1
|
Smith-Ryan AE, DelBiondo GM, Brown AF, Kleiner SM, Tran NT, Ellery SJ. Creatine in women's health: bridging the gap from menstruation through pregnancy to menopause. J Int Soc Sports Nutr 2025; 22:2502094. [PMID: 40371844 PMCID: PMC12086928 DOI: 10.1080/15502783.2025.2502094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Creatine supplementation in women has gained attention for its potential benefits beyond muscle growth, including reproductive health, cognitive health and aging. Women exhibit distinct physiological differences from men, influenced by hormonal fluctuations during pre-menopause, pregnancy, and menopause, and these factors should be considered for their influence on creatine metabolism. OBJECTIVE This review aims to provide a historical evaluation of creatine supplementation in women, its potential applications across female-specific life stages, recent research highlights, and targets for future research. The review also considers the impact of hormonal changes on creatine metabolism and effectiveness as a dietary supplementation. METHODS This is a narrative overview of historical and recent research evaluating the effects of creatine in women. RESULTS Early studies demonstrated the benefits of creatine on exercise performance in women, though they often overlooked menstrual cycle variability. Recent research has begun to account for these hormonal fluctuations, enhancing the understanding of creatine's applications. Creatine supplementation has shown positive effects on muscle strength, exercise performance, and body composition, particularly when combined with resistance training. Additionally, creatine may improve mood and cognitive function, potentially alleviating symptoms of depression. Emerging evidence suggests creatine's benefits during pregnancy and post-menopause, though data on perimenopausal women remains limited. CONCLUSION Creatine supplementation presents a promising strategy for enhancing various aspects of women's health across the lifespan. Future research should focus on optimizing dosing strategies, understanding long-term health implications, and exploring creatine's effects during pregnancy and perimenopause.
Collapse
Affiliation(s)
- Abbie E. Smith-Ryan
- University of North Carolina at Chapel Hill, Applied Physiology Laboratory, Department of Exercise and Sport Science, Chapel Hill, NC, USA
| | - Gabrielle M. DelBiondo
- University of North Carolina at Chapel Hill, Applied Physiology Laboratory, Department of Exercise and Sport Science, Chapel Hill, NC, USA
| | - Ann F. Brown
- University of Idaho, Human Performance Laboratory, Exercise, Sport and Health Sciences, Moscow, ID, USA
| | | | - Nhi T. Tran
- Hudson Institute of Medical Research, The Ritchie Centre, Clayton, Australia
- Monash University, Department of Obstetrics and Gynecology, Clayton, Australia
| | - Stacey J. Ellery
- Hudson Institute of Medical Research, The Ritchie Centre, Clayton, Australia
- Monash University, Department of Obstetrics and Gynecology, Clayton, Australia
| |
Collapse
|
2
|
Tran NT, Ellery SJ, Kelly SB, Sévigny J, Chatton M, Lu H, Polglase GR, Snow RJ, Walker DW, Galinsky R. Prophylactic Fetal Creatine Supplementation Improves Post-Asphyxial EEG Recovery and Reduces Seizures in Fetal Sheep: Implications for Hypoxic-Ischemic Encephalopathy. Ann Neurol 2025; 97:673-687. [PMID: 39644170 PMCID: PMC11889532 DOI: 10.1002/ana.27150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE Hypoxic-ischemic encephalopathy (HIE) is a major cause of perinatal brain injury. Creatine is a dietary supplement that can increase intracellular phosphocreatine to improve the provision of intracellular adenosine triphosphate (ATP) to meet the increase in metabolic demand of oxygen deprivation. Here, we assessed prophylactic fetal creatine supplementation in reducing acute asphyxia-induced seizures, disordered electroencephalography (EEG) activity and cerebral inflammation and cell death histopathology. METHODS Fetal sheep (118 ± 1 days' gestational age [dGA]; 0.8 gestation) were implanted with electrodes to continuously record EEG and nuchal electromyogram activity. At 121 dGA, fetuses were randomly assigned to sham control (i.v. saline infusion without umbilical cord occlusion [UCO]; SalCon), continuous i.v. creatine infusion (6 mg/kg/h; CrUCO) or isovolumetric saline (SalUCO) followed by UCO at 128 ± 2 dGA that lasted until the mean arterial blood pressure reached 19 mmHg. Brain tissue was collected for histopathology after 72 hours of recovery. RESULTS Creatine supplementation had no effects on basal systemic or neurological physiology. UCO duration did not differ between CrUCO and SalUCO. After reperfusion, CrUCO fetuses had improved EEG power and frequency recovery and reduced electrographic seizure incidence (SalUCO, 86% vs CrUCO, 29%) and burden. At 72 hours after UCO, cell death in the cerebral cortex and astrogliosis in the periventricular white matter were reduced in CrUCO fetuses compared with SalUCO. INTERPRETATION Creatine supplementation reduced post-asphyxial seizures and improved EEG recovery. Improvements in functional recovery with creatine were associated with regional reductions in cell death and astrogliosis. Prophylactic creatine treatment has the potential to mitigate functional indices of HIE in the late gestation fetal brain. ANN NEUROL 2025;97:673-687.
Collapse
Affiliation(s)
- Nhi T. Tran
- The Ritchie Centre, Hudson Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Obstetrics and GynecologyMonash UniversityMelbourneVictoriaAustralia
| | - Stacey J. Ellery
- The Ritchie Centre, Hudson Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Obstetrics and GynecologyMonash UniversityMelbourneVictoriaAustralia
| | - Sharmony B. Kelly
- The Ritchie Centre, Hudson Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Obstetrics and GynecologyMonash UniversityMelbourneVictoriaAustralia
| | - Juliane Sévigny
- The Ritchie Centre, Hudson Institute of Medical ResearchMelbourneVictoriaAustralia
- University of SherbrookeQuebecCanada
| | - Madeleine Chatton
- The Ritchie Centre, Hudson Institute of Medical ResearchMelbourneVictoriaAustralia
| | - Hui Lu
- The Ritchie Centre, Hudson Institute of Medical ResearchMelbourneVictoriaAustralia
| | - Graeme R. Polglase
- The Ritchie Centre, Hudson Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of PediatricsMonash UniversityMelbourneVictoriaAustralia
| | - Rod J. Snow
- Institute for Physical Activity and NutritionDeakin UniversityMelbourneVictoriaAustralia
| | - David W. Walker
- The Ritchie Centre, Hudson Institute of Medical ResearchMelbourneVictoriaAustralia
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Obstetrics and GynecologyMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
3
|
Choi YJ, Williams E, Dahl MJ, Amos SE, James C, Bautista AP, Kurup V, Musk GC, Kershaw H, Arthur PG, Kicic A, Choi YS, Terrill JR, Pillow JJ. Antenatal creatine supplementation reduces persistent fetal lung inflammation and oxidative stress in an ovine model of chorioamnionitis. Am J Physiol Lung Cell Mol Physiol 2024; 327:L40-L53. [PMID: 38712443 DOI: 10.1152/ajplung.00241.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024] Open
Abstract
Chorioamnionitis is a common antecedent of preterm birth and induces inflammation and oxidative stress in the fetal lungs. Reducing inflammation and oxidative stress in the fetal lungs may improve respiratory outcomes in preterm infants. Creatine is an organic acid with known anti-inflammatory and antioxidant properties. The objective of the study was to evaluate the efficacy of direct fetal creatine supplementation to reduce inflammation and oxidative stress in fetal lungs arising from an in utero proinflammatory stimulus. Fetal lambs (n = 51) were instrumented at 90 days gestation to receive a continuous infusion of creatine monohydrate (6 mg·kg-1·h-1) or saline for 17 days. Maternal chorioamnionitis was induced with intra-amniotic lipopolysaccharide (LPS; 1 mg, O55:H6) or saline 7 days before delivery at 110 days gestation. Tissue creatine content was assessed with capillary electrophoresis, and inflammatory markers were analyzed with Luminex Magpix and immunohistochemistry. Oxidative stress was measured as the level of protein thiol oxidation. The effects of LPS and creatine were analyzed using a two-way ANOVA. Fetal creatine supplementation increased lung creatine content by 149% (PCr < 0.0001) and had no adverse effects on lung morphology. LPS-exposed groups showed increased levels of interleukin-8 in the bronchoalveolar lavage (PLPS < 0.0001) and increased levels of CD45+ leukocytes (PLPS < 0.0001) and MPO+ (PLPS < 0.0001) cells in the lung parenchyma. Creatine supplementation significantly reduced the levels of CD45+ (PCr = 0.045) and MPO+ cells (PCr = 0.012) in the lungs and reduced thiol oxidation in plasma (PCr < 0.01) and lung tissue (PCr = 0.02). In conclusion, fetal creatine supplementation reduced markers of inflammation and oxidative stress in the fetal lungs arising from chorioamnionitis.NEW & NOTEWORTHY We evaluated the effect of antenatal creatine supplementation to reduce pulmonary inflammation and oxidative stress in the fetal lamb lungs arising from lipopolysaccharide (LPS)-induced chorioamnionitis. Fetal creatine supplementation increased lung creatine content and had no adverse effects on systemic fetal physiology and overall lung architecture. Importantly, fetuses that received creatine had significantly lower levels of inflammation and oxidative stress in the lungs, suggesting an anti-inflammatory and antioxidant benefit of creatine.
Collapse
Affiliation(s)
- Y Jane Choi
- School of Human Sciences, University of Western Australia, Perth, Western Australia, Australia
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Ellen Williams
- School of Human Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Mar Janna Dahl
- School of Human Sciences, University of Western Australia, Perth, Western Australia, Australia
- Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Sebastian E Amos
- School of Human Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Christopher James
- School of Molecular Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Angelo P Bautista
- School of Molecular Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Veena Kurup
- School of Human Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Gabrielle C Musk
- School of Human Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Helen Kershaw
- Animal Care Services, University of Western Australia, Perth, Western Australia, Australia
| | - Peter G Arthur
- School of Molecular Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
- School of Population Health, Curtin University, Perth, Western Australia, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Perth, Western Australia, Australia
- Center for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
| | - Yu Suk Choi
- School of Human Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Jessica R Terrill
- School of Molecular Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - J Jane Pillow
- School of Human Sciences, University of Western Australia, Perth, Western Australia, Australia
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
| |
Collapse
|
4
|
Nasri H, Ghotbeddin Z, Rahimi K, Tabandeh MR. The effects of MEPaL on oxidative stress and motor function in the rats affected by prenatal hypoxia. Brain Behav 2024; 14:e3539. [PMID: 38849974 PMCID: PMC11161390 DOI: 10.1002/brb3.3539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/23/2024] [Accepted: 04/19/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Maternal hypoxia disrupts neural development and subsequently leads to cerebral palsy and epilepsy in newborns. Hypoxia plays a role in neurodegeneration by increasing oxidative stress. Pistacia atlantica is known as an important antioxidant, and its anti-inflammatory and antioxidant effects have been shown in various studies. This study aims to investigate the effects of methanolic extract of P. atlantica leaves (MEPaLs) on the oxidative parameters in the serum of rats affected by maternal hypoxia. MATERIAL AND METHODS In this study, eight pregnant rats were used. The newborns were divided into four groups, including the control and the hypoxia groups, which are affected by maternal hypoxia, hypoxia + MEPaL 100 mg/kg, and hypoxia + MEPaL 150 mg/kg. MEPaL was injected (i.p) for 21 days into the neonatal rats after the lactation period. Hypoxia was induced by keeping pregnant rats in a hypoxic chamber with 7% oxygen and 93% nitrogen intensity for 3 h on the 20th day of pregnancy. Behavioral changes were measured using open-field and rotarod tests. Finally, biomarkers of oxidative stress, nitric oxide (NO), glutathione (GSH), GSSG, TAS, TOS, and oxidative stress index (OSI) were measured in the experimental groups. RESULTS Behavioral results showed that the anxiety behavior in the hypoxia group increased, but the motor activity (moved distance and movement speed) decreased. Moreover, the amount of time spent maintaining balance on the rotarod rod was significantly decreased in the hypoxia group. The concentration of NO in the group of hypoxia + MEPaL 100 mg/kg showed a significant decrease, and MEPaL 100, and 150 mg/kg + hypoxia also increased the concentration of GSH and decreased GSSG. In addition, MEPaL100 and 150 mg/kg caused a significant increase in the ratio of GSH to GSSG and decreased OSI and total oxidant capacity. CONCLUSIONS Oxidative stress increased in the rats affected by maternal hypoxia and may be the main mechanism for motor activity impairment and balance disturbance, whereas MELaL improved motor performance by decreasing oxidative stress.
Collapse
Affiliation(s)
- Hadis Nasri
- Department of Basic Sciences, Faculty of Veterinary MedicineShahid Chamran University of AhvazAhvazIran
| | - Zohreh Ghotbeddin
- Department of Basic Sciences, Faculty of Veterinary MedicineShahid Chamran University of AhvazAhvazIran
- Stem Cell and Transgenic Technology Research CenterShahid Chamran University of AhvazAhvazIran
| | - Kaveh Rahimi
- Department of Basic Sciences, Faculty of Veterinary MedicineShahid Chamran University of AhvazAhvazIran
| | - Mohammad Reza Tabandeh
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary MedicineShahid Chamran University of AhvazAhvazIran
- Stem Cell and Transgenic Technology Research CenterShahid Chamran University of AhvazAhvazIran
| |
Collapse
|
5
|
Davidson JO, Gunn AJ, Dean JM. Back to the beginning: can we stop brain injury before it starts? J Physiol 2022; 600:3013-3014. [PMID: 35652911 PMCID: PMC9328369 DOI: 10.1113/jp283330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Joanne O Davidson
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, New Zealand
| | - Justin M Dean
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, New Zealand
| |
Collapse
|
6
|
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: 10] [Impact Index Per Article: 3.3] [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
|
7
|
Seifert AW, Temple-Smith P. A remarkable rodent: Regeneration and reproduction in spiny mice (Acomys). Curr Top Dev Biol 2022; 147:659-707. [PMID: 35337466 DOI: 10.1016/bs.ctdb.2021.12.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although certain organisms are chosen and employed to better understand a specific problem in biology (so-called model organisms), sometimes an animal model reveals its' biomedical importance by happenstance. In many ways, the advent of spiny mice (Acomys) as an emerging model to study regeneration and menstruation stands as a case study in scientific pseudoserendipity (Diaz de Chumaceiro, 1995). As we recount in this chapter, the discovery of these phenotypes, while not entirely accidental, was nonetheless unexpected. In addition to recounting how we uncovered these unusual mammalian traits, we outline recent work by our groups and others that has begun to outline the cellular and genetic mechanisms underlying bonafide mammalian tissue regeneration and a human-like mode of reproduction in spiny mice.
Collapse
Affiliation(s)
- Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, KY, United States; Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya.
| | - Peter Temple-Smith
- Department of Obstetrics & Gynecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
8
|
McKenna J, Bellofiore N, Temple-Smith P. Good things come to those who mate: analysis of the mating behaviour in the menstruating rodent, Acomys cahirinus. BMC ZOOL 2022; 7:13. [PMID: 37170145 PMCID: PMC10127372 DOI: 10.1186/s40850-022-00112-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/14/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The Egyptian spiny mouse (Acomys cahirinus) is the only known rodent to exhibit true, human-like menstruation and postpartum ovulation, and is an important new model for reproductive studies. Spiny mice do not produce a visible copulatory plug, and calculation of gestational age is therefore restricted by the need to use mated postpartum dams. The current inefficient method of monitoring until parturition to provide a subsequent estimate of gestational age increases study duration and costs. This study addressed this issue by comparing the mating behaviour of spiny mice across the menstrual cycle and proposes a more accurate method for staging and pairing animals that provides reliable estimates of gestational age. In experiment 1, mating behaviour was recorded overnight to collect data on mounting, intromission, and ejaculation (n = 5 pairs per stage) in spiny mice paired at menses and at early and late follicular and luteal phases of the menstrual cycle. In experiment 2, female spiny mice were paired at the follicular or luteal phases of the menstrual cycle to determine any effect on the pairing-birth interval (n = 10 pairs).
Results
We report a broad mating window of ~ 3 days during the follicular phase and early luteal phase of spiny mice. Males displayed a discrete ‘foot twitch’ behaviour during intromission and a brief copulatory lock during ejaculation. Litters were delivered after 40–43 days if pairing occurred during the mating window, compared with 46–48 days for spiny mice paired in the late luteal phase. When pairing occurred during the late luteal phase or menses no mating activity was observed during the recording period.
Conclusion
This study clearly demonstrates an effect of the menstrual cycle on mating behaviour and pregnancy in the spiny mouse and provides a reliable and more effective protocol for estimating gestational age without the need for postpartum dams.
Collapse
|
9
|
The Effects of In Utero Fetal Hypoxia and Creatine Treatment on Mitochondrial Function in the Late Gestation Fetal Sheep Brain. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3255296. [PMID: 35132347 PMCID: PMC8817846 DOI: 10.1155/2022/3255296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/13/2021] [Accepted: 01/05/2022] [Indexed: 12/21/2022]
Abstract
Near-term acute hypoxia in utero can result in significant fetal brain injury, with some brain regions more vulnerable than others. As mitochondrial dysfunction is an underlying feature of the injury cascade following hypoxia, this study is aimed at characterizing mitochondrial function at a region-specific level in the near-term fetal brain after a period of acute hypoxia. We hypothesized that regional differences in mitochondrial function would be evident, and that prophylactic creatine treatment would mitigate mitochondrial dysfunction following hypoxia; thereby reducing fetal brain injury. Pregnant Border-Leicester/Merino ewes with singleton fetuses were surgically instrumented at 118 days of gestation (dGa; term is ~145 dGA). A continuous infusion of either creatine (n = 15; 6 mg/kg/h) or isovolumetric saline (n = 16; 1.5 ml/kg/h) was administered to the fetuses from 121 dGa. After 10 days of infusion, a subset of fetuses (8 saline-, 7 creatine-treated) were subjected to 10 minutes of umbilical cord occlusion (UCO) to induce a mild global fetal hypoxia. At 72 hours after UCO, the fetal brain was collected for high-resolution mitochondrial respirometry and molecular and histological analyses. The results show that the transient UCO-induced acute hypoxia impaired mitochondrial function in the hippocampus and the periventricular white matter and increased the incidence of cell death in the hippocampus. Creatine treatment did not rectify the changes in mitochondrial respiration associated with hypoxia, but there was a negative relationship between cell death and creatine content following treatment. Irrespective of UCO, creatine increased the proportion of cytochrome c bound to the inner mitochondrial membrane, upregulated the mRNA expression of the antiapoptotic gene Bcl2, and of PCG1-α, a driver of mitogenesis, in the hippocampus. We conclude that creatine treatment prior to brief, acute hypoxia does not fundamentally modify mitochondrial respiratory function, but may improve mitochondrial structural integrity and potentially increase mitogenesis and activity of antiapoptotic pathways.
Collapse
|
10
|
OUP accepted manuscript. Nutr Rev 2022; 80:2136-2153. [DOI: 10.1093/nutrit/nuac028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
11
|
Lespay-Rebolledo C, Tapia-Bustos A, Perez-Lobos R, Vio V, Casanova-Ortiz E, Farfan-Troncoso N, Zamorano-Cataldo M, Redel-Villarroel M, Ezquer F, Quintanilla ME, Israel Y, Morales P, Herrera-Marschitz M. Sustained Energy Deficit Following Perinatal Asphyxia: A Shift towards the Fructose-2,6-bisphosphatase (TIGAR)-Dependent Pentose Phosphate Pathway and Postnatal Development. Antioxidants (Basel) 2021; 11:74. [PMID: 35052577 PMCID: PMC8773255 DOI: 10.3390/antiox11010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Labor and delivery entail a complex and sequential metabolic and physiologic cascade, culminating in most circumstances in successful childbirth, although delivery can be a risky episode if oxygen supply is interrupted, resulting in perinatal asphyxia (PA). PA causes an energy failure, leading to cell dysfunction and death if re-oxygenation is not promptly restored. PA is associated with long-term effects, challenging the ability of the brain to cope with stressors occurring along with life. We review here relevant targets responsible for metabolic cascades linked to neurodevelopmental impairments, that we have identified with a model of global PA in rats. Severe PA induces a sustained effect on redox homeostasis, increasing oxidative stress, decreasing metabolic and tissue antioxidant capacity in vulnerable brain regions, which remains weeks after the insult. Catalase activity is decreased in mesencephalon and hippocampus from PA-exposed (AS), compared to control neonates (CS), in parallel with increased cleaved caspase-3 levels, associated with decreased glutathione reductase and glutathione peroxidase activity, a shift towards the TIGAR-dependent pentose phosphate pathway, and delayed calpain-dependent cell death. The brain damage continues long after the re-oxygenation period, extending for weeks after PA, affecting neurons and glial cells, including myelination in grey and white matter. The resulting vulnerability was investigated with organotypic cultures built from AS and CS rat newborns, showing that substantia nigra TH-dopamine-positive cells from AS were more vulnerable to 1 mM of H2O2 than those from CS animals. Several therapeutic strategies are discussed, including hypothermia; N-acetylcysteine; memantine; nicotinamide, and intranasally administered mesenchymal stem cell secretomes, promising clinical translation.
Collapse
Affiliation(s)
- Carolyne Lespay-Rebolledo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Andrea Tapia-Bustos
- School of Pharmacy, Faculty of Medicine, Universidad Andres Bello, Santiago 8370149, Chile;
| | - Ronald Perez-Lobos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Valentina Vio
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Emmanuel Casanova-Ortiz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Nancy Farfan-Troncoso
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Marta Zamorano-Cataldo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Martina Redel-Villarroel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Maria Elena Quintanilla
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Yedy Israel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| |
Collapse
|
12
|
Tran NT, Kelly SB, Snow RJ, Walker DW, Ellery SJ, Galinsky R. Assessing Creatine Supplementation for Neuroprotection against Perinatal Hypoxic-Ischaemic Encephalopathy: A Systematic Review of Perinatal and Adult Pre-Clinical Studies. Cells 2021; 10:2902. [PMID: 34831126 PMCID: PMC8616304 DOI: 10.3390/cells10112902] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/09/2022] Open
Abstract
There is an important unmet need to develop interventions that improve outcomes of hypoxic-ischaemic encephalopathy (HIE). Creatine has emerged as a promising neuroprotective agent. Our objective was to systematically evaluate the preclinical animal studies that used creatine for perinatal neuroprotection, and to identify knowledge gaps that need to be addressed before creatine can be considered for pragmatic clinical trials for HIE. METHODS We reviewed preclinical studies up to 20 September 2021 using PubMed, EMBASE and OVID MEDLINE databases. The SYRCLE risk of bias assessment tool was utilized. RESULTS Seventeen studies were identified. Dietary creatine was the most common administration route. Cerebral creatine loading was age-dependent with near term/term-equivalent studies reporting higher increases in creatine/phosphocreatine compared to adolescent-adult equivalent studies. Most studies did not control for sex, study long-term histological and functional outcomes, or test creatine post-HI. None of the perinatal studies that suggested benefit directly controlled core body temperature (a known confounder) and many did not clearly state controlling for potential study bias. CONCLUSION Creatine is a promising neuroprotective intervention for HIE. However, this systematic review reveals key knowledge gaps and improvements to preclinical studies that must be addressed before creatine can be trailed for neuroprotection of the human fetus/neonate.
Collapse
Affiliation(s)
- Nhi Thao Tran
- School of Health & Biomedical Sciences, STEM College, RMIT University, Melbourne 3083, Australia; (N.T.T.); (D.W.W.)
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia; (S.B.K.); (S.J.E.)
| | - Sharmony B. Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia; (S.B.K.); (S.J.E.)
- Department of Obstetrics & Gynecology, Monash University, Melbourne 3168, Australia
| | - Rod J. Snow
- Institute for Physical Activity & Nutrition, Deakin University, Melbourne 3125, Australia;
| | - David W. Walker
- School of Health & Biomedical Sciences, STEM College, RMIT University, Melbourne 3083, Australia; (N.T.T.); (D.W.W.)
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia; (S.B.K.); (S.J.E.)
| | - Stacey J. Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia; (S.B.K.); (S.J.E.)
- Department of Obstetrics & Gynecology, Monash University, Melbourne 3168, Australia
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia; (S.B.K.); (S.J.E.)
- Department of Obstetrics & Gynecology, Monash University, Melbourne 3168, Australia
| |
Collapse
|
13
|
Tran NT, Muccini AM, Snow RJ, Nitsos I, Hale N, Walker DW, Ellery SJ. The physiological effects of creatine supplementation in fetal sheep before, during and after umbilical cord occlusion and global hypoxia. J Appl Physiol (1985) 2021; 131:1088-1099. [PMID: 34382841 DOI: 10.1152/japplphysiol.00092.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to investigate the effects of direct creatine infusion on fetal systemic metabolic and cardiovascular responses to mild acute in utero hypoxia. Pregnant ewes (n=28) were surgically instrumented at 118 days gestation (dGa). A constant intravenous infusion of creatine (6 mg.kg-1.h-1) or isovolumetric saline (1.5 ml.h-1) began at 121 dGa. After 10 days, fetuses were subjected to 10-minute umbilical cord occlusion (UCO) to induce mild global hypoxia (saline-UCO, n=8; creatine-UCO, n=7) or sham UCO (saline-control, n=6; creatine-control, n=7). Cardiovascular, arterial blood gases and metabolites, and plasma creatine were monitored prior to, during, and then for 72 hours following the UCO. Total creatine content in discrete fetal brain regions was also measured. Fetal creatine infusion increased plasma concentrations 5-fold but had no significant effects on any measurement pre-UCO. Creatine did not alter fetal physiology during the UCO or in the early recovery stage, up to 24 hours after UCO. During the late recovery stage, 24-72 hours after UCO, there was a significant reduction in the arterial oxygen pressure and saturation in creatine fetuses (PUCO x TREATMENT = 0.02 and0.04, respectively). At 72 hours after UCO, significant creatine loading was detected in cortical grey matter, hippocampus, thalamus and striatum (PTREATMENT = 0.01-0.001). In the striatum, the UCO itself increased total creatine content (PUCO = 0.019). Overall, fetal creatine supplementation may alter oxygen flux following an acute hypoxic insult. Increasing total creatine content in the striatum may also be a fetal adaptation to acute oxygen deprivation.
Collapse
Affiliation(s)
- Nhi Thao Tran
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Faculty of Health Science, RMIT University, Melbourne, Victoria, Australia
| | - Anna M Muccini
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Rod J Snow
- Institute for Physical Activity and Nutrition, Deakin University, Burwood, Melbourne, Victoria, Australia
| | - Ilias Nitsos
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Nadia Hale
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - David W Walker
- Faculty of Health Science, RMIT University, Melbourne, Victoria, Australia
| | - Stacey J Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Stevens S, Mohan S. Opioid withdrawal behavior in spiny mice: A novel preclinical model of neonatal opioid withdrawal syndrome (NOWS). Heliyon 2021; 7:e06694. [PMID: 33898824 PMCID: PMC8056230 DOI: 10.1016/j.heliyon.2021.e06694] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 11/15/2022] Open
Abstract
As the opioid epidemic continues to grow, opioid use among pregnant women is increasing significantly. This has led to a steady rise in the number of infants born with neonatal opioid withdrawal syndrome (NOWS). Although short-term withdrawal symptoms associated with NOWS are well characterized, there are many gaps in our understanding of the short and long-term effects of prenatal opioid exposure. Current animal models of NOWS are limited by shortened gestational periods, large litter sizes, and primary organogenesis occurring after birth. This often leads to postnatal treatment to mimic drug exposure during third-trimester development. Using the unique rodent species Acomys cahirinus, more commonly known as spiny mice, which have an extended 40-day gestation period, small litter sizes, and increased in utero organogenesis we aim to study the short-term effects of prenatal morphine exposure by assessing withdrawal behavior. To model maternal opioid use, dams were treated daily with morphine (10 and 30 mg/kg S.C.) beginning on gestation day 19 until the day of birth; this resulted in a cumulative exposure of 19-21 days. Withdrawal behaviors for each pup were recorded daily between postnatal days 0-7 (PND 0-7). Our study found that prenatal morphine exposure in spiny mice led to an increase in withdrawal behavior throughout the early postnatal period and validated the use of this species as a novel pre-clinical model of NOWS. We are hopeful this rodent model will further our understanding of the short and long-term consequences of prenatal opioid exposure on neurodevelopment and behavior.
Collapse
Affiliation(s)
- Sarah Stevens
- Department of Pharmaceutical Science and Research, Marshall University, School of Pharmacy, Huntington, WV 25701, USA
| | - Shekher Mohan
- Department of Pharmaceutical Sciences, Manchester University, College of Pharmacy, Fort Wayne, IN 46845, USA.,Department of Integrative Physiology and Pharmacology, Liberty University, College of Osteopathic Medicine, Lynchburg, VA 24502, USA
| |
Collapse
|
15
|
β-Hydroxy-β-Methylbutyrate (HMB) Supplementation Prevents Bone Loss during Pregnancy-Novel Evidence from a Spiny Mouse ( Acomys cahirinus) Model. Int J Mol Sci 2021; 22:ijms22063047. [PMID: 33802646 PMCID: PMC8002460 DOI: 10.3390/ijms22063047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/23/2022] Open
Abstract
The aim of this study was to determine the effects of ß-hydroxy-ß-methylbutyrate (HMB) supplementation during pregnancy on postpartum bone tissue quality by assessing changes in trabecular and compact bone as well as in hyaline and epiphyseal cartilage. The experiment was carried out on adult 6-month-old female spiny mice (Acomys cahirinus) divided into three groups: pregnant control (PregCont), pregnant HMB-treated (supplemented with 0.02 g/kg b.w of HMB during the second trimester of pregnancy, PregHMB), and non-pregnant females (NonPreg). Cross-sectional area and cortical index of the femoral mid-shaft, stiffness, and Young modulus were significantly greater in the PregHMB group. Whole-bone mineral density was similar in all groups, and HMB supplementation increased trabecular number. Growth plate cartilage was the thinnest, while the articular cartilage was the thickest in the PregHMB group. HMB supplementation increased the content of proteoglycans in the articular cartilage and the percentage of immature collagen content in metaphyseal trabeculae and compact bone. In summary, dietary HMB supplementation during the second trimester of pregnancy intensifies bone metabolic processes and prevents bone loss during pregnancy.
Collapse
|
16
|
Creatine Supplementation in Women's Health: A Lifespan Perspective. Nutrients 2021; 13:nu13030877. [PMID: 33800439 PMCID: PMC7998865 DOI: 10.3390/nu13030877] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
Despite extensive research on creatine, evidence for use among females is understudied. Creatine characteristics vary between males and females, with females exhibiting 70–80% lower endogenous creatine stores compared to males. Understanding creatine metabolism pre- and post-menopause yields important implications for creatine supplementation for performance and health among females. Due to the hormone-related changes to creatine kinetics and phosphocreatine resynthesis, supplementation may be particularly important during menses, pregnancy, post-partum, during and post-menopause. Creatine supplementation among pre-menopausal females appears to be effective for improving strength and exercise performance. Post-menopausal females may also experience benefits in skeletal muscle size and function when consuming high doses of creatine (0.3 g·kg−1·d−1); and favorable effects on bone when combined with resistance training. Pre-clinical and clinical evidence indicates positive effects from creatine supplementation on mood and cognition, possibly by restoring brain energy levels and homeostasis. Creatine supplementation may be even more effective for females by supporting a pro-energetic environment in the brain. The purpose of this review was to highlight the use of creatine in females across the lifespan with particular emphasis on performance, body composition, mood, and dosing strategies.
Collapse
|
17
|
Postpartum ovulation and early pregnancy in the menstruating spiny mouse, Acomys cahirinus. Sci Rep 2021; 11:5344. [PMID: 33674629 PMCID: PMC7935856 DOI: 10.1038/s41598-021-84361-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/16/2021] [Indexed: 11/09/2022] Open
Abstract
Egyptian spiny mice are the only known species to have human-like menstruation and a postpartum ovulation. Unfortunately, no endocrine or morphological evidence has been provided for a postpartum ovulation in spiny mice, and while later stages of pregnancy have been well studied, early events including embryo implantation and spiral artery remodelling have not been reported. This study compared the sex steroid endocrinology and reproductive tract morphology of dams at eight timepoints (n = 40) postpartum to determine the timing of ovulation and the timing and invasiveness of embryo implantation in A. cahirinus. Reproductive tracts were fixed and stained for histology and immunohistochemistry, and plasma was prepared for enzyme-linked immunosorbent assay. Ovarian histology and estradiol-17B concentrations indicate ovulation within 48 h of parturition and then immediate resumption of follicular growth. Uterine histology and immunohistochemistry revealed progressive epithelial repair, endometrial growth and spiral artery assembly and remodelling in dams postpartum. Blastocysts were seen in the uterine lumen at day 4-5 postpartum and embryos had implanted superficially with minimal stromal invasion by day 5-6. This study provides further evidence for the unique, humanesque reproductive biology of spiny mice and for a postpartum ovulation using endocrine and morphological changes observed during early pregnancy. Taken together, our data suggest that spiny mice may act as appropriate models of human pregnancy disorders such as implantation failure or pre-eclampsia.
Collapse
|
18
|
Reyes-Corral M, Sola-Idígora N, de la Puerta R, Montaner J, Ybot-González P. Nutraceuticals in the Prevention of Neonatal Hypoxia-Ischemia: A Comprehensive Review of their Neuroprotective Properties, Mechanisms of Action and Future Directions. Int J Mol Sci 2021; 22:2524. [PMID: 33802413 PMCID: PMC7959318 DOI: 10.3390/ijms22052524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/22/2022] Open
Abstract
Neonatal hypoxia-ischemia (HI) is a brain injury caused by oxygen deprivation to the brain due to birth asphyxia or reduced cerebral blood perfusion, and it often leads to lifelong limiting sequelae such as cerebral palsy, seizures, or mental retardation. HI remains one of the leading causes of neonatal mortality and morbidity worldwide, and current therapies are limited. Hypothermia has been successful in reducing mortality and some disabilities, but it is only applied to a subset of newborns that meet strict inclusion criteria. Given the unpredictable nature of the obstetric complications that contribute to neonatal HI, prophylactic treatments that prevent, rather than rescue, HI brain injury are emerging as a therapeutic alternative. Nutraceuticals are natural compounds present in the diet or used as dietary supplements that have antioxidant, anti-inflammatory, or antiapoptotic properties. This review summarizes the preclinical in vivo studies, mostly conducted on rodent models, that have investigated the neuroprotective properties of nutraceuticals in preventing and reducing HI-induced brain damage and cognitive impairments. The natural products reviewed include polyphenols, omega-3 fatty acids, vitamins, plant-derived compounds (tanshinones, sulforaphane, and capsaicin), and endogenous compounds (melatonin, carnitine, creatine, and lactate). These nutraceuticals were administered before the damage occurred, either to the mothers as a dietary supplement during pregnancy and/or lactation or to the pups prior to HI induction. To date, very few of these nutritional interventions have been investigated in humans, but we refer to those that have been successful in reducing ischemic stroke in adults. Overall, there is a robust body of preclinical evidence that supports the neuroprotective properties of nutraceuticals, and these may represent a safe and inexpensive nutritional strategy for the prevention of neonatal HI encephalopathy.
Collapse
Affiliation(s)
- Marta Reyes-Corral
- Neurodevelopment Research Group, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain; (M.R.-C.); (N.S.-I.); (P.Y.-G.)
| | - Noelia Sola-Idígora
- Neurodevelopment Research Group, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain; (M.R.-C.); (N.S.-I.); (P.Y.-G.)
| | - Rocío de la Puerta
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain;
| | - Joan Montaner
- Neurovascular Research Lab, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain
- Department of Neurology and Neurophysiology, Hospital Universitario Virgen Macarena, 41009 Seville, Spain
| | - Patricia Ybot-González
- Neurodevelopment Research Group, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain; (M.R.-C.); (N.S.-I.); (P.Y.-G.)
- Department of Neurology and Neurophysiology, Hospital Universitario Virgen Macarena, 41009 Seville, Spain
| |
Collapse
|
19
|
Antonio J, Candow DG, Forbes SC, Gualano B, Jagim AR, Kreider RB, Rawson ES, Smith-Ryan AE, VanDusseldorp TA, Willoughby DS, Ziegenfuss TN. Common questions and misconceptions about creatine supplementation: what does the scientific evidence really show? J Int Soc Sports Nutr 2021; 18:13. [PMID: 33557850 PMCID: PMC7871530 DOI: 10.1186/s12970-021-00412-w] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/28/2021] [Indexed: 01/01/2023] Open
Abstract
Supplementing with creatine is very popular amongst athletes and exercising individuals for improving muscle mass, performance and recovery. Accumulating evidence also suggests that creatine supplementation produces a variety of beneficial effects in older and patient populations. Furthermore, evidence-based research shows that creatine supplementation is relatively well tolerated, especially at recommended dosages (i.e. 3-5 g/day or 0.1 g/kg of body mass/day). Although there are over 500 peer-refereed publications involving creatine supplementation, it is somewhat surprising that questions regarding the efficacy and safety of creatine still remain. These include, but are not limited to: 1. Does creatine lead to water retention? 2. Is creatine an anabolic steroid? 3. Does creatine cause kidney damage/renal dysfunction? 4. Does creatine cause hair loss / baldness? 5. Does creatine lead to dehydration and muscle cramping? 6. Is creatine harmful for children and adolescents? 7. Does creatine increase fat mass? 8. Is a creatine 'loading-phase' required? 9. Is creatine beneficial for older adults? 10. Is creatine only useful for resistance / power type activities? 11. Is creatine only effective for males? 12. Are other forms of creatine similar or superior to monohydrate and is creatine stable in solutions/beverages? To answer these questions, an internationally renowned team of research experts was formed to perform an evidence-based scientific evaluation of the literature regarding creatine supplementation.
Collapse
Affiliation(s)
- Jose Antonio
- Department of Health and Human Performance, Nova Southeastern University, Davie, Florida, USA.
| | - Darren G Candow
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, Canada
| | - Scott C Forbes
- Department of Physical Education, Faculty of Education, Brandon University, Brandon, MB, Canada
| | - Bruno Gualano
- Applied Physiology & Nutrition Research Group; School of Medicine, FMUSP, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Andrew R Jagim
- Sports Medicine Department, Mayo Clinic Health System, La Crosse, WI, USA
| | - Richard B Kreider
- Exercise & Sport Nutrition Lab, Human Clinical Research Facility, Department of Health & Kinesiology, Texas A&M University, College Station, USA
| | - Eric S Rawson
- Department of Health, Nutrition, and Exercise Science, Messiah University, Mechanicsburg, PA, USA
| | - Abbie E Smith-Ryan
- Department of Exercise and Sport Science, University of North Carolina, Chapel Hill, NC, USA
| | - Trisha A VanDusseldorp
- Department of Exercise Science and Sport Management, Kennesaw State University, Kennesaw, GA, USA
| | - Darryn S Willoughby
- School of Exercise and Sport Science, University of Mary Hardin-Baylor, Belton, TX, USA
| | | |
Collapse
|
20
|
Creatine Metabolism in Female Reproduction, Pregnancy and Newborn Health. Nutrients 2021; 13:nu13020490. [PMID: 33540766 PMCID: PMC7912953 DOI: 10.3390/nu13020490] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 12/12/2022] Open
Abstract
Creatine metabolism is an important component of cellular energy homeostasis. Via the creatine kinase circuit, creatine derived from our diet or synthesized endogenously provides spatial and temporal maintenance of intracellular adenosine triphosphate (ATP) production; this is particularly important for cells with high or fluctuating energy demands. The use of this circuit by tissues within the female reproductive system, as well as the placenta and the developing fetus during pregnancy is apparent throughout the literature, with some studies linking perturbations in creatine metabolism to reduced fertility and poor pregnancy outcomes. Maternal dietary creatine supplementation during pregnancy as a safeguard against hypoxia-induced perinatal injury, particularly that of the brain, has also been widely studied in pre-clinical in vitro and small animal models. However, there is still no consensus on whether creatine is essential for successful reproduction. This review consolidates the available literature on creatine metabolism in female reproduction, pregnancy and the early neonatal period. Creatine metabolism is discussed in relation to cellular bioenergetics and de novo synthesis, as well as the potential to use dietary creatine in a reproductive setting. We highlight the apparent knowledge gaps and the research “road forward” to understand, and then utilize, creatine to improve reproductive health and perinatal outcomes.
Collapse
|
21
|
Kreider RB, Stout JR. Creatine in Health and Disease. Nutrients 2021; 13:nu13020447. [PMID: 33572884 PMCID: PMC7910963 DOI: 10.3390/nu13020447] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022] Open
Abstract
Although creatine has been mostly studied as an ergogenic aid for exercise, training, and sport, several health and potential therapeutic benefits have been reported. This is because creatine plays a critical role in cellular metabolism, particularly during metabolically stressed states, and limitations in the ability to transport and/or store creatine can impair metabolism. Moreover, increasing availability of creatine in tissue may enhance cellular metabolism and thereby lessen the severity of injury and/or disease conditions, particularly when oxygen availability is compromised. This systematic review assesses the peer-reviewed scientific and medical evidence related to creatine's role in promoting general health as we age and how creatine supplementation has been used as a nutritional strategy to help individuals recover from injury and/or manage chronic disease. Additionally, it provides reasonable conclusions about the role of creatine on health and disease based on current scientific evidence. Based on this analysis, it can be concluded that creatine supplementation has several health and therapeutic benefits throughout the lifespan.
Collapse
Affiliation(s)
- Richard B. Kreider
- Human Clinical Research Facility, Exercise & Sport Nutrition Lab, Department of Health & Kinesiology, Texas A&M University, College Station, TX 77843, USA
- Correspondence:
| | - Jeffery R. Stout
- Physiology of Work and Exercise Response (POWER) Laboratory, Institute of Exercise Physiology and Rehabilitation Science, School of Kinesiology and Physical Therapy, University of Central Florida, 12494 University Blvd., Orlando, FL 32816, USA;
| |
Collapse
|
22
|
A human-based assisted reproduction protocol for the menstruating spiny mouse, Acomys cahirinus. PLoS One 2020; 15:e0244411. [PMID: 33370773 PMCID: PMC7769615 DOI: 10.1371/journal.pone.0244411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/08/2020] [Indexed: 11/19/2022] Open
Abstract
The Egyptian or Common spiny mouse (A. cahirinus) is the first rodent species to show human-like menstruation and spontaneous decidualisation. We consider from these, and its other, human-like characteristics that this species will be a more useful and appropriate small animal model for human reproductive studies. Based on this, there is a need to develop specific laboratory-based assisted reproduction protocols including superovulation, in-vitro fertilisation, embryo cryopreservation and transfer to expand and make this model more relevant. Because standard rodent superovulation has not been successful in the spiny mouse, we have selected to test a human protocol. Female spiny mice will receive a subcutaneous GnRH agonist implant and be allowed to recover. Menstrual cycle lengths will then be allowed to stabilize prior to ovarian stimulation. After recovery, females will be injected IP once a day for 4 days with a FSH analogue, to induce follicular growth, and on day 5 will be injected IP with a hCG analogue to trigger ovulation. Females will either be culled 36hrs after trigger to collect oocytes or immediately paired with a stud male and two cell embryos collected 48hrs later. Mature oocytes will be inseminated using fresh spiny mouse spermatozoa and all in-vitro grown and in-vivo collected two cell embryos will be cryopreserved using methods developed in a close spiny mouse relative, the Mongolian gerbil. For embryo transfer, vitrified embryos will be rapidly warmed and non-surgically transferred to surrogate mice. Surrogates will be monitored until pregnancy is apparent (roughly 30 days) and then left undisturbed until birth, 38-40 days after transfer. By successfully developing robust assisted reproduction protocols in A. cahirinus we will be able to use this rodent as a more effective model for human reproduction.
Collapse
|
23
|
de Guingand DL, Palmer KR, Snow RJ, Davies-Tuck ML, Ellery SJ. Risk of Adverse Outcomes in Females Taking Oral Creatine Monohydrate: A Systematic Review and Meta-Analysis. Nutrients 2020; 12:E1780. [PMID: 32549301 PMCID: PMC7353222 DOI: 10.3390/nu12061780] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
Creatine Monohydrate (CrM) is a dietary supplement routinely used as an ergogenic aid for sport and training, and as a potential therapeutic aid to augment different disease processes. Despite its increased use in recent years, studies reporting potential adverse outcomes of CrM have been mostly derived from male or mixed sex populations. A systematic search was conducted, which included female participants on CrM, where adverse outcomes were reported, with meta-analysis performed where appropriate. Six hundred and fifty-six studies were identified where creatine supplementation was the primary intervention; fifty-eight were female only studies (9%). Twenty-nine studies monitored for adverse outcomes, with 951 participants. There were no deaths or serious adverse outcomes reported. There were no significant differences in total adverse events, (risk ratio (RR) 1.24 (95% CI 0.51, 2.98)), gastrointestinal events, (RR 1.09 (95% CI 0.53, 2.24)), or weight gain, (mean difference (MD) 1.24 kg pre-intervention, (95% CI -0.34, 2.82)) to 1.37 kg post-intervention (95% CI -0.50, 3.23)), in CrM supplemented females, when stratified by dosing regimen and subject to meta-analysis. No statistically significant difference was reported in measures of renal or hepatic function. In conclusion, mortality and serious adverse events are not associated with CrM supplementation in females. Nor does the use of creatine supplementation increase the risk of total adverse outcomes, weight gain or renal and hepatic complications in females. However, all future studies of creatine supplementation in females should consider surveillance and comprehensive reporting of adverse outcomes to better inform participants and health professionals involved in future trials.
Collapse
Affiliation(s)
- Deborah L. de Guingand
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia; (M.L.D.-T.); (S.J.E.)
| | - Kirsten R. Palmer
- Department of Obstetrics and Gynaecology, Monash University, Melbourne 3168, Australia;
- Monash Health, Monash Medical Centre, Melbourne 3168, Australia
| | - Rodney J. Snow
- Institute of Physical Activity and Nutrition, Deakin University, Melbourne 3125, Australia;
| | - Miranda L. Davies-Tuck
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia; (M.L.D.-T.); (S.J.E.)
| | - Stacey J. Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne 3168, Australia; (M.L.D.-T.); (S.J.E.)
- Department of Obstetrics and Gynaecology, Monash University, Melbourne 3168, Australia;
| |
Collapse
|
24
|
de Guingand DL, Palmer KR, Bilardi JE, Ellery SJ. Acceptability of dietary or nutritional supplementation in pregnancy (ADONS) - Exploring the consumer's perspective on introducing creatine monohydrate as a pregnancy supplement. Midwifery 2019; 82:102599. [PMID: 31877396 DOI: 10.1016/j.midw.2019.102599] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND Pre-clinical studies suggest maternal dietary creatine supplementation during pregnancy could protect babies against hypoxic intrapartum events, however creatine has not been used as a supplement in pregnancy. The aim of this study was to explore pregnant women and healthcare professional's general knowledge, behaviours, and attitudes toward nutritional supplements, and their thoughts on introducing creatine as a pregnancy supplement. METHODS Pregnant women (n = 42) and partners (n = 23), attending a tertiary care pregnancy service in Melbourne, Australia, participated in focus groups or semi-structured interviews. Health professionals (n = 100), completed a semi-structured online survey. Descriptive data were analyzed using SPSS 25.0 and qualitative data was managed using NVivo 22.0. RESULTS Use of branded nutritional supplements in pregnancy was commonplace and acceptable. All primary healthcare respondents discussed supplements with their patients at first consultation. Supplements consumed corresponded closely to those recommended. Women had good general awareness of commonly recommended nutritional supplements, however, were less aware of the rationale for supplement use. This aligned with health professional's perceptions. Women would consider taking creatine if recommended by their health professional. Health professionals would require detailed safety, beneficence, and efficacy information before recommending creatine supplementation. They would also be more likely to recommend a new supplement in higher-risk pregnancies, where benefits may outweigh any perceived side-effects. CONCLUSION There is high acceptance of current recommended nutritional supplements in pregnancy. Implementing creatine as a new supplement will require substantive empirical evidence and changes to clinical guidelines. Public awareness and education would also be essential to consumer acceptability of creatine.
Collapse
Affiliation(s)
- Deborah L de Guingand
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia; Department of Obstetrics & Gynaecology, Monash University, Melbourne, Australia.
| | - Kirsten R Palmer
- Department of Obstetrics & Gynaecology, Monash University, Melbourne, Australia; Department of Obstetrics & Gynaecology, Monash Health, Melbourne, Australia
| | - Jade E Bilardi
- Melbourne Sexual Health Centre, Central Clinical School, Monash University, Melbourne, Australia
| | - Stacey J Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia; Department of Obstetrics & Gynaecology, Monash University, Melbourne, Australia
| |
Collapse
|
25
|
Abstract
Perinatal brain injury is a major cause of neurological disability in both premature and term infants. In this review, we summarize the evidence behind some established neuroprotective practices such as administration of antenatal steroids, intrapartum magnesium for preterm delivery, and therapeutic hypothermia. In addition, we examine emerging practices such as delayed cord clamping, postnatal magnesium administration, recombinant erythropoietin, and non-steroidal anti-inflammatory agents and finally inform the reader about novel interventions, some of which are currently in trials, such as xenon, melatonin, topiramate, allopurinol, creatine, and autologous cord cell therapy.
Collapse
Affiliation(s)
- Samata Singhi
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA
- Department of Pediatric Neurology, Johns Hopkins Medicine, Baltimore, MD, 21287, USA
| | - Michael Johnston
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA
| |
Collapse
|
26
|
Zhang L, Li Y, Lin X, Jia C, Yu X. Liquid Chromatography/Mass Spectrometry based serum metabolomics study on recurrent abortion women with antiphospholipid syndrome. PLoS One 2019; 14:e0225463. [PMID: 31751406 PMCID: PMC6874207 DOI: 10.1371/journal.pone.0225463] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/05/2019] [Indexed: 12/05/2022] Open
Abstract
Objective The antiphospholipid syndrome (APS) is an important cause of acquired thromboembolic complications and pregnancy morbidity. The pathogenic mechanisms that damage the fetal–maternal unit and cause abnormal placental development are incompletely understood in APS patients. Liquid Chromatography/Mass Spectrometry (LC/MS) based metabolomics are applied for the mechanism of disease and further supporting the research of diagnosis and management in recent years. The aim of this research was to investigate the difference of serum metabolic profiles in recurrent abortion women with APS and healthy women to explore the mechanism of this disease. Methods Serum samples of 25 recurrent abortion women with APS and 25 healthy women were collected and analyzed by LC/MS in this study. Potential biomarkers were discovered by multivariate statistical analysis and then identified based on analysis results. Results Totally, we identified five biomarkers that involved in different metabolic pathway such as purine metabolism, amino acid metabolism and tyrosine metabolism. These biomarkers showed different roles in disease development. Conclusion Metabolomics was proved as a powerful tool in understanding the mechanism of recurrent abortion caused by APS.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Ultrasonography, First Hospital, Jilin University, Changchun, China
| | - Ying Li
- Department of Neonatology, First Hospital, Jilin University, Changchun, China
| | - Xiuying Lin
- Center for Reproductive Medicine, Jilin Province People's Hospital, Changchun, China
| | - Chunshu Jia
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
- * E-mail: (CJ); (XY)
| | - Xiaowei Yu
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
- * E-mail: (CJ); (XY)
| |
Collapse
|
27
|
Sartini S, Lattanzi D, Di Palma M, Savelli D, Eusebi S, Sestili P, Cuppini R, Ambrogini P. Maternal Creatine Supplementation Positively Affects Male Rat Hippocampal Synaptic Plasticity in Adult Offspring. Nutrients 2019; 11:nu11092014. [PMID: 31461895 PMCID: PMC6770830 DOI: 10.3390/nu11092014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/16/2019] [Accepted: 08/23/2019] [Indexed: 01/16/2023] Open
Abstract
Creatine plays a crucial role in developing the brain, so much that its genetic deficiency results in mental dysfunction and cognitive impairments. Moreover, creatine supplementation is currently under investigation as a preventive measure to protect the fetus against oxidative stress during difficult pregnancies. Although creatine use is considered safe, posing minimal risk to clinical health, we found an alteration in morpho-functional maturation of neurons when male rats were exposed to creatine loads during brain development. In particular, increased excitability and enhanced long-term potentiation (LTP) were observed in the hippocampal pyramidal neurons of weaning pups. Since these effects were observed a long time after creatine treatment had been terminated, long-lasting modifications persisting into adulthood were hypothesized. Such modifications were investigated in the present study using morphological, electrophysiological, and calcium imaging techniques applied to hippocampal Cornu Ammonis 1 (CA1) neurons of adult rats born from dams supplemented with creatine. When compared to age-matched controls, the treated adult offspring were found to retain enhanced neuron excitability and an improved LTP, the best-documented neuronal substrate for memory formation. While translating data from rats to humans does have limitations, our findings suggest that prenatal creatine supplementation could have positive effects on adult cognitive abilities.
Collapse
Affiliation(s)
- Stefano Sartini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, I-61029 Urbino, Italy.
| | - Davide Lattanzi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, I-61029 Urbino, Italy
| | - Michael Di Palma
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, I-61029 Urbino, Italy
| | - David Savelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, I-61029 Urbino, Italy
| | - Silvia Eusebi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, I-61029 Urbino, Italy
| | - Piero Sestili
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, I-61029 Urbino, Italy
| | - Riccardo Cuppini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, I-61029 Urbino, Italy
| | - Patrizia Ambrogini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, I-61029 Urbino, Italy
| |
Collapse
|
28
|
Finch-Edmondson M, Morgan C, Hunt RW, Novak I. Emergent Prophylactic, Reparative and Restorative Brain Interventions for Infants Born Preterm With Cerebral Palsy. Front Physiol 2019; 10:15. [PMID: 30745876 PMCID: PMC6360173 DOI: 10.3389/fphys.2019.00015] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/08/2019] [Indexed: 12/13/2022] Open
Abstract
Worldwide, an estimated 15 million babies are born preterm (<37 weeks' gestation) every year. Despite significant improvements in survival rates, preterm infants often face a lifetime of neurodevelopmental disability including cognitive, behavioral, and motor impairments. Indeed, prematurity remains the largest risk factor for the development of cerebral palsy. The developing brain of the preterm infant is particularly fragile; preterm babies exhibit varying severities of cerebral palsy arising from reductions in both cerebral white and gray matter volumes, as well as altered brain microstructure and connectivity. Current intensive care therapies aim to optimize cardiovascular and respiratory function to protect the brain from injury by preserving oxygenation and blood flow. If a brain injury does occur, definitive diagnosis of cerebral palsy in the first few hours and weeks of life is difficult, especially when the lesions are subtle and not apparent on cranial ultrasound. However, early diagnosis of mildly affected infants is critical, because these are the patients most likely to respond to emergent treatments inducing neuroplasticity via high-intensity motor training programs and regenerative therapies involving stem cells. A current controversy is whether to test universal treatment in all infants at risk of brain injury, accepting that some patients never required treatment, because the perceived potential benefits outweigh the risk of harm. Versus, waiting for a diagnosis before commencing targeted treatment for infants with a brain injury, and potentially missing the therapeutic window. In this review, we discuss the emerging prophylactic, reparative, and restorative brain interventions for infants born preterm, who are at high risk of developing cerebral palsy. We examine the current evidence, considering the timing of the intervention with relation to the proposed mechanism/s of action. Finally, we consider the development of novel markers of preterm brain injury, which will undoubtedly lead to improved diagnostic and prognostic capability, and more accurate instruments to assess the efficacy of emerging interventions for this most vulnerable group of infants.
Collapse
Affiliation(s)
- Megan Finch-Edmondson
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Catherine Morgan
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Rod W. Hunt
- Department of Neonatal Medicine, The Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
- Neonatal Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Iona Novak
- The Discipline of Child and Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney Medical School, Sydney, NSW, Australia
- Cerebral Palsy Alliance Research Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
29
|
De Guingand DL, Ellery SJ, Davies-Tuck ML, Dickinson H. Creatine and pregnancy outcomes, a prospective cohort study in low-risk pregnant women: study protocol. BMJ Open 2019; 9:e026756. [PMID: 30647050 PMCID: PMC6340624 DOI: 10.1136/bmjopen-2018-026756] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION The creatine kinase circuit is central to the regulation of high-energy phosphate metabolism and the maintenance of cellular energy turnover. This circuit is fuelled by creatine, an amino acid derivative that can be obtained from a diet containing animal products, and by synthesis in the body de novo. A recent retrospective study conducted in a cohort of 287 pregnant women determined that maternal excreted levels of creatine may be associated with fetal growth. This prospective study aims to overcome some of the limitations associated with the previous study and thoroughly characterise creatine homeostasis throughout gestation in a low-risk pregnant population. METHODS AND ANALYSIS This study is recruiting women with a singleton low-risk pregnancy who are attending Monash Health, in Melbourne, Australia. Maternal blood and urine samples, along with dietary surveys, are collected at five time points during pregnancy and then at delivery. Cord blood and placenta (including membranes and cord) are collected at birth. A biobank of tissue samples for future research is being established. Primary outcome measures will include creatine, creatine kinase and associated metabolites in antenatal bloods and urine, cord bloods and placenta, along with molecular analysis of the creatine transporter (SLC6A8) and synthesising enzymes L - arginine:glycine amidinotransferase (AGAT) and guanidinoacetate methyltransferase (GAMT) in placental tissues. Secondary outcome measures include dietary protein intake over pregnancy and any associations with maternal creatine, pregnancy events and birth outcomes. ETHICS AND DISSEMINATION Ethical approval was granted in August 2015 from Monash Health (Ref: 14140B) and Monash University (Ref: 7785). Study outcomes will be disseminated at international conferences and published in peer-reviewed scientific journals. TRIAL REGISTRATION NUMBER ACTRN12618001558213; Pre-results.
Collapse
Affiliation(s)
- Deborah L De Guingand
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Stacey J Ellery
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Miranda L Davies-Tuck
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Hayley Dickinson
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
30
|
Bellofiore N, Evans J. Monkeys, mice and menses: the bloody anomaly of the spiny mouse. J Assist Reprod Genet 2019; 36:811-817. [PMID: 30610663 DOI: 10.1007/s10815-018-1390-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 12/17/2018] [Indexed: 01/01/2023] Open
Abstract
The common spiny mouse (Acomys cahirinus) is the only known rodent to demonstrate a myriad of physiological processes unseen in their murid relatives. The most recently discovered of these uncharacteristic traits: spontaneous decidual transformation of the uterus in virgin females, preceding menstruation. Menstruation occurring without experimental intervention in rodents has not been documented elsewhere to date, and natural menstruation is indeed rare in the animal kingdom outside of higher order primates. This review briefly summarises the current knowledge of spiny mouse biology and taxonomy, and explores their endocrinology which may aid in our understanding of the evolution of menstruation in this species. We propose that DHEA, synthesised by the spiny mouse (but not other rodents), humans and other menstruating primates, is integral in spontaneous decidualisation and therefore menstruation. We discuss both physiological and behavioural attributes across the menstrual cycle in the spiny mouse analogous to those observed in other menstruating species, including premenstrual syndrome. We further encourage the use of the spiny mouse as a small animal model of menstruation and female reproductive biology.
Collapse
Affiliation(s)
- Nadia Bellofiore
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, 3168, Australia. .,Obstetrics and Gynaecology, Monash University, 246 Clayton Rd, Clayton, 3168, Australia.
| | - Jemma Evans
- Centre for Reproductive Health, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, 3168, Australia
| |
Collapse
|
31
|
Fleiss B, Wong F, Brownfoot F, Shearer IK, Baud O, Walker DW, Gressens P, Tolcos M. Knowledge Gaps and Emerging Research Areas in Intrauterine Growth Restriction-Associated Brain Injury. Front Endocrinol (Lausanne) 2019; 10:188. [PMID: 30984110 PMCID: PMC6449431 DOI: 10.3389/fendo.2019.00188] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/06/2019] [Indexed: 12/16/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is a complex global healthcare issue. Concerted research and clinical efforts have improved our knowledge of the neurodevelopmental sequelae of IUGR which has raised the profile of this complex problem. Nevertheless, there is still a lack of therapies to prevent the substantial rates of fetal demise or the constellation of permanent neurological deficits that arise from IUGR. The purpose of this article is to highlight the clinical and translational gaps in our knowledge that hamper our collective efforts to improve the neurological sequelae of IUGR. Also, we draw attention to cutting-edge tools and techniques that can provide novel insights into this disorder, and technologies that offer the potential for better drug design and delivery. We cover topics including: how we can improve our use of crib-side monitoring options, what we still need to know about inflammation in IUGR, the necessity for more human post-mortem studies, lessons from improved integrated histology-imaging analyses regarding the cell-specific nature of magnetic resonance imaging (MRI) signals, options to improve risk stratification with genomic analysis, and treatments mediated by nanoparticle delivery which are designed to modify specific cell functions.
Collapse
Affiliation(s)
- Bobbi Fleiss
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- NeuroDiderot, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, United Kingdom
- *Correspondence: Bobbi Fleiss
| | - Flora Wong
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Paediatrics, Monash University, Clayton, VIC, Australia
- Monash Newborn, Monash Children's Hospital, Clayton, VIC, Australia
| | - Fiona Brownfoot
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Heidelberg, VIC, Australia
| | - Isabelle K. Shearer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Olivier Baud
- NeuroDiderot, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Division of Neonatal Intensive Care, University Hospitals of Geneva, Children's Hospital, University of Geneva, Geneva, Switzerland
| | - David W. Walker
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Pierre Gressens
- NeuroDiderot, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, United Kingdom
- PremUP, Paris, France
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
32
|
Ellery SJ, Goss MG, Brew N, Dickinson H, Hale N, LaRosa DA, Walker DW, Wong FY. Evaluation of 3K3A-Activated Protein C to Treat Neonatal Hypoxic Ischemic Brain Injury in the Spiny Mouse. Neurotherapeutics 2019; 16:231-243. [PMID: 30225791 PMCID: PMC6361063 DOI: 10.1007/s13311-018-0661-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) resulting from intrapartum asphyxia is a global problem that causes severe disabilities and up to 1 million deaths annually. A variant form of activated protein C, 3K3A-APC, has cytoprotective properties that attenuate brain injury in models of adult stroke. In this study, we compared the ability of 3K3A-APC and APC (wild-type (wt)) to attenuate neonatal brain injury, using the spiny mouse (Acomys cahirinus) model of intrapartum asphyxia. Pups were delivered at 38 days of gestation (term = 39 days), with an intrapartum hypoxic insult of 7.5 min (intrapartum asphyxia cohort), or immediate removal from the uterus (control cohort). After 1 h, pups received a subcutaneous injection of 3K3A-APC or wild-type APC (wtAPC) at 7 mg/kg, or vehicle (saline). At 24 h of age, pups were killed and brain tissue was collected for measurement of inflammation and cell death using RT-qPCR and histopathology. Intrapartum asphyxia increased weight loss, inflammation, and apoptosis/necrosis in the newborn brain. 3K3A-APC administration maintained body weight and ameliorated an asphyxia-induced increase of TGFβ1 messenger RNA expression in the cerebral cortex, immune cell aggregation in the corpus callosum, and cell death in the deep gray matter and hippocampus. In the cortex, 3K3A-APC appeared to exacerbate the immune response to the hypoxic ischemic insult. While wtAPC reduced cell death in the corpus callosum and hippocampus following intrapartum asphyxia, it increased markers of neuro-inflammation and cell death in control pups. These findings suggest 3K3A-APC administration may be a useful therapy to reduce cell death and neonatal brain injury associated with HIE.
Collapse
Affiliation(s)
- Stacey J Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia.
| | - Madeleine G Goss
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
| | - Nadine Brew
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
| | - Hayley Dickinson
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - Nadia Hale
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
| | - Domenic A LaRosa
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
- Women and Infants Hospital, Alpert Medical School, Brown University, Providence, RI, USA
| | - David W Walker
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Flora Y Wong
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Melbourne, 3168, Australia
- Department of Paediatrics, Monash University, Clayton, Australia
- Monash Newborn, Monash Medical Centre, Clayton, Melbourne, Australia
| |
Collapse
|
33
|
Preventing childhood and lifelong disability: Maternal dietary supplementation for perinatal brain injury. Pharmacol Res 2018; 139:228-242. [PMID: 30227261 DOI: 10.1016/j.phrs.2018.08.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 07/29/2018] [Accepted: 08/24/2018] [Indexed: 12/30/2022]
Abstract
The majority of brain injuries that lead to cerebral palsy, developmental disability, and mental health disorders have their onset in utero. These lifelong conditions come with great economic and emotional burden as they impact function in nearly all domains of affected individuals' lives. Unfortunately, current therapeutic options are limited. There remains a focus on rescue, rehabilitation, and regeneration after the injury has occurred, rather than aiming to prevent the initial injury. Prevention would imply treating the mother during pregnancy to alter the fetal environment and in turn, treat the fetus. Fear of harming the developing fetus remains as a result of errors of the past such as the release of thalidomide. In this review, we outline evidence from animal studies and clinical trials that have explored maternal dietary supplementation with natural health products (including nutraceuticals and functional foods) for perinatal brain injury prevention. Namely, we discuss magnesium sulphate, creatine, choline, melatonin, resveratrol and broccoli sprouts/sulforaphane. Although clinical trials have only been completed in this realm for magnesium sulphate, results in animal models have been promising, suggesting that this is a productive avenue for further research. Natural health products may provide safe, effective, affordable, and easily accessible prevention of fetal brain injury and resulting lifelong disabilities.
Collapse
|
34
|
Ellery SJ, Kelleher M, Grigsby P, Burd I, Derks JB, Hirst J, Miller SL, Sherman LS, Tolcos M, Walker DW. Antenatal prevention of cerebral palsy and childhood disability: is the impossible possible? J Physiol 2018; 596:5593-5609. [PMID: 29928763 DOI: 10.1113/jp275595] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 05/15/2018] [Indexed: 12/11/2022] Open
Abstract
This review covers our current knowledge of the causes of perinatal brain injury leading to cerebral palsy-like outcomes, and argues that much of this brain damage is preventable. We review the experimental evidence that there are treatments that can be safely administered to women in late pregnancy that decrease the likelihood and extent of perinatal brain damage that occurs because of acute and severe hypoxia that arises during some births, and the additional impact of chronic fetal hypoxia, infection, inflammation, growth restriction and preterm birth. We discuss the types of interventions required to ameliorate or even prevent apoptotic and necrotic cell death, and the vulnerability of all the major cell types in the brain (neurons, astrocytes, oligodendrocytes, microglia, cerebral vasculature) to hypoxia/ischaemia, and whether a pan-protective treatment given to the mother before birth is a realistic prospect.
Collapse
Affiliation(s)
- Stacey J Ellery
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Meredith Kelleher
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Peta Grigsby
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Irina Burd
- Department of Gynecology & Obstetrics, Johns Hopkins University, Baltimore, MD, USA
| | - Jan B Derks
- Department of Perinatal Medicine University Medical Center Utrecht, The Netherlands, Gynaecology, Monash University, Melbourne, Australia
| | - Jon Hirst
- University of Newcastle, Newcastle, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Larry S Sherman
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Mary Tolcos
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Melbourne, Australia
| | - David W Walker
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia.,School of Health & Biomedical Sciences, RMIT University, Bundoora, Melbourne, Australia
| |
Collapse
|
35
|
Netto CA, Sanches EF, Odorcyk F, Duran-Carabali LE, Sizonenko SV. Pregnancy as a valuable period for preventing hypoxia-ischemia brain damage. Int J Dev Neurosci 2018; 70:12-24. [PMID: 29920306 DOI: 10.1016/j.ijdevneu.2018.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/11/2018] [Accepted: 06/15/2018] [Indexed: 12/16/2022] Open
Abstract
Neonatal brain Hypoxia-Ischemia (HI) is one of the major causes of infant mortality and lifelong neurological disabilities. The knowledge about the physiopathological mechanisms involved in HI lesion have increased in recent years, however these findings have not been translated into clinical practice. Current therapeutic approaches remain limited; hypothermia, used only in term or near-term infants, is the golden standard. Epidemiological evidence shows a link between adverse prenatal conditions and increased risk for diseases, health problems, and psychological outcomes later in life, what makes pregnancy a relevant period for preventing future brain injury. Here, we review experimental literature regarding preventive interventions used during pregnancy, i.e., previous to the HI injury, encompassing pharmacological, nutritional and/or behavioral strategies. Literature review used PubMed database. A total of forty one studies reported protective properties of maternal treatments preventing perinatal hypoxia-ischemia injury in rodents. Pharmacological agents and dietary supplementation showed mainly anti-excitotoxicity, anti-oxidant or anti-apoptotic properties. Interestingly, maternal preconditioning, physical exercise and environmental enrichment seem to engage the same referred mechanisms in order to protect neonatal brain against injury. This construct must be challenged by further studies to clearly define the main mechanisms responsible for neuroprotection to be explored in experimental context, as well as to test their potential in clinical settings.
Collapse
Affiliation(s)
- C A Netto
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, Brazil.
| | - E F Sanches
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, Brazil
| | - F Odorcyk
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, Brazil
| | - L E Duran-Carabali
- Biochemistry Department, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, Brazil
| | - S V Sizonenko
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| |
Collapse
|
36
|
Pinheiro G, Prata DF, Araújo IM, Tiscornia G. The African spiny mouse ( Acomys spp.) as an emerging model for development and regeneration. Lab Anim 2018; 52:565-576. [PMID: 29699452 DOI: 10.1177/0023677218769921] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The African spiny mouse ( Acomys spp.) is an emerging animal model with remarkable biological characteristics that make it a subject of interest for a broad range of research fields. Typically a desert species adapted to a low-calorie diet, spiny mice develop diabetes-related symptoms when switched to high-energy diets. Spiny mice undergo relatively long gestation periods and have small litters of highly developed pups, making them an adequate model for late organogenesis and perinatal biology. Recently, they have been shown to have remarkable healing and regeneration capabilities, which make them unique among mammals. In this work, we describe our experience in housing a colony of African spiny mice and cover all basic aspects of feeding, maintenance and breeding for research purposes.
Collapse
Affiliation(s)
- Gonçalo Pinheiro
- 1 Center for Biomedical Research (CBMR), University of Algarve, Faro, Portugal.,2 Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
| | - Diogo Filipe Prata
- 1 Center for Biomedical Research (CBMR), University of Algarve, Faro, Portugal.,2 Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
| | - Inês Maria Araújo
- 1 Center for Biomedical Research (CBMR), University of Algarve, Faro, Portugal.,2 Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal.,3 Algarve Biomedical Center, University of Algarve, Faro, Portugal
| | - Gustavo Tiscornia
- 1 Center for Biomedical Research (CBMR), University of Algarve, Faro, Portugal.,3 Algarve Biomedical Center, University of Algarve, Faro, Portugal.,4 Clínica Eugin, Research and Innovation Department, Spain
| |
Collapse
|
37
|
Santacruz L, Arciniegas AJL, Darrabie M, Mantilla JG, Baron RM, Bowles DE, Mishra R, Jacobs DO. Hypoxia decreases creatine uptake in cardiomyocytes, while creatine supplementation enhances HIF activation. Physiol Rep 2018; 5:5/16/e13382. [PMID: 28821596 PMCID: PMC5582266 DOI: 10.14814/phy2.13382] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 07/18/2017] [Indexed: 12/23/2022] Open
Abstract
Creatine (Cr), phosphocreatine (PCr), and creatine kinases (CK) comprise an energy shuttle linking ATP production in mitochondria with cellular consumption sites. Myocytes cannot synthesize Cr: these cells depend on uptake across the cell membrane by a specialized creatine transporter (CrT) to maintain intracellular Cr levels. Hypoxia interferes with energy metabolism, including the activity of the creatine energy shuttle, and therefore affects intracellular ATP and PCr levels. Here, we report that exposing cultured cardiomyocytes to low oxygen levels rapidly diminishes Cr transport by decreasing Vmax and Km. Pharmacological activation of AMP‐activated kinase (AMPK) abrogated the reduction in Cr transport caused by hypoxia. Cr supplementation increases ATP and PCr content in cardiomyocytes subjected to hypoxia, while also significantly augmenting the cellular adaptive response to hypoxia mediated by HIF‐1 activation. Our results indicate that: (1) hypoxia reduces Cr transport in cardiomyocytes in culture, (2) the cytoprotective effects of Cr supplementation are related to enhanced adaptive physiological responses to hypoxia mediated by HIF‐1, and (3) Cr supplementation increases the cellular ATP and PCr content in RNCMs exposed to hypoxia.
Collapse
Affiliation(s)
- Lucia Santacruz
- Department of Molecular Biology and Biochemistry, The University of Texas Medical Branch, Galveston, Texas .,Department of Natural Sciences, Bowie State University, Bowie, Maryland
| | - Antonio Jose Luis Arciniegas
- Department of Medicine, Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - Rebecca M Baron
- Department of Medicine, Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dawn E Bowles
- Duke University Medical Center, Durham, North Carolina
| | | | - Danny O Jacobs
- Department of Surgery, The University of Texas Medical Branch, Galveston, Texas.,Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
38
|
Herrera-Marschitz M, Perez-Lobos R, Lespay-Rebolledo C, Tapia-Bustos A, Casanova-Ortiz E, Morales P, Valdes JL, Bustamante D, Cassels BK. Targeting Sentinel Proteins and Extrasynaptic Glutamate Receptors: a Therapeutic Strategy for Preventing the Effects Elicited by Perinatal Asphyxia? Neurotox Res 2018; 33:461-473. [PMID: 28844085 PMCID: PMC5766721 DOI: 10.1007/s12640-017-9795-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/29/2022]
Abstract
Perinatal asphyxia (PA) is a relevant cause of death at the time of labour, and when survival is stabilised, associated with short- and long-term developmental disabilities, requiring inordinate care by health systems and families. Its prevalence is high (1 to 10/1000 live births) worldwide. At present, there are few therapeutic options, apart from hypothermia, that regrettably provides only limited protection if applied shortly after the insult.PA implies a primary and a secondary insult. The primary insult relates to the lack of oxygen, and the secondary one to the oxidative stress triggered by re-oxygenation, formation of reactive oxygen (ROS) and reactive nitrogen (RNS) species, and overactivation of glutamate receptors and mitochondrial deficiencies. PA induces overactivation of a number of sentinel proteins, including hypoxia-induced factor-1α (HIF-1α) and the genome-protecting poly(ADP-ribose) polymerase-1 (PARP-1). Upon activation, PARP-1 consumes high amounts of ATP at a time when this metabolite is scarce, worsening in turn the energy crisis elicited by asphyxia. The energy crisis also impairs ATP-dependent transport, including glutamate re-uptake by astroglia. Nicotinamide, a PARP-1 inhibitor, protects against the metabolic cascade elicited by the primary stage, avoiding NAD+ exhaustion and the energetic crisis. Upon re-oxygenation, however, oxidative stress leads to nuclear translocation of the NF-κB subunit p65, overexpression of the pro-inflammatory cytokines IL-1β and TNF-α, and glutamate-excitotoxicity, due to impairment of glial-glutamate transport, extracellular glutamate overflow, and overactivation of NMDA receptors, mainly of the extrasynaptic type. This leads to calcium influx, mitochondrial impairment, and inactivation of antioxidant enzymes, increasing further the activity of pro-oxidant enzymes, thereby making the surviving neonate vulnerable to recurrent metabolic insults whenever oxidative stress is involved. Here, we discuss evidence showing that (i) inhibition of PARP-1 overactivation by nicotinamide and (ii) inhibition of extrasynaptic NMDA receptor overactivation by memantine can prevent the short- and long-term consequences of PA. These hypotheses have been evaluated in a rat preclinical model of PA, aiming to identify the metabolic cascades responsible for the long-term consequences induced by the insult, also assessing postnatal vulnerability to recurrent oxidative insults. Thus, we present and discuss evidence demonstrating that PA induces long-term changes in metabolic pathways related to energy and oxidative stress, priming vulnerability of cells with both the neuronal and the glial phenotype. The effects induced by PA are region dependent, the substantia nigra being particularly prone to cell death. The issue of short- and long-term consequences of PA provides a framework for addressing a fundamental issue referred to plasticity of the CNS, since the perinatal insult triggers a domino-like sequence of events making the developing individual vulnerable to recurrent adverse conditions, decreasing his/her coping repertoire because of a relevant insult occurring at birth.
Collapse
Affiliation(s)
- Mario Herrera-Marschitz
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Ronald Perez-Lobos
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
- Escuela de Tecnologia Medica, Facultad de Medicina, Universidad Andres Bello, PO Box 8370146, Santiago, Chile
| | - Carolyne Lespay-Rebolledo
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Andrea Tapia-Bustos
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Emmanuel Casanova-Ortiz
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Paola Morales
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
- Faculty of Sciences, University of Chile, Santiago, Chile
| | | | - Diego Bustamante
- Programme of Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Av. Independencia, PO Box 8389100, 1027 Santiago, Chile
| | - Bruce K. Cassels
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
39
|
Yavuz A, Sezik M, Ozmen O, Asci H. Fingolimod against endotoxin-induced fetal brain injury in a rat model. J Obstet Gynaecol Res 2017; 43:1708-1713. [DOI: 10.1111/jog.13444] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/16/2017] [Accepted: 06/10/2017] [Indexed: 12/01/2022]
Affiliation(s)
- And Yavuz
- Department of Obstetrics and Gynecology; Süleyman Demirel University School of Medicine, SDU Campus; Isparta Turkey
| | - Mekin Sezik
- Department of Obstetrics and Gynecology; Süleyman Demirel University School of Medicine, SDU Campus; Isparta Turkey
| | - Ozlem Ozmen
- Department of Pathology; Mehmet Akif Ersoy University Faculty of Veterinary Medicine; Burdur Turkey
| | - Halil Asci
- Department of Pharmacology; Süleyman Demirel University School of Medicine, SDU Campus; Isparta Turkey
| |
Collapse
|
40
|
Leaw B, Nair S, Lim R, Thornton C, Mallard C, Hagberg H. Mitochondria, Bioenergetics and Excitotoxicity: New Therapeutic Targets in Perinatal Brain Injury. Front Cell Neurosci 2017; 11:199. [PMID: 28747873 PMCID: PMC5506196 DOI: 10.3389/fncel.2017.00199] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/26/2017] [Indexed: 12/30/2022] Open
Abstract
Injury to the fragile immature brain is implicated in the manifestation of long-term neurological disorders, including childhood disability such as cerebral palsy, learning disability and behavioral disorders. Advancements in perinatal practice and improved care mean the majority of infants suffering from perinatal brain injury will survive, with many subtle clinical symptoms going undiagnosed until later in life. Hypoxic-ischemia is the dominant cause of perinatal brain injury, and constitutes a significant socioeconomic burden to both developed and developing countries. Therapeutic hypothermia is the sole validated clinical intervention to perinatal asphyxia; however it is not always neuroprotective and its utility is limited to developed countries. There is an urgent need to better understand the molecular pathways underlying hypoxic-ischemic injury to identify new therapeutic targets in such a small but critical therapeutic window. Mitochondria are highly implicated following ischemic injury due to their roles as the powerhouse and main energy generators of the cell, as well as cell death processes. While the link between impaired mitochondrial bioenergetics and secondary energy failure following loss of high-energy phosphates is well established after hypoxia-ischemia (HI), there is emerging evidence that the roles of mitochondria in disease extend far beyond this. Indeed, mitochondrial turnover, including processes such as mitochondrial biogenesis, fusion, fission and mitophagy, affect recovery of neurons after injury and mitochondria are involved in the regulation of the innate immune response to inflammation. This review article will explore these mitochondrial pathways, and finally will summarize past and current efforts in targeting these pathways after hypoxic-ischemic injury, as a means of identifying new avenues for clinical intervention.
Collapse
Affiliation(s)
- Bryan Leaw
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia
| | - Syam Nair
- Perinatal Center, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical ResearchClayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University ClaytonClayton, VIC, Australia
| | - Claire Thornton
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' HospitalLondon, United Kingdom
| | - Carina Mallard
- Perinatal Center, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Henrik Hagberg
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' HospitalLondon, United Kingdom.,Perinatal Center, Department of Clinical Sciences, Sahlgrenska Academy, Gothenburg UniversityGothenburg, Sweden
| |
Collapse
|
41
|
Baharom S, De Matteo R, Ellery S, Della Gatta P, Bruce CR, Kowalski GM, Hale N, Dickinson H, Harding R, Walker D, Snow RJ. Does maternal-fetal transfer of creatine occur in pregnant sheep? Am J Physiol Endocrinol Metab 2017; 313:E75-E83. [PMID: 28325734 DOI: 10.1152/ajpendo.00450.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/17/2017] [Accepted: 03/18/2017] [Indexed: 01/03/2023]
Abstract
Our aim was to determine the disposition of creatine in ovine pregnancy and whether creatine is transferred across the placenta from mother to fetus. Pregnant ewes received either 1) a continuous intravenous infusion of creatine monohydrate or saline from 122 to 131 days gestation, with maternal and fetal arterial blood and amniotic fluid samples collected daily for creatine analysis and fetal tissues collected at necropsy at 133 days for analysis of creatine content, or 2) a single systemic bolus injection of [13C]creatine monohydrate at 130 days of gestation, with maternal and fetal arterial blood, uterine vein blood, and amniotic fluid samples collected before and for 4 h after injection and analyzed for creatine, creatine isotopic enrichment, and guanidinoacetic acid (GAA; precursor of creatine) concentrations. Presence of the creatine transporter-1 (SLC6A8) and l-arginine:glycine amidinotransferase (AGAT; the enzyme synthesizing GAA) proteins were determined by Western blots of placental cotyledons. The 10-day creatine infusion increased maternal plasma creatine concentration three- to fourfold (P < 0.05) without significantly changing fetal arterial, amniotic fluid, fetal tissues, or placental creatine content. Maternal arterial 13C enrichment was increased (P < 0.05) after bolus [13C]creatine injection without change of fetal arterial 13C enrichment. SLC6A8 and AGAT proteins were identified in placental cotyledons, and GAA concentration was significantly higher in uterine vein than maternal artery plasma. Despite the presence of SLC6A8 protein in cotyledons, these results suggest that creatine is not transferred from mother to fetus in near-term sheep and that the ovine utero-placental unit releases GAA into the maternal circulation.
Collapse
Affiliation(s)
- Syed Baharom
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
| | - Robert De Matteo
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Stacey Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
| | - Paul Della Gatta
- Institute for Physical Activity and Nutrition, Deakin University, Melbourne, Australia
| | - Clinton R Bruce
- Institute for Physical Activity and Nutrition, Deakin University, Melbourne, Australia
| | - Greg M Kowalski
- Institute for Physical Activity and Nutrition, Deakin University, Melbourne, Australia
| | - Nadia Hale
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
| | - Hayley Dickinson
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
| | - Richard Harding
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - David Walker
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia; and
| | - Rodney J Snow
- Institute for Physical Activity and Nutrition, Deakin University, Melbourne, Australia;
| |
Collapse
|
42
|
Kreider RB, Kalman DS, Antonio J, Ziegenfuss TN, Wildman R, Collins R, Candow DG, Kleiner SM, Almada AL, Lopez HL. International Society of Sports Nutrition position stand: safety and efficacy of creatine supplementation in exercise, sport, and medicine. J Int Soc Sports Nutr 2017; 14:18. [PMID: 28615996 PMCID: PMC5469049 DOI: 10.1186/s12970-017-0173-z] [Citation(s) in RCA: 381] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 05/30/2017] [Indexed: 12/16/2022] Open
Abstract
Creatine is one of the most popular nutritional ergogenic aids for athletes. Studies have consistently shown that creatine supplementation increases intramuscular creatine concentrations which may help explain the observed improvements in high intensity exercise performance leading to greater training adaptations. In addition to athletic and exercise improvement, research has shown that creatine supplementation may enhance post-exercise recovery, injury prevention, thermoregulation, rehabilitation, and concussion and/or spinal cord neuroprotection. Additionally, a number of clinical applications of creatine supplementation have been studied involving neurodegenerative diseases (e.g., muscular dystrophy, Parkinson's, Huntington's disease), diabetes, osteoarthritis, fibromyalgia, aging, brain and heart ischemia, adolescent depression, and pregnancy. These studies provide a large body of evidence that creatine can not only improve exercise performance, but can play a role in preventing and/or reducing the severity of injury, enhancing rehabilitation from injuries, and helping athletes tolerate heavy training loads. Additionally, researchers have identified a number of potentially beneficial clinical uses of creatine supplementation. These studies show that short and long-term supplementation (up to 30 g/day for 5 years) is safe and well-tolerated in healthy individuals and in a number of patient populations ranging from infants to the elderly. Moreover, significant health benefits may be provided by ensuring habitual low dietary creatine ingestion (e.g., 3 g/day) throughout the lifespan. The purpose of this review is to provide an update to the current literature regarding the role and safety of creatine supplementation in exercise, sport, and medicine and to update the position stand of International Society of Sports Nutrition (ISSN).
Collapse
Affiliation(s)
- Richard B. Kreider
- Exercise & Sport Nutrition Lab, Human Clinical Research Facility, Department of Health & Kinesiology, Texas A&M University, College Station, TX 77843-4243 USA
| | - Douglas S. Kalman
- Nutrition Research Unit, QPS, 6141 Sunset Drive Suite 301, Miami, FL 33143 USA
| | - Jose Antonio
- Department of Health and Human Performance, Nova Southeastern University, Davie, FL 33328 USA
| | - Tim N. Ziegenfuss
- The Center for Applied Health Sciences, 4302 Allen Road, STE 120, Stow, OH 44224 USA
| | - Robert Wildman
- Post Active Nutrition, 111 Leslie St, Dallas, TX 75208 USA
| | - Rick Collins
- Collins Gann McCloskey & Barry, PLLC, 138 Mineola Blvd., Mineola, NY 11501 USA
| | - Darren G. Candow
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, SK S4S 0A2 Canada
| | | | | | - Hector L. Lopez
- The Center for Applied Health Sciences, 4302 Allen Road, STE 120, Stow, OH 44224 USA
- Supplement Safety Solutions, LLC, Bedford, MA 01730 USA
| |
Collapse
|
43
|
Ellery SJ, LaRosa DA, Cullen-McEwen LA, Brown RD, Snow RJ, Walker DW, Kett MM, Dickinson H. Renal dysfunction in early adulthood following birth asphyxia in male spiny mice, and its amelioration by maternal creatine supplementation during pregnancy. Pediatr Res 2017; 81:646-653. [PMID: 27997529 DOI: 10.1038/pr.2016.268] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/26/2016] [Indexed: 11/09/2022]
Abstract
BACKGROUND Acute kidney injury affects ~70% of asphyxiated newborns, and increases their risk of developing chronic kidney disease later in life. Acute kidney injury is driven by renal oxygen deprivation during asphyxia, thus we hypothesized that creatine administered antenatally would protect the kidney from the long-term effects of birth asphyxia. METHODS Pregnant spiny mice were fed standard chow or chow supplemented with 5% creatine from 20-d gestation (midgestation). One day prior to term (37-d gestation), pups were delivered by caesarean or subjected to intrauterine asphyxia. Litters were allocated to one of two time-points. Kidneys were collected at 1 mo of age to estimate nephron number (stereology). Renal function (excretory profile and glomerular filtration rate) was measured at 3 mo of age, and kidneys then collected for assessment of glomerulosclerosis. RESULTS Compared with controls, at 1 mo of age male (but not female) birth-asphyxia offspring had 20% fewer nephrons (P < 0.05). At 3 mo of age male birth-asphyxia offspring had 31% lower glomerular filtration rate (P < 0.05) and greater glomerular collagen IV content (P < 0.01). Antenatal creatine prevented these renal injuries arising from birth asphyxia. CONCLUSION Maternal creatine supplementation during pregnancy may be an effective prophylactic to prevent birth asphyxia induced acute kidney injury and the emergence of chronic kidney disease.
Collapse
Affiliation(s)
- Stacey J Ellery
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash University, Melbourne, Australia
| | - Domenic A LaRosa
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash University, Melbourne, Australia
| | - Luise A Cullen-McEwen
- Department of Anatomy and Developmental Biology, Monash University, Clayton Campus, Melbourne, Australia
| | - Russell D Brown
- Department of Physiology, Monash University, Clayton Campus, Melbourne, Victoria, Australia
| | - Rod J Snow
- Institute for Physical Activity and Nutrition, Deakin University, Burwood Campus, Melbourne, Australia
| | - David W Walker
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash University, Melbourne, Australia
| | - Michelle M Kett
- Department of Physiology, Monash University, Clayton Campus, Melbourne, Victoria, Australia
| | - Hayley Dickinson
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics & Gynaecology, Monash Medical Centre, Monash University, Melbourne, Australia
| |
Collapse
|
44
|
LaRosa DA, Ellery SJ, Walker DW, Dickinson H. Understanding the Full Spectrum of Organ Injury Following Intrapartum Asphyxia. Front Pediatr 2017; 5:16. [PMID: 28261573 PMCID: PMC5313537 DOI: 10.3389/fped.2017.00016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/23/2017] [Indexed: 11/13/2022] Open
Abstract
Birth asphyxia is a significant global health problem, responsible for ~1.2 million neonatal deaths each year worldwide. Those who survive often suffer from a range of health issues including brain damage-manifesting as cerebral palsy (CP)-respiratory insufficiency, cardiovascular collapse, and renal dysfunction, to name a few. Although the majority of research is directed toward reducing the brain injury that results from intrapartum birth asphyxia, the multi-organ injury observed in surviving neonates is of equal importance. Despite the advent of hypothermia therapy for the treatment of hypoxic-ischemic encephalopathy (HIE), treatment options following asphyxia at birth remain limited, particularly in low-resource settings where the incidence of birth asphyxia is highest. Furthermore, although cooling of the neonate results in improved neurological outcomes for a small proportion of treated infants, it does not provide any benefit to the other organ systems affected by asphyxia at birth. The aim of this review is to summarize the current knowledge of the multi-organ effects of intrapartum asphyxia, with particular reference to the findings from our laboratory using the precocial spiny mouse to model birth asphyxia. Furthermore, we reviewed the current treatments available for neonates who have undergone intrapartum asphyxia, and highlight the emergence of maternal dietary creatine supplementation as a preventative therapy, which has been shown to provide multi-organ protection from birth asphyxia-induced injury in our preclinical studies. This cheap and effective nutritional supplement may be the key to reducing birth asphyxia-induced death and disability, particularly in low-resource settings where current treatments are unavailable.
Collapse
Affiliation(s)
- Domenic A LaRosa
- Ritchie Centre, Department of Obstetrics and Gynaecology, Hudson Institute of Medical Research, Monash University, Melbourne, VIC, Australia; Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Stacey J Ellery
- Ritchie Centre, Department of Obstetrics and Gynaecology, Hudson Institute of Medical Research, Monash University , Melbourne, VIC , Australia
| | - David W Walker
- Ritchie Centre, Department of Obstetrics and Gynaecology, Hudson Institute of Medical Research, Monash University , Melbourne, VIC , Australia
| | - Hayley Dickinson
- Ritchie Centre, Department of Obstetrics and Gynaecology, Hudson Institute of Medical Research, Monash University , Melbourne, VIC , Australia
| |
Collapse
|
45
|
Maternal creatine supplementation during pregnancy prevents acute and long-term deficits in skeletal muscle after birth asphyxia: a study of structure and function of hind limb muscle in the spiny mouse. Pediatr Res 2016; 80:852-860. [PMID: 27466898 DOI: 10.1038/pr.2016.153] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/10/2016] [Indexed: 11/08/2022]
Abstract
BACKGROUND Maternal antenatal creatine supplementation protects the brain, kidney, and diaphragm against the effects of birth asphyxia in the spiny mouse. In this study, we examined creatine's potential to prevent damage to axial skeletal muscles. METHODS Pregnant spiny mice were fed a control or creatine-supplemented diet from mid-pregnancy, and 1 d before term (39 d), fetuses were delivered by c-section with or without 7.5 min of birth asphyxia. At 24 h or 33 ± 2 d after birth, gastrocnemius muscles were obtained for ex-vivo study of twitch-tension, muscle fatigue, and structural and histochemical analysis. RESULTS Birth asphyxia significantly reduced cross-sectional area of all muscle fiber types (P < 0.05), and increased fatigue caused by repeated tetanic contractions at 24 h of age (P < 0.05). There were fewer (P < 0.05) Type I and IIa fibers and more (P < 0.05) Type IIb fibers in male gastrocnemius at 33 d of age. Muscle oxidative capacity was reduced (P < 0.05) in males at 24 h and 33 d and in females at 24 h only. Maternal creatine treatment prevented all asphyxia-induced changes in the gastrocnemius, improved motor performance. CONCLUSION This study demonstrates that creatine loading before birth protects the muscle from asphyxia-induced damage at birth.
Collapse
|
46
|
Zou R, Mu DZ. [Prevention and treatment of energy failure in neonates with hypoxic-ischemic encephalopathy]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016; 18:915-920. [PMID: 27655554 PMCID: PMC7389965 DOI: 10.7499/j.issn.1008-8830.2016.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 06/27/2016] [Indexed: 06/06/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) in neonates is the brain injury caused by perinatal asphyxia or hypoxia and is a major cause of death in neonates and nervous system dysfunction in infants and young children. Although to a certain degree, mild hypothermia therapy reduces the mortality of infants with moderate to severe HIE, it cannot achieve the expected improvements in nervous system dysfunction. Hence, it is of vital importance to search for effective therapeutic methods for HIE. The search for more therapies and better preventive measures based on the pathogenesis of HIE has resulted in much research. As an important link in the course of HIE, energy failure greatly affects the development and progression of HIE. This article reviews the research advances in the treatment and prevention of energy failure in the course of HIE.
Collapse
Affiliation(s)
- Rong Zou
- Department of Pediatrics, West China Second Hospital, Sichuan University/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China.
| | | |
Collapse
|
47
|
Dickinson H, Davies-Tuck M, Ellery SJ, Grieger JA, Wallace EM, Snow RJ, Walker DW, Clifton VL. Maternal creatine in pregnancy: a retrospective cohort study. BJOG 2016; 123:1830-8. [PMID: 27550725 DOI: 10.1111/1471-0528.14237] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2016] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To estimate creatine concentrations in maternal plasma and urine, and establish relationships with maternal characteristics, diet and fetal growth. DESIGN Retrospective cohort study. SETTING Lyell McEwin Hospital, Adelaide, Australia. POPULATION A biobank of plasma and urine samples collected at 13, 18, 30 and 36 weeks' gestation from 287 pregnant women from a prospective cohort of asthmatic and non-asthmatic women. METHODS Creatine was measured by enzymatic analysis. Change in creatine over pregnancy was assessed using the Friedman test. Linear mixed models regression was used to determine associations between maternal factors and diet with creatine across pregnancy and between creatine with indices of fetal growth at birth. MAIN OUTCOME MEASURES Maternal creatine concentrations, associations between maternal factors and creatine and between creatine and fetal growth parameters. RESULTS Maternal smoking, body mass index, asthma and socio-economic status were positively and parity negatively associated with maternal plasma and/or urine creatine. Maternal urine creatine concentration was positively associated with birthweight centile and birth length. After adjustment, each μmol/l increase in maternal urinary creatine was associated with a 1.23 (95% CI 0.44-2.02) unit increase in birthweight centile and a 0.11-cm (95% CI 0.03-0.2) increase in birth length. CONCLUSIONS Maternal factors and fetal growth measures are associated with maternal plasma and urine creatine concentrations. TWEETABLE ABSTRACT Maternal creatine is altered by pregnancy; fetal growth measures are associated with maternal creatine concentrations.
Collapse
Affiliation(s)
- H Dickinson
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Vic., Australia. .,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Vic., Australia.
| | - M Davies-Tuck
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Vic., Australia
| | - S J Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Vic., Australia
| | - J A Grieger
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - E M Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Vic., Australia
| | - R J Snow
- School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition, Deakin University, Burwood, Vic., Australia
| | - D W Walker
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, Vic., Australia
| | - V L Clifton
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide, SA, Australia.,Mater Medical Research Institute and Translational Research Institute, University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
48
|
Koob M, Viola A, Le Fur Y, Viout P, Ratiney H, Confort-Gouny S, Cozzone PJ, Girard N. Creatine, Glutamine plus Glutamate, and Macromolecules Are Decreased in the Central White Matter of Premature Neonates around Term. PLoS One 2016; 11:e0160990. [PMID: 27547969 PMCID: PMC4993494 DOI: 10.1371/journal.pone.0160990] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 07/28/2016] [Indexed: 11/18/2022] Open
Abstract
Preterm birth represents a high risk of neurodevelopmental disabilities when associated with white-matter damage. Recent studies have reported cognitive deficits in children born preterm without brain injury on MRI at term-equivalent age. Understanding the microstructural and metabolic underpinnings of these deficits is essential for their early detection. Here, we used diffusion-weighted imaging and single-voxel 1H magnetic resonance spectroscopy (MRS) to compare brain maturation at term-equivalent age in premature neonates with no evidence of white matter injury on conventional MRI except diffuse excessive high-signal intensity, and normal term neonates. Thirty-two infants, 16 term neonates (mean post-conceptional age at scan: 39.8±1 weeks) and 16 premature neonates (mean gestational age at birth: 29.1±2 weeks, mean post-conceptional age at scan: 39.2±1 weeks) were investigated. The MRI/MRS protocol performed at 1.5T involved diffusion-weighted MRI and localized 1H-MRS with the Point RESolved Spectroscopy (PRESS) sequence. Preterm neonates showed significantly higher ADC values in the temporal white matter (P<0.05), the occipital white matter (P<0.005) and the thalamus (P<0.05). The proton spectrum of the centrum semiovale was characterized by significantly lower taurine/H2O and macromolecules/H2O ratios (P<0.05) at a TE of 30 ms, and reduced (creatine+phosphocreatine)/H2O and (glutamine+glutamate)/H2O ratios (P<0.05) at a TE of 135 ms in the preterm neonates than in full-term neonates. Our findings indicate that premature neonates with normal conventional MRI present a delay in brain maturation affecting the white matter and the thalamus. Their brain metabolic profile is characterized by lower levels of creatine, glutamine plus glutamate, and macromolecules in the centrum semiovale, a finding suggesting altered energy metabolism and protein synthesis.
Collapse
Affiliation(s)
- Meriam Koob
- Service de Neuroradiologie, AP-HM Timone, Aix-Marseille Université, Marseille, France
- Service de Radiopédiatrie-Imagerie 2, CHU de Strasbourg, Hôpital de Hautepierre, Strasbourg, France
- Laboratoire ICube, UMR 7357, FMTS, Université de Strasbourg-CNRS, Strasbourg, France
| | - Angèle Viola
- Aix-Marseille Université, CNRS, Centre de Résonance Magnétique Biologique et Médicale, UMR 7339, Faculté de Médecine la Timone, Marseille, France
- * E-mail: (NG); (AV)
| | - Yann Le Fur
- Aix-Marseille Université, CNRS, Centre de Résonance Magnétique Biologique et Médicale, UMR 7339, Faculté de Médecine la Timone, Marseille, France
| | - Patrick Viout
- Aix-Marseille Université, CNRS, Centre de Résonance Magnétique Biologique et Médicale, UMR 7339, Faculté de Médecine la Timone, Marseille, France
| | - Hélène Ratiney
- Laboratoire CREATIS, CNRS UMR 5220, Inserm U1044, Université Claude Bernard Lyon I, INSA-Lyon, Lyon, France
| | - Sylviane Confort-Gouny
- Aix-Marseille Université, CNRS, Centre de Résonance Magnétique Biologique et Médicale, UMR 7339, Faculté de Médecine la Timone, Marseille, France
| | - Patrick J. Cozzone
- Aix-Marseille Université, CNRS, Centre de Résonance Magnétique Biologique et Médicale, UMR 7339, Faculté de Médecine la Timone, Marseille, France
| | - Nadine Girard
- Service de Neuroradiologie, AP-HM Timone, Aix-Marseille Université, Marseille, France
- Aix-Marseille Université, CNRS, Centre de Résonance Magnétique Biologique et Médicale, UMR 7339, Faculté de Médecine la Timone, Marseille, France
- * E-mail: (NG); (AV)
| |
Collapse
|
49
|
Kitzenberg D, Colgan SP, Glover LE. Creatine kinase in ischemic and inflammatory disorders. Clin Transl Med 2016; 5:31. [PMID: 27527620 PMCID: PMC4987751 DOI: 10.1186/s40169-016-0114-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/02/2016] [Indexed: 12/20/2022] Open
Abstract
The creatine/phosphocreatine pathway plays a conserved and central role in energy metabolism. Compartmentalization of specific creatine kinase enzymes permits buffering of local high energy phosphates in a thermodynamically favorable manner, enabling both rapid energy storage and energy transfer within the cell. Augmentation of this metabolic pathway by nutritional creatine supplementation has been shown to elicit beneficial effects in a number of diverse pathologies, particularly those that incur tissue ischemia, hypoxia or oxidative stress. In these settings, creatine and phosphocreatine prevent depletion of intracellular ATP and internal acidification, enhance post-ischemic recovery of protein synthesis and promote free radical scavenging and stabilization of cellular membranes. The creatine kinase energy system is itself further regulated by hypoxic signaling, highlighting the existence of endogenous mechanisms in mammals that can enhance creatine metabolism during oxygen deprivation to promote tissue resolution and homeostasis. Here, we review recent insights into the creatine kinase pathway, and provide rationale for dietary creatine supplementation in human ischemic and inflammatory pathologies.
Collapse
Affiliation(s)
- David Kitzenberg
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, 12700 East 19th Ave. MS B-146, Aurora, CO, 80045, USA.,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Sean P Colgan
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, 12700 East 19th Ave. MS B-146, Aurora, CO, 80045, USA.,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Louise E Glover
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, 12700 East 19th Ave. MS B-146, Aurora, CO, 80045, USA. .,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
50
|
Maternal Creatine Supplementation during Pregnancy Prevents Long-Term Changes in Diaphragm Muscle Structure and Function after Birth Asphyxia. PLoS One 2016; 11:e0149840. [PMID: 26930669 PMCID: PMC4773130 DOI: 10.1371/journal.pone.0149840] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 01/14/2016] [Indexed: 11/03/2022] Open
Abstract
Using a model of birth asphyxia, we previously reported significant structural and functional deficits in the diaphragm muscle in spiny mice, deficits that are prevented by supplementing the maternal diet with 5% creatine from mid-pregnancy. The long-term effects of this exposure are unknown. Pregnant spiny mice were fed control or 5% creatine-supplemented diet for the second half of pregnancy, and fetuses were delivered by caesarean section with or without 7.5 min of in-utero asphyxia. Surviving pups were raised by a cross-foster dam until 33±2 days of age when they were euthanized to obtain the diaphragm muscle for ex-vivo study of twitch tension and muscle fatigue, and for structural and enzymatic analyses. Functional analysis of the diaphragm revealed no differences in single twitch contractile parameters between any groups. However, muscle fatigue, induced by stimulation of diaphragm strips with a train of pulses (330 ms train/sec, 40 Hz) for 300 sec, was significantly greater for asphyxia pups compared with controls (p<0.05), and this did not occur in diaphragms of creatine + asphyxia pups. Birth asphyxia resulted in a significant increase in the proportion of glycolytic, fast-twitch fibres and a reduction in oxidative capacity of Type I and IIb fibres in male offspring, as well as reduced cross-sectional area of all muscle fibre types (Type I, IIa, IIb/d) in both males and females at 33 days of age. None of these changes were observed in creatine + asphyxia animals. Thus, the changes in diaphragm fatigue and structure induced by birth asphyxia persist long-term but are prevented by maternal creatine supplementation.
Collapse
|