1
|
Wang Q, Guo C, Wang T, Shuai P, Wu W, Huang S, Li Y, Zhao P, Zeng C, Yi L. Drug protection against radiation-induced neurological injury: mechanisms and developments. Arch Toxicol 2025; 99:851-863. [PMID: 39724149 DOI: 10.1007/s00204-024-03933-w] [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: 09/27/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
In daily life, individuals are frequently exposed to various forms of radiation, which, when adhering to safety standards, typically result in relatively minor health effects. However, accidental exposure to radiation levels that exceed these safety standards can lead to significant health consequences. This study focuses on the analysis of radiation-induced damage to the nervous system and the mechanisms of pharmacological protection. The findings indicate that radiation can adversely affect neural structures, memory, and neurobehaviour. A range of pharmacological agents, including traditional Chinese medicine, Western medicine, and other therapeutic drugs, can be employed to safeguard the nervous system from radiation damage. The primary protective mechanisms of these agents encompass antioxidant effects, attenuation of apoptosis, and reduction of neurogenesis. A comprehensive review of these topics will offer new insights for the development and investigation of drugs aimed at mitigating radiation-induced damage to the nervous system.
Collapse
Affiliation(s)
- Qingyu Wang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Caimao Guo
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tiantian Wang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Peimeng Shuai
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wenyu Wu
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Shuqi Huang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yuanyuan Li
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pei Zhao
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Chengkai Zeng
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Lan Yi
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
2
|
Buratovic S, Philippot G, Stenerlöw B, Lönnqvist PA. Exposure to lidocaine in early life does not cause negative long-term behavioural changes in mice. Basic Clin Pharmacol Toxicol 2024; 135:210-216. [PMID: 38898535 DOI: 10.1111/bcpt.14045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/27/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND The local anaesthetic lidocaine is widely used in the neonatal intensive unit to treat seizures in premature babies. However, other antiepileptics administered during early development in various animal models have shown negative long-term behavioural effects. Since no long-term behavioural data so far exist regarding lidocaine exposure at an early age, we decided to perform this extended follow-up study using a sensitive behavioural test. METHODS Neonatal mice received a subcutaneous administration of saline or one dose of lidocaine (0.5, 4, or 12 mg kg-1) on postnatal day 10 (P10; peak of the Brain Growth Spurt). A well-established test to detect long-term behavioural alterations was conducted at 2 and 6 months of age, corresponding to early and late adulthood in humans. RESULTS All animal survived to later testing. No signs of acute toxicity were observed. Lidocaine exposure did not result in any negative behavioural effects during habituation to a new home environment at any of the two studied time points, compared to saline placebo. CONCLUSIONS Lidocaine does not by itself produce any negative long-term behavioural effects in mice exposed in early life (P10) despite long-term follow-up. This is reassuring regarding the current practice of treating seizures in premature babies with intravenous lidocaine.
Collapse
Affiliation(s)
- Sonja Buratovic
- Toxicology and Drug Safety, Department of Pharmaceutical biosciences, Uppsala University, Uppsala, Sweden
| | - Gaetan Philippot
- Toxicology and Drug Safety, Department of Pharmaceutical biosciences, Uppsala University, Uppsala, Sweden
| | - Bo Stenerlöw
- Cancer Precision Medicine, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Per-Arne Lönnqvist
- Section of Anaesthesiology & Intensive Care, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Millevert C, Vidas-Guscic N, Vanherp L, Jonckers E, Verhoye M, Staelens S, Bertoglio D, Weckhuysen S. Resting-State Functional MRI and PET Imaging as Noninvasive Tools to Study (Ab)Normal Neurodevelopment in Humans and Rodents. J Neurosci 2023; 43:8275-8293. [PMID: 38073598 PMCID: PMC10711730 DOI: 10.1523/jneurosci.1043-23.2023] [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: 09/18/2023] [Revised: 06/09/2023] [Accepted: 09/13/2023] [Indexed: 12/18/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) are a group of complex neurologic and psychiatric disorders. Functional and molecular imaging techniques, such as resting-state functional magnetic resonance imaging (rs-fMRI) and positron emission tomography (PET), can be used to measure network activity noninvasively and longitudinally during maturation in both humans and rodent models. Here, we review the current knowledge on rs-fMRI and PET biomarkers in the study of normal and abnormal neurodevelopment, including intellectual disability (ID; with/without epilepsy), autism spectrum disorder (ASD), and attention deficit hyperactivity disorder (ADHD), in humans and rodent models from birth until adulthood, and evaluate the cross-species translational value of the imaging biomarkers. To date, only a few isolated studies have used rs-fMRI or PET to study (abnormal) neurodevelopment in rodents during infancy, the critical period of neurodevelopment. Further work to explore the feasibility of performing functional imaging studies in infant rodent models is essential, as rs-fMRI and PET imaging in transgenic rodent models of NDDs are powerful techniques for studying disease pathogenesis, developing noninvasive preclinical imaging biomarkers of neurodevelopmental dysfunction, and evaluating treatment-response in disease-specific models.
Collapse
Affiliation(s)
- Charissa Millevert
- Applied & Translational Neurogenomics Group, Vlaams Instituut voor Biotechnology (VIB) Center for Molecular Neurology, VIB, Antwerp 2610, Belgium
- Department of Neurology, University Hospital of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Nicholas Vidas-Guscic
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Liesbeth Vanherp
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Elisabeth Jonckers
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Daniele Bertoglio
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Sarah Weckhuysen
- Applied & Translational Neurogenomics Group, Vlaams Instituut voor Biotechnology (VIB) Center for Molecular Neurology, VIB, Antwerp 2610, Belgium
- Department of Neurology, University Hospital of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp 2610, Belgium
| |
Collapse
|
4
|
Giovannini D, De Angelis C, Astorino MD, Fratini E, Cisbani E, Bazzano G, Ampollini A, Piccinini M, Nichelatti E, Trinca E, Nenzi P, Mancuso M, Picardi L, Marino C, Ronsivalle C, Pazzaglia S. In Vivo Radiobiological Investigations with the TOP-IMPLART Proton Beam on a Medulloblastoma Mouse Model. Int J Mol Sci 2023; 24:ijms24098281. [PMID: 37175984 PMCID: PMC10179102 DOI: 10.3390/ijms24098281] [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: 03/28/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Protons are now increasingly used to treat pediatric medulloblastoma (MB) patients. We designed and characterized a setup to deliver proton beams for in vivo radiobiology experiments at a TOP-IMPLART facility, a prototype of a proton-therapy linear accelerator developed at the ENEA Frascati Research Center, with the goal of assessing the feasibility of TOP-IMPLART for small animal proton therapy research. Mice bearing Sonic-Hedgehog (Shh)-dependent MB in the flank were irradiated with protons to test whether irradiation could be restricted to a specific depth in the tumor tissue and to compare apoptosis induced by the same dose of protons or photons. In addition, the brains of neonatal mice at postnatal day 5 (P5), representing a very small target, were irradiated with 6 Gy of protons with two different collimated Spread-Out Bragg Peaks (SOBPs). Apoptosis was visualized by immunohistochemistry for the apoptotic marker caspase-3-activated, and quantified by Western blot. Our findings proved that protons could be delivered to the upper part while sparing the deepest part of MB. In addition, a comparison of the effectiveness of protons and photons revealed a very similar increase in the expression of cleaved caspase-3. Finally, by using a very small target, the brain of P5-neonatal mice, we demonstrated that the proton irradiation field reached the desired depth in brain tissue. Using the TOP-IMPLART accelerator we established setup and procedures for proton irradiation, suitable for translational preclinical studies. This is the first example of in vivo experiments performed with a "full-linac" proton-therapy accelerator.
Collapse
Affiliation(s)
- Daniela Giovannini
- ENEA SSPT-TECS-TEB, Casaccia Research Center, Division of Health Protection Technology (TECS), Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Via Anguillarese 301, 00123 Rome, Italy
| | - Cinzia De Angelis
- Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161 Rome, Italy
| | - Maria Denise Astorino
- ENEA FSN-TECFIS-APAM, Frascati Research Center, Via Enrico Fermi 45, 00044 Frascati, Italy
| | - Emiliano Fratini
- ENEA SSPT-TECS-TEB, Casaccia Research Center, Division of Health Protection Technology (TECS), Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Via Anguillarese 301, 00123 Rome, Italy
| | - Evaristo Cisbani
- Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161 Rome, Italy
| | - Giulia Bazzano
- ENEA FSN-TECFIS-APAM, Frascati Research Center, Via Enrico Fermi 45, 00044 Frascati, Italy
| | - Alessandro Ampollini
- ENEA FSN-TECFIS-APAM, Frascati Research Center, Via Enrico Fermi 45, 00044 Frascati, Italy
| | - Massimo Piccinini
- ENEA FSN-TECFIS-MNF, Frascati Research Center, Via Enrico Fermi 45, 00044 Frascati, Italy
| | - Enrico Nichelatti
- ENEA FSN-TECFIS-MNF, Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy
| | - Emiliano Trinca
- ENEA FSN-TECFIS-APAM, Frascati Research Center, Via Enrico Fermi 45, 00044 Frascati, Italy
| | - Paolo Nenzi
- ENEA FSN-TECFIS-APAM, Frascati Research Center, Via Enrico Fermi 45, 00044 Frascati, Italy
| | - Mariateresa Mancuso
- ENEA SSPT-TECS-TEB, Casaccia Research Center, Division of Health Protection Technology (TECS), Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Via Anguillarese 301, 00123 Rome, Italy
| | - Luigi Picardi
- ENEA FSN-TECFIS-APAM, Frascati Research Center, Via Enrico Fermi 45, 00044 Frascati, Italy
| | - Carmela Marino
- ENEA SSPT-TECS-TEB, Casaccia Research Center, Division of Health Protection Technology (TECS), Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Via Anguillarese 301, 00123 Rome, Italy
| | - Concetta Ronsivalle
- ENEA FSN-TECFIS-APAM, Frascati Research Center, Via Enrico Fermi 45, 00044 Frascati, Italy
| | - Simonetta Pazzaglia
- ENEA SSPT-TECS-TEB, Casaccia Research Center, Division of Health Protection Technology (TECS), Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Via Anguillarese 301, 00123 Rome, Italy
| |
Collapse
|
5
|
Mohammed MR, El-Bahkery AM, Shedid SM. The Influence of Different γ-Irradiation Patterns on Factors that May Affect Cell Cycle Progression in Male Rats. Dose Response 2022; 20:15593258221117898. [PMID: 35982824 PMCID: PMC9379971 DOI: 10.1177/15593258221117898] [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] [Indexed: 11/16/2022]
Abstract
Most studies of the biological effects of ionizing radiation have been done on a
single acute dose, while clinically and environmentally exposures occur under
chronic/repetitive conditions. It is important to study effects of different
patterns of ionizing radiation. In this study, a rat model was used to compare
the effects of repetitive and acute exposure. Groups: (I) control, (II, III)
were exposed to fractionated doses (1.5 GyX4) and (2 GyX4), respectively/24h
interval, and (IV, V) were exposed to 6 Gy and 8 Gy of whole-body gamma
irradiation, respectively. The gene expression of MAPT and tau phosphorylation
increased in all irradiated groups but the gene expression of PKN not affected.
TGFβ% increased at dose of 2 GyX4 only. In addition, the cell cycle was arrested
in S phase. Micronucleus (MN) increased and cell proliferation decreased. In
conclusion, the dose and pattern of ionizing radiation do not affect the MAPT
and PKN gene expression, but TGF-β, p-tau, MN assay and cell proliferation are
significantly affected. The dose of 2 GyX4 showed distinctive effect. Repetitive
exposure may increase TGF-β%, which causes radio-resistance and, G2/M delay.
Thus, the cell cycle could be regulated in a different manner according to the
dose and pattern of irradiation.
Collapse
|
6
|
Gauvrit T, Benderradji H, Buée L, Blum D, Vieau D. Early-Life Environment Influence on Late-Onset Alzheimer's Disease. Front Cell Dev Biol 2022; 10:834661. [PMID: 35252195 PMCID: PMC8891536 DOI: 10.3389/fcell.2022.834661] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/27/2022] [Indexed: 12/30/2022] Open
Abstract
With the expand of the population's average age, the incidence of neurodegenerative disorders has dramatically increased over the last decades. Alzheimer disease (AD) which is the most prevalent neurodegenerative disease is mostly sporadic and primarily characterized by cognitive deficits and neuropathological lesions such as amyloid -β (Aβ) plaques and neurofibrillary tangles composed of hyper- and/or abnormally phosphorylated Tau protein. AD is considered a complex disease that arises from the interaction between environmental and genetic factors, modulated by epigenetic mechanisms. Besides the well-described cognitive decline, AD patients also exhibit metabolic impairments. Metabolic and cognitive perturbations are indeed frequently observed in the Developmental Origin of Health and Diseases (DOHaD) field of research which proposes that environmental perturbations during the perinatal period determine the susceptibility to pathological conditions later in life. In this review, we explored the potential influence of early environmental exposure to risk factors (maternal stress, malnutrition, xenobiotics, chemical factors … ) and the involvement of epigenetic mechanisms on the programming of late-onset AD. Animal models indicate that offspring exposed to early-life stress during gestation and/or lactation increase both AD lesions, lead to defects in synaptic plasticity and finally to cognitive impairments. This long-lasting epigenetic programming could be modulated by factors such as nutriceuticals, epigenetic modifiers or psychosocial behaviour, offering thus future therapeutic opportunity to protect from AD development.
Collapse
Affiliation(s)
- Thibaut Gauvrit
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Hamza Benderradji
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Luc Buée
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Didier Vieau
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| |
Collapse
|
7
|
Wood TR, Parikh P, Comstock BA, Law JB, Bammler TK, Kuban KC, Mayock DE, Heagerty PJ, Juul S, for the PENUT Trial consortium. Early Biomarkers of Hypoxia and Inflammation and Two-Year Neurodevelopmental Outcomes in the Preterm Erythropoietin Neuroprotection (PENUT) Trial. EBioMedicine 2021; 72:103605. [PMID: 34619638 PMCID: PMC8498235 DOI: 10.1016/j.ebiom.2021.103605] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 08/24/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND In the Preterm Erythropoietin (Epo) NeUroproTection (PENUT) Trial, potential biomarkers of neurological injury were measured to determine their association with outcomes at two years of age and whether Epo treatment decreased markers of inflammation in extremely preterm (<28 weeks' gestation) infants. METHODS Plasma Epo was measured (n=391 Epo, n=384 placebo) within 24h after birth (baseline), 30min after study drug administration (day 7), 30min before study drug (day 9), and on day 14. A subset of infants (n=113 Epo, n=107 placebo) had interferon-gamma (IFN-γ), Interleukin (IL)-6, IL-8, IL-10, Tau, and tumour necrosis factor-α (TNF-α) levels evaluated at baseline, day 7 and 14. Infants were then evaluated at 2 years using the Bayley Scales of Infant and Toddler Development, 3rd Edition (BSID-III). FINDINGS Elevated baseline Epo was associated with increased risk of death or severe disability (BSID-III Motor and Cognitive subscales <70 or severe cerebral palsy). No difference in other biomarkers were seen between treatment groups at any time, though Epo appeared to mitigate the association between elevated baseline IL-6 and lower BSID-III scores in survivors. Elevated baseline, day 7 and 14 Tau concentrations were associated with worse BSID-III Cognitive, Motor, and Language skills at two years. INTERPRETATION Elevated Epo at baseline and elevated Tau in the first two weeks after birth predict poor outcomes in infants born extremely preterm. However, no clear prognostic cut-off values are apparent, and further work is required before these biomarkers can be widely implemented in clinical practice. FUNDING PENUT was funded by the National Institute of Neurological Disorders and Stroke (U01NS077955 and U01NS077953).
Collapse
Affiliation(s)
- Thomas R. Wood
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
| | - Pratik Parikh
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
| | | | - Janessa B. Law
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
| | - Theo K. Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Karl C. Kuban
- Department of Pediatrics, Boston University School of Medicine, Boston, MA
| | - Dennis E. Mayock
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
| | | | - Sandra Juul
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
| | - for the PENUT Trial consortium
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
- Department of Biostatistics, University of Washington, Seattle, WA
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
- Department of Pediatrics, Boston University School of Medicine, Boston, MA
| |
Collapse
|
8
|
Serrano C, Dos Santos M, Kereselidze D, Beugnies L, Lestaevel P, Poirier R, Durand C. Targeted Dorsal Dentate Gyrus or Whole Brain Irradiation in Juvenile Mice Differently Affects Spatial Memory and Adult Hippocampal Neurogenesis. BIOLOGY 2021; 10:biology10030192. [PMID: 33806303 PMCID: PMC8002088 DOI: 10.3390/biology10030192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 02/26/2021] [Indexed: 12/11/2022]
Abstract
The cognitive consequences of postnatal brain exposure to ionizing radiation (IR) at low to moderate doses in the adult are not fully established. Because of the advent of pediatric computed tomography scans used for head exploration, improving our knowledge of these effects represents a major scientific challenge. To evaluate how IR may affect the developing brain, models of either whole brain (WB) or targeted dorsal dentate gyrus (DDG) irradiation in C57Bl/6J ten-day-old male mice were previously developed. Here, using these models, we assessed and compared the effect of IR (doses range: 0.25-2 Gy) on long-term spatial memory in adulthood using a spatial water maze task. We then evaluated the effects of IR exposure on adult hippocampal neurogenesis, a form of plasticity involved in spatial memory. Three months after WB exposure, none of the doses resulted in spatial memory impairment. In contrast, a deficit in memory retrieval was identified after DDG exposure for the dose of 1 Gy only, highlighting a non-monotonic dose-effect relationship in this model. At this dose, a brain irradiated volume effect was also observed when studying adult hippocampal neurogenesis in the two models. In particular, only DDG exposure caused alteration in cell differentiation. The most deleterious effect observed in adult hippocampal neurogenesis after targeted DDG exposure at 1 Gy may contribute to the memory retrieval deficit in this model. Altogether these results highlight the complexity of IR mechanisms in the brain that can lead or not to cognitive disorders and provide new knowledge of interest for the radiation protection of children.
Collapse
Affiliation(s)
- Céline Serrano
- Laboratory of Experimental Radiotoxicology and Radiobiology (LRTOX), Research Department on the Biological and Health Effects of Ionizing Radiation (SESANE), Institute for Radiological Protection and Nuclear Safety (IRSN), 92260 Fontenay-aux-Roses, France; (C.S.); (D.K.); (L.B.); (P.L.)
| | - Morgane Dos Santos
- Laboratory of Radiobiology of Accidental Exposure (LRAcc), Research Department in Radiobiology and Regenerative Medicine (SERAMED), Institute for Radiological Protection and Nuclear Safety (IRSN), 92260 Fontenay-aux-Roses, France;
| | - Dimitri Kereselidze
- Laboratory of Experimental Radiotoxicology and Radiobiology (LRTOX), Research Department on the Biological and Health Effects of Ionizing Radiation (SESANE), Institute for Radiological Protection and Nuclear Safety (IRSN), 92260 Fontenay-aux-Roses, France; (C.S.); (D.K.); (L.B.); (P.L.)
| | - Louison Beugnies
- Laboratory of Experimental Radiotoxicology and Radiobiology (LRTOX), Research Department on the Biological and Health Effects of Ionizing Radiation (SESANE), Institute for Radiological Protection and Nuclear Safety (IRSN), 92260 Fontenay-aux-Roses, France; (C.S.); (D.K.); (L.B.); (P.L.)
| | - Philippe Lestaevel
- Laboratory of Experimental Radiotoxicology and Radiobiology (LRTOX), Research Department on the Biological and Health Effects of Ionizing Radiation (SESANE), Institute for Radiological Protection and Nuclear Safety (IRSN), 92260 Fontenay-aux-Roses, France; (C.S.); (D.K.); (L.B.); (P.L.)
| | - Roseline Poirier
- Paris-Saclay Neuroscience Institute (Neuro-PSI), University Paris-Saclay, UMR 9197 CNRS, F-91405 Orsay, France
- Correspondence: (R.P.); (C.D.)
| | - Christelle Durand
- Laboratory of Experimental Radiotoxicology and Radiobiology (LRTOX), Research Department on the Biological and Health Effects of Ionizing Radiation (SESANE), Institute for Radiological Protection and Nuclear Safety (IRSN), 92260 Fontenay-aux-Roses, France; (C.S.); (D.K.); (L.B.); (P.L.)
- Correspondence: (R.P.); (C.D.)
| |
Collapse
|
9
|
Pasqual E, Boussin F, Bazyka D, Nordenskjold A, Yamada M, Ozasa K, Pazzaglia S, Roy L, Thierry-Chef I, de Vathaire F, Benotmane MA, Cardis E. Cognitive effects of low dose of ionizing radiation - Lessons learned and research gaps from epidemiological and biological studies. ENVIRONMENT INTERNATIONAL 2021; 147:106295. [PMID: 33341586 DOI: 10.1016/j.envint.2020.106295] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/02/2020] [Accepted: 10/20/2020] [Indexed: 06/12/2023]
Abstract
The last decades have seen increased concern about the possible effects of low to moderate doses of ionizing radiation (IR) exposure on cognitive function. An interdisciplinary group of experts (biologists, epidemiologists, dosimetrists and clinicians) in this field gathered together in the framework of the European MELODI workshop on non-cancer effects of IR to summarise the state of knowledge on the topic and elaborate research recommendations for future studies in this area. Overall, there is evidence of cognitive effects from low IR doses both from biology and epidemiology, though a better characterization of effects and understanding of mechanisms is needed. There is a need to better describe the specific cognitive function or diseases that may be affected by radiation exposure. Such cognitive deficit characterization should consider the human life span, as effects might differ with age at exposure and at outcome assessment. Measurements of biomarkers, including imaging, will likely help our understanding on the mechanism of cognitive-related radiation induced deficit. The identification of loci of individual genetic susceptibility and the study of gene expression may help identify individuals at higher risk. The mechanisms behind the radiation induced cognitive effects are not clear and are likely to involve several biological pathways and different cell types. Well conducted research in large epidemiological cohorts and experimental studies in appropriate animal models are needed to improve the understanding of radiation-induced cognitive effects. Results may then be translated into recommendations for clinical radiation oncology and imaging decision making processes.
Collapse
Affiliation(s)
- Elisa Pasqual
- Barcelona Institute for Global Health (ISGlobal), Campus Mar, Barcelona Biomedical Research Park (PRBB), Dr Aiguader 88, 08003 Barcelona, Spain; University Pompeu Fabra, Barcelona, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain.
| | - François Boussin
- Université de Paris and Université Paris-Saclay, Inserm, LRP/iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
| | - Dimitry Bazyka
- National Research Center for Radiation Medicine, 53 Illenko str., Kyiv, Ukraine
| | - Arvid Nordenskjold
- Department of Clinical Neuroscience, Division of Neurology, Karolinska Institutet, Stockholm, Sweden
| | - Michiko Yamada
- Department of Clinical Studies, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Kotaro Ozasa
- Department of Epidemiology, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Simonetta Pazzaglia
- Laboratory of Biomedical Technologies, ENEA CR-Casaccia, Via Anguillarese 301, 00123 Rome, Italy
| | - Laurence Roy
- Department for Research on the Biological and Health Effects of Ionising Radiation. Institut of Radiation Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Isabelle Thierry-Chef
- Barcelona Institute for Global Health (ISGlobal), Campus Mar, Barcelona Biomedical Research Park (PRBB), Dr Aiguader 88, 08003 Barcelona, Spain; University Pompeu Fabra, Barcelona, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain
| | - Florent de Vathaire
- Radiation Epidemiology Teams, INSERM Unit 1018, University Paris Saclay, Gustave Roussy, 94800 Villejuif, France
| | | | - Elisabeth Cardis
- Barcelona Institute for Global Health (ISGlobal), Campus Mar, Barcelona Biomedical Research Park (PRBB), Dr Aiguader 88, 08003 Barcelona, Spain; University Pompeu Fabra, Barcelona, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain
| |
Collapse
|
10
|
Combined Treatment with Low-Dose Ionizing Radiation and Ketamine Induces Adverse Changes in CA1 Neuronal Structure in Male Murine Hippocampi. Int J Mol Sci 2019; 20:ijms20236103. [PMID: 31817026 PMCID: PMC6929167 DOI: 10.3390/ijms20236103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
In children, ketamine sedation is often used during radiological procedures. Combined exposure of ketamine and radiation at doses that alone did not affect learning and memory induced permanent cognitive impairment in mice. The aim of this study was to elucidate the mechanism behind this adverse outcome. Neonatal male NMRI mice were administered ketamine (7.5 mg kg−1) and irradiated (whole-body, 100 mGy or 200 mGy, 137Cs) one hour after ketamine exposure on postnatal day 10. The control mice were injected with saline and sham-irradiated. The hippocampi were analyzed using label-free proteomics, immunoblotting, and Golgi staining of CA1 neurons six months after treatment. Mice co-exposed to ketamine and low-dose radiation showed alterations in hippocampal proteins related to neuronal shaping and synaptic plasticity. The expression of brain-derived neurotrophic factor, activity-regulated cytoskeleton-associated protein, and postsynaptic density protein 95 were significantly altered only after the combined treatment (100 mGy or 200 mGy combined with ketamine, respectively). Increased numbers of basal dendrites and branching were observed only after the co-exposure, thereby constituting a possible reason for the displayed alterations in behavior. These data suggest that the risk of radiation-induced neurotoxic effects in the pediatric population may be underestimated if based only on the radiation dose.
Collapse
|
11
|
Peng S, Yang B, Duan MY, Liu ZW, Wang WF, Zhang XZ, Ren BX, Tang FR. The Disparity of Impairment of Neurogenesis and Cognition After Acute or Fractionated Radiation Exposure in Adolescent BALB/c Mice. Dose Response 2019; 17:1559325818822574. [PMID: 30670940 PMCID: PMC6327339 DOI: 10.1177/1559325818822574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 09/20/2018] [Accepted: 09/25/2018] [Indexed: 12/31/2022] Open
Abstract
The effect of acute X-ray irradiation with 2 Gy or fractionated exposure with 0.2 Gy continuously for 10 days (0.2 Gy × 10 = 2 Gy) was evaluated in the postnatal day 21 (P21) BALB/c mouse model. Both acute and fractionated irradiation induced impairment of cell proliferation and neurogenesis in the subgranular zone of the dentate gyrus labeled by Ki67 and doublecortin, respectively. Parvalbumin immunopositive interneurons in the subgranular zone were also reduced significantly. However, the 2 patterns of irradiation did not affect animal weight gain when measured at ages of P90 and P180 or 69 and 159 days after irradiation. Behavioral tests indicated that neither acute nor fractionated irradiation with a total dose of 2 Gy induced deficits in the contextual fear or spatial memory and memory for novel object recognition. Animal motor activity was also not affected in the open-field test. The disparity of the impairment of neurogenesis and unaffected cognition suggests that the severity of impairment of neurogenesis induced by acute or fractionated irradiation with a total dose of 2 Gy at P21 may not be worse enough to induce the deficit of cognition.
Collapse
Affiliation(s)
- Shuang Peng
- Health Center of Yangtze University, Jingzhou, Hubei, China
| | - Bo Yang
- Department of Medical Imaging Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Meng Yun Duan
- Health Center of Yangtze University, Jingzhou, Hubei, China
| | - Zi Wei Liu
- Health Center of Yangtze University, Jingzhou, Hubei, China
| | - Wei Feng Wang
- Health Center of Yangtze University, Jingzhou, Hubei, China
| | - Xiang Zhi Zhang
- Affiliated Hospital of Yangtze University, Jingzhou, Hubei, People's Republic of China
| | - Bo Xu Ren
- Health Center of Yangtze University, Jingzhou, Hubei, China
| | - Feng Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| |
Collapse
|
12
|
Dos Santos M, Kereselidze D, Gloaguen C, Benadjaoud MA, Tack K, Lestaevel P, Durand C. Development of whole brain versus targeted dentate gyrus irradiation model to explain low to moderate doses of exposure effects in mice. Sci Rep 2018; 8:17262. [PMID: 30467388 PMCID: PMC6250717 DOI: 10.1038/s41598-018-35579-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 10/16/2018] [Indexed: 12/23/2022] Open
Abstract
Evaluation of the consequences of low to moderate doses of ionizing radiation (IR) remains a societal challenge, especially for children exposed to CT scans. Appropriate experimental models are needed to improve scientific understanding of how exposure of the postnatal brain to IR affects behavioral functions and their related pathophysiological mechanisms, considering brain complex functional organization. In the brain, the dorsal and ventral hippocampal dentate gyrus can be involved in distinct major behavioral functions. To study the long term behavioral effects of brain exposure at low to moderate doses of IR (doses range 0.25–1 Gy), we developed three new experimental models in 10-day-old mice: a model of brain irradiation and two targeted irradiation models of the dorsal and ventral dentate gyrus. We used the technological properties of the SARRP coupled with MR imaging. Our irradiation strategy has been twofold endorsed. The millimetric ballistic specificity of our models was first validated by measuring gamma-H2AX increase after irradiation. We then demonstrated higher anxiety/depressive-like behavior, preferentially mediate by the ventral part of the dentate gyrus, in mice after brain and ventral dentate gyrus IR exposure. This work provides new tools to enhance scientific understanding of how to protect children exposed to IR.
Collapse
Affiliation(s)
- M Dos Santos
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department of RAdiobiology and regenerative MEDicine (SERAMED), Laboratory of Radiobiology of Accidental exposures (LRAcc), Fontenay-aux-Roses, France
| | - D Kereselidze
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department on the Biological and Health Effects of Ionizing Radiation (SESANE), Laboratory of experimental Radiotoxicology and Radiobiology (LRTOX), Fontenay aux Roses, France
| | - C Gloaguen
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department on the Biological and Health Effects of Ionizing Radiation (SESANE), Laboratory of experimental Radiotoxicology and Radiobiology (LRTOX), Fontenay aux Roses, France
| | - M A Benadjaoud
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department of RAdiobiology and regenerative MEDicine (SERAMED), Fontenay-aux-Roses, France
| | - K Tack
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department on the Biological and Health Effects of Ionizing Radiation (SESANE), Laboratory of experimental Radiotoxicology and Radiobiology (LRTOX), Fontenay aux Roses, France
| | - P Lestaevel
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department on the Biological and Health Effects of Ionizing Radiation (SESANE), Laboratory of experimental Radiotoxicology and Radiobiology (LRTOX), Fontenay aux Roses, France
| | - C Durand
- Institute for Radiological Protection and Nuclear Safety (IRSN), Research department on the Biological and Health Effects of Ionizing Radiation (SESANE), Laboratory of experimental Radiotoxicology and Radiobiology (LRTOX), Fontenay aux Roses, France.
| |
Collapse
|
13
|
Philippot G, Stenerlöw B, Fredriksson A, Sundell‐Bergman S, Eriksson P, Buratovic S. Developmental effects of neonatal fractionated co‐exposure to low‐dose gamma radiation and paraquat on behaviour in adult mice. J Appl Toxicol 2018; 39:582-589. [DOI: 10.1002/jat.3748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/26/2018] [Accepted: 10/07/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Gaëtan Philippot
- Department of Environmental ToxicologyUppsala University Uppsala Sweden
| | - Bo Stenerlöw
- Department of Immunology, Genetics and PathologyUppsala University Uppsala Sweden
| | | | - Synnöve Sundell‐Bergman
- Department of Soil and EnvironmentSwedish University of Agricultural Sciences Uppsala Sweden
| | - Per Eriksson
- Department of Environmental ToxicologyUppsala University Uppsala Sweden
| | - Sonja Buratovic
- Department of Environmental ToxicologyUppsala University Uppsala Sweden
| |
Collapse
|
14
|
Kempf SJ, Janik D, Barjaktarovic Z, Braga-Tanaka I, Tanaka S, Neff F, Saran A, Larsen MR, Tapio S. Chronic low-dose-rate ionising radiation affects the hippocampal phosphoproteome in the ApoE-/- Alzheimer's mouse model. Oncotarget 2018; 7:71817-71832. [PMID: 27708245 PMCID: PMC5342125 DOI: 10.18632/oncotarget.12376] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/20/2016] [Indexed: 12/22/2022] Open
Abstract
Accruing data indicate that radiation-induced consequences resemble pathologies of neurodegenerative diseases such as Alzheimer´s. The aim of this study was to elucidate the effect on hippocampus of chronic low-dose-rate radiation exposure (1 mGy/day or 20 mGy/day) given over 300 days with cumulative doses of 0.3 Gy and 6.0 Gy, respectively. ApoE deficient mutant C57Bl/6 mouse was used as an Alzheimer´s model. Using mass spectrometry, a marked alteration in the phosphoproteome was found at both dose rates. The radiation-induced changes in the phosphoproteome were associated with the control of synaptic plasticity, calcium-dependent signalling and brain metabolism. An inhibition of CREB signalling was found at both dose rates whereas Rac1-Cofilin signalling was found activated only at the lower dose rate. Similarly, the reduction in the number of activated microglia in the molecular layer of hippocampus that paralleled with reduced levels of TNFα expression and lipid peroxidation was significant only at the lower dose rate. Adult neurogenesis, investigated by Ki67, GFAP and NeuN staining, and cell death (activated caspase-3) were not influenced at any dose or dose rate. This study shows that several molecular targets induced by chronic low-dose-rate radiation overlap with those of Alzheimer´s pathology. It may suggest that ionising radiation functions as a contributing risk factor to this neurodegenerative disease.
Collapse
Affiliation(s)
- Stefan J Kempf
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Dirk Janik
- Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Zarko Barjaktarovic
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | | | | | - Frauke Neff
- Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Anna Saran
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l´Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| |
Collapse
|
15
|
Feng P, Chen Y, Zhang L, Qian CG, Xiao X, Han X, Shen QD. Near-Infrared Fluorescent Nanoprobes for Revealing the Role of Dopamine in Drug Addiction. ACS APPLIED MATERIALS & INTERFACES 2018; 10:4359-4368. [PMID: 29308644 DOI: 10.1021/acsami.7b12005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Brain imaging techniques enable visualizing the activity of central nervous system without invasive neurosurgery. Dopamine is an important neurotransmitter. Its fluctuation in brain leads to a wide range of diseases and disorders, like drug addiction, depression, and Parkinson's disease. We designed near-infrared fluorescence dopamine-responsive nanoprobes (DRNs) for brain activity imaging during drug abuse and addiction process. On the basis of light-induced electron transfer between DRNs and dopamine and molecular wire effect of the DRNs, we can track the dynamical change of the neurotransmitter level in the physiological environment and the releasing of the neurotransmitter in living dopaminergic neurons in response to nicotine stimulation. The functional near-infrared fluorescence imaging can dynamically track the dopamine level in the mice midbrain under normal or drug-activated condition and evaluate the long-term effect of addictive substances to the brain. This strategy has the potential for studying neural activity under physiological condition.
Collapse
Affiliation(s)
- Peijian Feng
- Department of Polymer Science and Engineering, Key Laboratory of High Performance Polymer Materials and Technology of MOE, Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210023, China
| | - Yulei Chen
- Department of Polymer Science and Engineering, Key Laboratory of High Performance Polymer Materials and Technology of MOE, Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210023, China
| | - Lei Zhang
- Department of Biomedical Engineering, College of Engineering and Applied Science, Nanjing University , Nanjing 210093, China
| | - Cheng-Gen Qian
- Department of Polymer Science and Engineering, Key Laboratory of High Performance Polymer Materials and Technology of MOE, Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210023, China
| | - Xuanzhong Xiao
- Department of Polymer Science and Engineering, Key Laboratory of High Performance Polymer Materials and Technology of MOE, Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210023, China
| | - Xu Han
- Department of Polymer Science and Engineering, Key Laboratory of High Performance Polymer Materials and Technology of MOE, Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210023, China
| | - Qun-Dong Shen
- Department of Polymer Science and Engineering, Key Laboratory of High Performance Polymer Materials and Technology of MOE, Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry & Chemical Engineering, Nanjing University , Nanjing 210023, China
| |
Collapse
|
16
|
Effects on adult cognitive function after neonatal exposure to clinically relevant doses of ionising radiation and ketamine in mice. Br J Anaesth 2018; 120:546-554. [PMID: 29452811 DOI: 10.1016/j.bja.2017.11.099] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 10/31/2017] [Accepted: 11/10/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Radiological methods for screening, diagnostics and therapy are frequently used in healthcare. In infants and children, anaesthesia/sedation is often used in these situations to relieve the patients' perception of stress or pain. Both ionising radiation (IR) and ketamine have been shown to induce developmental neurotoxic effects and this study aimed to identify the combined effects of these in a murine model. METHODS Male mice were exposed to a single dose of ketamine (7.5 mg kg-1 body weight) s.c. on postnatal day 10. One hour after ketamine exposure, mice were whole body irradiated with 50-200 mGy gamma radiation (137Cs). Behavioural observations were performed at 2, 4 and 5 months of age. At 6 months of age, cerebral cortex and hippocampus tissue were analysed for neuroprotein levels. RESULTS Animals co-exposed to IR and ketamine displayed significant (P≤0.01) lack of habituation in the spontaneous behaviour test, when compared with controls and single agent exposed mice. In the Morris Water Maze test, co-exposed animals showed significant (P≤0.05) impaired learning and memory capacity in both the spatial acquisition task and the relearning test compared with controls and single agent exposed mice. Furthermore, in co-exposed mice a significantly (P≤0.05) elevated level of tau protein in cerebral cortex was observed. Single agent exposure did not cause any significant effects on the investigated endpoints. CONCLUSION Co-exposure to IR and ketamine can aggravate developmental neurotoxic effects at doses where the single agent exposure does not impact on the measured variables. These findings show that estimation of risk after paediatric low-dose IR exposure, based upon radiation dose alone, may underestimate the consequences for this vulnerable population.
Collapse
|
17
|
Shinsuke K, Junya K, Tomonobu U, Yoshiko K, Izumo N, Takahiko S. Chronic irradiation with low-dose-rate 137Cs-γ rays inhibits NGF-induced neurite extension of PC12 cells via Ca2+/calmodulin-dependent kinase II activation. JOURNAL OF RADIATION RESEARCH 2017; 58:809-815. [PMID: 29106600 PMCID: PMC5710646 DOI: 10.1093/jrr/rrx032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 02/27/2017] [Indexed: 06/07/2023]
Abstract
Chronic irradiation with low-dose-rate 137Cs-γ rays inhibits the differentiation of human neural progenitor cells and influences the expression of proteins associated with several cellular functions. We aimed to determine whether such chronic irradiation influences the expression of proteins associated with PC12 cells. Chronic irradiation at 0.027 mGy/min resulted in inhibition of NGF-induced neurite extension. Furthermore, irradiation enhanced the nerve growth factor (NGF)-induced increase in the phosphorylation of extracellular signal-regulated kinase (ERK), but did not affect the phosphorylation of NGF receptors, suggesting that irradiation influences pathways unassociated with the activation of ERK. We then examined whether irradiation influenced the Akt-Rac1 pathway, which is unaffected by ERK activation. Chronic irradiation also enhanced the NGF-induced increase in Akt phosphorylation, but markedly inhibited the NGF-induced increase in Rac1 activity that is associated with neurite extension. These results suggest that the inhibitory effect of irradiation on neurite extension influences pathways unassociated with Akt activation. As Ca2+/calmodulin-dependent kinase II (CaMKII) is known to inhibit the NGF-induced neurite extension in PC12 cells, independent of ERK and Akt activation, we next examined the effects of irradiation on CaMKII activation. Chronic irradiation induced CaMKII activation, while application of KN-62 (a specific inhibitor of CaMKII), attenuated increases in CaMKII activation and recovered neurite extension and NGF-induced increases in Rac1 activity that was inhibited by irradiation. Our results suggest that chronic irradiation with low-dose-rate γ-rays inhibits Rac1 activity via CaMKII activation, thereby inhibiting NGF-induced neurite extension.
Collapse
Affiliation(s)
- Katoh Shinsuke
- Research Center for Radiation Science, Yokohama University of Pharmacy, 601 Matano-cho, Totsuka-ku, Yokohama 245-0066, Japan
| | - Kobayashi Junya
- Radiation Biology Center, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Umeda Tomonobu
- Research Center for Radiation Science, Yokohama University of Pharmacy, 601 Matano-cho, Totsuka-ku, Yokohama 245-0066, Japan
| | - Kobayashi Yoshiko
- Research Center for Radiation Science, Yokohama University of Pharmacy, 601 Matano-cho, Totsuka-ku, Yokohama 245-0066, Japan
| | - Nobuo Izumo
- General Health Medical Center, Yokohama University of Pharmacy, 601 Matano-cho, Totsuka-ku, Yokohama 245-0066, Japan
| | - Suzuki Takahiko
- Clinical Radiology, Faculty of Medical Technology, Teikyo University, 2-11-1 Kaga, Itabashi 173-8605, Japan
| |
Collapse
|
18
|
A transient insulin system dysfunction in newborn rat brain followed by neonatal intracerebroventricular administration of streptozotocin could be accompanied by a labile cognitive impairment. Neurosci Res 2017; 132:17-25. [PMID: 29055675 DOI: 10.1016/j.neures.2017.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 09/16/2017] [Accepted: 10/06/2017] [Indexed: 01/22/2023]
Abstract
The early postnatal period is a critical period of hippocampus development, which is highly dependent on insulin receptor (IR) signaling and very important in cognitive function. The present study was conducted in order to present a model of neonatal transient brain insulin system dysfunction through finding an appropriate dose of injection of streptozotocin (STZ) during the neonatal period. Sixty male Wistar rat pups were divided into 4 groups of 15 and received intracerebroventricular saline or STZ (icv-STZ) (15, 20 and 25μg/kg) on postnatal day 7. Gene expression of IR and target genes for IR signaling (choline acetyltransferase (ChAT) and Tau) were measured at the ages of 2 and 7 weeks. Behavioral tests were performed at the ages of 3 and 6 weeks to assess short- and long-term cognitive function. 20μg/kg dose of icv-STZ was estimated as the optimal dose causing transient alteration in gene expression of IR, ChAT and Tau. Additionally, cognitive function of the animals restored to normal level at the age of 6 weeks. Therefore, 20μg/kg dose of icv-STZ is proposed as a new approach to generating transient brain insulin system dysfunction associated with transient cognitive impairments at a critical postnatal period of brain development.
Collapse
|
19
|
Tang FR, Loke WK, Wong P, Khoo BC. Radioprotective effect of ursolic acid in radiation-induced impairment of neurogenesis, learning and memory in adolescent BALB/c mouse. Physiol Behav 2017; 175:37-46. [PMID: 28341234 DOI: 10.1016/j.physbeh.2017.03.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/13/2017] [Accepted: 03/20/2017] [Indexed: 10/19/2022]
Abstract
The effect of acute irradiation with 5Gy or fractionated exposure with 0.5Gy continuously for 10days (a total dose of 5Gy) was evaluated in an immature BALB/c mouse model. Radioprotective effect of ursolic acid (at 25mg/kg/daily administered 1h after acute or each of fractionated irradiations, and continuously for 30days) was also investigated. We found that both acute and fractionated irradiation at a total dose of 5Gy did not induce any mortality within 30days after exposure to postnatal day 26 (P26) BALB/c mice, but reduced animal weigh gain in the first few weeks. At 90days after irradiation, the weight of animals with acute irradiation was still significantly lower than the control group; no significant difference though was observed for those fractionatedly exposed mice compared to the control group. Behavioral tests indicated that acute irradiation at 5Gy induced deficits in learning and memory in the contextual fear conditioning test. The memory for novel object recognition was also impaired. Similar changes were not observed in mice with fractionated irradiation. Immunohistochemical study demonstrated clearly that acute and fractionated irradiations induced impairment of neurogenesis in the subgranular zone (SGZ) of the dentate gyrus although fractionated exposure induced much lesser loss of newly generated neurons. Ursolic acid administered at 25mg/kg/daily for 30days after irradiation greatly improved acute irradiation-induced deficits in contextual learning and memory and in novel object recognition memory although it exacerbated radiation-induced reduction of neurogenesis in SGZ.
Collapse
Affiliation(s)
- Feng Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety initiative, National University of Singapore, 1 CREATE Way #04-01, CREATE Tower, 138602, Singapore.
| | - Weng Keong Loke
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 11 Stockport Road, 11760, Singapore
| | - Peiyan Wong
- Neuroscience Phenotyping Core, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Boo Cheong Khoo
- Temasek Laboratories, National University of Singapore, 5A, Engineering Drive 1, 117411, Singapore
| |
Collapse
|
20
|
Exposure to low doses of 137cesium and nicotine during postnatal development modifies anxiety levels, learning, and spatial memory performance in mice. Food Chem Toxicol 2016; 97:82-88. [PMID: 27590783 DOI: 10.1016/j.fct.2016.08.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 12/18/2022]
Abstract
Radiation therapy is a major cause of long-term complications observed in survivors of pediatric brain tumors. However, the effects of low-doses of ionizing radiation (IR) to the brain are less studied. On the other hand, tobacco is one of the most heavily abused drugs in the world. Tobacco is not only a health concern for adults. It has also shown to exert deleterious effects on fetuses, newborns, children and adolescents. Exposure to nicotine (Nic) from smoking may potentiate the toxic effects induced by IR on brain development. In this study, we evaluated in mice the cognitive effects of concomitant exposure to low doses of internal radiation (137Cs) and Nic during neonatal brain development. On postnatal day 10 (PND10), two groups of C57BL/6J mice were subcutaneously exposed to 137-Cesium (137Cs) (4000 and 8000 Bq/kg) and/or Nic (100 μg/ml). At the age of two months, neurobehavior of mice was assessed. Results showed that exposure to IR-alone or in combination with Nic-increased the anxiety-like of the animals without changing the activity levels. Moreover, exposure to IR impaired learning and spatial memory. However, Nic administration was able to reverse this effect, but only at the low dose of 137Cs.
Collapse
|
21
|
Verreet T, Rangarajan JR, Quintens R, Verslegers M, Lo AC, Govaerts K, Neefs M, Leysen L, Baatout S, Maes F, Himmelreich U, D'Hooge R, Moons L, Benotmane MA. Persistent Impact of In utero Irradiation on Mouse Brain Structure and Function Characterized by MR Imaging and Behavioral Analysis. Front Behav Neurosci 2016; 10:83. [PMID: 27199692 PMCID: PMC4854899 DOI: 10.3389/fnbeh.2016.00083] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/13/2016] [Indexed: 01/29/2023] Open
Abstract
Prenatal irradiation is known to perturb brain development. Epidemiological studies revealed that radiation exposure during weeks 8-15 of pregnancy was associated with an increased occurrence of mental disability and microcephaly. Such neurological deficits were reproduced in animal models, in which rodent behavioral testing is an often used tool to evaluate radiation-induced defective brain functionality. However, up to now, animal studies suggested a threshold dose of around 0.30 Gray (Gy) below which no behavioral alterations can be observed, while human studies hinted at late defects after exposure to doses as low as 0.10 Gy. Here, we acutely irradiated pregnant mice at embryonic day 11 with doses ranging from 0.10 to 1.00 Gy. A thorough investigation of the dose-response relationship of altered brain function and architecture following in utero irradiation was achieved using a behavioral test battery and volumetric 3D T2-weighted magnetic resonance imaging (MRI). We found dose-dependent changes in cage activity, social behavior, anxiety-related exploration, and spatio-cognitive performance. Although behavioral alterations in low-dose exposed animals were mild, we did unveil that both emotionality and higher cognitive abilities were affected in mice exposed to ≥0.10 Gy. Microcephaly was apparent from 0.33 Gy onwards and accompanied by deviations in regional brain volumes as compared to controls. Of note, total brain volume and the relative volume of the ventricles, frontal and posterior cerebral cortex, cerebellum, and striatum were most strongly correlated to altered behavioral parameters. Taken together, we present conclusive evidence for persistent low-dose effects after prenatal irradiation in mice and provide a better understanding of the correlation between their brain size and performance in behavioral tests.
Collapse
Affiliation(s)
- Tine Verreet
- Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK•CEN)Mol, Belgium; Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, Faculty of Science, Katholieke Universiteit LeuvenLeuven, Belgium
| | - Janaki Raman Rangarajan
- Faculty of Medicine, Molecular Small Animal Imaging Center, Katholieke Universiteit LeuvenLeuven, Belgium; Department of Electrical Engineering (ESAT/PSI), Katholieke Universiteit Leuven and Medical Image Research Center, University Hospital LeuvenLeuven, Belgium
| | - Roel Quintens
- Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK•CEN) Mol, Belgium
| | - Mieke Verslegers
- Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK•CEN) Mol, Belgium
| | - Adrian C Lo
- Laboratory of Biological Psychology, Faculty of Psychology and Educational Sciences, Katholieke Universiteit Leuven Leuven, Belgium
| | - Kristof Govaerts
- Biomedical MRI Unit, Department of Imaging and Pathology, Faculty of Medicine, Katholieke Universiteit Leuven Leuven, Belgium
| | - Mieke Neefs
- Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK•CEN) Mol, Belgium
| | - Liselotte Leysen
- Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK•CEN) Mol, Belgium
| | - Sarah Baatout
- Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK•CEN) Mol, Belgium
| | - Frederik Maes
- Department of Electrical Engineering (ESAT/PSI), Katholieke Universiteit Leuven and Medical Image Research Center, University Hospital Leuven Leuven, Belgium
| | - Uwe Himmelreich
- Faculty of Medicine, Molecular Small Animal Imaging Center, Katholieke Universiteit LeuvenLeuven, Belgium; Biomedical MRI Unit, Department of Imaging and Pathology, Faculty of Medicine, Katholieke Universiteit LeuvenLeuven, Belgium
| | - Rudi D'Hooge
- Laboratory of Biological Psychology, Faculty of Psychology and Educational Sciences, Katholieke Universiteit Leuven Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, Faculty of Science, Katholieke Universiteit Leuven Leuven, Belgium
| | - Mohammed A Benotmane
- Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK•CEN) Mol, Belgium
| |
Collapse
|
22
|
Buratovic S, Stenerlöw B, Fredriksson A, Sundell-Bergman S, Eriksson P. Developmental effects of fractionated low-dose exposure to gamma radiation on behaviour and susceptibility of the cholinergic system in mice. Int J Radiat Biol 2016; 92:371-9. [PMID: 27043364 DOI: 10.3109/09553002.2016.1164911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE To investigate whether neonatal exposure to fractionated external gamma radiation and co-exposure to radiation and nicotine can affect/exacerbate developmental neurotoxic effects, including altered behavior/cognitive function and the susceptibility of the cholinergic system in adult male mice. MATERIALS AND METHODS Neonatal male Naval Medical Research Institute (NMRI) mice were irradiated with one 200 mGy fraction/day and/or exposed to nicotine (66 μg/kg b.w.) twice daily on postnatal day (PND) 10, 10-11, 10-12 or 10-13 (nicotine only). At 2 months of age the animals were tested for spontaneous behavior in a novel home environment, habituation capacity and nicotine-induced behavior. RESULTS Fractionated irradiation and co-exposure to radiation and nicotine on three consecutive days disrupted behavior and habituation and altered susceptibility of the cholinergic system. All observed effects were significantly more pronounced in mice co-exposed to both radiation and nicotine. CONCLUSIONS The fractionated irradiation regime affects behavior/cognitive function in a similar manner as has previously been observed for single-dose exposures. Neonatal co-exposure to radiation and nicotine, during a critical period of brain development in general and cholinergic system development in particular, enhance these behavioral defects suggesting that the cholinergic system can be a target system for this type of developmental neurotoxic effects.
Collapse
Affiliation(s)
- Sonja Buratovic
- a Department of Environmental Toxicology , Uppsala University , Uppsala, Sweden
| | - Bo Stenerlöw
- b Department of Immunology, Genetics and Pathology , Uppsala University , Uppsala, Sweden
| | - Anders Fredriksson
- a Department of Environmental Toxicology , Uppsala University , Uppsala, Sweden
| | - Synnöve Sundell-Bergman
- c Department of Soil and Environment , Swedish University of Agricultural Sciences , Uppsala , Sweden
| | - Per Eriksson
- a Department of Environmental Toxicology , Uppsala University , Uppsala, Sweden
| |
Collapse
|
23
|
Eriksson P, Buratovic S, Fredriksson A, Stenerlöw B, Sundell-Bergman S. Neonatal exposure to whole body ionizing radiation induces adult neurobehavioural defects: Critical period, dose--response effects and strain and sex comparison. Behav Brain Res 2016; 304:11-9. [PMID: 26876140 DOI: 10.1016/j.bbr.2016.02.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 02/03/2016] [Accepted: 02/07/2016] [Indexed: 02/06/2023]
Abstract
Development of the brain includes periods which can be critical for its normal maturation. The present study investigates specifically vulnerable peri-/postnatal periods in mice which are essential for understanding the etiology behind radiation induced neurotoxicity and functional defects, including evaluation of neurotoxicity between sexes or commonly used laboratory mouse strains following low/moderate doses of ionizing radiation (IR). Male Naval Medical Research Institute (NMRI) mice, whole body irradiated to a single 500 mGy IR dose, on postnatal day (PND) 3 or PND 10 showed an altered adult spontaneous behaviour and impaired habituation capacity, whereas irradiation on PND 19 did not have any impact on the studied variables. Both NMRI and C57bl/6 male and female mice showed an altered adult spontaneous behaviour and impaired habituation following a single whole body irradiation of 500 or 1000 mGy, but not after 20 or 100 mGy, on PND 10. The present study shows that exposure to low/moderate doses of IR during critical life stages might be involved in the induction of neurological/neurodegenerative disorder/disease. A specifically vulnerable period for radiation induced neurotoxicity seems to be around PND 3-10 in mice. Further studies are needed to investigate mechanisms involved in induction of developmental neurotoxicity following low-dose irradiation.
Collapse
Affiliation(s)
- Per Eriksson
- Department of Environmental Toxicology, Uppsala University, Norbyvägen 18A, SE-75236 Uppsala, Sweden.
| | - Sonja Buratovic
- Department of Environmental Toxicology, Uppsala University, Norbyvägen 18A, SE-75236 Uppsala, Sweden
| | - Anders Fredriksson
- Department of Environmental Toxicology, Uppsala University, Norbyvägen 18A, SE-75236 Uppsala, Sweden
| | - Bo Stenerlöw
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Synnöve Sundell-Bergman
- Department of Soil and Environment, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
24
|
Heredia L, Bellés M, LLovet MI, Domingo JL, Linares V. Behavioral effects in mice of postnatal exposure to low-doses of 137-cesium and bisphenol A. Toxicology 2016; 340:10-6. [DOI: 10.1016/j.tox.2015.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/02/2015] [Accepted: 12/17/2015] [Indexed: 12/23/2022]
|
25
|
Zakhvataev VE. Possible scenarios of the influence of low-dose ionizing radiation on neural functioning. Med Hypotheses 2015; 85:723-35. [PMID: 26526727 DOI: 10.1016/j.mehy.2015.10.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/05/2015] [Accepted: 10/20/2015] [Indexed: 12/30/2022]
Abstract
Possible scenarios of the influence of ionizing radiation on neural functioning and the CNS are suggested. We argue that the radiation-induced bystander mechanisms associated with Ca(2+) flows, reactive nitrogen and oxygen species, and cytokines might lead to modulation of certain neuronal signaling pathways. The considered scenarios of conjugation of the bystander signaling and the neuronal signaling might result in modulation of certain synaptic receptors, neurogenesis, neurotransmission, channel conductance, synaptic signaling, different forms of neural plasticity, memory formation and storage, and learning. On this basis, corresponding new possible strategies for treating neurodegenerative deceases and mental disorders are proposed. The mechanisms considered might also be associated with neuronal survival and relevant to the treatment for brain injuries. At the same time, these mechanisms might be associated with detrimental effects and might facilitate the development of some neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Vladimir E Zakhvataev
- Neuroinformatics Department, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; Laboratory of Biological Action of Low-Intensity Factors, Siberian Federal University, 79 Svobodny pr., 660041 Krasnoyarsk, Russia.
| |
Collapse
|
26
|
Kempf SJ, Moertl S, Sepe S, von Toerne C, Hauck SM, Atkinson MJ, Mastroberardino PG, Tapio S. Low-dose ionizing radiation rapidly affects mitochondrial and synaptic signaling pathways in murine hippocampus and cortex. J Proteome Res 2015; 14:2055-64. [PMID: 25807253 DOI: 10.1021/acs.jproteome.5b00114] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The increased use of radiation-based medical imaging methods such as computer tomography is a matter of concern due to potential radiation-induced adverse effects. Efficient protection against such detrimental effects has not been possible due to inadequate understanding of radiation-induced alterations in signaling pathways. The aim of this study was to elucidate the molecular mechanisms behind learning and memory deficits after acute low and moderate doses of ionizing radiation. Female C57BL/6J mice were irradiated on postnatal day 10 (PND10) with gamma doses of 0.1 or 0.5 Gy. This was followed by evaluation of the cellular proteome, pathway-focused transcriptome, and neurological development/disease-focused miRNAome of hippocampus and cortex 24 h postirradiation. Our analysis showed that signaling pathways related to mitochondrial and synaptic functions were changed by acute irradiation. This may lead to reduced mitochondrial function paralleled by enhanced number of dendritic spines and neurite outgrowth due to elevated long-term potentiation, triggered by increased phosphorylated CREB. This was predominately observed in the cortex at 0.1 and 0.5 Gy and in the hippocampus only at 0.5 Gy. Moreover, a radiation-induced increase in the expression of several neural miRNAs associated with synaptic plasticity was found. The early changes in signaling pathways related to memory formation may be associated with the acute neurocognitive side effects in patients after brain radiotherapy but might also contribute to late radiation-induced cognitive injury.
Collapse
Affiliation(s)
- Stefan J Kempf
- †Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Simone Moertl
- †Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Sara Sepe
- ‡Department of Genetics, Erasmus Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Christine von Toerne
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Stefanie M Hauck
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Michael J Atkinson
- †Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany.,∥Chair of Radiation Biology, Technical University Munich, Arcisstrasse 21, 80333 Munich, Germany
| | - Pier G Mastroberardino
- ‡Department of Genetics, Erasmus Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Soile Tapio
- †Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| |
Collapse
|
27
|
Heredia L, Bellés M, Llovet MI, Domingo JL, Linares V. Neurobehavioral effects of concurrent exposure to cesium-137 and paraquat during neonatal development in mice. Toxicology 2015; 329:73-9. [DOI: 10.1016/j.tox.2015.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/01/2015] [Accepted: 01/16/2015] [Indexed: 01/21/2023]
|