependently prevents acute ethanol-induced neurotoxicity in the developing rodent brain. J Neuroinflammation 2018; 15:119. [PMID: 29679979 PMCID: PMC5911370 DOI: 10.1186/s12974-018-1157-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Melatonin is a well-known potent endogenous antioxidant pharmacological agent with significant neuroprotective actions. Here in the current study, we explored the nuclear factor erythroid 2-related factor 2 (Nrf2) gene-dependent antioxidant mechanism underlying the neuroprotective effects of the acute melatonin against acute ethanol-induced elevated reactive oxygen species (ROS)-mediated neuroinflammation and neurodegeneration in the developing rodent brain. METHODS In vivo rat pups were co-treated with a single dose of acute ethanol (5 g/kg, subcutaneous (S.C.)) and a single dose of acute melatonin (20 mg/kg, intraperitoneal (I.P.)). Four hours after a single S.C. and I.P. injections, all of the rat pups were sacrificed for further biochemical (Western blotting, ROS- assay, LPO-assay, and immunohistochemical) analyses. In order to corroborate the in vivo results, we used the in vitro murine-hippocampal HT22 and microglial BV2 cells, which were subjected to knockdown with small interfering RNA (siRNA) of Nrf2 genes and exposed with melatonin (100 μM) and ethanol (100 mM) and proceed for further biochemical analyses. RESULTS Our biochemical, immunohistochemical, and immunofluorescence results demonstrate that acute melatonin significantly upregulated the master endogenous antioxidant Nrf2 and heme oxygenase-1, consequently reversing the acute ethanol-induced elevated ROS and oxidative stress in the developing rodent brain, and in the murine-hippocampal HT22 and microglial BV2 cells. In addition, acute melatonin subsequently reduced the activated MAPK-p-P38-JNK pathways and attenuated neuroinflammation by decreasing the expression of activated gliosis and downregulated the p-NF-K-B/p-IKKβ pathway and decreased the expression levels of other inflammatory markers in the developing rodent brain and BV2 cells. Of note, melatonin acted through the Nrf2-dependent mechanism to attenuate neuronal apoptosis in the postnatal rodent brain and HT22 cells. Immunohistofluorescence results also showed that melatonin prevented ethanol-induced neurodegeneration in the developing rodent brain. The in vitro results indicated that melatonin induced neuroprotection via Nrf2-dependent manner and reduced ethanol-induced neurotoxicity. CONCLUSIONS The pleiotropic and potent neuroprotective antioxidant characteristics of melatonin, together with our in vivo and in vitro findings, suppose that acute melatonin could be beneficial to prevent and combat the acute ethanol-induced neurotoxic effects, such as elevated ROS, neuroinflammation, and neurodegeneration in the developing rodent brain.
Collapse
Affiliation(s)
- Tahir Ali
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Shafiq Ur Rehman
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Fawad Ali Shah
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Myeong Ok Kim
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea.
| |
Collapse
|
46
|
Brain tissue oxygen regulation in awake and anesthetized neonates. Neuropharmacology 2018; 135:368-375. [PMID: 29580952 DOI: 10.1016/j.neuropharm.2018.03.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 12/16/2022]
Abstract
Inhaled general anesthetics are used commonly in adults and children, and a growing body of literature from animals and humans suggests that exposure to anesthesia at an early age can impact brain development. While the origin of these effects is not well understood, it is known that anesthesia can disrupt oxygen regulation in the brain, which is critically important for maintaining healthy brain function. Here we investigated how anesthesia affected brain tissue oxygen regulation in neonatal rabbits by comparing brain tissue oxygen and single unit activity in the awake and anesthetized states. We tested two common general anesthetics, isoflurane and sevoflurane, delivered in both air and 80% oxygen. Our findings show that general anesthetics can greatly increase brain tissue PO2 in neonates, especially when combined with supplemental oxygen. Although isoflurane and sevoflurane belong to the same class of anesthetics, notable differences were observed in their effects upon neuronal activity and spontaneous respiration. Our findings point to the need to consider the potential effects of hyperoxia when supplemental oxygen is utilized, particularly in children and neonates.
Collapse
|
47
|
Phytochemicals in Human Milk and Their Potential Antioxidative Protection. Antioxidants (Basel) 2018; 7:antiox7020032. [PMID: 29470421 PMCID: PMC5836022 DOI: 10.3390/antiox7020032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/13/2018] [Accepted: 02/17/2018] [Indexed: 01/07/2023] Open
Abstract
Diets contain secondary plant metabolites commonly referred to as phytochemicals. Many of them are believed to impact human health through various mechanisms, including protection against oxidative stress and inflammation, and decreased risks of developing chronic diseases. For mothers and other people, phytochemical intake occurs through the consumption of foods such as fruits, vegetables, and grains. Research has shown that some these phytochemicals are present in the mother’s milk and can contribute to its oxidative stability. For infants, human milk (HM) represents the primary and preferred source of nutrition because it is a complete food. Studies have reported that the benefit provided by HM goes beyond basic nutrition. It can, for example, reduce oxidative stress in infants, thereby reducing the risk of lung and intestinal diseases in infants. This paper summarizes the phytochemicals present in HM and their potential contribution to infant health.
Collapse
|
48
|
Panfoli I, Candiano G, Malova M, De Angelis L, Cardiello V, Buonocore G, Ramenghi LA. Oxidative Stress as a Primary Risk Factor for Brain Damage in Preterm Newborns. Front Pediatr 2018; 6:369. [PMID: 30555809 PMCID: PMC6281966 DOI: 10.3389/fped.2018.00369] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/12/2018] [Indexed: 12/28/2022] Open
Abstract
The risk of oxidative stress is high in preterm newborns. Room air exposure of an organism primed to develop in a hypoxic environment, lacking antioxidant defenses, and subjected to hyperoxia, hypoxia, and ischemia challenges the newborn with oxidative stress production. Free radicals can be generated by a multitude of other mechanisms, such as glutamate excitotoxicity, excess free iron, inflammation, and immune reactions. Free radical-induced damage caused by oxidative stress appears to be the major candidate for the pathogenesis of most of the complications of prematurity, brain being especially at risk, with short to long-term consequences. We review the role of free radical oxidative damage to the newborn brain and propose a mechanism of oxidative injury, taking into consideration the particular maturation-dependent vulnerability of the oligodendrocyte precursors. Prompted by our observation of an increase in plasma Adenosine concentrations significantly associated with brain white matter lesions in some premature infants, we discuss a possible bioenergetics hypothesis, correlated to the oxidative challenge of the premature infant. We aim at explaining both the oxidative stress generation and the mechanism promoting the myelination disturbances. Being white matter abnormalities among the most common lesions of prematurity, the use of Adenosine as a biomarker of brain damage appears promising in order to design neuroprotective strategies.
Collapse
Affiliation(s)
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Mariya Malova
- Neonatal Intensive Care Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Laura De Angelis
- Neonatal Intensive Care Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Valentina Cardiello
- Neonatal Intensive Care Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Luca A Ramenghi
- Neonatal Intensive Care Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| |
Collapse
|
49
|
Sinha B, Wu Q, Li W, Tu Y, Sirianni AC, Chen Y, Jiang J, Zhang X, Chen W, Zhou S, Reiter RJ, Manning SM, Patel NJ, Aziz-Sultan AM, Inder TE, Friedlander RM, Fu J, Wang X. Protection of melatonin in experimental models of newborn hypoxic-ischemic brain injury through MT1 receptor. J Pineal Res 2018; 64. [PMID: 28796402 DOI: 10.1111/jpi.12443] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 08/04/2017] [Indexed: 12/19/2022]
Abstract
The function of melatonin as a protective agent against newborn hypoxic-ischemic (H-I) brain injury is not yet well studied, and the mechanisms by which melatonin causes neuroprotection in neurological diseases are still evolving. This study was designed to investigate whether expression of MT1 receptors is reduced in newborn H-I brain injury and whether the protective action of melatonin is by alterations of the MT1 receptors. We demonstrated that there was significant reduction in MT1 receptors in ischemic brain of mouse pups in vivo following H-I brain injury and that melatonin offers neuroprotection through upregulation of MT1 receptors. The role of MT1 receptors was further supported by observation of increased mortality in MT1 knockout mice following H-I brain injury and the reversal of the inhibitory role of melatonin on mitochondrial cell death pathways by the melatonin receptor antagonist, luzindole. These data demonstrate that melatonin mediates its neuroprotective effect in mouse models of newborn H-I brain injury, at least in part, by the restoration of MT1 receptors, the inhibition of mitochondrial cell death pathways and the suppression of astrocytic and microglial activation.
Collapse
MESH Headings
- Animals
- Astrocytes/cytology
- Blotting, Western
- Cells, Cultured
- Female
- Genotype
- Hippocampus/cytology
- Hypoxia-Ischemia, Brain/drug therapy
- Hypoxia-Ischemia, Brain/metabolism
- Immunohistochemistry
- Male
- Melatonin/therapeutic use
- Membrane Potentials/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Theoretical
- Receptor, Melatonin, MT1/genetics
- Receptor, Melatonin, MT1/metabolism
Collapse
Affiliation(s)
- Bharati Sinha
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Division of Neonatology, Boston University School of Medicine, Boston, MA, USA
| | - Qiaofeng Wu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Li
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yanyang Tu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana C Sirianni
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yanchun Chen
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Jiying Jiang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Xinmu Zhang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wu Chen
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Clinical Laboratory, Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University Texas Health Science Center, San Antonio, TX, USA
| | - Simon M Manning
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nirav J Patel
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ali M Aziz-Sultan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Terrie E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert M Friedlander
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jianfang Fu
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- The Joslin Beth Israel Deaconess Foot Center, Harvard Medical School, Boston, MA, USA
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
50
|
Hobson SR, Lim R, Wallace EM. Phase I Pilot Clinical Trial of Antenatal Maternally Administered Melatonin to Decrease the Level of Oxidative Stress in Human Pregnancies Affected by Preeclampsia. Methods Mol Biol 2018; 1710:335-345. [PMID: 29197016 DOI: 10.1007/978-1-4939-7498-6_27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
This chapter describes the methodologies which may be used in the development of a phase I clinical trial investigating a therapy of choice in treating preeclampsia.
Collapse
Affiliation(s)
- Sebastian R Hobson
- Maternal Fetal Medicine Unit, Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash Health & Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia.
| | - Rebecca Lim
- Maternal Fetal Medicine Unit, Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash Health & Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
| | - Euan M Wallace
- Maternal Fetal Medicine Unit, Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash Health & Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
| |
Collapse
|