1
|
Kumar R, Kumari P, Kumar R. Central Nervous System Response Against Ionizing Radiation Exposure: Cellular, Biochemical, and Molecular Perspectives. Mol Neurobiol 2025; 62:7268-7295. [PMID: 39875779 DOI: 10.1007/s12035-025-04712-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Gamma radiation is known to induce several detrimental effects on the nervous system. The hippocampus region, specifically the dentate gyrus (DG) and subventricular zone (SVZ), have been identified as a radiation-sensitive neurogenic niche. Radiation alters the endogenous redox status of neural stem cells (NSCs) and other proliferative cells, especially in the hippocampus region, leading to oxidative stress, neuroinflammation, and cell death. Planned (i.e., radiotherapy of brain tumor patients) or unplanned radiation exposure (i.e., accidental radiation exposure) can induce nonspecific damage to neuronal tissues, resulting in chronic or acute radiation syndrome. Although anatomical alterations in the neuronal tissues have been reported at higher doses of gamma radiation, biochemical and molecular perturbations may be evident even at much lower radiation doses. They may manifest in the form of neuronal deficits and cognitive impairment. In the present review, several molecular events and signaling pathways, such as oxidative stress, neuroinflammation, apoptosis, cognition, neuroplasticity, and neurotoxicity induced in neuronal cells upon ionizing radiation exposure, are reviewed. Furthermore, brain-specific radioprotectors and mitigators that protect normal neuronal cells and tissues against ionizing radiation during radiotherapy of cancer patients or nuclear emergencies are also discussed.
Collapse
Affiliation(s)
- Ravi Kumar
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Pratibha Kumari
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Raj Kumar
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
2
|
Yadav P, Nasir F, Sivanandam TM. Neuroprotective effect of vitamin B 12 supplementation on cognitive functions and neuronal morphology at different time intervals after traumatic brain injury in male Swiss albino mice. Neurochem Int 2024; 180:105869. [PMID: 39332530 DOI: 10.1016/j.neuint.2024.105869] [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: 07/15/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Traumatic brain injury is a highly irreversible process that consists of primary as well as secondary injury which develops and progresses over months to years, leading to cognitive dysfunctions. Vitamin B12 received considerable interest due to its potential therapeutic properties. The pathways of vitamin B12 are closely related to neuronal survival but its effects on the pathophysiology of injury with respect to cognition is a relatively unexplored area of research. In this study, we investigated, the effect of vitamin B12 and its involvement in neuroprotection on TBI-induced pathophysiology in male Swiss albino mice. Our findings suggested that vitamin B12 supplementation improves TBI-mediated neurological impairments, spatial and recognition memory, and anxiety-like behavior. Furthermore, the oxidative stress was reduced by declined homocysteine level with vitamin B12 supplementation validating declined expression of astrocytes and TBI biomarkers. The studies on neuronal morphology revealed that vitamin B12 supplementation increases the dendritic arborization and density of mushroom and filopodia-shaped spines and further increases the expression of synaptic plasticity-related genes and proteins. Taken together, our findings reveal that, supplementation of vitamin B12 restored the TBI-induced downregulation of dendritic arborization, and spine density which ultimately increases synaptic plasticity, cell survival, and recovery of cognitive dysfunctions.
Collapse
Affiliation(s)
- Priyanka Yadav
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Farheen Nasir
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Thamil Mani Sivanandam
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
3
|
Raber J, Holden S, Kessler K, Glaeser B, McQuesten C, Chaudhari M, Stenzel F, Lenarczyk M, Leonard SW, Morré J, Choi J, Kronenberg A, Borg A, Kwok A, Stevens JF, Olsen C, Willey JS, Bobe G, Minnier J, Baker JE. Effects of photon irradiation in the presence and absence of hindlimb unloading on the behavioral performance and metabolic pathways in the plasma of Fischer rats. Front Physiol 2024; 14:1316186. [PMID: 38260101 PMCID: PMC10800373 DOI: 10.3389/fphys.2023.1316186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction: The space environment astronauts experience during space missions consists of multiple environmental challenges, including microgravity. In this study, we assessed the behavioral and cognitive performances of male Fisher rats 2 months after sham irradiation or total body irradiation with photons in the absence or presence of simulated microgravity. We analyzed the plasma collected 9 months after sham irradiation or total body irradiation for distinct alterations in metabolic pathways and to determine whether changes to metabolic measures were associated with specific behavioral and cognitive measures. Methods: A total of 344 male Fischer rats were irradiated with photons (6 MeV; 3, 8, or 10 Gy) in the absence or presence of simulated weightlessness achieved using hindlimb unloading (HU). To identify potential plasma biomarkers of photon radiation exposure or the HU condition for behavioral or cognitive performance, we performed regression analyses. Results: The behavioral effects of HU on activity levels in an open field, measures of anxiety in an elevated plus maze, and anhedonia in the M&M consumption test were more pronounced than those of photon irradiation. Phenylalanine, tyrosine, and tryptophan metabolism, and phenylalanine metabolism and biosynthesis showed very strong pathway changes, following photon irradiation and HU in animals irradiated with 3 Gy. Here, 29 out of 101 plasma metabolites were associated with 1 out of 13 behavioral measures. In the absence of HU, 22 metabolites were related to behavioral and cognitive measures. In HU animals that were sham-irradiated or irradiated with 8 Gy, one metabolite was related to behavioral and cognitive measures. In HU animals irradiated with 3 Gy, six metabolites were related to behavioral and cognitive measures. Discussion: These data suggest that it will be possible to develop stable plasma biomarkers of behavioral and cognitive performance, following environmental challenges like HU and radiation exposure.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Departments of Neurology, and Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health & Science University, Portland, OR, United States
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Sarah Holden
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Kat Kessler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Breanna Glaeser
- Neuroscience Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Chloe McQuesten
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Mitali Chaudhari
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Fiona Stenzel
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Marek Lenarczyk
- Radiation Biosciences Laboratory, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Scott Willem Leonard
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jeffrey Morré
- Mass Spectrometry Core, Oregon State University, Corvallis, OR, United States
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Amy Kronenberg
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Alexander Borg
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Andy Kwok
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jan Frederik Stevens
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Christopher Olsen
- Neuroscience Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jeffrey S. Willey
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- Department of Animal Sciences, Oregon State University, Corvallis, OR, United States
| | - Jessica Minnier
- Oregon Health & Science University-Portland State University School of Public Health, Knight Cancer Institute Biostatistics Shared Resource, The Knight Cardiovascular Institute, OR Health & Science University, Portland, OR, United States
| | - John E. Baker
- Neuroscience Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
4
|
Beeraka NM, Vikram PRH, Greeshma MV, Uthaiah CA, Huria T, Liu J, Kumar P, Nikolenko VN, Bulygin KV, Sinelnikov MY, Sukocheva O, Fan R. Recent Investigations on Neurotransmitters' Role in Acute White Matter Injury of Perinatal Glia and Pharmacotherapies-Glia Dynamics in Stem Cell Therapy. Mol Neurobiol 2022; 59:2009-2026. [PMID: 35041139 DOI: 10.1007/s12035-021-02700-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/10/2021] [Indexed: 02/05/2023]
Abstract
Periventricular leukomalacia (PVL) and cerebral palsy are two neurological disease conditions developed from the premyelinated white matter ischemic injury (WMI). The significant pathophysiology of these diseases is accompanied by the cognitive deficits due to the loss of function of glial cells and axons. White matter makes up 50% of the brain volume consisting of myelinated and non-myelinated axons, glia, blood vessels, optic nerves, and corpus callosum. Studies over the years have delineated the susceptibility of white matter towards ischemic injury especially during pregnancy (prenatal, perinatal) or immediately after child birth (postnatal). Impairment in membrane depolarization of neurons and glial cells by ischemia-invoked excitotoxicity is mediated through the overactivation of NMDA receptors or non-NMDA receptors by excessive glutamate influx, calcium, or ROS overload and has been some of the well-studied molecular mechanisms conducive to the injury of white matter. Expression of glutamate receptors (GluR) and transporters (GLT1, EACC1, and GST) has significant influence in glial and axonal-mediated injury of premyelinated white matter during PVL and cerebral palsy. Predominantly, the central premyelinated axons express extensive levels of functional NMDA GluR receptors to confer ischemic injury to premyelinated white matter which in turn invoke defects in neural plasticity. Several underlying molecular mechanisms are yet to be unraveled to delineate the complete pathophysiology of these prenatal neurological diseases for developing the novel therapeutic modalities to mitigate pathophysiology and premature mortality of newborn babies. In this review, we have substantially discussed the above multiple pathophysiological aspects of white matter injury along with glial dynamics, and the pharmacotherapies including recent insights into the application of MSCs as therapeutic modality in treating white matter injury.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - P R Hemanth Vikram
- Department of Pharmaceutical Chemistry, JSS Pharmacy College, Mysuru, Karnataka, India
| | - M V Greeshma
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
| | - Chinnappa A Uthaiah
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
| | - Tahani Huria
- Faculty of Medicine, Benghazi University, Benghazi, Libya
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, LE1 7RH, UK
| | - Junqi Liu
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China
| | - Pramod Kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER-Guwahati), SilaKatamur (Halugurisuk), Changsari, Kamrup, 781101, Assam, India
| | - Vladimir N Nikolenko
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
- Department of Normal and Topographic Anatomy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kirill V Bulygin
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Mikhail Y Sinelnikov
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
- Research Institute of Human Morphology, 3 Tsyurupy Street, Moscow, 117418, Russian Federation
| | - Olga Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia
| | - Ruitai Fan
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China.
| |
Collapse
|
5
|
High-frequency head impact causes chronic synaptic adaptation and long-term cognitive impairment in mice. Nat Commun 2021; 12:2613. [PMID: 33972519 PMCID: PMC8110563 DOI: 10.1038/s41467-021-22744-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 03/24/2021] [Indexed: 02/03/2023] Open
Abstract
Repeated head impact exposure can cause memory and behavioral impairments. Here, we report that exposure to non-damaging, but high frequency, head impacts can alter brain function in mice through synaptic adaptation. High frequency head impact mice develop chronic cognitive impairments in the absence of traditional brain trauma pathology, and transcriptomic profiling of mouse and human chronic traumatic encephalopathy brain reveal that synapses are strongly affected by head impact. Electrophysiological analysis shows that high frequency head impacts cause chronic modification of the AMPA/NMDA ratio in neurons that underlie the changes to cognition. To demonstrate that synaptic adaptation is caused by head impact-induced glutamate release, we pretreated mice with memantine prior to head impact. Memantine prevents the development of the key transcriptomic and electrophysiological signatures of high frequency head impact, and averts cognitive dysfunction. These data reveal synapses as a target of high frequency head impact in human and mouse brain, and that this physiological adaptation in response to head impact is sufficient to induce chronic cognitive impairment in mice.
Collapse
|
6
|
Gampierakis IA, Koutmani Y, Semitekolou M, Morianos I, Polissidis A, Katsouda A, Charalampopoulos I, Xanthou G, Gravanis A, Karalis KP. Hippocampal neural stem cells and microglia response to experimental inflammatory bowel disease (IBD). Mol Psychiatry 2021; 26:1248-1263. [PMID: 31969694 DOI: 10.1038/s41380-020-0651-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 12/20/2022]
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), is a disease associated with dysbiosis, resulting in compromised intestinal epithelial barrier and chronic mucosal inflammation. Patients with IBD present with increased incidence of psychiatric disorders and cognitive impairment. Hippocampus is a brain region where adult neurogenesis occurs with functional implications in mood control and cognition. Using a well-established model of experimental colitis based on the administration of dextran sodium sulfate (DSS) in the drinking water, we sought to characterize the short and long-term effects of colitis on neurogenesis and glia responses in the hippocampus. We show that acute DSS colitis enhanced neurogenesis but with deficits in cell cycle kinetics of proliferating progenitors in the hippocampus. Chronic DSS colitis was characterized by normal levels of neurogenesis but with deficits in the migration and integration of newborn neurons in the functional circuitry of the DG. Notably, we found that acute DSS colitis-induced enhanced infiltration of the hippocampus with macrophages and inflammatory myeloid cells from the periphery, along with elevated frequencies of inflammatory M1-like microglia and increased release of pro-inflammatory cytokines. In contrast, increased percentages of tissue-repairing M2-like microglia, along with elevated levels of the anti-inflammatory cytokine, IL-10 were observed in the hippocampus during chronic DSS colitis. These findings uncover key effects of acute and chronic experimental colitis on adult hippocampal neurogenesis and innate immune cell responses, highlighting the potential mechanisms underlying cognitive and mood dysfunction in patients with IBD.
Collapse
Affiliation(s)
- Ioannis-Alexandros Gampierakis
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Greece
| | - Yassemi Koutmani
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maria Semitekolou
- Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Ioannis Morianos
- Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Alexia Polissidis
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Antonia Katsouda
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- School of Pharmacy, University of Athens, Athens, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation of Research & Technology Hellas (FORTH), Heraklion, Greece
| | - Georgina Xanthou
- Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation of Research & Technology Hellas (FORTH), Heraklion, Greece
| | - Katia P Karalis
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
- Emulate, Inc., 27 Drydock Avenue, Boston, MA, 02210, USA.
- Endocrine Division, Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Institute for Fundamental Biomedical Research, Biomedical Science Research Centre "Alexander Fleming", Athens, Greece.
| |
Collapse
|
7
|
Martínez-Cué C, Rueda N. Cellular Senescence in Neurodegenerative Diseases. Front Cell Neurosci 2020; 14:16. [PMID: 32116562 PMCID: PMC7026683 DOI: 10.3389/fncel.2020.00016] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/21/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a homeostatic biological process characterized by a permanent state of cell cycle arrest that can contribute to the decline of the regenerative potential and function of tissues. The increased presence of senescent cells in different neurodegenerative diseases suggests the contribution of senescence in the pathophysiology of these disorders. Although several factors can induce senescence, DNA damage, oxidative stress, neuroinflammation, and altered proteostasis have been shown to play a role in its onset. Oxidative stress contributes to accelerated aging and cognitive dysfunction stages affecting neurogenesis, neuronal differentiation, connectivity, and survival. During later life stages, it is implicated in the progression of cognitive decline, synapse loss, and neuronal degeneration. Also, neuroinflammation exacerbates oxidative stress, synaptic dysfunction, and neuronal death through the harmful effects of pro-inflammatory cytokines on cell proliferation and maturation. Both oxidative stress and neuroinflammation can induce DNA damage and alterations in DNA repair that, in turn, can exacerbate them. Another important feature associated with senescence is altered proteostasis. Because of the disruption in the function and balance of the proteome, senescence can modify the proper synthesis, folding, quality control, and degradation rate of proteins producing, in some diseases, misfolded proteins or aggregation of abnormal proteins. There is an extensive body of literature that associates cellular senescence with several neurodegenerative disorders including Alzheimer’s disease (AD), Down syndrome (DS), and Parkinson’s disease (PD). This review summarizes the evidence of the shared neuropathological events in these neurodegenerative diseases and the implication of cellular senescence in their onset or aggravation. Understanding the role that cellular senescence plays in them could help to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| |
Collapse
|
8
|
Cucinotta FA, Cacao E. Risks of cognitive detriments after low dose heavy ion and proton exposures. Int J Radiat Biol 2019; 95:985-998. [PMID: 31120359 PMCID: PMC6606350 DOI: 10.1080/09553002.2019.1623427] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/16/2019] [Accepted: 04/25/2019] [Indexed: 12/15/2022]
Abstract
Purpose: Heavy ion and proton brain irradiations occur during space travel and in Hadron therapy for cancer. Heavy ions produce distinct patterns of energy deposition in neuron cells and brain tissues compared to X-rays leading to large uncertainties in risk estimates. We make a critical review of findings from research studies over the last 25 years for understanding risks at low dose. Conclusions: A large number of mouse and rat cognitive testing measures have been reported for a variety of particle species and energies for acute doses. However, tissue reactions occur above dose thresholds and very few studies were performed at the heavy ion doses to be encountered on space missions (<0.04 Gy/y) or considered dose-rate effects, such that threshold doses are not known in rodent models. Investigations of possible mechanisms for cognitive changes have been limited by experimental design with largely group specific and not subject specific findings reported. Persistent oxidative stress and activated microglia cells are common mechanisms studied, while impairment of neurogenesis, detriments in neuron morphology, and changes to gene and protein expression were each found to be important in specific studies. Future research should focus on estimating threshold doses carried out with experimental designs aimed at understating causative mechanisms, which will be essential for extrapolating rodent findings to humans and chronic radiation scenarios, while establishing if mitigation are needed.
Collapse
|
9
|
Jiang P, Chen L, Sun J, Li J, Xu J, Liu W, Feng F, Qu W. Chotosan ameliorates cognitive impairment and hippocampus neuronal loss in experimental vascular dementia via activating the Nrf2-mediated antioxidant pathway. J Pharmacol Sci 2018; 139:105-111. [PMID: 30642751 DOI: 10.1016/j.jphs.2018.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/14/2018] [Accepted: 12/04/2018] [Indexed: 10/27/2022] Open
Abstract
Recent studies suggested that Chotosan has ameliorative effects on vascular dementia through antioxidative pathways. Nevertheless, no systematic pharmacological research was conducted to evaluate the contribution of nuclear factor-E2-related factor 2 (Nrf2), a crucial regulator of antioxidative system, on Chotosan-induced neuroprotection invascular dementia. The present study aimed to investigate the neuroprotective effect of Chotosan on vascular dementia and reveal the possible molecular mechanism involving Nrf2. We found that Chotosan treatment could ameliorate memory impairment and reduce neuron cell loss induced by common carotid artery occlusion surgery. Furthermore, Chotosan could significantly reverse reactive oxygen species production, neuronal apoptosis and microglia over-activation in hippocampus. In addition, Chotosan enhanced Nrf2 expression and its nuclear translocation as well as its downstream antioxidant protein expression, NAD(P)H/quinone oxidoreductase 1 and heme oxygenase-1. These findings suggest that Chotosan exert neuroprotection in an animal model of vascular dementia via activating Nrf2-mediated antioxidant pathway. Chotosan may serve as a potential candidate and promising Nrf2 activator for treating vascular dementia.
Collapse
Affiliation(s)
- Pan Jiang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Lei Chen
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Jing Sun
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Jingsong Li
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Jian Xu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China; Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing, 211198, People's Republic of China; Jiangsu Food and Pharmaceutical Science College, Huaian 223003, People's Republic of China
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China; Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
10
|
Central Nervous System Responses to Simulated Galactic Cosmic Rays. Int J Mol Sci 2018; 19:ijms19113669. [PMID: 30463349 PMCID: PMC6275046 DOI: 10.3390/ijms19113669] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 12/20/2022] Open
Abstract
In preparation for lunar and Mars missions it is essential to consider the challenges to human health that are posed by long-duration deep space habitation via multiple stressors, including ionizing radiation, gravitational changes during flight and in orbit, other aspects of the space environment such as high level of carbon dioxide, and psychological stress from confined environment and social isolation. It remains unclear how these stressors individually or in combination impact the central nervous system (CNS), presenting potential obstacles for astronauts engaged in deep space travel. Although human spaceflight research only within the last decade has started to include the effects of radiation transmitted by galactic cosmic rays to the CNS, radiation is currently considered to be one of the main stressors for prolonged spaceflight and deep space exploration. Here we will review the current knowledge of CNS damage caused by simulated space radiation with an emphasis on neuronal and glial responses along with cognitive functions. Furthermore, we will present novel experimental approaches to integrate the knowledge into more comprehensive studies, including multiple stressors at once and potential translation to human functions. Finally, we will discuss the need for developing biomarkers as predictors for cognitive decline and therapeutic countermeasures to prevent CNS damage and the loss of cognitive abilities.
Collapse
|
11
|
Rueda N, Vidal V, García-Cerro S, Narcís JO, Llorens-Martín M, Corrales A, Lantigua S, Iglesias M, Merino J, Merino R, Martínez-Cué C. Anti-IL17 treatment ameliorates Down syndrome phenotypes in mice. Brain Behav Immun 2018; 73:235-251. [PMID: 29758264 DOI: 10.1016/j.bbi.2018.05.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/02/2018] [Accepted: 05/07/2018] [Indexed: 12/12/2022] Open
Abstract
Down syndrome (DS) is characterized by structural and functional anomalies that are present prenatally and that lead to intellectual disabilities. Later in life, the cognitive abilities of DS individuals progressively deteriorate due to the development of Alzheimer's disease (AD)-associated neuropathology (i.e., β-amyloid (Aβ) plaques, neurofibrillary tangles (NFTs), neurodegeneration, synaptic pathology, neuroinflammation and increased oxidative stress). Increasing evidence has shown that among these pathological processes, neuroinflammation plays a predominant role in AD etiopathology. In AD mouse models, increased neuroinflammation appears earlier than Aβ plaques and NFTs, and in DS and AD models, neuroinflammation exacerbates the levels of soluble and insoluble Aβ species, favoring neurodegeneration. The Ts65Dn (TS) mouse, the most commonly used murine model of DS, recapitulates many alterations present in both DS and AD individuals, including enhanced neuroinflammation. In this study, we observed an altered neuroinflammatory milieu in the hippocampus of the TS mouse model. Pro-inflammatory mediators that were elevated in the hippocampus of this model included pro-inflammatory cytokine IL17A, which has a fundamental role in mediating brain damage in neuroinflammatory processes. Here, we analyzed the ability of an anti-IL17A antibody to reduce the neuropathological alterations that are present in TS mice during early neurodevelopmental stages (i.e., hippocampal neurogenesis and hypocellularity) or that are aggravated in later-life stages (i.e., cognitive abilities, cholinergic neuronal loss and increased cellular senescence, APP expression, Aβ peptide expression and neuroinflammation). Administration of anti-IL17 for 5 months, starting at the age of 7 months, partially improved the cognitive abilities of the TS mice, reduced the expression of several pro-inflammatory cytokines and the density of activated microglia and normalized the APP and Aβ1-42 levels in the hippocampi of the TS mice. These results suggest that IL17-mediated neuroinflammation is involved in several AD phenotypes in TS mice and provide a new therapeutic target to reduce these pathological characteristics.
Collapse
Affiliation(s)
- Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Verónica Vidal
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Susana García-Cerro
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Josep Oriol Narcís
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - María Llorens-Martín
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa", CBMSO, CSICUAM, Madrid, Spain; Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
| | - Andrea Corrales
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Sara Lantigua
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Marcos Iglesias
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, USA
| | - Jesús Merino
- Department of Molecular Biology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Ramón Merino
- Institute of Biomedicine and Biotechnology of Cantabria, Consejo Superior de Investigaciones Científicas-University of Cantabria, Santander, Spain.
| | - Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain.
| |
Collapse
|
12
|
Alexander TC, Butcher H, Krager K, Kiffer F, Groves T, Wang J, Carter G, Allen AR. Behavioral Effects of Focal Irradiation in a Juvenile Murine Model. Radiat Res 2018; 189:605-617. [PMID: 29584587 DOI: 10.1667/rr14847.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Chemotherapy has been successfully used to reduce radiation dose and volume for most pediatric patients. However, because of the failure of chemotherapeutic agents to cross the blood-brain barrier and the lack of response of some brain tumors to these agents, radiation therapy is still used to treat many childhood cancers with CNS involvement. In this study, we investigated the radiation effects on cognition and dendritic structure in the hippocampus in juvenile male mice. Twenty-one-day-old male C57BL/6 mice were irradiated using the small animal radiation research platform (SARRP). Animals were exposed to either a 10 Gy single dose or 10 Gy × 2 fractionated doses of X-ray cranial radiation. Five weeks after irradiation, animals were tested for hippocampus-dependent cognitive performance in the Morris water maze. Significant impairment in spatial memory retention was observed in the probe trial after the first day of hidden-platform training (first probe trial) in animals that received either 10 Gy single-dose or 10 Gy × 2 fractionated doses. However, by day 5, mice that received a 10 Gy single dose showed spatial memory retention in the probe trials, whereas mice that received the 20 Gy fractionated doses remained impaired. During Y-maze testing, animals exposed to radiation were impaired; the irradiated mice were not able to distinguish among the three Y-maze arms and spent approximately the same amount of time in all three arms during the retention trial. Radiation significantly compromised the dendritic architecture and reduced spine density throughout the hippocampal trisynaptic network.
Collapse
Affiliation(s)
- Tyler C Alexander
- a Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205.,b Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Hannah Butcher
- a Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205.,b Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Kimberly Krager
- a Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205.,b Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Frederico Kiffer
- a Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205.,b Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Thomas Groves
- a Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205.,b Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205.,c Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Jing Wang
- a Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205.,b Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Gwendolyn Carter
- a Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205.,b Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Antiño R Allen
- a Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205.,b Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205.,c Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| |
Collapse
|
13
|
Tucker LB, Velosky AG, McCabe JT. Applications of the Morris water maze in translational traumatic brain injury research. Neurosci Biobehav Rev 2018; 88:187-200. [PMID: 29545166 DOI: 10.1016/j.neubiorev.2018.03.010] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/09/2018] [Accepted: 03/09/2018] [Indexed: 12/21/2022]
Abstract
Acquired traumatic brain injury (TBI) is frequently accompanied by persistent cognitive symptoms, including executive function disruptions and memory deficits. The Morris Water Maze (MWM) is the most widely-employed laboratory behavioral test for assessing cognitive deficits in rodents after experimental TBI. Numerous protocols exist for performing the test, which has shown great robustness in detecting learning and memory deficits in rodents after infliction of TBI. We review applications of the MWM for the study of cognitive deficits following TBI in pre-clinical studies, describing multiple ways in which the test can be employed to examine specific aspects of learning and memory. Emphasis is placed on dependent measures that are available and important controls that must be considered in the context of TBI. Finally, caution is given regarding interpretation of deficits as being indicative of dysfunction of a single brain region (hippocampus), as experimental models of TBI most often result in more diffuse damage that disrupts multiple neural pathways and larger functional networks that participate in complex behaviors required in MWM performance.
Collapse
Affiliation(s)
- Laura B Tucker
- Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA; Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301, Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Alexander G Velosky
- Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Joseph T McCabe
- Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA; Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301, Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
14
|
Montelukast ameliorates streptozotocin-induced cognitive impairment and neurotoxicity in mice. Neurotoxicology 2016; 57:214-222. [DOI: 10.1016/j.neuro.2016.09.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 08/24/2016] [Accepted: 09/30/2016] [Indexed: 12/22/2022]
|
15
|
Sweet TB, Hurley SD, Wu MD, Olschowka JA, Williams JP, O'Banion MK. Neurogenic Effects of Low-Dose Whole-Body HZE (Fe) Ion and Gamma Irradiation. Radiat Res 2016; 186:614-623. [PMID: 27905869 DOI: 10.1667/rr14530.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Understanding the dose-toxicity profile of radiation is critical when evaluating potential health risks associated with natural and man-made sources in our environment. The purpose of this study was to evaluate the effects of low-dose whole-body high-energy charged (HZE) iron (Fe) ions and low-energy gamma exposure on proliferation and differentiation of adult-born neurons within the dentate gyrus of the hippocampus, cells deemed to play a critical role in memory regulation. To determine the dose-response characteristics of the brain to whole-body Fe-ion vs. gamma-radiation exposure, C57BL/6J mice were irradiated with 1 GeV/n Fe ions or a static 137Cs source (0.662 MeV) at doses ranging from 0 to 300 cGy. The neurogenesis was analyzed at 48 h and one month postirradiation. These experiments revealed that whole-body exposure to either Fe ions or gamma radiation leads to: 1. An acute decrease in cell division within the dentate gyrus of the hippocampus, detected at doses as low as 30 and 100 cGy for Fe ions and gamma radiation, respectively; and 2. A reduction in newly differentiated neurons (DCX immunoreactivity) at one month postirradiation, with significant decreases detected at doses as low as 100 cGy for both Fe ions and gamma rays. The data presented here contribute to our understanding of brain responses to whole-body Fe ions and gamma rays and may help inform health-risk evaluations related to systemic exposure during a medical or radiologic/nuclear event or as a result of prolonged space travel.
Collapse
Affiliation(s)
- Tara B Sweet
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Sean D Hurley
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Michael D Wu
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - John A Olschowka
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Jacqueline P Williams
- b Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642.,c Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - M Kerry O'Banion
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642.,d Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
16
|
Giannakopoulou A, Lyras GA, Grigoriadis N. Long-term effects of autoimmune CNS inflammation on adult hippocampal neurogenesis. J Neurosci Res 2016; 95:1446-1458. [PMID: 27781303 DOI: 10.1002/jnr.23982] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 10/09/2016] [Accepted: 10/10/2016] [Indexed: 01/03/2023]
Abstract
Neurogenesis is a well-characterized phenomenon within the dentate gyrus (DG) of the adult hippocampus. Aging and chronic degenerative disorders have been shown to impair hippocampal neurogenesis, but the consequence of chronic inflammation remains controversial. In this study the chronic experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis was used to investigate the long-term effects of T cell-mediated central nervous system inflammation on hippocampal neurogenesis. 5-Bromodeoxyuridine (BrdU)-labeled subpopulations of hippocampal cells in EAE and control mice (coexpressing GFAP, doublecortin, NeuN, calretinin, and S100) were quantified at the recovery phase, 21 days after BrdU administration, to estimate alterations on the rate and differentiation pattern of the neurogenesis process. The core features of EAE mice DG are (i) elevated number of newborn (BrdU+) cells indicating vigorous proliferation, which in the long term subsided; (ii) enhanced migration of newborn cells into the granule cell layer; (iii) increased level of immature neuronal markers (including calretinin and doublecortin); (iv) trending decrease in the percentage of newborn mature neurons; and (v) augmented gliogenesis and differentiation of newborn neural precursor cells (NPCs) to mature astrocytes (BrdU+/S100+). Although the inflammatory environment in the brain of EAE mice enhances the proliferation of hippocampal NPCs, in the long term neurogenesis is progressively depleted, giving prominence to gliogenesis. The discrepancy between the high number of immature cells and the low number of mature newborn cells could be the result of a caused defect in the maturation pathway. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Aggeliki Giannakopoulou
- Laboratory of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George A Lyras
- Department of Historical Geology and Palaeontology, Faculty of Geology and Geoenvironment, University of Athens, Athens, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
17
|
Impey S, Jopson T, Pelz C, Tafessu A, Fareh F, Zuloaga D, Marzulla T, Riparip LK, Stewart B, Rosi S, Turker MS, Raber J. Short- and long-term effects of 56Fe irradiation on cognition and hippocampal DNA methylation and gene expression. BMC Genomics 2016; 17:825. [PMID: 27776477 PMCID: PMC5078898 DOI: 10.1186/s12864-016-3110-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/22/2016] [Indexed: 12/21/2022] Open
Abstract
Background Astronauts are exposed to 56Fe ions that may pose a significant health hazard during and following prolonged missions in deep space. We showed previously that object recognition requiring the hippocampus, a structure critical for cognitive function, is affected in 2-month-old mice irradiated with 56Fe ions. Here we examined object recognition in 6-month-old mice irradiated with 56Fe ions, a biological age more relevant to the typical ages of astronauts. Moreover, because the mechanisms mediating the detrimental effects of 56Fe ions on hippocampal function are unclear, we examined changes in hippocampal networks involved in synaptic plasticity and memory, gene expression, and epigenetic changes in cytosine methylation (5mC) and hydroxymethylation (5hmC) that could accompany changes in gene expression. We assessed the effects of whole body 56Fe ion irradiation at early (2 weeks) and late (20 weeks) time points on hippocampus-dependent memory and hippocampal network stability, and whether these effects are associated with epigenetic changes in hippocampal DNA methylation (both 5mC and 5hmC) and gene expression. Results At the two-week time point, object recognition and network stability were impaired following irradiation at the 0.1 and 0.4 Gy dose, but not following irradiation at the 0.2 Gy dose. No impairments in object recognition or network stability were seen at the 20-week time point at any irradiation dose used. Consistent with this pattern, the significance of pathways for gene categories for 5hmC was lower, though not eliminated, at the 20-week time point compared to the 2-week time point. Similarly, significant changes were observed for 5mC gene pathways at the 2-week time point, but no significant gene categories were observed at the 20-week time point. Only the 5hmC changes tracked with gene expression changes. Conclusions Dose- and time-dependent epigenomic remodeling in the hippocampus following 56Fe ion exposure correlates with behavioral changes. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3110-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Soren Impey
- Oregon Stem Cell Center and Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA. .,Department of Cell, Developmental Biology, and Cancer Biology, Oregon Health and Science University, Portland, OR, 97239, USA.
| | - Timothy Jopson
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, 94110, USA.,Departments of Physical Therapy Rehabilitation Science, University of California, San Francisco, San Francisco, CA, 94110, USA.,Neurological Surgery, University of California San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, 94110, USA
| | - Carl Pelz
- Oregon Stem Cell Center and Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Amanuel Tafessu
- Oregon Stem Cell Center and Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Fatema Fareh
- Oregon Stem Cell Center and Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Damian Zuloaga
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Tessa Marzulla
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Lara-Kirstie Riparip
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, 94110, USA.,Departments of Physical Therapy Rehabilitation Science, University of California, San Francisco, San Francisco, CA, 94110, USA.,Neurological Surgery, University of California San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, 94110, USA
| | - Blair Stewart
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Susanna Rosi
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, 94110, USA.,Departments of Physical Therapy Rehabilitation Science, University of California, San Francisco, San Francisco, CA, 94110, USA.,Neurological Surgery, University of California San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, 94110, USA
| | - Mitchell S Turker
- Oregon Institute of Occupational Health Sciences and Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, 97239, USA. .,Departments of Neurology and Radiation Medicine, Oregon Health and Science University, Portland, OR, 97239, USA. .,Division of Neuroscience ONPRC, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
18
|
Raber J, Allen AR, Sharma S, Allen B, Rosi S, Olsen RHJ, Davis MJ, Eiwaz M, Fike JR, Nelson GA. Effects of Proton and Combined Proton and 56Fe Radiation on the Hippocampus. Radiat Res 2015; 185:20-30. [DOI: 10.1667/rr14222.1] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
| | - Antiño R. Allen
- Brain and Spinal Injury Center, Department of Neurological Surgery,
| | - Sourabh Sharma
- Brain and Spinal Injury Center, Department of Neurological Surgery,
| | - Barrett Allen
- Brain and Spinal Injury Center, Department of Neurological Surgery,
| | - Susanna Rosi
- Brain and Spinal Injury Center, Department of Neurological Surgery,
| | | | | | | | - John R. Fike
- Brain and Spinal Injury Center, Department of Neurological Surgery,
| | - Gregory A. Nelson
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, California, 92350
| |
Collapse
|
19
|
Raber J, Marzulla T, Stewart B, Kronenberg A, Turker MS. 28Silicon Irradiation Impairs Contextual Fear Memory in B6D2F1 Mice. Radiat Res 2015; 183:708-12. [DOI: 10.1667/rr13951.1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
20
|
miR21 is Associated with the Cognitive Improvement Following Voluntary Running Wheel Exercise in TBI Mice. J Mol Neurosci 2015; 57:114-22. [DOI: 10.1007/s12031-015-0584-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 05/18/2015] [Indexed: 11/27/2022]
|
21
|
Miao W, Bao TH, Han JH, Yin M, Yan Y, Wang WW, Zhu YH. Voluntary exercise prior to traumatic brain injury alters miRNA expression in the injured mouse cerebral cortex. ACTA ACUST UNITED AC 2015; 48:433-9. [PMID: 25760028 PMCID: PMC4445667 DOI: 10.1590/1414-431x20144012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 12/05/2014] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) may be important mediators of the profound molecular and cellular
changes that occur after traumatic brain injury (TBI). However, the changes and
possible roles of miRNAs induced by voluntary exercise prior to TBI are still not
known. In this report, the microarray method was used to demonstrate alterations in
miRNA expression levels in the cerebral cortex of TBI mice that were pretrained on a
running wheel (RW). Voluntary RW exercise prior to TBI: i) significantly decreased
the mortality rate and improved the recovery of the righting reflex in TBI mice, and
ii) differentially changed the levels of several miRNAs, upregulating some and
downregulating others. Furthermore, we revealed global upregulation of miR-21,
miR-92a, and miR-874 and downregulation of miR-138, let-7c, and miR-124 expression
among the sham-non-runner, TBI-non-runner, and TBI-runner groups. Quantitative
reverse transcription polymerase chain reaction data (RT-qPCR) indicated good
consistency with the microarray results. Our microarray-based analysis of miRNA
expression in mice cerebral cortex after TBI revealed that some miRNAs such as
miR-21, miR-92a, miR-874, miR-138, let-7c, and miR-124 could be involved in the
prevention and protection afforded by voluntary exercise in a TBI model.
Collapse
Affiliation(s)
- W Miao
- Department of Neurology, Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - T H Bao
- Department of Neurology, Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - J H Han
- Department of Neurology, Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - M Yin
- Department of Neurology, Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Y Yan
- Department of Neurology, Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - W W Wang
- Department of Cardiology, Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Y H Zhu
- Department of Neurology, Second Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
22
|
Allen AR, Eilertson K, Sharma S, Baure J, Allen B, Leu D, Rosi S, Raber J, Huang TT, Fike JR. Delayed administration of alpha-difluoromethylornithine prevents hippocampus-dependent cognitive impairment after single and combined injury in mice. Radiat Res 2014; 182:489-98. [PMID: 25375198 PMCID: PMC4282164 DOI: 10.1667/rr13753.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Radiation exposure due to radiological terrorism and military circumstances are a continuing threat for the civilian population. In an uncontrolled radiation event, it is likely that there will be other types of injury involved, including trauma. While radiation combined injury is recognized as an area of great significance, overall there is a paucity of information regarding the mechanisms underlying the interactions between irradiation and other forms of injury, or what countermeasures might be effective in ameliorating such changes. The objective of this study was to determine if difluoromethylornithine (DFMO) could reduce the adverse effects of single or combined injury if administered beginning 24 h after exposure. Eight-week-old C57BL/J6 young-adult male mice received whole-body cesium-137 ((137)Cs) irradiation with 4 Gy. Immediately after irradiation, unilateral traumatic brain injury was induced using a controlled cortical impact system. Forty-four days postirradiation, animals were tested for hippocampus-dependent cognitive performance in the Morris water maze. After cognitive testing, animals were euthanized and their brains snap frozen for immunohistochemical assessment of neuroinflammation (activated microglia) and neurogenesis in the hippocampal dentate gyrus. Our data show that single and combined injuries induced variable degrees of hippocampus-dependent cognitive dysfunction, and when given 24 h post trauma, DFMO treatment ameliorated those effects. Cellular changes including neurogenesis and numbers of activated microglia were generally not associated with the cognitive changes. Further analyses also revealed that DFMO increased hippocampal protein levels of the antioxidants thioredoxin 1 and peroxiredoxin 3 compared to vehicle treated animals. While the mechanisms responsible for the improvement in cognition after DFMO treatment are not yet clear, these results constitute a basis for further development of DFMO as a countermeasure for ameliorating the of risks for cognitive dysfunction in individuals subjected to trauma and radiation combined injury.
Collapse
Affiliation(s)
- Antiño R. Allen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Kirsten Eilertson
- Bioinformatics Core, Gladstone Institutes San Francisco, California 94158
| | - Sourabh Sharma
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110
| | - Jennifer Baure
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110
| | - Barrett Allen
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110
| | - David Leu
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94304
| | - Susanna Rosi
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110
- Physical Therapy and Rehabilitation Science, University of California, San Francisco, California 94110
| | - Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, Oregon 97239
| | - Ting-Ting Huang
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94304
- Geriatric Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304
| | - John R. Fike
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110
- Department of Radiation Oncology, University of California, San Francisco, California 94110
| |
Collapse
|
23
|
Peripherally triggered and GSK-3β-driven brain inflammation differentially skew adult hippocampal neurogenesis, behavioral pattern separation and microglial activation in response to ibuprofen. Transl Psychiatry 2014; 4:e463. [PMID: 25313506 PMCID: PMC4350524 DOI: 10.1038/tp.2014.92] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/24/2014] [Accepted: 08/13/2014] [Indexed: 12/14/2022] Open
Abstract
Both familial and sporadic forms of Alzheimer disease (AD) present memory impairments. It has been proposed that these impairments are related to inflammation in relevant brain areas such as the hippocampus. Whether peripherally triggered and neuron-driven brain inflammation produce similar and equally reversible alterations is a matter of discussion. Here we studied the effects of ibuprofen administration on a familial AD mouse model overexpressing GSK-3β that presents severe brain inflammation. We compared these effects with those observed in a peripherally triggered brain inflammation model based on chronic lipopolysaccharide (LPS) administration. Both proinflammatory stimuli produced equivalent reversible morphological alterations in granule neurons; however, GSK-3β had a much more prominent role in newborn neuron connectivity, causing alterations that were not reversed by ibuprofen. Although both insults triggered similar behavioral impairments, ibuprofen rescued this defect in LPS-treated mice but did not produce any improvement in GSK-3β-overexpressing animals. This observation could be attributable to the different microglial phenotype induced by ibuprofen treatment. These data may be clinically relevant for AD therapies, as GSK-3β appears to determine the efficacy of ibuprofen treatment.
Collapse
|
24
|
DeCarolis NA, Rivera PD, Ahn F, Amaral WZ, LeBlanc JA, Malhotra S, Shih HY, Petrik D, Melvin N, Chen BP, Eisch AJ. 56Fe Particle Exposure Results in a Long-Lasting Increase in a Cellular Index of Genomic Instability and Transiently Suppresses Adult Hippocampal Neurogenesis in Vivo. LIFE SCIENCES IN SPACE RESEARCH 2014; 2:70-79. [PMID: 25170435 PMCID: PMC4142527 DOI: 10.1016/j.lssr.2014.06.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The high-LET HZE particles from galactic cosmic radiation pose tremendous health risks to astronauts, as they may incur sub-threshold brain injury or maladaptations that may lead to cognitive impairment. The health effects of HZE particles are difficult to predict and unfeasible to prevent. This underscores the importance of estimating radiation risks to the central nervous system as a whole as well as to specific brain regions like the hippocampus, which is central to learning and memory. Given that neurogenesis in the hippocampus has been linked to learning and memory, we investigated the response and recovery of neurogenesis and neural stem cells in the adult mouse hippocampal dentate gyrus after HZE particle exposure using two nestin transgenic reporter mouse lines to label and track radial glia stem cells (Nestin-GFP and Nestin-CreERT2/R26R:YFP mice, respectively). Mice were subjected to 56Fe particle exposure (0 or 1 Gy, at either 300 or 1000 MeV/n) and brains were harvested at early (24h), intermediate (7d), and/or long time points (2-3mo) post-irradiation. 56Fe particle exposure resulted in a robust increase in 53BP1+ foci at both the intermediate and long time points post-irradiation, suggesting long-term genomic instability in the brain. However, 56Fe particle exposure only produced a transient decrease in immature neuron number at the intermediate time point, with no significant decrease at the long time point post-irradiation. 56Fe particle exposure similarly produced a transient decrease in dividing progenitors, with fewer progenitors labeled at the early time point but equal number labeled at the intermediate time point, suggesting a recovery of neurogenesis. Notably, 56Fe particle exposure did not change the total number of nestin-expressing neural stem cells. These results highlight that despite the persistence of an index of genomic instability, 56Fe particle-induced deficits in adult hippocampal neurogenesis may be transient. These data support the regenerative capacity of the adult SGZ after HZE particle exposure and encourage additional inquiry into the relationship between radial glia stem cells and cognitive function after HZE particle exposure.
Collapse
Affiliation(s)
| | | | - Francisca Ahn
- Dept Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Junie A. LeBlanc
- Dept Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Shveta Malhotra
- Dept Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Hung-Ying Shih
- Dept Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - David Petrik
- Dept Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Neal Melvin
- Dept Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Benjamin P.C. Chen
- Dept Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
- Co-Corresponding Authors: Amelia J. Eisch, Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390-9070. . Benjamin P. C. Chen, Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390-9187.
| | - Amelia J. Eisch
- Dept Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
- Co-Corresponding Authors: Amelia J. Eisch, Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390-9070. . Benjamin P. C. Chen, Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390-9187.
| |
Collapse
|
25
|
Spontaneous Running Wheel Improves Cognitive Functions of Mouse Associated with miRNA Expressional Alteration in Hippocampus Following Traumatic Brain Injury. J Mol Neurosci 2014; 54:622-9. [DOI: 10.1007/s12031-014-0344-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 05/27/2014] [Indexed: 01/22/2023]
|
26
|
Gu YL, Zhang LW, Ma N, Ye LL, Wang DX, Gao X. Cognitive improvement of mice induced by exercise prior to traumatic brain injury is associated with cytochrome c oxidase. Neurosci Lett 2014; 570:86-91. [PMID: 24746931 DOI: 10.1016/j.neulet.2014.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/04/2014] [Accepted: 04/05/2014] [Indexed: 12/01/2022]
Abstract
Though the evidence demonstrated that voluntary exercise programs could be implemented to enhance recovery of cognitive function induced by traumatic brain injury (TBI), the exact mechanisms were still not known. We proposed that the cognitive improvement induced by exercise in TBI mice is associated with cytochrome c oxidase (COX). To demonstrate this hypothesis, adult mice were housed with or without access to a running wheel (RW) for three weeks followed by TBI operation. Acquisition of spatial learning and memory retention was assessed by using the Morris Water Maze (MWM) on days 15 post TBI. The synaptic density was measured by Golji staining. Immunohistochemistry (IHC) for NeuN, GFAP and growth associated protein 43 (GAP43) were also performed. Using Western blot, the expressions of COX I, II, III, BDNF, synapsin I, synaptophysin (SYP) and GAP43 in hippocampus of TBI mice were determinated. Lastly, CcO activity and ATP amount were also detected. Results showed that voluntary exercise prior TBI: (i) counteracted the cognitive deficits and neuron and synaptic density loss associated with the injury; (ii) increased the levels of COX I, II, III, BDNF, synapsin I, SYP and GAP43; (iii) switched the mitochondrial CcO activity and ATP amounts. These studies demonstrated that the COX plays an important role in exercise's cognitive effects in TBI model and also provide evidence that RW training is a promise exercise for traumatically injured mice.
Collapse
Affiliation(s)
- Ying Li Gu
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Li Wei Zhang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ning Ma
- The Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Lin Lin Ye
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - De Xin Wang
- Department of Neurology, Beichen Hospital of Traditional Chinese Medicine, Tianjin 300400, China
| | - Xu Gao
- The Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
27
|
Allen AR, Eilertson K, Chakraborti A, Sharma S, Baure J, Habdank-Kolaczkowski J, Allen B, Rosi S, Raber J, Fike JR. Radiation exposure prior to traumatic brain injury induces responses that differ as a function of animal age. Int J Radiat Biol 2014; 90:214-23. [PMID: 24164494 PMCID: PMC3971762 DOI: 10.3109/09553002.2014.859761] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Purpose: Uncontrolled radiation exposure due to radiological terrorism, industrial accidents or military circumstances is a continuing threat for the civilian population. Age plays a major role in the susceptibility to radiation; younger children are at higher risk of developing cognitive deterioration when compared to adults. Our objective was to determine if an exposure to radiation affected the vulnerability of the juvenile hippocampus to a subsequent moderate traumatic injury. Materials and methods: Three-week-old (juvenile) and eight-week-old young adult C57BL/J6 male mice received whole body cesium-137 (137Cs) irradiation with 4 gray (Gy). One month later, unilateral traumatic brain injury was induced using a controlled cortical impact system. Two months post-irradiation, animals were tested for hippocampus-dependent cognitive performance in the Morris water-maze. After cognitive testing, animals were euthanized and their brains frozen for immunohistochemical assessment of activated microglia and neurogenesis in the hippocampal dentate gyrus. Results: All animals were able to learn the water maze task; however, treatment effects were seen when spatial memory retention was assessed. Animals that received irradiation as juveniles followed by a moderate traumatic brain injury one month later did not show spatial memory retention, i.e., were cognitively impaired. In contrast, all groups of animals that were treated as adults showed spatial memory retention in the probe trials. Conclusion: Although the mechanisms involved are not clear, our results suggest that irradiation enhanced a young animal's vulnerability to develop cognitive injury following a subsequent traumatic injury.
Collapse
Affiliation(s)
- Antiño R Allen
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California , San Francisco, CA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
There are more than 3.17 million people coping with long-term disabilities due to traumatic brain injury (TBI) in the United States. The majority of TBI research is focused on developing acute neuroprotective treatments to prevent or minimize these long-term disabilities. Therefore, chronic TBI survivors represent a large, underserved population that could significantly benefit from a therapy that capitalizes on the endogenous recovery mechanisms occurring during the weeks to months following brain trauma. Previous studies have found that the hippocampus is highly vulnerable to brain injury, in both experimental models of TBI and during human TBI. Although often not directly mechanically injured by the head injury, in the weeks to months following TBI, the hippocampus undergoes atrophy and exhibits deficits in long-term potentiation (LTP), a persistent increase in synaptic strength that is considered to be a model of learning and memory. Decoding the chronic hippocampal LTP and cell signaling deficits after brain trauma will provide new insights into the molecular mechanisms of hippocampal-dependent learning impairments caused by TBI and facilitate the development of effective therapeutic strategies to improve hippocampal-dependent learning for chronic survivors of TBI.
Collapse
Affiliation(s)
- Coleen M Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
29
|
Rivera PD, Shih HY, Leblanc JA, Cole MG, Amaral WZ, Mukherjee S, Zhang S, Lucero MJ, Decarolis NA, Chen BPC, Eisch AJ. Acute and fractionated exposure to high-LET (56)Fe HZE-particle radiation both result in similar long-term deficits in adult hippocampal neurogenesis. Radiat Res 2013; 180:658-67. [PMID: 24320054 DOI: 10.1667/rr13480.1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astronauts on multi-year interplanetary missions will be exposed to a low, chronic dose of high-energy, high-charge particles. Studies in rodents show acute, nonfractionated exposure to these particles causes brain changes such as fewer adult-generated hippocampal neurons and stem cells that may be detrimental to cognition and mood regulation and thus compromise mission success. However, the influence of a low, chronic dose of these particles on neurogenesis and stem cells is unknown. To examine the influence of galactic cosmic radiation on neurogenesis, adult-generated stem and progenitor cells in Nestin-CreER(T2)/R26R-YFP transgenic mice were inducibly labeled to allow fate tracking. Mice were then sham exposed or given one acute 100 cGy (56)Fe-particle exposure or five fractionated 20 cGy (56)Fe-particle exposures. Adult-generated hippocampal neurons and stem cells were quantified 24 h or 3 months later. Both acute and fractionated exposure decreased the amount of proliferating cells and immature neurons relative to sham exposure. Unexpectedly, neither acute nor fractionated exposure decreased the number of adult neural stem cells relative to sham expsoure. Our findings show that single and fractionated exposures of (56)Fe-particle irradiation are similarly detrimental to adult-generated neurons. Implications for future missions and ground-based studies in space radiation are discussed.
Collapse
Affiliation(s)
- Phillip D Rivera
- a Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas 75390-9070; and
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
de Bartolomeis A, Sarappa C, Buonaguro EF, Marmo F, Eramo A, Tomasetti C, Iasevoli F. Different effects of the NMDA receptor antagonists ketamine, MK-801, and memantine on postsynaptic density transcripts and their topography: role of Homer signaling, and implications for novel antipsychotic and pro-cognitive targets in psychosis. Prog Neuropsychopharmacol Biol Psychiatry 2013; 46:1-12. [PMID: 23800465 DOI: 10.1016/j.pnpbp.2013.06.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/10/2013] [Accepted: 06/14/2013] [Indexed: 12/15/2022]
Abstract
Administration of NMDA receptor antagonists, such as ketamine and MK-801, may induce psychotic-like behaviors in preclinical models of schizophrenia. Ketamine has also been observed to exacerbate psychotic symptoms in schizophrenia patients. However, memantine, a non-competitive NMDA receptor antagonist approved for Alzheimer's disease and proposed for antipsychotic augmentation, may challenge this view. To date, the molecular mechanisms by which these NMDA receptor antagonists cause different neurochemical, behavioral, and clinical effects are still a matter of debate. Here, we investigated by molecular imaging whether these agents could differently modulate gene expression and topographical distribution of glutamatergic postsynaptic density (PSD) proteins. We focused on Homer1a/Homer1b/PSD-95 signaling network, which may be implicated in glutamate-dependent synaptic plasticity, as well as in psychosis pathophysiology and treatment. Ketamine (25 and 50mg/kg) and MK-801 (0.8mg/kg) significantly induced the transcripts of immediate-early genes (Arc, c-fos, and Homer1a) in cortical regions compared to vehicle, whereas they reduced Homer1b and PSD-95 expression in cortical and striatal regions. Differently, memantine (5mg/kg) did not increase Homer1a signal compared to vehicle, whereas it induced c-fos in the somatosensory and in the medial agranular cortices. Moreover, memantine did not affect Homer1b and PSD-95 expression. When compared to ketamine and MK-801, memantine significantly increased the expression of c-fos, Homer1b and PSD-95. Overall, ketamine and MK-801 prominently increased Homer1a/Homer1b expression ratio, whereas memantine elicited the opposite effect. These data may support the view that ketamine, MK-801 and memantine exert divergent effects on PSD transcripts, which may contribute to their partially different behavioral and clinical effects.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine "Federico II", Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
31
|
Belarbi K, Rosi S. Modulation of adult-born neurons in the inflamed hippocampus. Front Cell Neurosci 2013; 7:145. [PMID: 24046730 PMCID: PMC3764370 DOI: 10.3389/fncel.2013.00145] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/21/2013] [Indexed: 11/13/2022] Open
Abstract
Throughout life new neurons are continuously added to the hippocampal circuitry involved with spatial learning and memory. These new cells originate from neural precursors in the subgranular zone of the dentate gyrus, migrate into the granule cell layer, and integrate into neural networks encoding spatial and contextual information. This process can be influenced by several environmental and endogenous factors and is modified in different animal models of neurological disorders. Neuroinflammation, as defined by the presence of activated microglia, is a common key factor to the progression of neurological disorders. Analysis of the literature shows that microglial activation impacts not only the production, but also the migration and the recruitment of new neurons. The impact of microglia on adult-born neurons appears much more multifaceted than ever envisioned before, combining both supportive and detrimental effects that are dependent upon the activation phenotype and the factors being released. The development of strategies aimed to change microglia toward states that promote functional neurogenesis could therefore offer novel therapeutic opportunities against neurological disorders associated with cognitive deficits and neuroinflammation. The present review summarizes the current knowledge on how production, distribution, and recruitment of new neurons into behaviorally relevant neural networks are modified in the inflamed hippocampus.
Collapse
Affiliation(s)
- Karim Belarbi
- Brain and Spinal Injury Center, San Francisco General Hospital, University of California at San Francisco San Francisco, CA, USA ; Department of Physical Therapy and Rehabilitation Science, University of California at San Francisco San Francisco, CA, USA
| | | |
Collapse
|
32
|
Allen AR, Eilertson K, Sharma S, Schneider D, Baure J, Allen B, Rosi S, Raber J, Fike JR. Effects of radiation combined injury on hippocampal function are modulated in mice deficient in chemokine receptor 2 (CCR2). Radiat Res 2013; 180:78-88. [PMID: 23772926 DOI: 10.1667/rr3344.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Chemokines and their receptors play a crucial role in normal brain function as well as in pathological conditions such as injury and disease-associated neuroinflammation. Chemokine receptor-2 (CCR2), which mediates the recruitment of infiltrating and resident microglia to sites of central nervous system (CNS) inflammation, is upregulated by ionizing irradiation and traumatic brain injury. Our objective was to determine if a deficiency in CCR2 and subsequent effects on brain microglia affect neurogenesis and cognitive function after radiation combined injury (RCI). CCR2 knock-out ⁻/⁻ and wild-type (WT) mice received 4 Gy of whole body ¹³⁷Cs irradiation. Immediately after irradiation, unilateral traumatic brain injury was induced using a controlled cortical impact system. Forty-four days postirradiation, animals were tested for hippocampus-dependent cognitive performance in the Morris water-maze. After cognitive testing, animals were euthanized and their brains snap frozen for immunohistochemical assessment of neuroinflammation (activated microglia) and neurogenesis in the hippocampal dentate gyrus. All animals were able to locate the visible and hidden platform locations in the water maze; however, treatment effects were seen when spatial memory retention was assessed in the probe trials (no platform). In WT animals that received combined injury, a significant impairment in spatial memory retention was observed in the probe trial after the first day of hidden platform training (first probe trial). This impairment was associated with increased neurogenesis in the ipsilateral hemisphere of the dentate gyrus. In contrast, CCR2⁻/⁻ mice, independent of insult showed significant memory retention in the first probe trial and there were no differences in the numbers of newly born neurons in the animals receiving irradiation, trauma or combined injury. Although the mechanisms involved are not clear, our data suggests that CCR2 deficiency can exert a protective effect preventing the impairment of cognitive function after combined injury.
Collapse
Affiliation(s)
- Antiño R Allen
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Raber J, Allen AR, Rosi S, Sharma S, Dayger C, Davis MJ, Fike JR. Effects of whole body 56Fe radiation on contextual freezing and Arc-positive cells in the dentate gyrus. Behav Brain Res 2013; 246:162-7. [DOI: 10.1016/j.bbr.2013.02.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 02/14/2013] [Accepted: 02/19/2013] [Indexed: 12/21/2022]
|
34
|
Effects of (56)Fe radiation on hippocampal function in mice deficient in chemokine receptor 2 (CCR2). Behav Brain Res 2013; 246:69-75. [PMID: 23500678 DOI: 10.1016/j.bbr.2013.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 02/26/2013] [Accepted: 03/02/2013] [Indexed: 12/19/2022]
Abstract
(56)Fe irradiation affects hippocampus-dependent cognition. The underlying mechanisms may involve alterations in neurogenesis, expression of the plasticity-related immediate early gene Arc, and inflammation. Chemokine receptor-2 (CCR2), which mediates the recruitment of infiltrating and resident microglia to sites of CNS inflammation, is upregulated by (56)Fe irradiation. CCR2 KO and wild-type mice were used to compare effects of (56)Fe radiation (600MeV, 0.25Gy) on hippocampal function using contextual fear conditioning involving tone shock pairing during training (+/+) and exposure to the same environment without tone shock pairings (-/-). In the -/- condition, irradiation enhanced habituation in WT mice, but not CCR2 KO mice, suggesting that a lack of CCR2 was associated with reduced cognitive performance. In the +/+ condition, irradiation reduced freezing but there was no genotype differences. There were no significant correlations between the number of Arc-positive cells in the dentate gyrus and freezing in either genotype. While measures of neurogenesis and gliogenesis appeared to be modulated by CCR2, there were no effects of genotype on the total numbers of newly born activated microglia before or after irradiation, indicating that other mechanisms are involved in the genotype-dependent radiation response.
Collapse
|
35
|
Belarbi K, Jopson T, Arellano C, Fike JR, Rosi S. CCR2 deficiency prevents neuronal dysfunction and cognitive impairments induced by cranial irradiation. Cancer Res 2012; 73:1201-10. [PMID: 23243025 DOI: 10.1158/0008-5472.can-12-2989] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Cranial irradiation can lead to long-lasting cognitive impairments in patients receiving radiotherapy for the treatment of malignant brain tumors. Recent studies have suggested inflammation as a major contributor to these deficits; we determined if the chemokine (C-C motif) receptor 2 (CCR2) was a mediator of cognitive impairments induced by irradiation. Two-month-old male Ccr2 knockout (-/-) and wild-type mice received 10 Gy cranial irradiation or sham-treatment. One month after irradiation, bromodeoxyuridine was injected intraperitoneally for seven consecutive days to label newly generated cells. At two months postirradiation, cognitive function was assessed by novel object recognition and Morris water maze. Our results show that CCR2 deficiency prevented hippocampus-dependent spatial learning and memory impairments induced by cranial irradiation. Hippocampal gene expression analysis showed that irradiation induced CCR2 ligands such as CCL8 and CCR2 deficiency reduced this induction. Irradiation reduced the number of adult-born neurons in both wild-type and Ccr2(-/-) mice, but the distribution pattern of the adult-born neurons through the granule cell layer was only altered in wild-type mice. Importantly, CCR2 deficiency normalized the fraction of pyramidal neurons expressing the plasticity-related immediate early gene Arc. These data offer new insight into the mechanism(s) of radiation-injury and suggest that CCR2 is a critical mediator of hippocampal neuronal dysfunction and hippocampal cognitive impairments after irradiation. Targeting CCR2 signaling could conceivably provide an effective approach to reduce or prevent the incidence and severity of this serious side effect of ionizing irradiation.
Collapse
Affiliation(s)
- Karim Belarbi
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA 94110, USA
| | | | | | | | | |
Collapse
|
36
|
Rosi S, Ferguson R, Fishman K, Allen A, Raber J, Fike JR. The polyamine inhibitor alpha-difluoromethylornithine modulates hippocampus-dependent function after single and combined injuries. PLoS One 2012; 7:e31094. [PMID: 22299052 PMCID: PMC3267765 DOI: 10.1371/journal.pone.0031094] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 01/02/2012] [Indexed: 12/31/2022] Open
Abstract
Exposure to uncontrolled irradiation in a radiologic terrorism scenario, a natural disaster or a nuclear battlefield, will likely be concomitantly superimposed on other types of injury, such as trauma. In the central nervous system, radiation combined injury (RCI) involving irradiation and traumatic brain injury may have a multifaceted character. This may entail cellular and molecular changes that are associated with cognitive performance, including changes in neurogenesis and the expression of the plasticity-related immediate early gene Arc. Because traumatic stimuli initiate a characteristic early increase in polyamine metabolism, we hypothesized that treatment with the polyamine inhibitor alpha-difluoromethylornithine (DFMO) would reduce the adverse effects of single or combined injury on hippocampus structure and function. Hippocampal dependent cognitive impairments were quantified with the Morris water maze and showed that DFMO effectively reversed cognitive impairments after all injuries, particularly traumatic brain injury. Similar results were seen with respect to the expression of Arc protein, but not neurogenesis. Given that polyamines have been found to modulate inflammatory responses in the brain we also assessed the numbers of total and newly born activated microglia, and found reduced numbers of newly born cells. While the mechanisms responsible for the improvement in cognition after DFMO treatment are not yet clear, the present study provides new and compelling data regarding the potential use of DFMO as a potential countermeasure against the adverse effects of single or combined injury.
Collapse
Affiliation(s)
- Susanna Rosi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Ryan Ferguson
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Kelly Fishman
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Antino Allen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, United States of America
- Department of Neurology, Oregon Health and Science University, Portland, Oregon, United States of America
- Division of Neuroscience, Oregon National Primate Research Center (ONPRC), Oregon Health and Science University, Portland, Oregon, United States of America
| | - John R. Fike
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
37
|
Belarbi K, Jopson T, Tweedie D, Arellano C, Luo W, Greig NH, Rosi S. TNF-α protein synthesis inhibitor restores neuronal function and reverses cognitive deficits induced by chronic neuroinflammation. J Neuroinflammation 2012; 9:23. [PMID: 22277195 PMCID: PMC3298520 DOI: 10.1186/1742-2094-9-23] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/25/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic neuroinflammation is a hallmark of several neurological disorders associated with cognitive loss. Activated microglia and secreted factors such as tumor necrosis factor (TNF)-α are key mediators of neuroinflammation and may contribute to neuronal dysfunction. Our study was aimed to evaluate the therapeutic potential of a novel analog of thalidomide, 3,6'-dithiothalidomide (DT), an agent with anti-TNF-α activity, in a model of chronic neuroinflammation. METHODS Lipopolysaccharide or artificial cerebrospinal fluid was infused into the fourth ventricle of three-month-old rats for 28 days. Starting on day 29, animals received daily intraperitoneal injections of DT (56 mg/kg/day) or vehicle for 14 days. Thereafter, cognitive function was assessed by novel object recognition, novel place recognition and Morris water maze, and animals were euthanized 25 min following water maze probe test evaluation. RESULTS Chronic LPS-infusion was characterized by increased gene expression of the proinflammatory cytokines TNF-α and IL-1β in the hippocampus. Treatment with DT normalized TNF-α levels back to control levels but not IL-1β. Treatment with DT attenuated the expression of TLR2, TLR4, IRAK1 and Hmgb1, all genes involved in the TLR-mediated signaling pathway associated with classical microglia activation. However DT did not impact the numbers of MHC Class II immunoreactive cells. Chronic neuroinflammation impaired novel place recognition, spatial learning and memory function; but it did not impact novel object recognition. Importantly, treatment with DT restored cognitive function in LPS-infused animals and normalized the fraction of hippocampal neurons expressing the plasticity-related immediate-early gene Arc. CONCLUSION Our data demonstrate that the TNF-α synthesis inhibitor DT can significantly reverse hippocampus-dependent cognitive deficits induced by chronic neuroinflammation. These results suggest that TNF-α is a critical mediator of chronic neuroinflammation-induced neuronal dysfunction and cognitive impairment and targeting its synthesis could provide an effective therapeutic approach to several human neurodegenerative diseases.
Collapse
Affiliation(s)
- Karim Belarbi
- Brain and Spinal Injury Center, University of California, San Francisco, California, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Belarbi K, Arellano C, Ferguson R, Jopson T, Rosi S. Chronic neuroinflammation impacts the recruitment of adult-born neurons into behaviorally relevant hippocampal networks. Brain Behav Immun 2012; 26:18-23. [PMID: 21787860 PMCID: PMC3221820 DOI: 10.1016/j.bbi.2011.07.225] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 06/23/2011] [Accepted: 07/07/2011] [Indexed: 01/05/2023] Open
Abstract
Growing evidence suggests that adult-born granule cells integrate into hippocampal networks and are required for proper cognitive function. Although neuroinflammation is involved in many disorders associated with cognitive impairment, it remains unknown whether it impacts the recruitment of adult-born neurons into behaviorally relevant hippocampal networks. Under similar behavioral conditions, exploration-induced expression of the immediate-early gene Arc in hippocampal cells has been linked to cellular activity observed by electrophysiological recording. By detecting exploration-induced Arc protein expression, we investigated whether neuroinflammation alters the recruitment of adult-born neurons into behaviorally relevant hippocampal networks. Neuroinflammation was induced in rats by intra-cerebroventricular infusion of lipopolysaccharide for 28 days. Animals received bromodeoxyuridine injections starting on day 29 (5 days) and were euthanized two months later. Persistent lipopolysaccharide-induced neuroinflammation was reliably detected by microglial activation in the hippocampus. Neuroinflammation did not impact the number of adult-born neurons but did alter their migration pattern through the granule cell layer. There was a positive correlation between the density of activated microglia and alterations in the fraction of existing granule neurons expressing Arc, suggesting that neuroinflammation induced a long-term disruption of hippocampal network activity. The proportion of adult-born neurons expressing behaviorally induced Arc was significantly lower in lipopolysaccharide-treated rats than in controls. This observation supports the fact that neuroinflammation significantly impacts adult-born neurons recruitment into hippocampal networks encoding spatial information.
Collapse
Affiliation(s)
- Karim Belarbi
- Brain and Spinal Injury Center, University of California, San Francisco, CA, USA,Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA
| | - Carla Arellano
- Brain and Spinal Injury Center, University of California, San Francisco, CA, USA,Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA
| | - Ryan Ferguson
- Brain and Spinal Injury Center, University of California, San Francisco, CA, USA,Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA
| | - Timothy Jopson
- Brain and Spinal Injury Center, University of California, San Francisco, CA, USA,Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA
| | - Susanna Rosi
- Brain and Spinal Injury Center, University of California, San Francisco, CA, USA,Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, CA, USA,Neurological Surgery, University of California, San Francisco, CA, USA
| |
Collapse
|
39
|
Raber J, Rosi S, Chakraborti A, Fishman K, Dayger C, Davis MJ, Villasana L, Fike JR. Effects of56Fe-Particle Cranial Radiation on Hippocampus-Dependent Cognition Depend on the Salience of the Environmental Stimuli. Radiat Res 2011; 176:521-6. [DOI: 10.1667/rr2635.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
40
|
Rosi S. Neuroinflammation and the plasticity-related immediate-early gene Arc. Brain Behav Immun 2011; 25 Suppl 1:S39-49. [PMID: 21320587 PMCID: PMC3098296 DOI: 10.1016/j.bbi.2011.02.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 02/07/2011] [Accepted: 02/07/2011] [Indexed: 12/01/2022] Open
Abstract
Neurons exist within a microenvironment that significantly influences their function and survival. While there are many environmental factors that can potentially impact neuronal function, activation of the innate immune system (microglia) is an important element common to many neurological and pathological conditions associated with memory loss. Learning and memory processes rely on the ability of neurons to alter their transcriptional programs in response to synaptic input. Recent advances in cell-based imaging of plasticity-related immediate-early gene (IEG) expression have provided a tool to investigate plasticity-related changes across multiple brain regions. The activity-regulated, cytoskeleton-associated IEG Arc is a regulator of protein synthesis-dependent forms of synaptic plasticity, which are essential for memory formation. Visualisation of Arc provides cellular level resolution for the mapping of neuronal networks. Chronic activation of the innate immune system alters Arc activity patterns, and this may be a mechanism by which it induces the cognitive dysfunction frequently associated with neuroinflammatory conditions. This review discusses the use of Arc expression during activation of the innate immune system as a valid marker of altered plasticity and a predictor of cognitive dysfunction.
Collapse
Affiliation(s)
- Susanna Rosi
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, CA 94110, USA.
| |
Collapse
|