1
|
Endesfelder S. Caffeine: The Story beyond Oxygen-Induced Lung and Brain Injury in Neonatal Animal Models-A Narrative Review. Antioxidants (Basel) 2024; 13:1076. [PMID: 39334735 PMCID: PMC11429035 DOI: 10.3390/antiox13091076] [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: 07/31/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Caffeine is one of the most commonly used drugs in intensive care to stimulate the respiratory control mechanisms of very preterm infants. Respiratory instability, due to the degree of immaturity at birth, results in apnea of prematurity (AOP), hyperoxic, hypoxic, and intermittent hypoxic episodes. Oxidative stress cannot be avoided as a direct reaction and leads to neurological developmental deficits and even a higher prevalence of respiratory diseases in the further development of premature infants. Due to the proven antioxidant effect of caffeine in early use, largely protective effects on clinical outcomes can be observed. This is also impressively observed in experimental studies of caffeine application in oxidative stress-adapted rodent models of damage to the developing brain and lungs. However, caffeine shows undesirable effects outside these oxygen toxicity injury models. This review shows the effects of caffeine in hyperoxic, hypoxic/hypoxic-ischemic, and intermittent hypoxic rodent injury models, but also the negative effects on the rodent organism when caffeine is administered without exogenous oxidative stress. The narrative analysis of caffeine benefits in cerebral and pulmonary preterm infant models supports protective caffeine use but should be given critical consideration when considering caffeine treatment beyond the recommended corrected gestational age.
Collapse
Affiliation(s)
- Stefanie Endesfelder
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
2
|
Song Y, Yang C. Mechanistic advances of hyperoxia-induced immature brain injury. Heliyon 2024; 10:e30005. [PMID: 38694048 PMCID: PMC11058899 DOI: 10.1016/j.heliyon.2024.e30005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/11/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024] Open
Abstract
The impact of hyperoxia-induced brain injury in preterm infants is being increasingly investigated. However, the parameters and protocols used to study this condition in animal models lack consistency. Research is further hampered by the fact that hyperoxia exerts both direct and indirect effects on oligodendrocytes and neurons, with the precise underlying mechanisms remaining unclear. In this article, we aim to provide a comprehensive overview of the conditions used to induce hyperoxia in animal models of immature brain injury. We discuss what is known regarding the mechanisms underlying hyperoxia-induced immature brain injury, focusing on the effects on oligodendrocytes and neurons, and briefly describe therapies that may counteract the effects of hyperoxia. We also identify further studies required to fully elucidate the effects of hyperoxia on the immature brain as well as discuss the leading therapeutic options.
Collapse
Affiliation(s)
- Yue Song
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, China
- Department of Clinical Medicine, The Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| | - Changqiang Yang
- Department of Cardiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan Province, China
- Department of Clinical Medicine, The Chengdu Medical College, Chengdu 610500, Sichuan Province, China
| |
Collapse
|
3
|
Hencz AJ, Magony A, Thomas C, Kovacs K, Szilagyi G, Pal J, Sik A. Short-term hyperoxia-induced functional and morphological changes in rat hippocampus. Front Cell Neurosci 2024; 18:1376577. [PMID: 38686017 PMCID: PMC11057248 DOI: 10.3389/fncel.2024.1376577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/27/2024] [Indexed: 05/02/2024] Open
Abstract
Excess oxygen (O2) levels may have a stimulating effect, but in the long term, and at high concentrations of O2, it is harmful to the nervous system. The hippocampus is very sensitive to pathophysiological changes and altered O2 concentrations can interfere with hippocampus-dependent learning and memory functions. In this study, we investigated the hyperoxia-induced changes in the rat hippocampus to evaluate the short-term effect of mild and severe hyperoxia. Wistar male rats were randomly divided into control (21% O2), mild hyperoxia (30% O2), and severe hyperoxia groups (100% O2). The O2 exposure lasted for 60 min. Multi-channel silicon probes were used to study network oscillations and firing properties of hippocampal putative inhibitory and excitatory neurons. Neural damage was assessed using the Gallyas silver impregnation method. Mild hyperoxia (30% O2) led to the formation of moderate numbers of silver-impregnated "dark" neurons in the hippocampus. On the other hand, exposure to 100% O2 was associated with a significant increase in the number of "dark" neurons located mostly in the hilus. The peak frequency of the delta oscillation decreased significantly in both mild and severe hyperoxia in urethane anesthetized rats. Compared to normoxia, the firing activity of pyramidal neurons under hyperoxia increased while it was more heterogeneous in putative interneurons in the cornu ammonis area 1 (CA1) and area 3 (CA3). These results indicate that short-term hyperoxia can change the firing properties of hippocampal neurons and network oscillations and damage neurons. Therefore, the use of elevated O2 concentration inhalation in hospitals (i.e., COVID treatment and surgery) and in various non-medical scenarios (i.e., airplane emergency O2 masks, fire-fighters, and high altitude trekkers) must be used with extreme caution.
Collapse
Affiliation(s)
| | - Andor Magony
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| | - Chloe Thomas
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Krisztina Kovacs
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gabor Szilagyi
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary
| | - Jozsef Pal
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| | - Attila Sik
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
4
|
Jackson W, Gonzalez D, Greenberg RG, Lee YZ, Laughon MM. A phase I trial of caffeine to evaluate safety in infants with hypoxic-ischemic encephalopathy. J Perinatol 2024; 44:508-512. [PMID: 37587184 PMCID: PMC10869636 DOI: 10.1038/s41372-023-01752-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
OBJECTIVE Caffeine provides neuroprotection following hypoxic-ischemic injury in animals. We characterized the safety of escalating doses of caffeine in infants with hypoxic-ischemic encephalopathy (HIE) receiving therapeutic hypothermia. STUDY DESIGN Phase I trial of infants undergoing therapeutic hypothermia for HIE receiving IV caffeine 20 mg/kg followed by up to two daily doses of 5 mg/kg (n = 9) or 10 mg/kg (n = 8). Safety was evaluated based on adverse events and frequency of pre-specified outcomes compared to data from the Whole-Body Hypothermia for HIE trial (Shankaran, 2005). RESULTS Twelve of 17 (71%) infants had ≥1 adverse event during the study period. The frequency of clinical outcomes related to HIE were not statistically different from outcomes in infants receiving hypothermia in the Whole-Body Hypothermia for HIE trial. CONCLUSION Caffeine administration was well tolerated. A larger study is required to determine the optimal dose and evaluate drug safety and efficacy. CLINICAL TRIAL ClinicalTrials.gov Identifier: NCT03913221.
Collapse
Affiliation(s)
- Wesley Jackson
- Department of Pediatrics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rachel G Greenberg
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Yueh Z Lee
- Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew M Laughon
- Department of Pediatrics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
5
|
Xue-Jiao H, Jian-Hua F. A review of the effects of early postnatal hyperoxia exposure on the immature brain. Exp Neurol 2023; 370:114550. [PMID: 37774766 DOI: 10.1016/j.expneurol.2023.114550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023]
Abstract
Preterm birth is a public health priority worldwide, with approximately 15 million premature babies born each year. Oxygen supplementation is one of the most common interventions for preterm infants. However, prolonged oxygen inhalation at supraphysiological concentrations can lead to the development of bronchopulmonary dysplasia (BPD). In addition to lifelong pulmonary sequelae, clinical evidence suggests that BPD is associated with adverse neurodevelopmental outcomes, such as motor impairment, cognitive impairment, and behavioral deficits, severely affecting the quality of life of preterm infants. However, the mechanisms underlying the combination of neurodevelopmental impairment with BPD remain unclear. Therefore, in recent years, attention has also been focused on the effects of hyperoxia on brain development in preterm infants. In this review, we outline the pathophysiological mechanisms of brain injury caused by developmental hyperoxia exposure in current animal models and briefly describe the pharmacological therapies that may be applicable to the associated brain injury. Overall, more studies are needed to assess the effects of hyperoxia on the immature brain, particularly combined analyses of the lungs and brain in the same experimental setting, to elucidate the potential causes of combined neurodevelopmental impairment in BPD.
Collapse
Affiliation(s)
- Huang Xue-Jiao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fu Jian-Hua
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
6
|
Shankar N, Thapa S, Shrestha AK, Sarkar P, Gaber MW, Barrios R, Shivanna B. Hyperoxia Disrupts Lung Lymphatic Homeostasis in Neonatal Mice. Antioxidants (Basel) 2023; 12:620. [PMID: 36978868 PMCID: PMC10045755 DOI: 10.3390/antiox12030620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Inflammation causes bronchopulmonary dysplasia (BPD), a common lung disease of preterm infants. One reason this disease lacks specific therapies is the paucity of information on the mechanisms regulating inflammation in developing lungs. We address this gap by characterizing the lymphatic phenotype in an experimental BPD model because lymphatics are major regulators of immune homeostasis. We hypothesized that hyperoxia (HO), a major risk factor for experimental and human BPD, disrupts lymphatic endothelial homeostasis using neonatal mice and human dermal lymphatic endothelial cells (HDLECs). Exposure to 70% O2 for 24-72 h decreased the expression of prospero homeobox 1 (Prox1) and vascular endothelial growth factor c (Vegf-c) and increased the expression of heme oxygenase 1 and NAD(P)H dehydrogenase [quinone]1 in HDLECs, and reduced their tubule formation ability. Next, we determined Prox1 and Vegf-c mRNA levels on postnatal days (P) 7 and 14 in neonatal murine lungs. The mRNA levels of these genes increased from P7 to P14, and 70% O2 exposure for 14 d (HO) attenuated this physiological increase in pro-lymphatic factors. Further, HO exposure decreased VEGFR3+ and podoplanin+ lymphatic vessel density and lymphatic function in neonatal murine lungs. Collectively, our results validate the hypothesis that HO disrupts lymphatic endothelial homeostasis.
Collapse
Affiliation(s)
- Nithyapriya Shankar
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Shyam Thapa
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Amrit Kumar Shrestha
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Poonam Sarkar
- Division of Hematology-Oncology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - M. Waleed Gaber
- Division of Hematology-Oncology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Binoy Shivanna
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| |
Collapse
|
7
|
Heise J, Schmitz T, Bührer C, Endesfelder S. Protective Effects of Early Caffeine Administration in Hyperoxia-Induced Neurotoxicity in the Juvenile Rat. Antioxidants (Basel) 2023; 12:295. [PMID: 36829854 PMCID: PMC9952771 DOI: 10.3390/antiox12020295] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
High-risk preterm infants are affected by a higher incidence of cognitive developmental deficits due to the unavoidable risk factor of oxygen toxicity. Caffeine is known to have a protective effect in preventing bronchopulmonary dysplasia associated with improved neurologic outcomes, although very early initiation of therapy is controversial. In this study, we used newborn rats in an oxygen injury model to test the hypothesis that near-birth caffeine administration modulates neuronal maturation and differentiation in the hippocampus of the developing brain. For this purpose, newborn Wistar rats were exposed to 21% or 80% oxygen on the day of birth for 3 or 5 days and treated with vehicle or caffeine (10 mg/kg/48 h). Postnatal exposure to 80% oxygen resulted in a drastic reduction of associated neuronal mediators for radial glia, mitotic/postmitotic neurons, and impaired cell-cycle regulation, predominantly persistent even after recovery to room air until postnatal day 15. Systemic caffeine administration significantly counteracted the effects of oxygen insult on neuronal maturation in the hippocampus. Interestingly, under normoxia, caffeine inhibited the transcription of neuronal mediators of maturing and mature neurons. The early administration of caffeine modulated hyperoxia-induced decreased neurogenesis in the hippocampus and showed neuroprotective properties in the neonatal rat oxygen toxicity model.
Collapse
Affiliation(s)
| | | | | | - Stefanie Endesfelder
- Department of Neonatology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
8
|
McLeod RM, Rosenkrantz TS, Fitch RH, Koski RR. Sex Differences in Microglia Activation in a Rodent Model of Preterm Hypoxic Ischemic Injury with Caffeine Treatment. Biomedicines 2023; 11:biomedicines11010185. [PMID: 36672692 PMCID: PMC9855625 DOI: 10.3390/biomedicines11010185] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
Preterm infants are often treated with caffeine as a respiratory stimulant. However, follow-up data shows caffeine may also have neuroprotective potential. There are several theories as to how caffeine might protect the brain, but none have been proven. This study looked at caffeine effects on microglial activation in rodent brains post hypoxic ischemic (HI) injury. Rat pups underwent either sham or HI surgery on P6, followed by treatment with either caffeine or saline. Forty-eight hours post-injury, brains were collected and underwent paraffin embedding and sectioning followed by immunofluorescence staining. Ionized calcium binding adaptor molecule 1 (Iba-1) was used to label microglia, and 4',6-diamindino-2-phenylindole (DAPI) was used to label DNA. Cell size measurements of microglia were obtained to gauge microglia activation, and chromatin condensation (DAPI optical density) was used as an index of neuronal cell death. Results suggest that caffeine does offer protective effects, based on significantly increased levels of cell death in HI-saline animals not seen in caffeine-treated HI males and females. However, the mechanism of action may be different. Male HI animals showed marginally reduced microglial activation following caffeine treatment, whereas females did not. Results indicate that though caffeine may act protectively in both sexes by reducing cell death, the benefits may be mediated by different mechanisms.
Collapse
Affiliation(s)
- Ruth Mae McLeod
- Behavioral Neuroscience Division, Department of Psychological Sciences, University of Connecticut, Storrs, CT 06269, USA
- Correspondence:
| | - Ted S. Rosenkrantz
- Department of Pediatrics, University of Connecticut Health Center and Connecticut Children’s Hospital, Farmington, CT 06030, USA
| | - Roslyn Holly Fitch
- Behavioral Neuroscience Division, Department of Psychological Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Rachel R. Koski
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA
| |
Collapse
|
9
|
Klein L, Van Steenwinckel J, Fleiss B, Scheuer T, Bührer C, Faivre V, Lemoine S, Blugeon C, Schwendimann L, Csaba Z, Bokobza C, Vousden DA, Lerch JP, Vernon AC, Gressens P, Schmitz T. A unique cerebellar pattern of microglia activation in a mouse model of encephalopathy of prematurity. Glia 2022; 70:1699-1719. [PMID: 35579329 PMCID: PMC9545095 DOI: 10.1002/glia.24190] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/24/2022]
Abstract
Preterm infants often show pathologies of the cerebellum, which are associated with impaired motor performance, lower IQ and poor language skills at school ages. Using a mouse model of inflammation-induced encephalopathy of prematurity driven by systemic administration of pro-inflammatory IL-1β, we sought to uncover causes of cerebellar damage. In this model, IL-1β is administered between postnatal day (P) 1 to day 5, a timing equivalent to the last trimester for brain development in humans. Structural MRI analysis revealed that systemic IL-1β treatment induced specific reductions in gray and white matter volumes of the mouse cerebellar lobules I and II (5% false discovery rate [FDR]) from P15 onwards. Preceding these MRI-detectable cerebellar volume changes, we observed damage to oligodendroglia, with reduced proliferation of OLIG2+ cells at P10 and reduced levels of the myelin proteins myelin basic protein (MBP) and myelin-associated glycoprotein (MAG) at P10 and P15. Increased density of IBA1+ cerebellar microglia were observed both at P5 and P45, with evidence for increased microglial proliferation at P5 and P10. Comparison of the transcriptome of microglia isolated from P5 cerebellums and cerebrums revealed significant enrichment of pro-inflammatory markers in microglia from both regions, but cerebellar microglia displayed a unique type I interferon signaling dysregulation. Collectively, these data suggest that perinatal inflammation driven by systemic IL-1β leads to specific cerebellar volume deficits, which likely reflect oligodendrocyte pathology downstream of microglial activation. Further studies are now required to confirm the potential of protective strategies aimed at preventing sustained type I interferon signaling driven by cerebellar microglia as an important therapeutic target.
Collapse
Affiliation(s)
- Luisa Klein
- Department of NeonatologyCharité University Medicine BerlinBerlinGermany
| | | | - Bobbi Fleiss
- NeuroDiderot, InsermUniversité de ParisParisFrance
- School of Health and Biomedical SciencesRMIT UniversityMelbourneVictoriaAustralia
| | - Till Scheuer
- Department of NeonatologyCharité University Medicine BerlinBerlinGermany
| | - Christoph Bührer
- Department of NeonatologyCharité University Medicine BerlinBerlinGermany
| | | | - Sophie Lemoine
- Genomics Core Facility, Département de Biologie, École Normale Supérieure, Institut de Biologie de l'ENS (IBENS), CNRS, INSERMUniversité PSLParisFrance
| | - Corinne Blugeon
- Genomics Core Facility, Département de Biologie, École Normale Supérieure, Institut de Biologie de l'ENS (IBENS), CNRS, INSERMUniversité PSLParisFrance
| | | | - Zsolt Csaba
- NeuroDiderot, InsermUniversité de ParisParisFrance
| | | | - Dulcie A. Vousden
- Mouse Imaging CentreThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Jason P. Lerch
- Mouse Imaging CentreThe Hospital for Sick ChildrenTorontoOntarioCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioCanada
- Wellcome Trust Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
| | - Anthony C. Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
| | | | - Thomas Schmitz
- Department of NeonatologyCharité University Medicine BerlinBerlinGermany
| |
Collapse
|
10
|
Hu J, Cheng Y, Chen P, Huang Z, Yang L. Caffeine Citrate Protects Against Sepsis-Associated Encephalopathy and Inhibits the UCP2/NLRP3 Axis in Astrocytes. J Interferon Cytokine Res 2022; 42:267-278. [PMID: 35420462 DOI: 10.1089/jir.2021.0241] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a diffuse brain dysfunction without overt central nervous system infection. Caffeine citrate has therapeutic effect on different brain diseases, while its role in SAE remains unclear. The expression levels of interleukin (IL)-18 and IL-1β were upregulated in the cerebrospinal fluid of the subjects. In this study, a rat model of SAE was established by cecal ligation and puncture. Caffeine citrate inhibited SAE-induced neuronal apoptosis and astrocytic activation, decreased reactive oxygen species (ROS) generation, and elevated mitochondrial membrane potential (MMP) level in the cerebral cortex. In vitro, primary astrocytes were isolated from rat cerebral cortex and incubated with lipopolysaccharide (LPS) and interferon-γ (IFN-γ). Caffeine citrate reduced ROS and MMP levels and mitochondrial complex enzyme activities in LPS plus IFN-γ-induced astrocytes. Moreover, caffeine citrate inhibited the activation of nucleotide-binding and oligomerization domain (NOD)-like receptor (NLRP3) inflammasome and decreased the production of IL-1β and IL-18 in vivo and in vitro. Notably, caffeine citrate promoted UCP2 expression in astrocytes. The neuroprotective role of UCP2 has been reported in several experimental brain diseases. These results suggest that caffeine citrate inhibits neuronal apoptosis, astrocytic activation, mitochondrial dysfunction in rat cerebral cortex, thereby alleviating SAE. The protection of caffeine citrate against SAE may be achieved by the UCP2-mediated NLRP3 pathway inhibition in astrocytes.
Collapse
Affiliation(s)
- Jing Hu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Yan Cheng
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Ping Chen
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Zhaoqi Huang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Liqi Yang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
11
|
Giszas V, Strauß E, Bührer C, Endesfelder S. The Conflicting Role of Caffeine Supplementation on Hyperoxia-Induced Injury on the Cerebellar Granular Cell Neurogenesis of Newborn Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5769784. [PMID: 35693697 PMCID: PMC9175096 DOI: 10.1155/2022/5769784] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/04/2022] [Accepted: 04/28/2022] [Indexed: 11/28/2022]
Abstract
Preterm birth disrupts cerebellar development, which may be mediated by systemic oxidative stress that damages neuronal developmental stages. Impaired cerebellar neurogenesis affects several downstream targets important for cognition, emotion, and speech. In this study, we demonstrate that oxidative stress induced with high oxygen (80%) for three or five postnatal days (P3/P5) could significantly damage neurogenesis and proliferative capacity of granular cell precursor and Purkinje cells in rat pups. Reversal of cellular neuronal damage after recovery to room air (P15) was augmented by treatment with caffeine. However, downstream transcripts important for migration and differentiation of postmitotic granular cells were irreversibly reduced by hyperoxia, without rescue by caffeine. Protective effects of caffeine in the cerebellum were limited to neuronal survival but failed to restore important transcript signatures.
Collapse
Affiliation(s)
- Vivien Giszas
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Evelyn Strauß
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
12
|
Neonatal Oxidative Stress Impairs Cortical Synapse Formation and GABA Homeostasis in Parvalbumin-Expressing Interneurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8469756. [PMID: 35663195 PMCID: PMC9159830 DOI: 10.1155/2022/8469756] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/08/2022] [Indexed: 11/28/2022]
Abstract
Neonatal brain injury is often caused by preterm birth. Brain development is vulnerable to increased environmental stress, including oxidative stress challenges. Due to a premature change of the fetal living environment from low oxygen in utero into postnatal high-oxygen room air conditions ex utero, the immature preterm brain is exposed to a relative hyperoxia, which can induce oxidative stress and impair neuronal cell development. To simulate the drastic increase of oxygen exposure in the immature brain, 5-day-old C57BL/6 mice were exposed to hyperoxia (80% oxygen) for 48 hours or kept in room air (normoxia, 21% oxygen) and mice were analyzed for maturational alterations of cortical GABAergic interneurons. As a result, oxidative stress was indicated by elevated tyrosine nitration of proteins. We found perturbation of perineuronal net formation in line with decreased density of parvalbumin-expressing (PVALB) cortical interneurons in hyperoxic mice. Moreover, maturational deficits of cortical PVALB+ interneurons were obtained by decreased glutamate decarboxylase 67 (GAD67) protein expression in Western blot analysis and lower gamma-aminobutyric acid (GABA) fluorescence intensity in immunostaining. Hyperoxia-induced oxidative stress affected cortical synaptogenesis by decreasing synapsin 1, synapsin 2, and synaptophysin expression. Developmental delay of synaptic marker expression was demonstrated together with decreased PI3K-signaling as a pathway being involved in synaptogenesis. These results elucidate that neonatal oxidative stress caused by increased oxygen exposure can lead to GABAergic interneuron damage which may serve as an explanation for the high incidence of psychiatric and behavioral alterations found in preterm infants.
Collapse
|
13
|
Perinatal Hyperoxia and Developmental Consequences on the Lung-Brain Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5784146. [PMID: 35251477 PMCID: PMC8894035 DOI: 10.1155/2022/5784146] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022]
Abstract
Approximately 11.1% of all newborns worldwide are born preterm. Improved neonatal intensive care significantly increased survival rates over the last decades but failed to reduce the risk for the development of chronic lung disease (i.e., bronchopulmonary dysplasia (BPD)) and impaired neurodevelopment (i.e., encephalopathy of prematurity (EoP)), two major long-term sequelae of prematurity. Premature infants are exposed to relative hyperoxia, when compared to physiological in-utero conditions and, if needed to additional therapeutic oxygen supplementation. Both are associated with an increased risk for impaired organ development. Since the detrimental effects of hyperoxia on the immature retina are known for many years, lung and brain have come into focus in the last decade. Hyperoxia-induced excessive production of reactive oxygen species leading to oxidative stress and inflammation contribute to pulmonary growth restriction and abnormal neurodevelopment, including myelination deficits. Despite a large body of studies, which unraveled important pathophysiological mechanisms for both organs at risk, the majority focused exclusively either on lung or on brain injury. However, considering that preterm infants suffering from BPD are at higher risk for poor neurodevelopmental outcome, an interaction between both organs seems plausible. This review summarizes recent findings regarding mechanisms of hyperoxia-induced neonatal lung and brain injury. We will discuss common pathophysiological pathways, which potentially link both injured organ systems. Furthermore, promises and needs of currently suggested therapies, including pharmacological and regenerative cell-based treatments for BPD and EoP, will be emphasized. Limited therapeutic approaches highlight the urgent need for a better understanding of the mechanisms underlying detrimental effects of hyperoxia on the lung-brain axis in order to pave the way for the development of novel multimodal therapies, ideally targeting both severe preterm birth-associated complications.
Collapse
|
14
|
Scheuer T, dem Brinke EA, Grosser S, Wolf SA, Mattei D, Sharkovska Y, Barthel PC, Endesfelder S, Friedrich V, Bührer C, Vida I, Schmitz T. Reduction of cortical parvalbumin-expressing GABAergic interneurons in a rodent hyperoxia model of preterm birth brain injury with deficits in social behavior and cognition. Development 2021; 148:272278. [PMID: 34557899 DOI: 10.1242/dev.198390] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 09/17/2021] [Indexed: 12/18/2022]
Abstract
The inhibitory GABAergic system in the brain is involved in the etiology of various psychiatric problems, including autism spectrum disorders (ASD), attention deficit hyperactivity disorder (ADHD) and others. These disorders are influenced not only by genetic but also by environmental factors, such as preterm birth, although the underlying mechanisms are not known. In a translational hyperoxia model, exposing mice pups at P5 to 80% oxygen for 48 h to mimic a steep rise of oxygen exposure caused by preterm birth from in utero into room air, we documented a persistent reduction of cortical mature parvalbumin-expressing interneurons until adulthood. Developmental delay of cortical myelin was observed, together with decreased expression of oligodendroglial glial cell-derived neurotrophic factor (GDNF), a factor involved in interneuronal development. Electrophysiological and morphological properties of remaining interneurons were unaffected. Behavioral deficits were observed for social interaction, learning and attention. These results demonstrate that neonatal oxidative stress can lead to decreased interneuron density and to psychiatric symptoms. The obtained cortical myelin deficit and decreased oligodendroglial GDNF expression indicate that an impaired oligodendroglial-interneuronal interplay contributes to interneuronal damage.
Collapse
Affiliation(s)
- Till Scheuer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Elena Auf dem Brinke
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Sabine Grosser
- Institute for Integrative Neuroanatomy, NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Susanne A Wolf
- Cellular Neurocience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany.,Department of Experimental Ophthalmology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Daniele Mattei
- Cellular Neurocience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany.,Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich CH-8057, Switzerland
| | - Yuliya Sharkovska
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany.,Institute for Cell and Neurobiology, Center for Anatomy, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany.,Berlin Institute of Health (BIH), Berlin 10178, Germany
| | - Paula C Barthel
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany.,Institute for Cell and Neurobiology, Center for Anatomy, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Stefanie Endesfelder
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Vivien Friedrich
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany.,Berlin Institute of Health (BIH), Berlin 10178, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Imre Vida
- Institute for Integrative Neuroanatomy, NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Thomas Schmitz
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| |
Collapse
|
15
|
Takechi R, Mamo J, Das S, Graneri L, D'Alonzo Z, Nesbit M, Junaldi E, Lam V. Short-term consumption of alcohol (vodka) mixed with energy drink (AMED) attenuated alcohol-induced cerebral capillary disturbances and neuroinflammation in adult wild-type mice. Nutr Neurosci 2021; 25:2398-2407. [PMID: 34549671 DOI: 10.1080/1028415x.2021.1975364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Background: The ingestion of combinatory Alcohol Mixed with Energy Drink (AMED) beverages continues to increase markedly, particularly among young adults. Some studies suggest detrimental health effects related to the combination of alcohol with energy drink formulations; however, the consumption of AMED has not been investigated in context of the cerebral microvasculature or neuroinflammation. We hypothesized that cerebral capillary integrity and glial cells are particularly vulnerable to the combination of AMED.Methods:12-week old wild-type C57BL/6J mice were orally gavaged with either vehicle (water), alcohol (vodka), an energy drink (MotherTM), or a combination AMED, daily for five days. Thereafter, mice were sacrificed, blood alcohol concentrations were analysed and cryosections of brain specimens were subjected to confocal immunofluorescent analysis for measures of cerebral capillary integrity via immunoglobulin G (IgG), and markers of neuroinflammation, ionized-calcium-binding-adaptor-molecule 1 (Iba1) and Glial-Fibrillary-Acidic-Protein (GFAP). Proinflammatory cytokines, IL-2, IL-17A, IFN-ϒ, and anti-inflammatory cytokines, IL-4, IL-6 and IL-10, were also measured in serum.Results: Consistent with previous studies, cerebral capillary dysfunction and astroglial cell activation were markedly greater in the alcohol-only group (AO); however, the AO-induced effects were profoundly attenuated with the AMED combination. Mice maintained on AO and AMED interventions exhibited a moderate increase in microglial recruitment. There were no significant changes in pro-inflammatory nor anti-inflammatory cytokines in ED or AMED treated mice.Conclusion: This study suggests that paradoxically the acute detrimental effects of alcohol on cerebral capillary integrity and astrogliosis are counteracted with the co-provision of an ED, rich in caffeine and taurine and containing B-group vitamins.
Collapse
Affiliation(s)
- Ryusuke Takechi
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Australia.,School of Population Health, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - John Mamo
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Australia.,School of Population Health, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Sukanya Das
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Liam Graneri
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Australia.,Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Zachary D'Alonzo
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Australia.,Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Michael Nesbit
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Australia.,School of Population Health, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Edwin Junaldi
- School of Population Health, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Virginie Lam
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Australia.,School of Population Health, Faculty of Health Sciences, Curtin University, Perth, Australia
| |
Collapse
|
16
|
Robertson SM, Edwards SH, Doran GS, Friend MA. Maternal caffeine administration to ewes does not affect perinatal lamb survival. Anim Reprod Sci 2021; 231:106799. [PMID: 34225237 DOI: 10.1016/j.anireprosci.2021.106799] [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: 01/28/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 11/24/2022]
Abstract
Perinatal mortality of lambs is the major source of reproductive loss in extensive sheep production systems. Treatment with caffeine has reduced intra-partum mortality and/or improved metabolic indicators in other species following hypoxia. This study was conducted to evaluate the efficacy of caffeine for improving perinatal lamb survival. Experiment 1 comprised group-fed Merino ewes grazing pasture and offered 1.8 g/day (estimated 20 mg/kg live weight) caffeine throughout a 4-week lambing period, and a control without caffeine. The survival of lambs to marking (vaccinated, tail docked, males castrated) age in the caffeine treatment group (0.81) did not differ (P = 0.199) from that of control lambs (0.73; total born n = 877). Experiment 2 comprised Merino ewes lambing from three successive weekly joining groups. Treated ewes were drenched with an aqueous caffeine solution at a dose rate of 10 mg/kg live weight from the day before anticipated lambing, until the individual lambed. Control ewes were drenched with water. The proportion of lambs born dead (0.07) and the survival of lambs to marking age (caffeine 0.61; control 0.62) were similar between treatment groups (total born n = 1158). In both experiments, ewe mortality and the weight of lambs at marking were not altered by caffeine treatments. The results from this large-scale field study indicate caffeine is not an effective therapeutic agent to increase either intra-partum or perinatal survival, or lamb growth rates.
Collapse
Affiliation(s)
- S M Robertson
- School of Animal and Veterinary Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW, 2678, Australia; Graham Centre for Agricultural Innovation (NSW Department of Primary Industries and Charles Sturt University), Albert Pugsley Place, Wagga Wagga, NSW, 2650, Australia; Fred Morley Centre, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW, 2678, Australia.
| | - S H Edwards
- School of Animal and Veterinary Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW, 2678, Australia
| | - G S Doran
- Graham Centre for Agricultural Innovation (NSW Department of Primary Industries and Charles Sturt University), Albert Pugsley Place, Wagga Wagga, NSW, 2650, Australia; School of Agricultural and Wine Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW, 2678, Australia
| | - M A Friend
- Office of the Pro Vice-Chancellor (Research and Innovation) Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW, 2678, Australia
| |
Collapse
|
17
|
Vieira AJ, Gaspar EM, Santos PM. Mechanisms of potential antioxidant activity of caffeine. Radiat Phys Chem Oxf Engl 1993 2020. [DOI: 10.1016/j.radphyschem.2020.108968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
18
|
Truttmann AC, Ginet V, Puyal J. Current Evidence on Cell Death in Preterm Brain Injury in Human and Preclinical Models. Front Cell Dev Biol 2020; 8:27. [PMID: 32133356 PMCID: PMC7039819 DOI: 10.3389/fcell.2020.00027] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/14/2020] [Indexed: 12/19/2022] Open
Abstract
Despite tremendous advances in neonatal intensive care over the past 20 years, prematurity carries a high burden of neurological morbidity lasting lifelong. The term encephalopathy of prematurity (EoP) coined by Volpe in 2009 encompasses all aspects of the now known effects of prematurity on the immature brain, including altered and disturbed development as well as specific lesional hallmarks. Understanding the way cells are damaged is crucial to design brain protective strategies, and in this purpose, preclinical models largely contribute to improve the comprehension of the cell death mechanisms. While neuronal cell death has been deeply investigated and characterized in (hypoxic–ischemic) encephalopathy of the newborn at term, little is known about the types of cell death occurring in preterm brain injury. Three main different morphological cell death types are observed in the immature brain, specifically in models of hypoxic–ischemic encephalopathy, namely, necrotic, apoptotic, and autophagic cell death. Features of all three types may be present in the same dying neuron. In preterm brain injury, description of cell death types is sparse, and cell loss primarily concerns immature oligodendrocytes and, infrequently, neurons. In the present review, we first shortly discuss the different main severe preterm brain injury conditions that have been reported to involve cell death, including periventricular leucomalacia (PVL), diffuse white matter injury (dWMI), and intraventricular hemorrhages, as well as potentially harmful iatrogenic conditions linked to premature birth (anesthesia and caffeine therapy). Then, we present an overview of current evidence concerning cell death in both clinical human tissue data and preclinical models by focusing on studies investigating the presence of cell death allowing discriminating between the types of cell death involved. We conclude that, to improve brain protective strategies, not only apoptosis but also other cell death (such as regulated necrotic and autophagic) pathways now need to be investigated together in order to consider all cell death mechanisms involved in the pathogenesis of preterm brain damage.
Collapse
Affiliation(s)
- Anita C Truttmann
- Clinic of Neonatology, Department of Women, Mother and Child, University Hospital Center of Vaud, Lausanne, Switzerland
| | - Vanessa Ginet
- Clinic of Neonatology, Department of Women, Mother and Child, University Hospital Center of Vaud, Lausanne, Switzerland.,Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,CURML, University Center of Legal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
19
|
Mateus JM, Ribeiro FF, Alonso-Gomes M, Rodrigues RS, Marques JM, Sebastião AM, Rodrigues RJ, Xapelli S. Neurogenesis and Gliogenesis: Relevance of Adenosine for Neuroregeneration in Brain Disorders. J Caffeine Adenosine Res 2019. [DOI: 10.1089/caff.2019.0010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Joana M. Mateus
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Filipa F. Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Marta Alonso-Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rui S. Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joana M. Marques
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Ana M. Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ricardo J. Rodrigues
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
20
|
Detrimental Impact of Energy Drink Compounds on Developing Oligodendrocytes and Neurons. Cells 2019; 8:cells8111381. [PMID: 31684159 PMCID: PMC6912672 DOI: 10.3390/cells8111381] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022] Open
Abstract
The consumption of energy drinks is continuously rising, particularly in children and adolescents. While risks for adverse health effects, like arrhythmia, have been described, effects on neural cells remain elusive. Considering that neurodevelopmental processes like myelination and neuronal network formation peak in childhood and adolescence we hypothesized that developing oligodendrocytes and neurons are particularly vulnerable to main energy drink components. Immature oligodendrocytes and hippocampal neurons were isolated from P0-P1 Wistar rats and were incubated with 0.3 mg/mL caffeine and 4 mg/mL taurine alone or in combination for 24 h. Analysis was performed immediately after treatment or after additional three days under differentiating conditions for oligodendrocytes and standard culture for neurons. Oligodendrocyte degeneration, proliferation, and differentiation were assessed via immunocytochemistry and immunoblotting. Neuronal integrity was investigated following immunocytochemistry by analysis of dendrite outgrowth and axonal morphology. Caffeine and taurine induced an increased degeneration and inhibited proliferation of immature oligodendrocytes accompanied by a decreased differentiation capacity. Moreover, dendritic branching and axonal integrity of hippocampal neurons were negatively affected by caffeine and taurine treatment. The negative impact of caffeine and taurine on developing oligodendrocytes and disturbed neuronal morphology indicates a high risk for disturbed neurodevelopment in children and adolescents by excessive energy drink consumption.
Collapse
|
21
|
Tsang JKW, Liu J, Lo ACY. Vascular and Neuronal Protection in the Developing Retina: Potential Therapeutic Targets for Retinopathy of Prematurity. Int J Mol Sci 2019; 20:E4321. [PMID: 31484463 PMCID: PMC6747312 DOI: 10.3390/ijms20174321] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/21/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a common retinal disease in preterm babies. To prolong the lives of preterm babies, high oxygen is provided to mimic the oxygen level in the intrauterine environment for postnatal organ development. However, hyperoxia-hypoxia induced pathological events occur when babies return to room air, leading to ROP with neuronal degeneration and vascular abnormality that affects retinal functions. With advances in neonatal intensive care, it is no longer uncommon for increased survival of very-low-birth-weight preterm infants, which, therefore, increased the incidence of ROP. ROP is now a major cause of preventable childhood blindness worldwide. Current proven treatment for ROP is limited to invasive retinal ablation, inherently destructive to the retina. The lack of pharmacological treatment for ROP creates a great need for effective and safe therapies in these developing infants. Therefore, it is essential to identify potential therapeutic agents that may have positive ROP outcomes, especially in preserving retinal functions. This review gives an overview of various agents in their efficacy in reducing retinal damages in cell culture tests, animal experiments and clinical studies. New perspectives along the neuroprotective pathways in the developing retina are also reviewed.
Collapse
Affiliation(s)
- Jessica K W Tsang
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jin Liu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Amy C Y Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
22
|
Abstract
Rates of bronchopulmonary dysplasia (BPD) are increasing. After preterm birth, there are important developmental periods in which neonates are more vulnerable to stressful events. These periods are opportunities for pharmacologic interventions. Many drugs remain inadequately tested and no new drugs have been approved in more than 25 years for BPD prevention or therapy. More progress is needed in defining appropriate end points based on the pathophysiology of BPD and postdischarge chronic pulmonary insufficiency of prematurity and to develop effective new drugs. In addition, much work is needed to better define perinatal factors, early postnatal findings, and physiologic phenotypes or endotypes.
Collapse
|
23
|
Endesfelder S, Strauß E, Scheuer T, Schmitz T, Bührer C. Antioxidative effects of caffeine in a hyperoxia-based rat model of bronchopulmonary dysplasia. Respir Res 2019; 20:88. [PMID: 31077204 PMCID: PMC6511176 DOI: 10.1186/s12931-019-1063-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023] Open
Abstract
Background While additional oxygen supply is often required for the survival of very premature infants in intensive care, this also brings an increasing risk of progressive lung diseases and poor long-term lung outcomes. Caffeine is administered to neonates in neonatal intensive care for the prevention and treatment of apneas and has been shown to reduce BPD incidence and the need for mechanical ventilation, although it is still unclear whether this is due to a direct pulmonary action via antagonism of adenosine receptors and/or an indirect action. This experimental study aims to investigate the action of caffeine on the oxidative stress response in pulmonary tissue in a hyperoxia-based model of bronchopulmonary dysplasia in newborn rats. Methods Newborn Wistar rats were exposed to 21% or 80% oxygen for 3 (P3) or 5 (P5) postnatal days with or without recovery on room air until postnatal day 15 (P15) and treated with vehicle or caffeine (10 mg/kg) every 48 h beginning on the day of birth. The lung tissue of the rat pups was examined for oxidative stress response at P3 and P5 immediately after oxygen exposure or after recovery in ambient air (P15) by immunohistological staining and analysis of lung homogenates by ELISA and qPCR. Results Lungs of newborn rats, corresponding to the saccular stage of lung development and to the human lung developmental stage of preterms, showed increased rates of total glutathione and hydrogen peroxide, oxidative damage to DNA and lipids, and induction of second-phase mediators of antioxidative stress response (superoxide dismutase, heme oxygenase-1, and the Nrf2/Keap1 system) in response to hyperoxia. Caffeine reduced oxidative DNA damage and had a protective interference with the oxidative stress response. Conclusion In addition to the pharmacological antagonism of adenosine receptors, caffeine appears to be a potent antioxidant and modulates the hyperoxia-induced pulmonary oxidative stress response and thus protective properties in the BPD-associated animal model. Free-radical-induced damage caused by oxidative stress seems to be a biological mechanism progress of newborn diseases. New aspects of antioxidative therapeutic strategies to passivate oxidative stress-related injury should be in focus of further investigations. Electronic supplementary material The online version of this article (10.1186/s12931-019-1063-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stefanie Endesfelder
- Department of Neonatology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Evelyn Strauß
- Department of Neonatology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Till Scheuer
- Department of Neonatology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Thomas Schmitz
- Department of Neonatology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| |
Collapse
|
24
|
Scheuer T, Klein LS, Bührer C, Endesfelder S, Schmitz T. Transient Improvement of Cerebellar Oligodendroglial Development in a Neonatal Hyperoxia Model by PDGFA Treatment. Dev Neurobiol 2019; 79:222-235. [PMID: 30674088 DOI: 10.1002/dneu.22667] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/10/2019] [Accepted: 01/12/2019] [Indexed: 12/13/2022]
Abstract
In preterm infants, the changes from fetal life to ex-utero conditions often coincide with reduced growth and white matter damage of the cerebellum. The premature increase in arterial oxygen tension caused by preterm birth may dysregulate cerebellar development. In a hyperoxia rat model of white matter damage to mimic a steep increase in oxygen levels by 24 h exposure to 80% O2 from postnatal day 6 (P6) to day 7, we analyzed growth factor (GF) synthesis of cerebellar astrocytes. Determination of GF production was performed in astrocytes after Magnetic-activated cell sorting (MACS) isolation from cerebelli after hyperoxia exposure ex vivo, and also in astroglial cultures. Oligodendrocyte progenitor cell (OPC) function was analyzed in cerebellar OPCs isolated by MACS after hyperoxia. Administration of PDGFA from P6 to P11, during hyperoxia and during 4 days recovery, was finally tested for protection of oligodendroglia and myelination. As a result, expression of the GFs Pdgfa, Fgf2, and Bdnf was diminished in cerebellar astrocytes in vitro and in vivo. Gene expression of Olig1, Olig2, Sox9, Sox10, and Cnp was reduced in OPCs in vivo. Nasal PDGFA application improved oligodendroglial proliferation after hyperoxia at P7. However, this treatment effect vanished until P9. Impaired MBP expression after hyperoxia was attenuated by PDGFA treatment until P11, but not beyond when PDGFA supply was stopped. In this study on neonatal cerebellar injury, it is documented for the first time that improvement of oligodendroglial proliferation and of myelination can be achieved by PDGFA treatment. However, the treatment benefit is not maintained long term.
Collapse
Affiliation(s)
- Till Scheuer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Luisa Sophie Klein
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Christoph Bührer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | | | - Thomas Schmitz
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| |
Collapse
|
25
|
Colombo R, Papetti A. An outlook on the role of decaffeinated coffee in neurodegenerative diseases. Crit Rev Food Sci Nutr 2019; 60:760-779. [PMID: 30614247 DOI: 10.1080/10408398.2018.1550384] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - Adele Papetti
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
26
|
Neuroinflammation in preterm babies and autism spectrum disorders. Pediatr Res 2019; 85:155-165. [PMID: 30446768 DOI: 10.1038/s41390-018-0208-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 12/23/2022]
Abstract
Genetic anomalies have a role in autism spectrum disorders (ASD). Each genetic factor is responsible for a small fraction of cases. Environment factors, like preterm delivery, have an important role in ASD. Preterm infants have a 10-fold higher risk of developing ASD. Preterm birth is often associated with maternal/fetal inflammation, leading to a fetal/neonatal inflammatory syndrome. There are demonstrated experimental links between fetal inflammation and the later development of behavioral symptoms consistent with ASD. Preterm infants have deficits in connectivity. Most ASD genes encode synaptic proteins, suggesting that ASD are connectivity pathologies. Microglia are essential for normal synaptogenesis. Microglia are diverted from homeostatic functions towards inflammatory phenotypes during perinatal inflammation, impairing synaptogenesis. Preterm infants with ASD have a different phenotype from term born peers. Our original hypothesis is that exposure to inflammation in preterm infants, combined with at risk genetic background, deregulates brain development leading to ASD.
Collapse
|
27
|
Does duration of caffeine therapy in preterm infants born ≤1250 g at birth influence neurodevelopmental (ND) outcomes at 3 years of age? J Perinatol 2018; 38:889-899. [PMID: 29740190 DOI: 10.1038/s41372-018-0106-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/28/2018] [Accepted: 03/06/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To evaluate the effect of duration of caffeine use on long-term neurodevelopmental (ND) outcomes at 3 years corrected age (CA) in preterm infants with birthweights (BW) ≤ 1250 g. DESIGN/METHODS All surviving infants with BW ≤ 1250 g admitted to the Foothills Medical Center neonatal intensive care unit (NICU) from January 2002 to December 2009 who received the first dose of caffeine in the first week of life and were followed up at three years CA were included in the study. Demographics and follow-up outcomes were compared based on early cessation of caffeine ≤ 14 days (ECC), intermediate cessation of caffeine 15-30 days (ICC), and late cessation of caffeine >30 days (LCC). The primary outcome of ND impairment was present if a child had any one of the following: cerebral palsy, cognitive delay, visual impairment, or hearing impairment or deafness. Univariate and logistic regression analyses were performed. RESULTS Of the 508 eligible infants, 448 (88%) were seen at 3 years CA at follow-up. ECC (n = 139), ICC (n = 122) and LCC (n = 187) groups had a median (range) BW of 979 (560-1250), 1010 (530-1250), and 980 (520-1250) g (p = 0.524) and median (range) gestational age (GA) of 27 (23-33), 28 (24-33), and 27 (24-32) weeks, respectively (p = 0.034). In logistic regression models adjusting for GA, maternal smoking, and each neonatal risk factor separately (IVH, NEC, sepsis, blood transfusions, BPD, postnatal dexamethasone, SNAP-II, and ventilator days), none of the models showed a statistically significant association between caffeine duration and ND impairment. CONCLUSION The duration of caffeine use in premature infants in the NICU does not impact on long-term ND outcomes at 3 years CA.
Collapse
|
28
|
Liu Y, Dong WB. [Preventive effect of caffeine on bronchopulmonary dysplasia in preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:598-602. [PMID: 30022766 PMCID: PMC7389204 DOI: 10.7499/j.issn.1008-8830.2018.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 06/05/2018] [Indexed: 06/08/2023]
Abstract
With the increase in the rescue success rate of critically ill preterm infants and extremely preterm infants, the incidence rate of bronchopulmonary dysplasia (BPD) is increasing year by year. BPD has a high mortality rate and high possibility of sequelae, which greatly affects the quality of life of preterm infants and brings a heavy burden to their families, and so the treatment of BPD is of vital importance. At present, no consensus has been reached on the treatment measures for BPD. However, recent studies have shown that early application of caffeine can prevent BPD. With reference to the latest studies on the effect of caffeine in the prevention of BPD, this article reviews the mechanism of action of caffeine in reducing pulmonary inflammation, improving morphological abnormalities of lung injury, reducing oxidative stress injury, and improving pulmonary function.
Collapse
MESH Headings
- Animals
- Bronchopulmonary Dysplasia/genetics
- Bronchopulmonary Dysplasia/metabolism
- Bronchopulmonary Dysplasia/physiopathology
- Bronchopulmonary Dysplasia/prevention & control
- Caffeine/administration & dosage
- Humans
- Infant, Premature/growth & development
- Infant, Premature/metabolism
- Infant, Premature, Diseases/genetics
- Infant, Premature, Diseases/metabolism
- Infant, Premature, Diseases/physiopathology
- Infant, Premature, Diseases/prevention & control
- Oxidative Stress/drug effects
Collapse
Affiliation(s)
- Yang Liu
- Department of Neonatology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | | |
Collapse
|
29
|
Serdar M, Herz J, Kempe K, Winterhager E, Jastrow H, Heumann R, Felderhoff-Müser U, Bendix I. Protection of Oligodendrocytes Through Neuronal Overexpression of the Small GTPase Ras in Hyperoxia-Induced Neonatal Brain Injury. Front Neurol 2018; 9:175. [PMID: 29619004 PMCID: PMC5871665 DOI: 10.3389/fneur.2018.00175] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/06/2018] [Indexed: 12/29/2022] Open
Abstract
Prematurely born infants are highly susceptible to various environmental factors, such as inflammation, drug exposure, and also high environmental oxygen concentrations. Hyperoxia induces perinatal brain injury affecting white and gray matter development. It is well known that mitogen-activated protein kinase signaling is involved in cell survival, proliferation, and differentiation. Therefore, we aim to elucidate cell-specific responses of neuronal overexpression of the small GTPase Ras on hyperoxia-mediated brain injury. Six-day-old (P6) synRas mice (neuronal Ras overexpression under the synapsin promoter) or wild-type littermates were kept under hyperoxia (80% oxygen) or room air (21% oxygen) for 24 h. Apoptosis was analyzed by Western blot of cleaved Caspase-3 and neuronal and oligodendrocyte degeneration via immunohistochemistry. Short-term differentiation capacity of oligodendrocytes was assessed by quantification of myelin basic protein expression at P11. Long-lasting changes of hyperoxia-induced alteration of myelin structures were evaluated via transmission electron microscopy in young adult animals (P42). Western blot analysis of active Caspase-3 demonstrates a significant upregulation in wild-type littermates exposed to hyperoxia whereas synRas mice did not show any marked alteration of cleaved Caspase-3 protein levels. Immunohistochemistry revealed a protective effect of neuronal Ras overexpression on neuron and oligodendrocyte survival. Hyperoxia-induced hypomyelination in wild-type littermates was restored in synRas mice. These short-term protective effects through promotion of neuronal survival translated into long-lasting improvement of ultrastructural alterations of myelin sheaths in mice with neuronal overexpression of Ras compared with hyperoxic wild-type mice. Our data suggest that transgenic increase of neuronal Ras activity in the immature brain results in secondary protection of oligodendrocytes from hyperoxia-induced white matter brain injury.
Collapse
Affiliation(s)
- Meray Serdar
- Department of Pediatrics I, Neonatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Pediatrics I, Neonatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Karina Kempe
- Department of Pediatrics I, Neonatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Elke Winterhager
- Imaging Center Essen, EM Unit, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Holger Jastrow
- Imaging Center Essen, EM Unit, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Institute of Anatomy, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Rolf Heumann
- Biochemistry II, Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Ursula Felderhoff-Müser
- Department of Pediatrics I, Neonatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Ivo Bendix
- Department of Pediatrics I, Neonatology, University Hospital, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
30
|
Per S, Kose M, Ozdemir A, Pandir D. Hepatoprotective effects of capping protein gelsolin against hyperoxia-induced hepatotoxicity, oxidative stress and DNA damage in neonatal rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 58:189-195. [PMID: 29408761 DOI: 10.1016/j.etap.2018.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 06/07/2023]
Abstract
Tissues and organs get exposed to high oxygen (O2) supply in hyperoxia conditions. The goal of this research was to investigate the protective effect of actin binding protein gelsolin on hyperoxia-induced hepatotoxicity through histopathology and measurement of oxidative stress parameters and DNA damage in a neonatal Wistar albino rats. The pups were randomly separated to four equal groups such as: normoxia control group (NC), normoxia plus gelsolin group (NG, 10 ng/kg bw/day gelsolin), hyperoxia (≥85% O2) group (HC), hyperoxia plus gelsolin group (HG, ≥85% O2; 10 ng/kg bw/day gelsolin). Histopathological changes of pups in hyperoxia condition were revealed in the form of severe leukocyte infiltration, vascular congestion, necrosis, vacuolar degeneration, binucleated hepatocytes and hemorrhage in the liver tissue. SOD, CAT, GPx and GST activities decreased and MDA level increased in the hyperoxia-induced group in liver tissue (P < 0.05). Tail DNA%, tail length and moment indicating DNA damage statistically increased in hyperoxia treatment groups when compared to controls. Treatment of rats with hyperoxia plus gelsolin prevented hyperoxia-induced changes in tissue structure, antioxidant enzyme activities and MDA level, mean tail DNA% and length. Based on these findings, gelsolin restored these changing to near normal levels but it does not protect completely in the hyperoxia conditions.
Collapse
Affiliation(s)
- Sedat Per
- Department of Biology, Bozok University, Yozgat, Turkey.
| | - Mehmet Kose
- Department of Pediatrics, Division of Pediatric Pulmonology Unit, Erciyes University, Kayseri, Turkey
| | - Ahmet Ozdemir
- Department of Pediatrics, Division of Neonatology, Erciyes University, Kayseri, Turkey
| | - Dilek Pandir
- Department of Biology, Bozok University, Yozgat, Turkey
| |
Collapse
|
31
|
Endesfelder S, Weichelt U, Schiller C, Winter K, von Haefen C, Bührer C. Caffeine Protects Against Anticonvulsant-Induced Impaired Neurogenesis in the Developing Rat Brain. Neurotox Res 2018; 34:173-187. [PMID: 29417440 DOI: 10.1007/s12640-018-9872-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/08/2018] [Accepted: 01/23/2018] [Indexed: 02/06/2023]
Abstract
In preterm infants, phenobarbital is the first-line antiepileptic drug for neonatal seizures while caffeine is used for the treatment of apnea. Data from experimental animals suggest that phenobarbital and other anticonvulsants are toxic for the developing brain, while neuroprotective effects have been reported for caffeine both in newborn rodents and preterm human infants. To characterize the interaction of phenobarbital and caffeine in the hippocampus of the developing rodent brain, we examined the effects of both drugs given separately or together on postnatal neurogenesis after administration to neonatal rats throughout postnatal day (P) 4 to P6. Phenobarbital treatment (50 mg/kg) resulted in a significant decrease of proliferative capacity in the dentate gyrus. Phenobarbital also reduced expression of neuronal markers (doublecortin (DCX), calretinin, NeuN), neuronal transcription factors (Pax6, Sox2, Tbr1/2, Prox1), and neurotrophins (NGF, BDNF, NT-3) up to 24 h after the last administration. The phenobarbital-mediated impairment of neurogenesis was largely ameliorated by preconditioning with caffeine (10 mg/kg). In contrast, caffeine alone reduced proliferative capacity and expression of the neuronal markers DCX and NeuN at 6 h, but increased expression of neurotrophins and neuronal transcription factors at 6 and 12 h. These results indicate that administration of phenobarbital during the vulnerable phase of brain development negatively interferes with neuronal development, which can be prevented in part by co-administration of caffeine.
Collapse
Affiliation(s)
- Stefanie Endesfelder
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Ulrike Weichelt
- Endowed Professorship of Immunotechnology, Institute of Biochemistry and Biology, University of Potsdam, Campus Golm, Karl-Liebknechtstraße 24-25, 14476, Potsdam - Golm, Germany
| | - Cornelia Schiller
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Katja Winter
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| |
Collapse
|
32
|
Deliktaş M, Ergin H, Demiray A, Akça H, Özdemir ÖMA, Özdemir MB. Caffeine prevents bilirubin-induced cytotoxicity in cultured newborn rat astrocytes. J Matern Fetal Neonatal Med 2018; 32:1813-1819. [PMID: 29295636 DOI: 10.1080/14767058.2017.1419175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Unconjugated bilirubin (UCB) may cause neurotoxicity in preterm neonates due to immaturity of UGT1A1 leading to bilirubin accumulation in the brain. Caffeine used in the treatment of apnea of prematurity was reported to decrease mechanical ventilation requirement, the frequencies of bronchopulmonary dysplasia, patent ductus arteriosus, cerebral palsy and neurodevelopmental disorders in very low birth weight infants. However, the effect of caffeine on hyperbilirubinemia was not yet clarified. METHODS We used astrocyte cell cultures obtained from 2-day-old Wistar albino rats via modified Cole and de Vellis method. UCB concentration toxic to 50% of astrocytes, and caffeine concentration increasing cell viability 100% were used in experiments. While no medication was applied to the control group, UCB (50 μM) and caffeine (100 μM) were applied to the bilirubin and caffeine groups for 24 h. Prophylactic and therapeutic caffeine groups were treated with caffeine 4 h before and after UCB exposure. The effects of caffeine were investigated in rat astrocytes exposed to UCB in terms of cell viability, apoptosis, antioxidant defense, proinflammatory cytokines, and Toll-like receptor (TLR)s. RESULTS Compared to the control group, UCB increased apoptosis, malondialdehyde (MDA), tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, total nitrate/nitrite, and TLR4 levels, and decreased cell viability, catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD) activities, glutathione, and TLR9 levels (for all p < .001). Conversely, prophylactic and therapeutic caffeine improved the detrimental effects of UCB. CONCLUSIONS Caffeine seems encouraging for the prevention and treatment of bilirubin neurotoxicity in rats by means of its antiapoptotic, antioxidant, anti-inflammatory, anti-nitrosative, and anti-TLR-4 properties.
Collapse
Affiliation(s)
- Mehmet Deliktaş
- a Department of Pediatrics, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| | - Hacer Ergin
- a Department of Pediatrics, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| | - Aydın Demiray
- b Department of Medical Biology, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| | - Hakan Akça
- b Department of Medical Biology, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| | - Özmert M A Özdemir
- a Department of Pediatrics, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| | - Mehmet Bülent Özdemir
- c Department of Anatomy, Division of Neonatology, Faculty of Medicine , Pamukkale University , Denizli , Turkey
| |
Collapse
|
33
|
|
34
|
Viaroli F, Cheung PY, O'Reilly M, Polglase GR, Pichler G, Schmölzer GM. Reducing Brain Injury of Preterm Infants in the Delivery Room. Front Pediatr 2018; 6:290. [PMID: 30386757 PMCID: PMC6198082 DOI: 10.3389/fped.2018.00290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/19/2018] [Indexed: 11/13/2022] Open
Abstract
Cerebrovascular injury is one of the major detrimental consequences of preterm birth. Recent studies have focused their attention on factors that contribute to the development of brain lesions immediately after birth. Among those factors, hypothermia and lower cerebral oxygen saturation during delivery room resuscitation and high tidal volumes delivered during respiratory support are associated with increased risk of severe neurologic injury. In preterm infants, knowledge about causes and prevention of brain injury must be applied before and at birth. Preventive and therapeutic approaches, including correct timing of cord clamping, monitoring of physiological changes during delivery room resuscitation using pulse oximetry, respiratory function monitoring, near infrared spectroscopy, and alpha EEG, may minimize brain injury, Furthermore, postnatal administration of caffeine or other potential novel treatments (e.g., proangiogenic therapies, antioxidants, hormones, or stem cells) might improve long-term neurodevelopmental outcomes in preterm infants.
Collapse
Affiliation(s)
- Francesca Viaroli
- Centre for the Studies of Asphyxia and Resuscitation, Royal Alexandra Hospital, Edmonton, AB, Canada.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Po-Yin Cheung
- Centre for the Studies of Asphyxia and Resuscitation, Royal Alexandra Hospital, Edmonton, AB, Canada.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Megan O'Reilly
- Centre for the Studies of Asphyxia and Resuscitation, Royal Alexandra Hospital, Edmonton, AB, Canada.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Gerhard Pichler
- Department of Pediatrics, Medical University Graz, Graz, Austria
| | - Georg M Schmölzer
- Centre for the Studies of Asphyxia and Resuscitation, Royal Alexandra Hospital, Edmonton, AB, Canada.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
35
|
Curran CP, Marczinski CA. Taurine, caffeine, and energy drinks: Reviewing the risks to the adolescent brain. Birth Defects Res 2017; 109:1640-1648. [PMID: 29251842 PMCID: PMC5737830 DOI: 10.1002/bdr2.1177] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 11/02/2017] [Indexed: 01/04/2023]
Abstract
Energy drinks are emerging as a major component of the beverage market with sales projected to top $60 billion globally in the next five years. Energy drinks contain a variety of ingredients, but many of the top-selling brands include high doses of caffeine and the amino acid taurine. Energy drink consumption by children has raised concerns, due to potential caffeine toxicity. An additional risk has been noted among college-aged consumers of energy drinks who appear at higher risk of over-consumption of alcohol when the two drinks are consumed together. The differential and combinatorial effects of caffeine and taurine on the developing brain are reviewed here with an emphasis on the adolescent brain, which is still maturing. Key data from animal studies are summarized to highlight both reported benefits and adverse effects reported following acute and chronic exposures. The data suggest that age is an important factor in both caffeine and taurine toxicity. Although the aged or diseased brain might benefit from taurine or caffeine supplementation, it appears that adolescents are not likely to benefit from supplementation and may, in fact, suffer ill effects from chronic ingestion of high doses. Additional work is needed though to address gaps in our understanding of how taurine affects females, since the majority of animal studies focused exclusively on male subjects.
Collapse
Affiliation(s)
- Christine Perdan Curran
- Department of Biological Sciences, Northern Kentucky University, Highland Heights, KY, USA 41099
| | - Cecile A. Marczinski
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY, USA 41099
| |
Collapse
|
36
|
Yamakawa GR, Lengkeek C, Salberg S, Spanswick SC, Mychasiuk R. Behavioral and pathophysiological outcomes associated with caffeine consumption and repetitive mild traumatic brain injury (RmTBI) in adolescent rats. PLoS One 2017; 12:e0187218. [PMID: 29108016 PMCID: PMC5673214 DOI: 10.1371/journal.pone.0187218] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/16/2017] [Indexed: 12/31/2022] Open
Abstract
Given that caffeine consumption is exponentially rising in adolescents and they are at increased risk for repetitive mild traumatic brain injury (RmTBI), we sought to examine the pathophysiological outcomes associated with early life caffeine consumption and RmTBI. Adolescent male and female Sprague Dawley rats received either caffeine in the drinking water or normal water and were then randomly assigned to 3 mild injuries using our lateral impact device or 3 sham procedures. Following injury induction, behavioral outcomes were measured with a test battery designed to examine symptoms consistent with clinical manifestation of PCS (balance and motor coordination, anxiety, short-term working memory, and depressive-like behaviours). In addition, pathophysiological outcomes were examined with histological measures of volume and cellular proliferation in the dentate gyrus, as well as microglia activation in the ventromedial hypothalamus. Finally, modifications to expression of 12 genes (Adora2a, App, Aqp4, Bdnf, Bmal1, Clock, Cry, Gfap, Orx1, Orx2, Per, Tau), in the prefrontal cortex, hippocampus, and/or the hypothalamus were assessed. We found that chronic caffeine consumption in adolescence altered normal developmental trajectories, as well as recovery from RmTBI. Of particular importance, many of the outcomes exhibited sex-dependent responses whereby the sex of the animal modified response to caffeine, RmTBI, and the combination of the two. These results suggest that caffeine consumption in adolescents at high risk for RmTBI should be monitored.
Collapse
Affiliation(s)
- Glenn R. Yamakawa
- University of Calgary, Department of Psychology, Calgary, Alberta, Canada
| | - Connor Lengkeek
- University of Calgary, Department of Psychology, Calgary, Alberta, Canada
| | - Sabrina Salberg
- University of Calgary, Department of Psychology, Calgary, Alberta, Canada
| | - Simon C. Spanswick
- University of Calgary, Department of Psychology, Calgary, Alberta, Canada
| | - Richelle Mychasiuk
- University of Calgary, Department of Psychology, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
37
|
|
38
|
Paston SV, Polyanichko AM, Shulenina OV. Study of DNA interactions with Cu2+ and Mg2+ ions in the presence of caffeine. J STRUCT CHEM+ 2017. [DOI: 10.1134/s0022476617020263] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
39
|
Scheuer T, Sharkovska Y, Tarabykin V, Marggraf K, Brockmöller V, Bührer C, Endesfelder S, Schmitz T. Neonatal Hyperoxia Perturbs Neuronal Development in the Cerebellum. Mol Neurobiol 2017; 55:3901-3915. [PMID: 28547531 DOI: 10.1007/s12035-017-0612-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/11/2017] [Indexed: 12/22/2022]
Abstract
Impaired postnatal brain development of preterm infants often results in neurological deficits. Besides pathologies of the forebrain, maldeveolopment of the cerebellum is increasingly recognized to contribute to psychomotor impairments of many former preterm infants. However, causes are poorly defined. We used a hyperoxia model to define neonatal damage in cerebellar granule cell precursors (GCPs) and in Purkinje cells (PCs) known to be essential for interaction with GCPs during development. We exposed newborn rats to 24 h 80% O2 from age P6 to P7 to identify postnatal and long-term damage in cerebellar GCPs at age P7 after hyperoxia and also after recovery in room air thereafter until P11 and P30. We determined proliferation and apoptosis of GCPs and immature neurons by immunohistochemistry, quantified neuronal damage by qPCR and Western blots for neuronal markers, and measured dendrite outgrowth of PCs by CALB1 immunostainings and by Sholl analysis of Golgi stainings. After hyperoxia, proliferation of PAX6+ GCPs was decreased at P7, while DCX + CASP3+ cells were increased at P11. Neuronal markers Pax6, Tbr2, and Prox1 were downregulated at P11 and P30. Neuronal damage was confirmed by reduced NeuN protein expression at P30. Sonic hedgehog (SHH) was significantly decreased at P7 and P11 after hyperoxia and coincided with lower CyclinD2 and Hes1 expression at P7. The granule cell injury was accompanied by hampered PC maturation with delayed dendrite formation and impaired branching. Neonatal injury induced by hyperoxia inhibits PC functioning and impairs granule cell development. As a conclusion, maldevelopment of the cerebellar neurons found in preterm infants could be caused by postnatal oxygen toxicity.
Collapse
Affiliation(s)
- Till Scheuer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany. .,Institute of Bioanalytics, Technische Universität Berlin, 13355, Berlin, Germany. .,Klinik für Neonatologie, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Yuliya Sharkovska
- Department for Neonatology, Charité University Medical Center, Berlin, Germany.,Institute for Cell and Neurobiology, Center for Anatomy, Charité University Medical Center, Berlin, Germany
| | - Victor Tarabykin
- Institute for Cell and Neurobiology, Center for Anatomy, Charité University Medical Center, Berlin, Germany
| | - Katharina Marggraf
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Vivien Brockmöller
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Christoph Bührer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | | | - Thomas Schmitz
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| |
Collapse
|
40
|
Endesfelder S, Makki H, von Haefen C, Spies CD, Bührer C, Sifringer M. Neuroprotective effects of dexmedetomidine against hyperoxia-induced injury in the developing rat brain. PLoS One 2017; 12:e0171498. [PMID: 28158247 PMCID: PMC5291450 DOI: 10.1371/journal.pone.0171498] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/20/2017] [Indexed: 12/19/2022] Open
Abstract
Dexmedetomidine (DEX) is a highly selective agonist of α2-receptors with sedative, anxiolytic, and analgesic properties. Neuroprotective effects of dexmedetomidine have been reported in various brain injury models. In the present study, we investigated the effects of dexmedetomidine on hippocampal neurogenesis, specifically the proliferation capacity and maturation of neurons and neuronal plasticity following the induction of hyperoxia in neonatal rats. Six-day old sex-matched Wistar rats were exposed to 80% oxygen or room air for 24 h and treated with 1, 5 or 10 μg/kg of dexmedetomidine or normal saline. A single pretreatment with DEX attenuated the hyperoxia-induced injury in terms of neurogenesis and plasticity. In detail, both the proliferation capacity (PCNA+ cells) as well as the expression of neuronal markers (Nestin+, PSA-NCAM+, NeuN+ cells) and transcription factors (SOX2, Tbr1/2, Prox1) were significantly reduced under hyperoxia compared to control. Furthermore, regulators of neuronal plasticity (Nrp1, Nrg1, Syp, and Sema3a/f) were also drastically decreased. A single administration of dexmedetomidine prior to oxygen exposure resulted in a significant up-regulation of expression-profiles compared to hyperoxia. Our results suggest that dexmedetomidine may have neuroprotective effects in an acute hyperoxic model of the neonatal rat.
Collapse
Affiliation(s)
- Stefanie Endesfelder
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Hanan Makki
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia D Spies
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Sifringer
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
41
|
Endesfelder S, Weichelt U, Strauß E, Schlör A, Sifringer M, Scheuer T, Bührer C, Schmitz T. Neuroprotection by Caffeine in Hyperoxia-Induced Neonatal Brain Injury. Int J Mol Sci 2017; 18:E187. [PMID: 28106777 PMCID: PMC5297819 DOI: 10.3390/ijms18010187] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/04/2017] [Accepted: 01/12/2017] [Indexed: 12/11/2022] Open
Abstract
Sequelae of prematurity triggered by oxidative stress and free radical-mediated tissue damage have coined the term "oxygen radical disease of prematurity". Caffeine, a potent free radical scavenger and adenosine receptor antagonist, reduces rates of brain damage in preterm infants. In the present study, we investigated the effects of caffeine on oxidative stress markers, anti-oxidative response, inflammation, redox-sensitive transcription factors, apoptosis, and extracellular matrix following the induction of hyperoxia in neonatal rats. The brain of a rat pups at postnatal Day 6 (P6) corresponds to that of a human fetal brain at 28-32 weeks gestation and the neonatal rat is an ideal model in which to investigate effects of oxidative stress and neuroprotection of caffeine on the developing brain. Six-day-old Wistar rats were pre-treated with caffeine and exposed to 80% oxygen for 24 and 48 h. Caffeine reduced oxidative stress marker (heme oxygenase-1, lipid peroxidation, hydrogen peroxide, and glutamate-cysteine ligase catalytic subunit (GCLC)), promoted anti-oxidative response (superoxide dismutase, peroxiredoxin 1, and sulfiredoxin 1), down-regulated pro-inflammatory cytokines, modulated redox-sensitive transcription factor expression (Nrf2/Keap1, and NFκB), reduced pro-apoptotic effectors (poly (ADP-ribose) polymerase-1 (PARP-1), apoptosis inducing factor (AIF), and caspase-3), and diminished extracellular matrix degeneration (matrix metalloproteinases (MMP) 2, and inhibitor of metalloproteinase (TIMP) 1/2). Our study affirms that caffeine is a pleiotropic neuroprotective drug in the developing brain due to its anti-oxidant, anti-inflammatory, and anti-apoptotic properties.
Collapse
Affiliation(s)
- Stefanie Endesfelder
- Department of Neonatology, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Ulrike Weichelt
- Department of Physiology, Charité, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Evelyn Strauß
- Department of Neonatology, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Anja Schlör
- Department of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany.
| | - Marco Sifringer
- Department of Anesthesiology and Intensive Care Medicine, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Till Scheuer
- Department of Neonatology, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Christoph Bührer
- Department of Neonatology, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Thomas Schmitz
- Department of Neonatology, Charité, Universitätsmedizin Berlin, 13353 Berlin, Germany.
| |
Collapse
|
42
|
Neuroprotection and neurotoxicity in the developing brain: an update on the effects of dexmedetomidine and xenon. Neurotoxicol Teratol 2017; 60:102-116. [PMID: 28065636 DOI: 10.1016/j.ntt.2017.01.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 12/30/2016] [Accepted: 01/04/2017] [Indexed: 12/13/2022]
Abstract
Growing and consistent preclinical evidence, combined with early clinical epidemiological observations, suggest potentially neurotoxic effects of commonly used anesthetic agents in the developing brain. This has prompted the FDA to issue a safety warning for all sedatives and anesthetics approved for use in children under three years of age. Recent studies have identified dexmedetomidine, the potent α2-adrenoceptor agonist, and xenon, the noble gas, as effective anesthetic adjuvants that are both less neurotoxic to the developing brain, and also possess neuroprotective properties in neonatal and other settings of acute ongoing neurologic injury. Dexmedetomidine and xenon are effective anesthetic adjuvants that appear to be less neurotoxic than other existing agents and have the potential to be neuroprotective in the neonatal and pediatric settings. Although results from recent clinical trials and case reports have indicated the neuroprotective potential of xenon and dexmedetomidine, additional randomized clinical trials corroborating these studies are necessary. By reviewing both the existing preclinical and clinical evidence on the neuroprotective effects of dexmedetomidine and xenon, we hope to provide insight into the potential clinical efficacy of these agents in the management of pediatric surgical patients.
Collapse
|
43
|
Erythropoietin Restores Long-Term Neurocognitive Function Involving Mechanisms of Neuronal Plasticity in a Model of Hyperoxia-Induced Preterm Brain Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9247493. [PMID: 27493706 PMCID: PMC4963567 DOI: 10.1155/2016/9247493] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/31/2016] [Accepted: 06/13/2016] [Indexed: 12/22/2022]
Abstract
Cerebral white and grey matter injury is the leading cause of an adverse neurodevelopmental outcome in prematurely born infants. High oxygen concentrations have been shown to contribute to the pathogenesis of neonatal brain damage. Here, we focused on motor-cognitive outcome up to the adolescent and adult age in an experimental model of preterm brain injury. In search of the putative mechanisms of action we evaluated oligodendrocyte degeneration, myelination, and modulation of synaptic plasticity-related molecules. A single dose of erythropoietin (20,000 IU/kg) at the onset of hyperoxia (24 hours, 80% oxygen) in 6-day-old Wistar rats improved long-lasting neurocognitive development up to the adolescent and adult stage. Analysis of white matter structures revealed a reduction of acute oligodendrocyte degeneration. However, erythropoietin did not influence hypomyelination occurring a few days after injury or long-term microstructural white matter abnormalities detected in adult animals. Erythropoietin administration reverted hyperoxia-induced reduction of neuronal plasticity-related mRNA expression up to four months after injury. Thus, our findings highlight the importance of erythropoietin as a neuroregenerative treatment option in neonatal brain injury, leading to improved memory function in adolescent and adult rats which may be linked to increased neuronal network connectivity.
Collapse
|
44
|
Aversa S, Marseglia L, Manti S, D'Angelo G, Cuppari C, David A, Chirico G, Gitto E. Ventilation strategies for preventing oxidative stress-induced injury in preterm infants with respiratory disease: an update. Paediatr Respir Rev 2016; 17:71-79. [PMID: 26572937 DOI: 10.1016/j.prrv.2015.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 06/16/2015] [Accepted: 08/23/2015] [Indexed: 10/22/2022]
Abstract
Reactive oxygen and nitrogen species are produced by several inflammatory and structural cells of the airways. The lungs of preterm newborns are susceptible to oxidative injury induced by both reactive oxygen and nitrogen species. Increased oxidative stress and imbalance in antioxidant enzymes may play a role in the pathogenesis of inflammatory pulmonary diseases. Preterm infants are frequently exposed to high oxygen concentrations, infections or inflammation; they have reduced antioxidant defense and high free iron levels which enhance toxic radical generation. Multiple ventilation strategies have been studied to reduce injury and improve outcomes in preterm infants. Using lung protective strategies, there is the need to reach a compromise between satisfaction of gas exchange and potential toxicities related to over-distension, derecruitment of lung units and high oxygen concentrations. In this review, the authors summarize scientific evidence concerning oxidative stress as it relates to resuscitation in the delivery room and to the strategies of ventilation.
Collapse
Affiliation(s)
- Salvatore Aversa
- Neonatal Intensive Care Unit, Children Hospital, Spedali Civili of Brescia, Brescia, Italy, PhD course in Intensive Care, University of Messina, Messina, Italy
| | - Lucia Marseglia
- Department of Pediatrics, University of Messina, Messina, Italy.
| | - Sara Manti
- Department of Pediatrics, University of Messina, Messina, Italy
| | | | | | - Antonio David
- Department of Neurosciences, Psychiatric and Anesthesiological Sciences, University of Messina, Messina, Italy
| | - Gaetano Chirico
- Neonatal Intensive Care Unit, Children Hospital, Spedali Civili of Brescia, Brescia, Italy
| | - Eloisa Gitto
- Department of Pediatrics, University of Messina, Messina, Italy
| |
Collapse
|
45
|
Sengoku T, Murray KM, Wilson ME. Neonatal hyperoxia induces alterations in neurotrophin gene expression. Int J Dev Neurosci 2015; 48:31-7. [PMID: 26592967 DOI: 10.1016/j.ijdevneu.2015.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/13/2015] [Accepted: 11/14/2015] [Indexed: 01/16/2023] Open
Abstract
Each year in the United States, nearly 500,000 infants a year are born prematurely. Babies born before 35 weeks gestation are often placed on ventilators and/or given supplemental oxygen. This increase in oxygen, while critical for survival, can cause long-term damage to lungs, retinas and brains. In particular, hyperoxia causes apoptosis in neurons and alters glial activity. Brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) are members of the neurotrophin family of proteins that function to promote the growth, differentiation and development of the nervous system. We hypothesized that hyperoxia can alter the regulation of these genes and by doing so adversely affect the development of the brain. We predicted that mice exposed to hyperoxic conditions would have differences in BDNF and GDNF mRNA expression and relative level of methylated promoter regions coinciding with differences in the relative levels of DNMT1 and DNMT3a mRNA expression. To test this hypothesis, newborn C57Bl/6 mice and their littermates were placed in hyperoxic or normoxic conditions from postnatal day 7 to 12. There were significant decreases in BDNF mRNA expression in the prefrontal cortex following hyperoxia, but a significant increase in the isocortex. GDNF mRNA expression was significantly increased in both the isocortex and prefrontal cortex following hyperoxia. DNMT1 mRNA expression was significantly decreased in the isocortex but significantly increased in the prefrontal following hyperoxia. Together these data suggest that short-term exposure to hyperoxic conditions can affect the regulation and expression of BDNF and GDNF potentially leading to alterations in neural development.
Collapse
Affiliation(s)
- T Sengoku
- University of Kentucky, Department of Physiology, 800 Rose Street, MS 508, Lexington, KY 40536, USA
| | - K M Murray
- University of Kentucky, Department of Physiology, 800 Rose Street, MS 508, Lexington, KY 40536, USA
| | - M E Wilson
- University of Kentucky, Department of Physiology, 800 Rose Street, MS 508, Lexington, KY 40536, USA.
| |
Collapse
|
46
|
Scheuer T, Brockmöller V, Blanco Knowlton M, Weitkamp JH, Ruhwedel T, Mueller S, Endesfelder S, Bührer C, Schmitz T. Oligodendroglial maldevelopment in the cerebellum after postnatal hyperoxia and its prevention by minocycline. Glia 2015; 63:1825-39. [PMID: 25964099 DOI: 10.1002/glia.22847] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/08/2015] [Indexed: 12/11/2022]
Abstract
According to recent research, brain injury after premature birth often includes impaired growth of the cerebellum. However, causes of cerebellar injury in this population are poorly understood. In this study, we analyzed whether postnatal hyperoxia perturbs white matter development of the cerebellum, and whether cerebellar glial damage can be prevented by minocycline. We used a hyperoxia model in neonatal rats providing 24 h exposure to fourfold increased oxygen concentration (80% O2) from P6 to P7, followed by recovery in room air until P9, P11, P15, P30. Injections with minocycline were performed at the beginning and 12 h into hyperoxia exposure. Hyperoxia induced oxidative stress in the cerebellum at P7 as evidenced by increased nitrotyrosine concentrations. Numbers of proliferating, NG2+Ki67+ oligodendroglial precursor cells were decreased at P7 after hyperoxia and at P11 following recovery in room air. Numbers of mature, CC1+ oligodendrocytes were diminished in recovering hyperoxia rats, and myelin basic protein expression was still decreased at P30. Electron microscopy analysis of myelinated fibers at P30 revealed thinner myelin sheath after hyperoxia. Long-term injury of the cerebellum by neonatal hyperoxia was confirmed by reduced volumes in MRI measurements at P30. In response to 80% O2, expression of platelet-derived growth factor (PDGF)-A was largely reduced in cerebellar tissue and also in cultured cerebellar astrocytes. Treatment with minocycline during hyperoxia prevented oxidative stress, attenuated oligodendroglial injury, and improved astroglial PDGF-A levels. In conclusion, early hyperoxia causes white matter damage in the cerebellum with astroglial dysfunction being involved, and both can be prevented by treatment with minocycline. Neonatal exposure to hyperoxia causes hypomyelination of the cerebellum. Reduced astroglial growth factor production but not microglial inflammation seems to contribute to oligodendroglial damage, and minocycline rescues oligodendroglia development in the cerebellum after hyperoxia.
Collapse
Affiliation(s)
- Till Scheuer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany.,Institute of Bioanalytics, Technische Universität Berlin, Berlin, 13353, Germany
| | - Vivien Brockmöller
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | | | | | - Torben Ruhwedel
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| | - Susanne Mueller
- Center for Stroke Research, Charité University Medical Center, Berlin, Germany
| | | | - Christoph Bührer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Thomas Schmitz
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| |
Collapse
|
47
|
Neuroprotective effect of dexmedetomidine on hyperoxia-induced toxicity in the neonatal rat brain. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:530371. [PMID: 25653737 PMCID: PMC4310240 DOI: 10.1155/2015/530371] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/10/2014] [Indexed: 11/19/2022]
Abstract
Dexmedetomidine is a highly selective agonist of α2-receptors with sedative, anxiolytic, analgesic, and anesthetic properties. Neuroprotective effects of dexmedetomidine have been reported in various brain injury models. In the present study, we investigated the effects of dexmedetomidine on neurodegeneration, oxidative stress markers, and inflammation following the induction of hyperoxia in neonatal rats. Six-day-old Wistar rats received different concentrations of dexmedetomidine (1, 5, or 10 µg/kg bodyweight) and were exposed to 80% oxygen for 24 h. Sex-matched littermates kept in room air and injected with normal saline or dexmedetomidine served as controls. Dexmedetomidine pretreatment significantly reduced hyperoxia-induced neurodegeneration in different brain regions of the neonatal rat. In addition, dexmedetomidine restored the reduced/oxidized glutathione ratio and attenuated the levels of malondialdehyde, a marker of lipid peroxidation, after exposure to high oxygen concentration. Moreover, administration of dexmedetomidine induced downregulation of IL-1β on mRNA and protein level in the developing rat brain. Dexmedetomidine provides protections against toxic oxygen induced neonatal brain injury which is likely associated with oxidative stress signaling and inflammatory cytokines. Our results suggest that dexmedetomidine may have a therapeutic potential since oxygen administration to neonates is sometimes inevitable.
Collapse
|
48
|
Effects of selected dietary secondary metabolites on reactive oxygen species production caused by iron(II) autoxidation. Molecules 2014; 19:20023-33. [PMID: 25470272 PMCID: PMC4351905 DOI: 10.3390/molecules191220023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/22/2014] [Accepted: 11/24/2014] [Indexed: 11/16/2022] Open
Abstract
Iron is an essential co-factor for many enzymes that catalyze electron transfer reactions. It is well known that so-called “poorly liganded” iron can increase ROS concentrations and trigger oxidative stress that is capable of initiating apoptosis. Conversely, controlled ROS production has been recognized as an integral part of cellular signaling. Elevated ROS concentrations are associated with aging, inflammatory and degenerative diseases. Anti-aging properties have been attributed especially to antioxidant phenolic plant metabolites that represent food additives in our diet. Consequently, this study explores the effects of flavonoids (quercetin and rutin), several phenolic acids (caffeic, chlorogenic, and protocatechuic acid), and the alkaloid caffeine on iron(II) autoxidation and ROS production in comparison to the standard antioxidants ascorbic acid and Trolox. The iron(II) autoxidation assay was carried out in pH 6.0 (plant apoplast and inflamed human tissue) and 7.4 (cell cytoplasm and human blood plasma). The obtained results accentuate phenolic acids as the more specific antioxidants compared to ascorbic acid and Trolox. Flavonoid redox chemistry depends more on the chemical milieu, specifically on pH. In vivo, the presence of iron cannot be ruled out and “wrongly” or “poorly” complexed iron has been pointed out as causative agent of various age-related diseases.
Collapse
|
49
|
Tiwari KK, Chu C, Couroucli X, Moorthy B, Lingappan K. Differential concentration-specific effects of caffeine on cell viability, oxidative stress, and cell cycle in pulmonary oxygen toxicity in vitro. Biochem Biophys Res Commun 2014; 450:1345-50. [PMID: 24997337 DOI: 10.1016/j.bbrc.2014.06.132] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 06/27/2014] [Indexed: 12/21/2022]
Abstract
Caffeine is used to prevent bronchopulmonary dysplasia (BPD) in premature neonates. Hyperoxia contributes to the development of BPD, inhibits cell proliferation and decreases cell survival. The mechanisms responsible for the protective effect of caffeine in pulmonary oxygen toxicity remain largely unknown. A549 and MLE 12 pulmonary epithelial cells were exposed to hyperoxia or maintained in room air, in the presence of different concentrations (0, 0.05, 0.1 and 1mM) of caffeine. Caffeine had a differential concentration-specific effect on cell cycle progression, oxidative stress and viability, with 1mM concentration being deleterious and 0.05 mM being protective. Reactive oxygen species (ROS) generation during hyperoxia was modulated by caffeine in a similar concentration-specific manner. Caffeine at 1mM, but not at the 0.05 mM concentration decreased the G2 arrest in these cells. Taken together this study shows the novel funding that caffeine has a concentration-specific effect on cell cycle regulation, ROS generation, and cell survival in hyperoxic conditions.
Collapse
Affiliation(s)
- Kirti Kumar Tiwari
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, 1102 Bates Avenue, MC: FC530.01, Houston, TX 77030, USA
| | - Chun Chu
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, 1102 Bates Avenue, MC: FC530.01, Houston, TX 77030, USA
| | - Xanthi Couroucli
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, 1102 Bates Avenue, MC: FC530.01, Houston, TX 77030, USA
| | - Bhagavatula Moorthy
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, 1102 Bates Avenue, MC: FC530.01, Houston, TX 77030, USA
| | - Krithika Lingappan
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, 1102 Bates Avenue, MC: FC530.01, Houston, TX 77030, USA.
| |
Collapse
|