1
|
Li Q, Zheng T, Chen J, Li B, Zhang Q, Yang S, Shao J, Guan W, Zhang S. Exploring melatonin's multifaceted role in female reproductive health: From follicular development to lactation and its therapeutic potential in obstetric syndromes. J Adv Res 2024:S2090-1232(24)00168-1. [PMID: 38692429 DOI: 10.1016/j.jare.2024.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/25/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Melatonin is mainly secreted by the pineal gland during darkness and regulates biological rhythms through its receptors in the suprachiasmatic nucleus of the hypothalamus. In addition, it also plays a role in the reproductive system by affecting the function of the hypothalamic-pituitary-gonadal axis, and by acting as a free radical scavenger thus contributing to the maintenance of the optimal physiological state of the gonads. Besides, melatonin can freely cross the placenta to influence fetal development. However, there is still a lack of overall understanding of the role of melatonin in the reproductive cycle of female mammals. AIM OF REVIEW Here we focus the role of melatonin in female reproduction from follicular development to delivery as well as the relationship between melatonin and lactation. We further summarize the potential role of melatonin in the treatment of preeclampsia, polycystic ovary syndrome, endometriosis, and ovarian aging. KEY SCIENTIFIC CONCEPTS OF REVIEW Understanding the physiological role of melatonin in female reproductive processes will contribute to the advancement of human fertility and reproductive medicine research.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiaming Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiayuan Shao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
2
|
Häusler S, Robertson NJ, Golhen K, van den Anker J, Tucker K, Felder TK. Melatonin as a Therapy for Preterm Brain Injury: What Is the Evidence? Antioxidants (Basel) 2023; 12:1630. [PMID: 37627625 PMCID: PMC10451719 DOI: 10.3390/antiox12081630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/28/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Despite significant improvements in survival following preterm birth in recent years, the neurodevelopmental burden of prematurity, with its long-term cognitive and behavioral consequences, remains a significant challenge in neonatology. Neuroprotective treatment options to improve neurodevelopmental outcomes in preterm infants are therefore urgently needed. Alleviating inflammatory and oxidative stress (OS), melatonin might modify important triggers of preterm brain injury, a complex combination of destructive and developmental abnormalities termed encephalopathy of prematurity (EoP). Preliminary data also suggests that melatonin has a direct neurotrophic impact, emphasizing its therapeutic potential with a favorable safety profile in the preterm setting. The current review outlines the most important pathomechanisms underlying preterm brain injury and correlates them with melatonin's neuroprotective potential, while underlining significant pharmacokinetic/pharmacodynamic uncertainties that need to be addressed in future studies.
Collapse
Affiliation(s)
- Silke Häusler
- Division of Neonatology, Department of Pediatrics, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Nicola J. Robertson
- EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK; (N.J.R.); (K.T.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Klervi Golhen
- Pediatric Pharmacology and Pharmacometrics, University Children’s Hospital Basel (UKBB), University of Basel, 4001 Basel, Switzerland; (K.G.); (J.v.d.A.)
| | - John van den Anker
- Pediatric Pharmacology and Pharmacometrics, University Children’s Hospital Basel (UKBB), University of Basel, 4001 Basel, Switzerland; (K.G.); (J.v.d.A.)
- Division of Clinical Pharmacology, Children’s National Hospital, Washington, DC 20001, USA
| | - Katie Tucker
- EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK; (N.J.R.); (K.T.)
| | - Thomas K. Felder
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria;
| |
Collapse
|
3
|
Rubinstein MR, Burgueño AL, Quiroga S, Wald MR, Genaro AM. Current Understanding of the Roles of Gut-Brain Axis in the Cognitive Deficits Caused by Perinatal Stress Exposure. Cells 2023; 12:1735. [PMID: 37443769 PMCID: PMC10340286 DOI: 10.3390/cells12131735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
The term 'perinatal environment' refers to the period surrounding birth, which plays a crucial role in brain development. It has been suggested that dynamic communication between the neuro-immune system and gut microbiota is essential in maintaining adequate brain function. This interaction depends on the mother's status during pregnancy and/or the newborn environment. Here, we show experimental and clinical evidence that indicates that the perinatal period is a critical window in which stress-induced immune activation and altered microbiota compositions produce lasting behavioral consequences, although a clear causative relationship has not yet been established. In addition, we discuss potential early treatments for preventing the deleterious effect of perinatal stress exposure. In this sense, early environmental enrichment exposure (including exercise) and melatonin use in the perinatal period could be valuable in improving the negative consequences of early adversities. The evidence presented in this review encourages the realization of studies investigating the beneficial role of melatonin administration and environmental enrichment exposure in mitigating cognitive alteration in offspring under perinatal stress exposure. On the other hand, direct evidence of microbiota restoration as the main mechanism behind the beneficial effects of this treatment has not been fully demonstrated and should be explored in future studies.
Collapse
Affiliation(s)
- Mara Roxana Rubinstein
- Laboratorio de Psiconeuroendocrinoinmunologia, Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)—Pontificia Universidad Católica Argentina, Buenos Aires C1107AFF, Argentina; (A.L.B.); (S.Q.); (M.R.W.)
| | | | | | | | - Ana María Genaro
- Laboratorio de Psiconeuroendocrinoinmunologia, Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)—Pontificia Universidad Católica Argentina, Buenos Aires C1107AFF, Argentina; (A.L.B.); (S.Q.); (M.R.W.)
| |
Collapse
|
4
|
McLafferty LP, Spada M, Gopalan P. Pharmacologic Treatment of Sleep Disorders in Pregnancy. Sleep Med Clin 2022; 17:445-452. [PMID: 36150806 DOI: 10.1016/j.jsmc.2022.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Pregnancy is a unique physiologic state whose characteristics often predispose women to new-onset sleep disturbances or exacerbations of preexisting sleep disorders. Pregnancy-related factors that can disrupt sleep include heartburn, nocturnal oxytocin secretion, nocturia, and fetal movement. Sleep disorders in pregnancy include insomnia (primary and secondary), restless legs syndrome, and narcolepsy.
Collapse
Affiliation(s)
- Laura P McLafferty
- Department of Psychiatry and Human Behavior, Thomas Jefferson University, Thompson Building, Suite 1652, 1020 Sansom Street, Philadelphia, PA 19107, USA.
| | - Meredith Spada
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh Medical Center, 3811 O'Hara Street, Pittsburgh, PA 15213, USA
| | - Priya Gopalan
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh Medical Center, 3811 O'Hara Street, Pittsburgh, PA 15213, USA
| |
Collapse
|
5
|
Monayo SM, Liu X. The Prospective Application of Melatonin in Treating Epigenetic Dysfunctional Diseases. Front Pharmacol 2022; 13:867500. [PMID: 35668933 PMCID: PMC9163742 DOI: 10.3389/fphar.2022.867500] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/14/2022] [Indexed: 01/09/2023] Open
Abstract
In the past, different human disorders were described by scientists from the perspective of either environmental factors or just by genetically related mechanisms. The rise in epigenetic studies and its modifications, i.e., heritable alterations in gene expression without changes in DNA sequences, have now been confirmed in diseases. Modifications namely, DNA methylation, posttranslational histone modifications, and non-coding RNAs have led to a better understanding of the coaction between epigenetic alterations and human pathologies. Melatonin is a widely-produced indoleamine regulator molecule that influences numerous biological functions within many cell types. Concerning its broad spectrum of actions, melatonin should be investigated much more for its contribution to the upstream and downstream mechanistic regulation of epigenetic modifications in diseases. It is, therefore, necessary to fill the existing gaps concerning corresponding processes associated with melatonin with the physiological abnormalities brought by epigenetic modifications. This review outlines the findings on melatonin’s action on epigenetic regulation in human diseases including neurodegenerative diseases, diabetes, cancer, and cardiovascular diseases. It summarizes the ability of melatonin to act on molecules such as proteins and RNAs which affect the development and progression of diseases.
Collapse
|
6
|
Deng C, Zhao L, Yang Z, Shang JJ, Wang CY, Shen MZ, Jiang S, Li T, Di WC, Chen Y, Li H, Cheng YD, Yang Y. Targeting HMGB1 for the treatment of sepsis and sepsis-induced organ injury. Acta Pharmacol Sin 2022; 43:520-528. [PMID: 34040166 PMCID: PMC8888646 DOI: 10.1038/s41401-021-00676-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 04/01/2021] [Indexed: 02/05/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that is present in almost all cells and regulates the activity of innate immune responses in both intracellular and extracellular settings. Current evidence suggests that HMGB1 plays a pivotal role in human pathological and pathophysiological processes such as the inflammatory response, immune reactions, cell migration, aging, and cell death. Sepsis is a systemic inflammatory response syndrome (SIRS) that occurs in hosts in response to microbial infections with a proven or suspected infectious etiology and is the leading cause of death in intensive care units worldwide, particularly in the aging population. Dysregulated systemic inflammation is a classic characteristic of sepsis, and suppression of HMGB1 may ameliorate inflammation and improve patient outcomes. Here, we focus on the latest breakthroughs regarding the roles of HMGB1 in sepsis and sepsis-related organ injury, the ways by which HMGB1 are released, and the signaling pathways and therapeutics associated with HMGB1. This review highlights recent advances related to HMGB1: the regulation of HMBG1 might be helpful for both basic research and drug development for the treatment of sepsis and sepsis-related organ injury.
Collapse
Affiliation(s)
- Chao Deng
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Department of Orthopaedics, Huaian Medical District of Jingling Hospital, Medical School of Nanjing University, Huaian, 213001, China
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lin Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Zhi Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Jia-Jia Shang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Chang-Yu Wang
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
| | - Ming-Zhi Shen
- Hainan Hospital of PLA General Hospital, The Second School of Clinical Medicine, Southern Medical University, Sanya, 572013, China
| | - Shuai Jiang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Tian Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
| | - Wen-Cheng Di
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518100, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - He Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Ye-Dong Cheng
- Department of Orthopaedics, Huaian Medical District of Jingling Hospital, Medical School of Nanjing University, Huaian, 213001, China.
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China.
| |
Collapse
|
7
|
Amniotic LPS-Induced Apoptosis in the Fetal Brain Is Suppressed by Vaginal LPS Preconditioning but Is Promoted by Continuous Ischemic Reperfusion. Int J Mol Sci 2022; 23:ijms23031787. [PMID: 35163709 PMCID: PMC8836254 DOI: 10.3390/ijms23031787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
Chorioamnionitis (CAM) is an increasingly common disease affecting pregnant women which derives from bacterial vaginosis. In different clinical cases, it has been shown that CAM can cause multiple risk factors for fetal brain damage, such as infection, and intra-uterine asphyxia. However, the molecular mechanism remains unknown. In this study, we established a novel CAM mouse model by exposing pregnant mice to a combination of three risk factors: vaginal lipopolysaccharides (LPS), amniotic LPS, and ischemic reperfusion. We found amniotic LPS caused Parkinson's disease-like fetal brain damage, in a dose and time-dependent manner. Moreover, the mechanism of this fetal brain damage is apoptosis induced by amniotic LPS but it was inhibited by being pretreated with a vaginal LPS challenge before amniotic LPS injection. In contrast, amniotic LPS with continuous ischemic reperfusion caused a higher level of apoptotic cell death than amniotic LPS alone. In particular, a potential neuroprotective biomarker phosphorylation (p)-CREB (ser133) appeared in only vaginal LPS preconditioned before amniotic LPS, whereas ischemic reperfusion triggered IKK phosphorylation after amniotic LPS. Despite the need for many future investigations, this study also discussed a developed understanding of the molecular mechanism of how these phenotypes occurred.
Collapse
|
8
|
Bao M, Hofsink N, Plösch T. LPS vs. Poly I:C Model: Comparison of Long-Term Effects of Bacterial and Viral Maternal Immune Activation (MIA) on the Offspring. Am J Physiol Regul Integr Comp Physiol 2021; 322:R99-R111. [PMID: 34874190 PMCID: PMC8782664 DOI: 10.1152/ajpregu.00087.2021] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A prominent health issue nowadays is the COVID-19 pandemic, which poses acute risks to human health. However, the long-term health consequences are largely unknown and cannot be neglected. An especially vulnerable period for infection is pregnancy, when infections could have long-term health effect on the child. Evidence suggests that maternal immune activation (MIA) induced by either bacteria or viruses presents various effects on the offspring, leading to adverse phenotypes in many organ systems. This review compares the mechanisms of bacterial and viral MIA and the possible long-term outcomes for the offspring by summarizing the outcome in animal LPS and Poly I:C models. Both models are activated immune responses mediated by Toll-like receptors. The outcomes for MIA offspring include neurodevelopment, immune response, circulation, metabolism, and reproduction. Some of these changes continue to exist until later life. Besides different doses and batches of LPS and Poly I:C, the injection day, administration route, and also different animal species influence the outcomes. Here, we specifically aim to support colleagues when choosing their animal models for future studies.
Collapse
Affiliation(s)
- Mian Bao
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Naomi Hofsink
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Perinatal Neurobiology, Department of Human Medicine, School of Medicine and Health Sciences Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
9
|
Favrais G, Saliba E, Savary L, Bodard S, Gulhan Z, Gressens P, Chalon S. Partial protective effects of melatonin on developing brain in a rat model of chorioamnionitis. Sci Rep 2021; 11:22167. [PMID: 34773065 PMCID: PMC8589852 DOI: 10.1038/s41598-021-01746-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/01/2021] [Indexed: 12/27/2022] Open
Abstract
Melatonin has shown promising neuroprotective effects due to its anti-oxidant, anti-inflammatory and anti-apoptotic properties, making it a candidate drug for translation to humans in conditions that compromise the developing brain. Our study aimed to explore the impact of prenatal melatonin in an inflammatory/infectious context on GABAergic neurons and on oligodendrocytes (OLs), key cells involved in the encephalopathy of prematurity. An inflammatory/infectious agent (LPS, 300 μg/kg) was injected intraperitoneally (i.p.) to pregnant Wistar rats at gestational day 19 and 20. Melatonin (5 mg/kg) was injected i.p. following the same schedule. Immunostainings focusing on GABAergic neurons, OL lineage and myelination were performed on pup brain sections. Melatonin succeeded in preventing the LPS-induced decrease of GABAergic neurons within the retrospenial cortex, and sustainably promoted GABAergic neurons within the dentate gyrus in the inflammatory/infectious context. However, melatonin did not effectively prevent the LPS-induced alterations on OLs and myelination. Therefore, we demonstrated that melatonin partially prevented the deleterious effects of LPS according to the cell type. The timing of exposure related to the cell maturation stage is likely to be critical to achieve an efficient action of melatonin. Furthermore, it can be speculated that melatonin exerts a modest protective effect on extremely preterm infant brains.
Collapse
Affiliation(s)
- Geraldine Favrais
- i-Brain Team- UMR INSERM U1253, UFR de Médecine, Université de Tours, Bâtiment Thérèse Planiol, 10 Bd Tonnellé, BP 3223, 37032, Tours Cedex 1, France. .,Neonatology Unit, CHRU de Tours, Tours, France.
| | - Elie Saliba
- i-Brain Team- UMR INSERM U1253, UFR de Médecine, Université de Tours, Bâtiment Thérèse Planiol, 10 Bd Tonnellé, BP 3223, 37032, Tours Cedex 1, France
| | - Léa Savary
- i-Brain Team- UMR INSERM U1253, UFR de Médecine, Université de Tours, Bâtiment Thérèse Planiol, 10 Bd Tonnellé, BP 3223, 37032, Tours Cedex 1, France
| | - Sylvie Bodard
- i-Brain Team- UMR INSERM U1253, UFR de Médecine, Université de Tours, Bâtiment Thérèse Planiol, 10 Bd Tonnellé, BP 3223, 37032, Tours Cedex 1, France
| | - Zuhal Gulhan
- i-Brain Team- UMR INSERM U1253, UFR de Médecine, Université de Tours, Bâtiment Thérèse Planiol, 10 Bd Tonnellé, BP 3223, 37032, Tours Cedex 1, France
| | | | - Sylvie Chalon
- i-Brain Team- UMR INSERM U1253, UFR de Médecine, Université de Tours, Bâtiment Thérèse Planiol, 10 Bd Tonnellé, BP 3223, 37032, Tours Cedex 1, France
| |
Collapse
|
10
|
Melatonin and Myo-Inositol: Supporting Reproduction from the Oocyte to Birth. Int J Mol Sci 2021; 22:ijms22168433. [PMID: 34445135 PMCID: PMC8395120 DOI: 10.3390/ijms22168433] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022] Open
Abstract
Human pregnancy is a sequence of events finely tuned by several molecular interactions that come with a new birth. The precise interlocking of these events affecting the reproductive system guarantees safe embryo formation and fetal development. In this scenario, melatonin and myo-inositol seem to be pivotal not only in the physiology of the reproduction process, but also in the promotion of positive gestational outcomes. Evidence demonstrates that melatonin, beyond the role of circadian rhythm management, is a key controller of human reproductive functions. Similarly, as the most representative member of the inositol’s family, myo-inositol is essential in ensuring correct advancing of reproductive cellular events. The molecular crosstalk mediated by these two species is directly regulated by their availability in the human body. To date, biological implications of unbalanced amounts of melatonin and myo-inositol in each pregnancy step are growing the idea that these molecules actively contribute to reduce negative outcomes and improve the fertilization rate. Clinical data suggest that melatonin and myo-inositol may constitute an optimal dietary supplementation to sustain safe human gestation and a new potential way to prevent pregnancy-associated pathologies.
Collapse
|
11
|
Kim JM, Lee SY, Lee JY. Melatonin for the prevention of fetal injury associated with intrauterine inflammation. Am J Reprod Immunol 2021; 86:e13402. [PMID: 33583108 DOI: 10.1111/aji.13402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 01/06/2023] Open
Abstract
Intrauterine inflammation is shown to be associated with preterm birth, fetal inflammatory response syndrome, and other pregnancy-related comorbidities such as central nervous system diseases including cerebral palsy and periventricular leukomalacia, pulmonary diseases such as bronchopulmonary dysplasia and respiratory distress syndrome, and necrotizing enterocolitis, to name a few. Many animal studies on intrauterine inflammation demonstrate that ascending infection of reproductive organs or the production of proinflammatory cytokines by some stimuli in utero results in such manifestations. Melatonin, known for its primary function in maintaining circadian rhythm, is now recognized as one of the most potent antioxidant and anti-inflammatory drugs. In some studies, melatonin injection in pregnant animals with intrauterine inflammation significantly reduced the number of preterm births, the severity of structural disintegration of the fetal lungs observed in bronchopulmonary dysplasia, and perinatal brain injuries with improvement in neuromotor function. These implicated benefits of melatonin in pregnant women with intrauterine inflammation seem promising in many research studies, strongly supporting the hypothesis that melatonin has antioxidative and anti-inflammatory properties that can potentially be taken by pregnant women who are at risk of having intrauterine inflammation. In this review, the potential of melatonin for improving outcomes of the pregnancies with intrauterine inflammation will be discussed.
Collapse
Affiliation(s)
- Jang Mee Kim
- Department of Medicine, CHA University School of Medicine, Pocheon, Korea
| | - Seung-Yun Lee
- Educational Competence Support Center, Hanshin University, Osan, Korea
| | - Ji Yeon Lee
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| |
Collapse
|
12
|
Schander JA, Marvaldi C, Correa F, Wolfson ML, Cella M, Aisemberg J, Jensen F, Franchi AM. Maternal environmental enrichment modulates the immune response against an inflammatory challenge during gestation and protects the offspring. J Reprod Immunol 2021; 144:103273. [PMID: 33515908 DOI: 10.1016/j.jri.2021.103273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/11/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023]
Abstract
The production of pro-inflammatory cytokines during inflammatory processes has been associated with preterm birth (PTB) and fetal injury in humans and mice. We previously demonstrated that exposition to an enriched environment (EE), defined as a noninvasive and biological significant stimulus of the sensory pathway combined with voluntary physical activity, prevented PTB and perinatal death induced by the systemic administration of bacterial lipopolysaccharide (LPS) in mice. This work aimed to analyze whether EE modulates the immune response to the inflammatory process induced by LPS in peripheral blood and the amniotic fluid (AF). We observed that EE modulated maternal white blood cell count and its response to LPS. Furthermore, we found higher levels of IL-10 and a higher percentage of B cells in AF from EE exposed mothers compared to controls. Albeit LPS significantly increased IL-6 levels in AF from both groups, it was 3.6 times higher in control environment (CE) exposed group when compared to EE. Similarly, levels of IL-22 were significantly increased by LPS in both groups, but it was 6.7 times higher in EE group. Interestingly, levels of PGE2 in AF were only increased in the EE-LPS treated group, and a positive correlation between IL-22 and PGE2 levels was observed. During lactation, EE prevented LPS-induced delay in physical landmarks analyzed to assess offspring development. Our results suggest that EE modulates the immune response to systemic LPS-administration protecting the offspring. We propose that an EE-like protocol could be designed for pregnant women aiming at preventing the sequelae present in premature children.
Collapse
Affiliation(s)
- Julieta Aylen Schander
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos, CONICET-UBA, Buenos Aires, Argentina.
| | - Carolina Marvaldi
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos, CONICET-UBA, Buenos Aires, Argentina
| | - Fernando Correa
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos, CONICET-UBA, Buenos Aires, Argentina
| | - Manuel Luis Wolfson
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos, CONICET-UBA, Buenos Aires, Argentina
| | - Maximiliano Cella
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos, CONICET-UBA, Buenos Aires, Argentina
| | - Julieta Aisemberg
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos, CONICET-UBA, Buenos Aires, Argentina
| | - Federico Jensen
- Laboratorio de Inmunología de la Reproducción, Centro de Estudios Farmacológicos y Botánicos, CONICET-UBA, Buenos Aires, Argentina
| | - Ana María Franchi
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos, CONICET-UBA, Buenos Aires, Argentina
| |
Collapse
|
13
|
Melatonin-Mediated Pak2 Activation Reduces Cardiomyocyte Death Through Suppressing Hypoxia Reoxygenation Injury-Induced Endoplasmic Reticulum Stress. J Cardiovasc Pharmacol 2020; 74:20-29. [PMID: 31274839 DOI: 10.1097/fjc.0000000000000678] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cardiac reperfusion injury has been found to be associated with endoplasmic reticulum (ER) stress. Recently, p21-activated kinase 2 (Pak2) has been identified as a primary mediator of ER stress in chronic myocardial injury. Melatonin, a biological clock-related hormone, has been demonstrated to attenuate heart reperfusion burden by modulating ER stress and mitochondrial function. The aim of our study was to explore whether reperfusion-induced ER stress is modulated by melatonin through Pak2. Hypoxia reoxygenation (HR) was used in vitro to mimic reperfusion injury in cardiomyocytes. ER stress, oxidative stress, calcium overload, and cell death were measured through Western blotting, enzyme-linked immunosorbent assay, quantitative polymerase chain reaction, and immunofluorescence with the assistance of siRNA transfection and pathway blocker treatment. The results of our study demonstrated that HR decreased the levels of Pak2 in cardiomyocytes in vitro, and inactivation of Pak2 was associated with ER stress, oxidative stress, calcium overload, caspase-12 activation, and cardiomyocytes apoptosis in vitro. Interestingly, melatonin treatment attenuated HR-mediated ER stress, redox imbalance, calcium overload, and caspase-12-related cardiomyocytes apoptosis, and these protective effects were dependent on Pak2 upregulation. Knockdown of Pak2 abolished the beneficial actions exerted by melatonin on HR-treated cardiomyocytes in vitro. Finally, we found that melatonin reversed Pak2 expression by activating the AMPK pathway and blockade of the AMPK pathway suppressed Pak2 upregulation and cardiomyocytes survival induced by melatonin in the presence of HR stress. Overall, our study reports that the AMPK-Pak2 axis, a novel signaling pathway modulated by melatonin, sends prosurvival signals for cardiomyocytes reperfusion injury through attenuation of ER stress in vitro.
Collapse
|
14
|
Gao J, Su G, Liu J, Zhang J, Zhou J, Liu X, Tian Y, Zhang Z. Mechanisms of Inhibition of Excessive Microglial Activation by Melatonin. J Mol Neurosci 2020; 70:1229-1236. [PMID: 32222896 DOI: 10.1007/s12031-020-01531-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/12/2020] [Indexed: 12/22/2022]
Abstract
As the innate immune cells that permanently reside in the central nervous system (CNS), microglia play an increasingly important role in maintaining brain function. Normally, microglia act as resting phenotype, which can be activated by various types of stimuli and release a variety of inflammatory mediators. Melatonin is an endogenous rhythmic hormone secreted principally by the pineal gland. Increasing evidence suggests that melatonin can detoxify reactive oxygen species (ROS) and prevent microglia from over-activation. This review summarizes the mechanisms of melatonin in inhibiting excessive activation of microglia and demonstrates the feasibility of melatonin in the treatment of diseases related to microglial over-activation.
Collapse
Affiliation(s)
- Juan Gao
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Jifei Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Jiajia Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Juanping Zhou
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Xiaoyan Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Ye Tian
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Zhenchang Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
15
|
Hassinger AB, Bletnisky N, Dudekula R, El-Solh AA. Selecting a pharmacotherapy regimen for patients with chronic insomnia. Expert Opin Pharmacother 2020; 21:1035-1043. [PMID: 32202451 DOI: 10.1080/14656566.2020.1743265] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Chronic insomnia, whether it is primary or in combination with another medical or psychiatric disorder, is a prevalent condition associated with significant morbidity, reduced productivity, increased risk of accidents, and poor quality of life. Pharmacologic and behavioral treatments have equivalent efficacy with each having its own advantages and limitations. AREAS COVERED The purpose of this perspective is to delineate the limitations encountered in implementing cognitive behavioral therapy (CBT) and to review the pharmacological treatments designed to target the different phenotypes of insomnia. The discussions address how to choose the optimal medication or combination thereof based on patients' characteristics, available medications, and the presence of comorbid conditions. Selective nonbenzodiazepine sedative 'Z-drug' hypnotics, melatonin receptor agonist-ramelteon, and low-dose doxepin are the agents of choice for treatment of primary and comorbid insomnia. EXPERT OPINION A pharmacological intervention should be offered if cognitive behavioral therapy for insomnia is not available or has failed to achieve its goals. Increasing evidence of the significant adverse consequences of long-term benzodiazepines should limit the prescription of these agents to specific conditions. Testing novel dosing regimens with a combination of hypnotic classes augmented with CBT deserve further investigation.
Collapse
Affiliation(s)
- Amanda B Hassinger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo, NY, USA.,VA Western New York Healthcare System , Buffalo, NY, USA
| | - Nikolas Bletnisky
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo, NY, USA.,VA Western New York Healthcare System , Buffalo, NY, USA
| | - Rizwan Dudekula
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo, NY, USA.,VA Western New York Healthcare System , Buffalo, NY, USA
| | - Ali A El-Solh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo, NY, USA.,VA Western New York Healthcare System , Buffalo, NY, USA.,Department of Epidemiology and Environmental Health, Research and Development, School of Public Health, University at Buffalo , Buffalo, NY, USA
| |
Collapse
|
16
|
Sleep Pharmacotherapy for Common Sleep Disorders in Pregnancy and Lactation. Chest 2019; 157:184-197. [PMID: 31622589 DOI: 10.1016/j.chest.2019.09.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/03/2019] [Accepted: 09/21/2019] [Indexed: 12/18/2022] Open
Abstract
Sleep disturbances are common in pregnancy, and sleep disorders may worsen or present de novo in the course of gestation. Managing a pregnant patient is complicated by the risk of teratogenicity, pharmacokinetic changes, and the dynamic nature of pregnancy. Although nonpharmacologic interventions are likely safest, they are often ineffective, and a patient is left dealing with frustrations of the sleep disturbance, as well as the negative outcomes of poor sleep in pregnancy. As with any other condition in pregnancy, management requires an understanding of pregnancy physiology, knowledge of the impact of a given condition on pregnancy or fetal and neonatal outcomes, and an ability to weigh the risk of the exposure to an untreated, or poorly treated condition, against the risk of a given drug. In partnership with the pregnant patient or couple, options for therapy should be reviewed in the context of the impact of the condition on pregnancy and offspring outcomes, while understanding that data (positive or negative) on the impact of therapy on perinatal outcomes are lacking. This article reviews the epidemiology of sleep disorders in pregnancy, general principles of prescribing in pregnancy and lactation, and safety surrounding therapeutic options in pregnancy.
Collapse
|
17
|
Lee JY, Song H, Dash O, Park M, Shin NE, McLane MW, Lei J, Hwang JY, Burd I. Administration of melatonin for prevention of preterm birth and fetal brain injury associated with premature birth in a mouse model. Am J Reprod Immunol 2019; 82:e13151. [PMID: 31131935 DOI: 10.1111/aji.13151] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/25/2019] [Accepted: 05/19/2019] [Indexed: 12/14/2022] Open
Abstract
PROBLEM Maternal inflammation leads to preterm birth and perinatal brain injury. Melatonin, through its anti-inflammatory effects, has been shown to be protective against inflammation-induced perinatal adverse effects. However, the immunomodulatory effects of melatonin on preterm birth and prematurity-related morbidity remain unknown. We wanted to investigate the effects of maternally administered melatonin on preterm birth and perinatal brain injury in a mouse model of maternal inflammation. METHOD OF STUDY A model of maternal inflammation employing lipopolysaccharide (LPS) was used to mimic the most common clinical scenario of preterm birth, that of maternal inflammation. Mice were randomly divided into the following groups: control, LPS, and LPS with melatonin pre-treatment. Doppler ultrasonography was used to obtain fetal and maternal hemodynamic measurements in utero. Placenta and fetal brains were harvested and analyzed for proinflammatory markers and signs of perinatal brain injury, respectively. Surviving offspring were assessed for neuromotor outcomes. RESULTS Melatonin pre-treatment lowered the level of proinflammatory cytokines in the uterus and the placenta, significantly improved LPS-induced acute fetal neuroinflammation and perinatal brain injury, as well as significantly upregulated the SIRT1/Nrf2 signaling pathway to reduce LPS-induced inflammation. Melatonin also prevented adverse neuromotor outcomes in offspring exposed to maternal inflammation. CONCLUSION Maternally administered melatonin modulated immune responses to maternal inflammation and decreased preterm birth and perinatal brain injury. These results suggest that melatonin, a safe treatment during pregnancy, may be used as an experimental therapeutic in clinical trials.
Collapse
Affiliation(s)
- Ji Yeon Lee
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea.,Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Haengseok Song
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | - Oyunbileg Dash
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| | - Mira Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | - Na E Shin
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael W McLane
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jun Lei
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jong Yun Hwang
- Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Irina Burd
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
18
|
Gonzalez-Candia A, Veliz M, Carrasco-Pozo C, Castillo RL, Cárdenas JC, Ebensperger G, Reyes RV, Llanos AJ, Herrera EA. Antenatal melatonin modulates an enhanced antioxidant/pro-oxidant ratio in pulmonary hypertensive newborn sheep. Redox Biol 2019; 22:101128. [PMID: 30771751 PMCID: PMC6375064 DOI: 10.1016/j.redox.2019.101128] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 01/07/2023] Open
Abstract
Chronic hypobaric hypoxia during fetal and neonatal life induces neonatal pulmonary hypertension. Hypoxia and oxidative stress are driving this condition, which implies an increase generation of reactive oxygen species (ROS) and/or decreased antioxidant capacity. Melatonin has antioxidant properties that decrease oxidative stress and improves pulmonary vascular function when administered postnatally. However, the effects of an antenatal treatment with melatonin in the neonatal pulmonary function and oxidative status are unknown. Therefore, we hypothesized that an antenatal therapy with melatonin improves the pulmonary arterial pressure and antioxidant status in high altitude pulmonary hypertensive neonates. Twelve ewes were bred at high altitude (3600 m); 6 of them were used as a control group (vehicle 1.4% ethanol) and 6 as a melatonin treated group (10 mg d-1 melatonin in vehicle). Treatments were given once daily during the last third of gestation (100-150 days). Lambs were born and raised with their mothers until 12 days old, and neonatal pulmonary arterial pressure and resistance, plasma antioxidant capacity and the lung oxidative status were determined. Furthermore, we measured the pulmonary expression and activity for the antioxidant enzymes superoxide dismutase, catalase and glutathione peroxidase, and the oxidative stress markers 8-isoprostanes, 4HNE and nitrotyrosine. Finally, we assessed pulmonary pro-oxidant sources by the expression and function of NADPH oxidase, mitochondria and xanthine oxidase. Melatonin decreased the birth weight. However, melatonin enhanced the plasma antioxidant capacity and decreased the pulmonary antioxidant activity, associated with a diminished oxidative stress during postnatal life. Interestingly, melatonin also decreased ROS generation at the main pro-oxidant sources. Our findings suggest that antenatal administration of melatonin programs an enhanced antioxidant/pro-oxidant status, modulating ROS sources in the postnatal lung.
Collapse
Affiliation(s)
- Alejandro Gonzalez-Candia
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile
| | - Marcelino Veliz
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile
| | - Catalina Carrasco-Pozo
- Departamento de Nutrición, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia, Santiago, Chile
| | - Rodrigo L Castillo
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile; Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - J Cesar Cárdenas
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Germán Ebensperger
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile
| | - Roberto V Reyes
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile
| | - Aníbal J Llanos
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Baquedano s/n, Putre, Chile
| | - Emilio A Herrera
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Av. Salvador 486, Providencia 7500922, Santiago, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Baquedano s/n, Putre, Chile.
| |
Collapse
|
19
|
Melatonin enhances TNF-α-mediated cervical cancer HeLa cells death via suppressing CaMKII/Parkin/mitophagy axis. Cancer Cell Int 2019; 19:58. [PMID: 30923460 PMCID: PMC6419493 DOI: 10.1186/s12935-019-0777-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/08/2019] [Indexed: 12/11/2022] Open
Abstract
Background Tumor necrosis factor-α (TNF-α) immunotherapy controls the progression of human cervical cancer. Here, we explored the detailed molecular mechanisms played by melatonin in human cervical cancer (HeLa cells) death in the presence of TNF-α injury, with a particular attention to the mitochondrial homeostasis. Methods HeLa cells were incubated with TNFα and then cell death was determined via MTT assay, TUNEL staining, caspase ELISA assay and western blotting. Mitochondrial function was detected via analyzing mitochondrial membrane potential using JC-1 staining, mitochondrial oxidative stress using flow cytometry and mitochondrial apoptosis using western blotting. Results Our data exhibited that treatment with HeLa cells using melatonin in the presence of TNF-α further triggered cancer cell cellular death. Molecular investigation demonstrated that melatonin enhanced the caspase-9 mitochondrion death, repressed mitochondrial potential, increased ROS production, augmented mPTP opening rate and elevated cyt-c expression in the nucleus. Moreover, melatonin application further suppressed mitochondrial ATP generation via reducing the expression of mitochondrial respiratory complex. Mechanistically, melatonin augmented the response of HeLa cells to TNF-α-mediated cancer death via repressing mitophagy. TNF-α treatment activated mitophagy via elevating Parkin expression and excessive mitophagy blocked mitochondrial apoptosis, ultimately alleviating the lethal action of TNF-α on HeLa cell. However, melatonin supplementation could prevent TNF-α-mediated mitophagy activation via inhibiting Parkin in a CaMKII-dependent manner. Interestingly, reactivation of CaMKII abolished the melatonin-mediated mitophagy arrest and HeLa cell death. Conclusions Overall, our data highlight that melatonin enhances TNF-α-induced human cervical cancer HeLa cells mitochondrial apoptosis via inactivating the CaMKII/Parkin/mitophagy axis.
Collapse
|
20
|
Ouyang H, Zhou E, Wang H. Mst1-Hippo pathway triggers breast cancer apoptosis via inducing mitochondrial fragmentation in a manner dependent on JNK-Drp1 axis. Onco Targets Ther 2019; 12:1147-1159. [PMID: 30809096 PMCID: PMC6376886 DOI: 10.2147/ott.s193787] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Mst1-Hippo pathway and mitochondrial fragmentation participate in the progression of several types of cancers. However, their roles in breast cancer requires investigation. The aim of our study is to determine whether Mst1 overexpression regulates the viability of breast cancer cells via modulating mitochondrial fragmentation. MATERIALS AND METHODS TUNEL staining, MTT assay and Western blotting were used to detect cancer cell death. Adenovirus-loaded Mst1 was transfected into cells to overexpress Mst1. Mitochondrial fragmentation was observed via immunofluorescence staining and Western blotting. Pathway blocker was used to detect whether Mst1 modulated cell death and mitochondrial fragmentation via JNK signaling pathway. RESULTS Our data showed that Mst1 overexpression promoted breast cancer cell death in a manner dependent on mitochondrial apoptosis. Mitochondrial oxidative stress, energy metabolism disorder, mitochondrial cyt-c liberation and mitochondrial apoptosis activation were observed after Mst1 overexpression. Furthermore, we demonstrated that Mst1 overexpression activated mitochondrial stress via triggering Drp1-related mitochondrial fragmentation, and that inhibition of Drp1-related mitochondrial fragmentation abrogated the proapoptotic effect of Mst1 overexpression on breast cancer cells. To this end, we found that Mst1 modulated Drp1 expression via the JNK signaling pathway, and that blockade of the JNK pathway attenuated mitochondrial stress and repressed apoptosis in Mst1-overexpressed cells. CONCLUSION Altogether, our results identified a tumor suppressive role for Mst1 overexpression in breast cancer via activation of the JNK-Drp1 axis and subsequent initiation of fatal mitochondrial fragmentation. Given these findings, strategies to enhance Mst1 activity and elevate the JNK-Drp1-mitochondrial fragmentation cascade have clinical benefits for patients with breast cancer.
Collapse
Affiliation(s)
- Hui Ouyang
- Department of Breast and Thyroid Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China,
| | - Enxiang Zhou
- Department of Breast and Thyroid Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China,
| | - Huan Wang
- Department of Breast and Thyroid Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China,
| |
Collapse
|
21
|
Chen H, Tang Y, Wang H, Chen W, Jiang H. Curcumin alleviates lipopolysaccharide-induced neuroinflammation in fetal mouse brain. Restor Neurol Neurosci 2018; 36:583-592. [PMID: 30010156 DOI: 10.3233/rnn-180834] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND/OBJECTIVE Curcumin exerts multiple functions including antioxidant and anti-inflammation, and has been shown protective potential on neurological disorders. Maternal or intrauterine infection/inflammation is one of the major factors underlying perinatal brain damage. This study aimed to determine whether maternal administration of curcumin has attenuation on neuroinflammation in fetal brain caused by lipopolysaccharide (LPS) administration. METHODS LPS was used to establish mouse fetal brain injury model, and we investigated the effects of curcumin (40 mg/kg) on the fetal mouse brain by evaluating the morphological change of the neuronal cells and the expression of different pro-inflammatory cytokines and chemokines at protein and mRNA levels in the fetal brains, the maternal serum and amniotic fluid. RESULTS Our results demonstrated that maternal administration of curcumin has attenuation on neuroinflammation in the fetal brain induced by LPS. Pretreatment of curcumin in the LPS-induced mice effectively reestablished the neuronal cell morphology, attenuated the expression of soluble intercellular adhesion molecule-1, sE-Selectin, macrophage chemoattractant protein-1 and cytokine-induced neutrophil chemoattractant-1 in the maternal serum, decreased the expression of cyclooxygenase-2, interleukin-1 beta and chemokine (C-C motif) ligand 2 in the brain, and suppressed interleukin-6 (IL-6) mRNA transcription in the amniotic fluid. In addition, curcumin suppressed the LPS-induced microglia activation. CONCLUSIONS Our study in animal models indicates that maternal administration of curcumin alleviates neuroinflammation in the fetal brain caused by LPS. Long-term consumption of curcumin might improve the neurological outcomes of premature neonates delivered from dams suffering from infection/ inflammation.
Collapse
Affiliation(s)
- Hongyan Chen
- Department of Pediatric, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Pediatric, People's Hospital of Rizhao City, Rizhao, Shandong, China
| | - Yingli Tang
- Department of Pediatric, People's Hospital of Rizhao City, Rizhao, Shandong, China
| | - Hanyan Wang
- Department of Pediatric, People's Hospital of Rizhao City, Rizhao, Shandong, China
| | - Weibing Chen
- Department of Pediatric, People's Hospital of Rizhao City, Rizhao, Shandong, China
| | - Hong Jiang
- Department of Pediatric, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
22
|
Corpas R, Griñán-Ferré C, Palomera-Ávalos V, Porquet D, García de Frutos P, Franciscato Cozzolino SM, Rodríguez-Farré E, Pallàs M, Sanfeliu C, Cardoso BR. Melatonin induces mechanisms of brain resilience against neurodegeneration. J Pineal Res 2018; 65:e12515. [PMID: 29907977 DOI: 10.1111/jpi.12515] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/06/2018] [Indexed: 12/20/2022]
Abstract
Melatonin is an endogenous pleiotropic molecule which orchestrates regulatory functions and protective capacity against age-related ailments. The increase in circulating levels of melatonin through dietary supplements intensifies its health benefits. Investigations in animal models have shown that melatonin protects against Alzheimer's disease (AD)-like pathology, although clinical studies have not been conclusive. We hypothesized that melatonin induces changes in the brain that prevent or attenuate AD by increasing resilience. Therefore, we treated healthy nontransgenic (NoTg) and AD transgenic (3xTg-AD) 6-month-old mice with a daily dose of 10 mg/kg of melatonin until 12 months of age. As expected, melatonin reversed cognitive impairment and dementia-associated behaviors of anxiety and apathy and reduced amyloid and tau burden in 3xTg-AD mice. Remarkably, melatonin induced cognitive enhancement and higher wellness level-related behavior in NoTg mice. At the mechanism level, NF-κB and proinflammatory cytokine expressions were decreased in both NoTg and 3xTg-AD mice. The SIRT1 pathway of longevity and neuroprotection was also activated in both mouse strains after melatonin dosing. Furthermore, we explored new mechanisms and pathways not previously associated with melatonin treatment such as the ubiquitin-proteasome proteolytic system and the recently proposed neuroprotective Gas6/TAM pathway. The upregulation of proteasome activity and the modulation of Gas6 and its receptors by melatonin were similarly displayed by both NoTg and 3xTg-AD mice. Therefore, these results confirm the potential of melatonin treatment against AD pathology, by way of opening new pathways in its mechanisms of action, and demonstrating that melatonin induces cognitive enhancement and brain resilience against neurodegenerative processes.
Collapse
Affiliation(s)
- Rubén Corpas
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC and IDIBAPS, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Christian Griñán-Ferré
- Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona and CIBERNED, Barcelona, Spain
| | - Verónica Palomera-Ávalos
- Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona and CIBERNED, Barcelona, Spain
| | - David Porquet
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC and IDIBAPS, Barcelona, Spain
| | - Pablo García de Frutos
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC and IDIBAPS, Barcelona, Spain
| | - Silvia M Franciscato Cozzolino
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eduard Rodríguez-Farré
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC and IDIBAPS, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Mercè Pallàs
- Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona and CIBERNED, Barcelona, Spain
| | - Coral Sanfeliu
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC and IDIBAPS, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Bárbara R Cardoso
- Institute for Physical Activity and Nutrition Research (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Vic., Australia
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Vic., Australia
| |
Collapse
|
23
|
Robinson S, Conteh FS, Oppong AY, Yellowhair TR, Newville JC, Demerdash NE, Shrock CL, Maxwell JR, Jett S, Northington FJ, Jantzie LL. Extended Combined Neonatal Treatment With Erythropoietin Plus Melatonin Prevents Posthemorrhagic Hydrocephalus of Prematurity in Rats. Front Cell Neurosci 2018; 12:322. [PMID: 30319361 PMCID: PMC6167494 DOI: 10.3389/fncel.2018.00322] [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: 08/04/2018] [Accepted: 09/06/2018] [Indexed: 12/15/2022] Open
Abstract
Posthemorrhagic hydrocephalus of prematurity (PHHP) remains a global challenge. Early preterm infants (<32 weeks gestation), particularly those exposed to chorioamnionitis (CAM), are prone to intraventricular hemorrhage (IVH) and PHHP. We established an age-appropriate, preclinical model of PHHP with progressive macrocephaly and ventriculomegaly to test whether non-surgical neonatal treatment could modulate PHHP. We combined prenatal CAM and postnatal day 1 (P1, equivalent to 30 weeks human gestation) IVH in rats, and administered systemic erythropoietin (EPO) plus melatonin (MLT), or vehicle, from P2 to P10. CAM-IVH rats developed progressive macrocephaly through P21. Macrocephaly was accompanied by ventriculomegaly at P5 (histology), and P21 (ex vivo MRI). CAM-IVH rats showed impaired performance of cliff aversion, a neonatal neurodevelopmental test. Neonatal EPO+MLT treatment prevented macrocephaly and cliff aversion impairment, and significantly reduced ventriculomegaly. EPO+MLT treatment prevented matted or missing ependymal motile cilia observed in vehicle-treated CAM-IVH rats. EPO+MLT treatment also normalized ependymal yes-associated protein (YAP) mRNA levels, and reduced ependymal GFAP-immunolabeling. Vehicle-treated CAM-IVH rats exhibited loss of microstructural integrity on diffusion tensor imaging, which was normalized in EPO+MLT-treated CAM-IVH rats. In summary, combined prenatal systemic inflammation plus early postnatal IVH caused progressive macrocephaly, ventriculomegaly and delayed development of cliff aversion reminiscent of PHHP. Neonatal systemic EPO+MLT treatment prevented multiple hallmarks of PHHP, consistent with a clinically viable, non-surgical treatment strategy.
Collapse
Affiliation(s)
- Shenandoah Robinson
- Division of Pediatric Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Fatu S Conteh
- Division of Pediatric Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Akosua Y Oppong
- Division of Pediatric Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Tracylyn R Yellowhair
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Jessie C Newville
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.,Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Nagat El Demerdash
- Division of Pediatric Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Christine L Shrock
- Division of Pediatric Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Jessie R Maxwell
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Stephen Jett
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Frances J Northington
- Division of Neonatology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Lauren L Jantzie
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.,Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
24
|
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: 2.0] [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
|
25
|
Liu Y, Choe J, Lee JJ, Kim J, Campbell JM, Polo J, Crenshaw JD, Pettigrew JE, Song M. Spray-dried plasma attenuates inflammation and lethargic behaviors of pregnant mice caused by lipopolysaccharide. PLoS One 2018; 13:e0203427. [PMID: 30208071 PMCID: PMC6135513 DOI: 10.1371/journal.pone.0203427] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 08/21/2018] [Indexed: 01/15/2023] Open
Abstract
This study evaluated whether dietary spray-dried plasma (SDP) can ameliorate inflammation, lethargic behaviors, and impairment of reproduction caused by lipopolysaccharide (LPS) challenge during late pregnancy. Two experiments were conducted with 125 mated female mice (C57BL/6 strain) in each experiment. All mice were shipped from a vendor on the gestation day (GD) 1 and arrived at the laboratory on GD 3. Mice were randomly assigned to dietary treatments with or without 8% SDP in the diet. On GD 17, mice determined pregnant by BW and abdomen shape were randomly assigned to intraperitoneal injections with or without 2 μg LPS. In experiment 1, 17 mice (26.7 ± 1.7 g BW) were identified pregnant and euthanized 6 h after the LPS challenge to measure inflammatory responses in uterus and placenta. In experiment 2, 44 mice (26.0 ± 1.6 g BW) were identified pregnant and euthanized 24 h after the LPS challenge to assess behavior and late-term pregnancy loss. Growth performance and reproductive responses, such as loss of pregnancy, percentage of fetal death, and etc., were measured in all pregnant mice. The LPS challenge increased (P < 0.05) uterine and placental tumor necrosis factor-α and interferon-γ, late-term pregnancy loss, and lethargy score, and decreased (P < 0.05) uterine transforming growth factor-β1, moving time and number of rearing, and growth and feed intake. The SDP decreased (P < 0.05) concentrations of both pro-inflammatory and anti-inflammatory cytokines in one or both tissues, and the lethargy score, and increased (P < 0.05) moving time and number of rearing, growth of pregnant mice, and fetal weight. However, the SDP did not affect late-term pregnancy loss caused by the LPS challenge. Consequently, dietary SDP attenuated acute inflammation and lethargic behaviors of pregnant mice caused by the LPS challenge, but did not affect late-term pregnancy loss after the acute inflammation.
Collapse
Affiliation(s)
- Yanhong Liu
- Department of Animal Science, University of California, Davis, California, United States of America
| | - Jeehwan Choe
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong Jae Lee
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Junsu Kim
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | | | - Javier Polo
- APC Inc., Ankeny, Iowa, United States of America
| | | | - James E. Pettigrew
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, United States of America
- * E-mail: (JEP); (MS)
| | - Minho Song
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
- * E-mail: (JEP); (MS)
| |
Collapse
|
26
|
Abstract
Pregnancy often predisposes women to new-onset sleep disturbances, as well as exacerbations of preexisting sleep disorders. The goals of treating perinatal sleep disorders include the promotion of restorative sleep and the benefits it brings to both mother and fetus. The prescribing of any sleep aid in pregnancy must include consideration of the risks and benefits for both the patient and her fetus. Although data on the perinatal use of sleep aids is limited, there may be effects on fetal development, timing and duration of delivery, and postnatal outcomes.
Collapse
Affiliation(s)
- Laura P McLafferty
- Department of Psychiatry and Human Behavior, Thomas Jefferson University, Thompson Building, Suite 1652, 1020 Sansom Street, Philadelphia, PA 19107, USA.
| | - Meredith Spada
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh Medical Center, 3811 O'Hara St., Pittsburgh, PA 15213, USA
| | - Priya Gopalan
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh Medical Center, 3811 O'Hara St., Pittsburgh, PA 15213, USA
| |
Collapse
|
27
|
Jantzie LL, Oppong AY, Conteh FS, Yellowhair TR, Kim J, Fink G, Wolin AR, Northington FJ, Robinson S. Repetitive Neonatal Erythropoietin and Melatonin Combinatorial Treatment Provides Sustained Repair of Functional Deficits in a Rat Model of Cerebral Palsy. Front Neurol 2018; 9:233. [PMID: 29706928 PMCID: PMC5908903 DOI: 10.3389/fneur.2018.00233] [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: 12/09/2017] [Accepted: 03/26/2018] [Indexed: 12/21/2022] Open
Abstract
Cerebral palsy (CP) is the leading cause of motor impairment for children worldwide and results from perinatal brain injury (PBI). To test novel therapeutics to mitigate deficits from PBI, we developed a rat model of extreme preterm birth (<28 weeks of gestation) that mimics dual intrauterine injury from placental underperfusion and chorioamnionitis. We hypothesized that a sustained postnatal treatment regimen that combines the endogenous neuroreparative agents erythropoietin (EPO) and melatonin (MLT) would mitigate molecular, sensorimotor, and cognitive abnormalities in adults rats following prenatal injury. On embryonic day 18 (E18), a laparotomy was performed in pregnant Sprague–Dawley rats. Uterine artery occlusion was performed for 60 min to induce placental insufficiency via transient systemic hypoxia-ischemia, followed by intra-amniotic injections of lipopolysaccharide, and laparotomy closure. On postnatal day 1 (P1), approximately equivalent to 30 weeks of gestation, injured rats were randomized to an extended EPO + MLT treatment regimen, or vehicle (sterile saline) from P1 to P10. Behavioral assays were performed along an extended developmental time course (n = 6–29). Open field testing shows injured rats exhibit hypermobility and disinhibition and that combined neonatal EPO + MLT treatment repairs disinhibition in injured rats, while EPO alone does not. Furthermore, EPO + MLT normalizes hindlimb deficits, including reduced paw area and paw pressure at peak stance, and elevated percent shared stance after prenatal injury. Injured rats had fewer social interactions than shams, and EPO + MLT normalized social drive. Touchscreen operant chamber testing of visual discrimination and reversal shows that EPO + MLT at least partially normalizes theses complex cognitive tasks. Together, these data indicate EPO + MLT can potentially repair multiple sensorimotor, cognitive, and behavioral realms following PBI, using highly translatable and sophisticated developmental testing platforms.
Collapse
Affiliation(s)
- Lauren L Jantzie
- Department of Pediatrics, University of New Mexico School of Medicine, University of New Mexico, Albuquerque, NM, United States.,Department of Neurosciences, University of New Mexico School of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Akosua Y Oppong
- Pediatric Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| | - Fatu S Conteh
- Pediatric Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| | - Tracylyn R Yellowhair
- Department of Pediatrics, University of New Mexico School of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Joshua Kim
- Pediatric Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| | - Gabrielle Fink
- Pediatric Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| | - Adam R Wolin
- Pediatric Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| | - Frances J Northington
- Neonatology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Shenandoah Robinson
- Pediatric Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|