1
|
Melara RD, Root JC, Edelman JA, Estelle MC, Mohr I, Ahles TA. Effects of Breast Cancer Treatment on Neural Noise: a Longitudinal Design. Arch Clin Neuropsychol 2025; 40:52-62. [PMID: 39197121 DOI: 10.1093/arclin/acae066] [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: 01/07/2024] [Revised: 07/08/2024] [Accepted: 08/09/2024] [Indexed: 08/30/2024] Open
Abstract
OBJECTIVE Cognitive dysfunction has been observed consistently in a subset of breast cancer survivors. Yet the precise neurophysiological origins of cancer-related cognitive decline remain unknown. The current study assessed neural noise (1/f activity in electroencephalogram [EEG]) in breast cancer survivors as a potential contributor to observed cognitive dysfunction from pre- to post-treatment. METHODS We measured EEG in a longitudinal design during performance of the paired-click task and the revised Attention Network Test (ANT-R) to investigate pre- versus post-treatment effects of neural noise in breast cancer patients (n = 20 in paired click; n = 19 in ANT-R) compared with healthy controls (n = 32 in paired click; n = 29 in ANT-R). RESULTS In both paradigms, one sensory (paired click) and one cognitive (ANT-R), we found that neural noise was significantly elevated after treatment in patients, remaining constant from pretest to posttest in controls. In the ANT-R, patients responded more slowly than controls on invalid cuing trials. Increased neural noise was associated with poorer alerting and poorer inhibitory control of attention (as measured by behavioral network scores), particularly for patients after treatment. CONCLUSIONS The current study is the first to show a deleterious effect of breast cancer and/or cancer treatment on neural noise, pointing to alterations in the relative balance of excitatory and inhibitory synaptic inputs, while also suggesting promising approaches for cognitive rehabilitation.
Collapse
Affiliation(s)
- Robert D Melara
- Department of Psychology, The City College, City University of New York, 160 Convent Avenue, NAC 7-120, New York, NY 10031, USA
| | - James C Root
- Memorial Sloan Kettering Cancer Center, Department of Psychiatry and Behavioral Science Services, 641 Lexington Avenue, 7th Floor, New York, New York 10022, USA
| | - Jay A Edelman
- Department of Biology, The City College, City University of New York, 160 Convent Avenue, MR 526, New York, NY 10031, USA
| | - Maria Camilla Estelle
- Memorial Sloan Kettering Cancer Center, Department of Psychiatry and Behavioral Science Services, 641 Lexington Avenue, 7th Floor, New York, New York 10022, USA
| | - Isabella Mohr
- Memorial Sloan Kettering Cancer Center, Department of Psychiatry and Behavioral Science Services, 641 Lexington Avenue, 7th Floor, New York, New York 10022, USA
| | - Tim A Ahles
- Memorial Sloan Kettering Cancer Center, Department of Psychiatry and Behavioral Science Services, 641 Lexington Avenue, 7th Floor, New York, New York 10022, USA
| |
Collapse
|
2
|
Crown LM, Featherstone RE, Sobell JL, Parekh K, Siegel SJ. The Use of Event-Related Potentials in the Study of Schizophrenia: An Overview. ADVANCES IN NEUROBIOLOGY 2024; 40:285-319. [PMID: 39562449 DOI: 10.1007/978-3-031-69491-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Event-related potentials (ERPs) are small voltage changes in the brain that reliably occur in response to auditory or visual stimuli. ERPs have been extensively studied in both humans and animals to identify biomarkers, test pharmacological agents, and generate testable hypotheses about the physiological and genetic basis of schizophrenia. In this chapter, we discuss how ERPs are generated and recorded as well as review canonical ERP components in the context of schizophrenia research in humans. We then discuss what is known about rodent homologs of these components and how they are altered in common pharmacologic and genetic manipulations used in preclinical schizophrenia research. This chapter will also explore the relationship of ERPs to leading hypotheses about the pathophysiology of schizophrenia. We conclude with an evaluation of both the utility and limitations of ERPs in schizophrenia research and offer recommendations of future directions that may be beneficial to the field.
Collapse
Affiliation(s)
- Lindsey M Crown
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert E Featherstone
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Janet L Sobell
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Krishna Parekh
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Steven J Siegel
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
3
|
The effect of doxorubicin or cyclophosphamide treatment on auditory brainstem response in mice. Exp Brain Res 2022; 240:2907-2921. [PMID: 36123538 DOI: 10.1007/s00221-022-06463-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 09/07/2022] [Indexed: 11/04/2022]
Abstract
Clinical studies suggest that chemotherapy is associated with long-term cognitive impairment in some patients. Several underlying mechanisms have been proposed; however, the etiology of chemotherapy-related cognitive dysfunction remains relatively unknown. There is evidence that oligodendrocytes and white matter tracts within the CNS may be particularly vulnerable to chemotherapy-related damage and dysfunction. Auditory brainstem responses (ABRs) have been used to detect and measure functional integrity of myelin in a variety of animal models of autoimmune disorders and demyelinating diseases. Limited evidence suggests that increases in interpeak latencies, associated with disrupted impulse conduction, can be detected in ABRs following 5-fluorouracil administration in mice. It is unknown if similar functional disruptions can be detected following treatment with other chemotherapeutic compounds and the extent to which alterations in ABR signals represent robust and long-lasting impairments associated with chemotherapy-related cognitive impairment. Thus, C57BL/6 J mice were treated every 3rd day for a total of 3 injections with low or high dose cyclophosphamide, or doxorubicin. ABRs of mice were assessed on days 1, 7, 14, 56 and 6 months following completion of chemotherapy administration. There were timing and amplitude differences in the ABRs of the doxorubicin and the high dose cyclophosphamide groups relative to the control animals. However, despite significant toxic effects as assessed by weight loss, the changes in the ABR were transient.
Collapse
|
4
|
Melara RD, Ahles TA, Prashad N, Fernbach M, Edelman JA, Root J. Longitudinal Effects of Breast Cancer Treatment on Neural Correlates of Attention. Arch Clin Neuropsychol 2022; 38:12-24. [PMID: 35901461 PMCID: PMC9868529 DOI: 10.1093/arclin/acac051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2022] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVE Cognitive dysfunction has been observed consistently in a subset of breast cancer survivors. Yet, the precise physiological and processing origins of dysfunction remain unknown. The current study examined the utility of methods and procedures based on cognitive neuroscience to study cognitive change associated with cancer and cancer treatment. METHODS We used electroencephalogram and behavioral measures in a longitudinal design to investigate pre- versus post-treatment effects on attention performance in breast cancer patients (n = 15) compared with healthy controls (n = 24), as participants completed the revised Attention Network Test, a cognitive measure of alerting, orienting, and inhibitory control of attention. RESULTS We found no group differences in behavioral performance from pretest to posttest, but significant event-related potential effects of cancer treatment in processing cue validity: After treatment, patients revealed decreased N1 amplitude and increased P3 amplitude, suggesting a suppressed early (N1) response and an exaggerated late (P3) response to invalid cues. CONCLUSIONS The results suggest that treatment-related attentional disruption begins in early sensory/perceptual processing and extends to compensatory top-down executive processes.
Collapse
Affiliation(s)
- Robert D Melara
- Corresponding author at: Department of Psychology, City College, City University of New York, 160 Convent Avenue, NAC 7-120, New York, NY 10031, USA. Tel.: +1-212-650-5716. E-mail address: (R. D. Melara)
| | - Tim A Ahles
- Department of Psychiatry and Behavioral Science Services, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Neelam Prashad
- Department of Psychiatry and Behavioral Science Services, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Madalyn Fernbach
- Department of Psychiatry and Behavioral Science Services, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jay A Edelman
- Department of Biology, City College, City University of New York, New York, New York, USA
| | - James Root
- Department of Psychiatry and Behavioral Science Services, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
5
|
Sirichoat A, Anosri T, Kaewngam S, Aranarochana A, Pannangrong W, Wigmore P, Welbat JU. Neuroprotective properties of chrysin on decreases of cell proliferation, immature neurons and neuronal cell survival in the hippocampal dentate gyrus associated with cognition induced by methotrexate. Neurotoxicology 2022; 92:15-24. [PMID: 35779630 DOI: 10.1016/j.neuro.2022.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/03/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Methotrexate (MTX) is a drug widely used for chemotherapy and can reduce cancer cell production by inhibiting dihydrofolate reductase and decreasing cancer cell growth. MTX has a neurotoxic effect on neural stem and glial cells, leading to memory deficits. Chrysin is a natural flavonoid that contains essential biological activities, such as neuroprotective and cognitive-improving properties. Therefore, the aim of the present study was to investigate the protective effect of chrysin against MTX-induced memory impairments related to hippocampal neurogenesis. Seventy-two male Sprague Dawley rats were divided into six groups: control, MTX, chrysin (10 and 30 mg/kg), and MTX+ chrysin (10 and 30 mg/kg) groups. Chrysin (10 and 30 mg/kg) was administered by oral gavage for 15 days. MTX (75 mg/kg) was administered by intravenous injection on days 8 and 15. Spatial and recognition memories were evaluated using the novel object location (NOL) and novel object recognition (NOR) tests, respectively. Moreover, cell proliferation, neuronal cell survival, and immature neurons in the subgranular zone of the hippocampal dentate gyrus were quantified by Ki-67, bromodeoxyuridine/neuronal nuclear protein (BrdU/NeuN), and doublecortin (DCX) immunohistochemistry staining. The results of the MTX group demonstrated that spatial and recognition memories were both impaired. Furthermore, cell division reduction, neuronal cell survival reduction, and immature neuron decreases were detected in the MTX group and not observed in the co-administration groups. Therefore, these results revealed that chrysin could alleviate memory and neurogenesis impairments in MTX-treated rats.
Collapse
Affiliation(s)
- Apiwat Sirichoat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Tanaporn Anosri
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Soraya Kaewngam
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Anusara Aranarochana
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Wanassanun Pannangrong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Peter Wigmore
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, United Kingdom
| | - Jariya Umka Welbat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Neurogenesis Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
6
|
Groves T, Corley C, Byrum SD, Allen AR. The Effects of 5-Fluorouracil/Leucovorin Chemotherapy on Cognitive Function in Male Mice. Front Mol Biosci 2021; 8:762116. [PMID: 34778377 PMCID: PMC8581634 DOI: 10.3389/fmolb.2021.762116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/04/2021] [Indexed: 12/14/2022] Open
Abstract
5-Fluorouracil (5-Fu) and leucovorin (LV) are often given in combination to treat colorectal cancer. 5-Fu/LV prevents cell proliferation by inhibiting thymidylate synthase, which catalyzes the conversion of deoxyuridine monophosphate to deoxythymidine monophosphate. While 5-Fu has been shown to cause cognitive impairment, the synergistic effect of 5-Fu with LV has not been fully explored. The present investigation was designed to assess how the combination of 5-Fu and LV affect cognition in a murine model. Six-month-old male mice were used in this study; 15 mice received saline injections and 15 mice received 5-Fu/LV injections. One month after treatment, the elevated plus maze, Y-maze, and Morris water maze behavioral tasks were performed. Brains were then extracted, cryosectioned, and stained for CD68 to assay microglial activation and with tomato lectin to assay the vasculature. All animals were able to locate the visible and hidden platform locations in the water maze. However, a 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 5-Fu/LV, but these animals showed spatial memory retention by day 5. There were no significant increases in inflammation as measured by CD68, but 5-Fu/LV treatment did modulate blood vessel morphology. Tandem mass tag proteomics analysis identified 6,049 proteins, 7 of which were differentially expressed with a p-value of <0.05 and a fold change of >1.5. The present data demonstrate that 5-Fu/LV increases anxiety and significantly impairs spatial memory retention.
Collapse
Affiliation(s)
- Thomas Groves
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Christa Corley
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| | - Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
7
|
Abstract
Survivors of breast and other cancers often report protracted difficulty in performing tasks involving concentration and memory, even years after the completion of treatment. The current study investigated whether cancer and treatment history is associated with deficits in sensory filtering (gating out) and sensory memory (gating in), early processes in stimulus processing that may contribute to difficulties in later remembering. A group of breast cancer survivors and age-matched healthy control participants (mean age 54 years) underwent testing with paired-click and oddball tasks while electroencephalographic (EEG) signals were recorded. The survivors showed relatively poor inhibition of redundant sensory stimulation (P50 suppression). Dipole source analysis localized the survivors' impairment to the hippocampus, with preservation of function in gating mechanisms of the frontal lobe and auditory cortex. Survivors also showed disruption to sensory memory processes needed to register novel information in an otherwise uniform auditory environment (mismatch negativity). The findings suggest that survivors experience deficits in early, automatic mechanisms of sensory gating, which may trigger a cascade of later perceived attentional and memory deficits. If our account is accurate, ideal therapies might aim to restore early inhibitory processes, such as those gauged by P50 suppression.
Collapse
Affiliation(s)
- Robert D. Melara
- Department of Psychology, City College, City University of New York, New York, NY, USA
| | - James C. Root
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan- Kettering Cancer Center, New York, NY, USA
| | - Raquel Bibi
- Department of Psychology, City College, City University of New York, New York, NY, USA
| | - Tim A. Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan- Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
8
|
Gibson EM, Monje M. Microglia in Cancer Therapy-Related Cognitive Impairment. Trends Neurosci 2021; 44:441-451. [PMID: 33674135 PMCID: PMC8593823 DOI: 10.1016/j.tins.2021.02.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/20/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022]
Abstract
Millions of cancer survivors experience a persistent neurological syndrome that includes deficits in memory, attention, information processing, and mental health. Cancer therapy-related cognitive impairment can cause mild to severe disruptions to quality of life for these cancer survivors. Understanding the cellular and molecular underpinnings of this disorder will facilitate new therapeutic strategies aimed at ameliorating these long-lasting impairments. Accumulating evidence suggests that a range of cancer therapies induce persistent activation of the brain's resident immune cells, microglia. Cancer therapy-induced microglial activation disrupts numerous mechanisms of neuroplasticity, and emerging findings suggest that this impairment in plasticity is central to cancer therapy-related cognitive impairment. This review explores reactive microglial dysregulation of neural circuit structure and function following cancer therapy.
Collapse
Affiliation(s)
- Erin M Gibson
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA 94305, USA.
| | - Michelle Monje
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA 94305, USA; Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA 94305, USA; Department of Pathology, Stanford University, Palo Alto, CA 94305, USA; Stanford California Department of Pediatrics, Stanford University, Palo Alto, CA 94305, USA.
| |
Collapse
|
9
|
John J, Kinra M, Mudgal J, Viswanatha GL, Nandakumar K. Animal models of chemotherapy-induced cognitive decline in preclinical drug development. Psychopharmacology (Berl) 2021; 238:3025-3053. [PMID: 34643772 PMCID: PMC8605973 DOI: 10.1007/s00213-021-05977-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 08/31/2021] [Indexed: 12/23/2022]
Abstract
RATIONALE Chemotherapy-induced cognitive impairment (CICI), chemobrain, and chemofog are the common terms for mental dysfunction in a cancer patient/survivor under the influence of chemotherapeutics. CICI is manifested as short/long term memory problems and delayed mental processing, which interferes with a person's day-to-day activities. Understanding CICI mechanisms help in developing therapeutic interventions that may alleviate the disease condition. Animal models facilitate critical evaluation to elucidate the underlying mechanisms and form an integral part of verifying different treatment hypotheses and strategies. OBJECTIVES A methodical evaluation of scientific literature is required to understand cognitive changes associated with the use of chemotherapeutic agents in different preclinical studies. This review mainly emphasizes animal models developed with various chemotherapeutic agents individually and in combination, with their proposed mechanisms contributing to the cognitive dysfunction. This review also points toward the analysis of chemobrain in healthy animals to understand the mechanism of interventions in absence of tumor and in tumor-bearing animals to mimic human cancer conditions to screen potential drug candidates against chemobrain. RESULTS Substantial memory deficit as a result of commonly used chemotherapeutic agents was evidenced in healthy and tumor-bearing animals. Spatial and episodic cognitive impairments, alterations in neurotrophins, oxidative and inflammatory markers, and changes in long-term potentiation were commonly observed changes in different animal models irrespective of the chemotherapeutic agent. CONCLUSION Dyscognition exists as one of the serious side effects of cancer chemotherapy. Due to differing mechanisms of chemotherapeutic agents with differing tendencies to alter behavioral and biochemical parameters, chemotherapy may present a significant risk in resulting memory impairments in healthy as well as tumor-bearing animals.
Collapse
Affiliation(s)
- Jeena John
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka India 576104
| | - Manas Kinra
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka India 576104
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka India 576104
| | - G. L. Viswanatha
- Independent Researcher, Kengeri, Bangalore, Karnataka India 560060
| | - K. Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka India 576104
| |
Collapse
|
10
|
Mounier NM, Abdel-Maged AES, Wahdan SA, Gad AM, Azab SS. Chemotherapy-induced cognitive impairment (CICI): An overview of etiology and pathogenesis. Life Sci 2020. [DOI: https://doi.org/10.1016/j.lfs.2020.118071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
11
|
Mounier NM, Abdel-Maged AES, Wahdan SA, Gad AM, Azab SS. Chemotherapy-induced cognitive impairment (CICI): An overview of etiology and pathogenesis. Life Sci 2020; 258:118071. [PMID: 32673664 DOI: 10.1016/j.lfs.2020.118071] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/26/2020] [Accepted: 07/06/2020] [Indexed: 02/08/2023]
Abstract
Many cancer patients treated with chemotherapy develop chemotherapy-induced cognitive impairment (CICI), often referred to as chemo-brain, which manifest during or post-treatment with variable degrees, onset and duration thereby affecting the patients' quality of life. Several chemotherapeutic agents have been studied to determine its possible association with cognitive impairment and to fully comprehend their contribution to CICI. A vast number of studies have emerged proposing several candidate underlying mechanisms and etiologies contributing to CICI such as direct neurotoxicity, BBB disruption, decreased hippocampal neurogenesis, white matter abnormalities, secondary neuro-inflammatory response and increased oxidative stress; however, the exact underlying mechanisms are still not well defined. This review summarizes CICI associated with most commonly used chemotherapeutic agents with emphasizes the possible underlying pathogenesis in both animal and clinical studies.
Collapse
Affiliation(s)
- Noha M Mounier
- National Organization for Drug Control and Research (NODCAR), Cairo, Egypt
| | | | - Sara A Wahdan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amany M Gad
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt
| | - Samar S Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
12
|
Gaynor AM, Pergolizzi D, Alici Y, Ryan E, McNeal K, Ahles TA, Root JC. Impact of transcranial direct current stimulation on sustained attention in breast cancer survivors: Evidence for feasibility, tolerability, and initial efficacy. Brain Stimul 2020; 13:1108-1116. [PMID: 32353419 DOI: 10.1016/j.brs.2020.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/26/2020] [Accepted: 04/19/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND A significant subset of breast cancer survivors experience cognitive difficulties in attention and memory, which persist for years following treatment. Transcranial direct current stimulation (tDCS) has been shown to be effective in improving working memory, attention, processing speed, and other cognitive functions in both healthy and clinical populations. To date, no studies have examined tDCS for rehabilitation of cancer-related cognitive dysfunction. OBJECTIVE/HYPOTHESIS We aimed to provide preliminary evidence for feasibility, tolerability, acceptability, and efficacy of tDCS in improving performance on a measure of sustained attention. METHODS In a within-subjects design, 16 breast cancer survivors underwent 2 consecutive days of active tDCS over the prefrontal cortex, and 2 days of sham tDCS, counterbalanced for order of stimulation condition, while performing a continuous performance test. RESULTS Stimulation was feasible and tolerable, with 89% of participants completing all sessions, and none reporting more than mild to moderate discomfort. Analyses of efficacy showed that during active stimulation, participants had significantly lower standard errors of reaction times overall, indicating better sustained attention ability, as compared to sham stimulation (p < 0.05). Furthermore, the effect of stimulation on standard errors of reaction times differed by inter-stimulus interval (ISI): for 1 and 2 s ISIs, there was no significant difference in performance between sham and active tDCS conditions, but for 4 s ISIs, stimulation improved variability in response times relative to sham (p < 0.05). CONCLUSIONS Results suggest that tDCS is feasible, tolerable, and may be an effective intervention to improve sustained attention difficulties in survivors with cancer-related cognitive dysfunction.
Collapse
Affiliation(s)
- Alexandra M Gaynor
- Memorial Sloan Kettering Cancer Center, Department of Psychiatry and Behavioral Sciences, New York, NY, USA.
| | - Denise Pergolizzi
- Universitat Internacional de Catalunya, School of Medicine and Health Sciences, Barcelona, Spain
| | - Yesne Alici
- Memorial Sloan Kettering Cancer Center, Department of Psychiatry and Behavioral Sciences, New York, NY, USA
| | - Elizabeth Ryan
- Memorial Sloan Kettering Cancer Center, Department of Psychiatry and Behavioral Sciences, New York, NY, USA
| | - Katrazyna McNeal
- Memorial Sloan Kettering Cancer Center, Department of Psychiatry and Behavioral Sciences, New York, NY, USA
| | - Tim A Ahles
- Memorial Sloan Kettering Cancer Center, Department of Psychiatry and Behavioral Sciences, New York, NY, USA
| | - James C Root
- Memorial Sloan Kettering Cancer Center, Department of Psychiatry and Behavioral Sciences, New York, NY, USA
| |
Collapse
|
13
|
Jones RM, Pattwell SS. Future considerations for pediatric cancer survivorship: Translational perspectives from developmental neuroscience. Dev Cogn Neurosci 2019; 38:100657. [PMID: 31158802 PMCID: PMC6697051 DOI: 10.1016/j.dcn.2019.100657] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 04/26/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023] Open
Abstract
Breakthroughs in modern medicine have increased pediatric cancer survival rates throughout the last several decades. Despite enhanced cure rates, a subset of pediatric cancer survivors exhibit life-long psychological side effects. A large body of work has addressed potential mechanisms for secondary symptoms of anxiety, post-traumatic stress, impaired emotion regulation and cognitive deficits in adults. Yet, absent from many studies are the ways in which cancer treatment can impact the developing brain. Additionally, it remains less known whether typical neurobiological changes during adolescence and early adulthood may potentially buffer or exacerbate some of the known negative cancer survivorship outcomes. This review highlights genetic, animal, and human neuroimaging research across development. We focus on the neural circuitry associated with aversive learning, which matures throughout childhood, adolescence and early adulthood. We argue that along with other individual differences, the precise timing of oncological treatment insults on such neural circuitry may expose particular vulnerabilities for pediatric cancer patients. We also explore other moderators of treatment outcomes, including genetic polymorphisms and neural mechanisms underlying memory and cognitive control. We discuss how neural maturation extending into young adulthood may also provide a sensitive period for intervention to improve psychological and cognitive outcomes in pediatric cancer survivors.
Collapse
Affiliation(s)
- Rebecca M Jones
- The Sackler Institute for Developmental Psychobiology, Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, United States
| | - Siobhan S Pattwell
- Human Biology Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Mailstop C3-168, Seattle, WA 98109, United States.
| |
Collapse
|
14
|
Chemotherapy-induced cognitive impairments: A systematic review of the animal literature. Neurosci Biobehav Rev 2019; 102:382-399. [DOI: 10.1016/j.neubiorev.2019.05.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/02/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022]
|
15
|
Port RG, Berman JI, Liu S, Featherstone RE, Roberts TP, Siegel SJ. Parvalbumin Cell Ablation of NMDA-R1 Leads to Altered Phase, But Not Amplitude, of Gamma-Band Cross-Frequency Coupling. Brain Connect 2019; 9:263-272. [PMID: 30588822 PMCID: PMC6479236 DOI: 10.1089/brain.2018.0639] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Altered gamma-band electrophysiological activity in individuals with autism spectrum disorder (ASD) is well documented, and analogous gamma-band alterations are recapitulated in several preclinical murine models relevant to ASD. Such gamma-band activity is hypothesized to underlie local circuit processes. Gamma-band cross-frequency coupling (CFC), a related though distinct metric, interrogates local neural circuit signal integration. Several recent studies have observed perturbed gamma-band CFC in individuals with ASD, although the direction of change remains unresolved. It also remains unclear whether murine models relevant to ASD recapitulate this altered gamma-band CFC. As such, this study examined whether mice with parvalbumin (PV) cell-specific ablation of NMDA-R1 (PVcre/NR1fl/fl) demonstrated altered gamma-band CFC as compared with their control littermates (PVcre/NR1+/+-mice that do not have the PV cell-specific ablation of NMDA-R1). Ten mice of each genotype had 4 min of "resting" electroencephalography recorded and analyzed. First, resting electrophysiological power was parsed into the canonical frequency bands and genotype-related differences were subsequently explored so as to provide context for the subsequent CFC analyses. PVcre/NR1fl/fl mice exhibited an increase in resting power specific to the high gamma-band, but not other frequency bands, as compared with PVcre/NR1+/+. CFC analyses then examined both the standard magnitude (strength) of CFC and the novel metric PhaseMax-which denotes the phase of the lower frequency signal at which the peak higher frequency signal power occurred. PVcre/NR1fl/fl mice exhibited altered PhaseMax, but not strength, of gamma-band CFC as compared with PVcre/NR1+/+ mice. As such, this study suggests a potential novel metric to explore when studying neuropsychiatric disorders.
Collapse
Affiliation(s)
- Russell G. Port
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jeffrey I. Berman
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Song Liu
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Robert E. Featherstone
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Timothy P.L. Roberts
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Steven J. Siegel
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
16
|
Abstract
As the population of cancer survivors has grown into the millions, there has been increasing emphasis on understanding how the late effects of treatment affect survivors' ability to return to work/school, their capacity to function and live independently, and their overall quality of life. This review focuses on cognitive change associated with cancer and cancer treatments. Research in this area has progressed from a pharmacotoxicology perspective to a view of the cognitive change as a complex interaction of aspects of the treatment, vulnerability factors that increase risk for posttreatment cognitive decline, cancer biology, and the biology of aging. Methodological advances include the development of (a) measurement approaches that assess more fine-grained subcomponents of cognition based on cognitive neuroscience and (b) advanced statistical approaches. Conceptual issues that arise from this multidimensional perspective are described in relation to future directions, understanding of mechanisms, and development of innovative interventions.
Collapse
Affiliation(s)
- Tim A Ahles
- Neurocognitive Research Lab, Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10022, USA; ,
| | - James C Root
- Neurocognitive Research Lab, Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10022, USA; ,
| |
Collapse
|
17
|
Himmel LE, Lustberg MB, DeVries AC, Poi M, Chen CS, Kulp SK. Minocycline, a putative neuroprotectant, co-administered with doxorubicin-cyclophosphamide chemotherapy in a xenograft model of triple-negative breast cancer. ACTA ACUST UNITED AC 2016; 68:505-515. [PMID: 27555377 DOI: 10.1016/j.etp.2016.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 07/15/2016] [Accepted: 08/08/2016] [Indexed: 12/25/2022]
Abstract
Minocycline is purported to have neuroprotective properties in experimental models of some human neurologic diseases, and has therefore been identified as a putative neuroprotectant for chemotherapy-induced cognitive impairment (CICI) in breast cancer patients. However, because its mechanism of action is believed to be mediated through anti-inflammatory, anti-apoptotic, and anti-oxidant pathways, co-administration of minocycline with chemotherapeutic agents has the potential to reduce the efficacy of anticancer drugs. The objective of this study is to evaluate the effect of minocycline on the activity of the AC chemotherapeutic regimen (Adriamycin [doxorubicin], Cytoxan [cyclophosphamide]) in in vitro and in vivo models of triple-negative breast cancer (TNBC). Clonogenic and methylthiazol tetrazolium (MTT) assays were used to assess survival and viability in two TNBC cell lines treated with increasing concentrations of AC in the presence or absence of minocycline. Biomarkers of apoptosis, cell stress, and DNA damage were evaluated by western blot. The in vivo effects of AC and minocycline, each alone and in combination, were assessed in a xenograft model of TNBC in female athymic nude mice by weekly tumor volume measurement, body and organ weight measurement, and histopathology. Apoptosis and proliferation were characterized by immunohistochemistry in the xenografts tumors. Brains from tumor-bearing mice were evaluated for microglial activation, glial scars, and the proportion of neural progenitor cells. Data from these in vitro and in vivo studies demonstrate that minocycline does not diminish the cytotoxic and tumor-suppressive effects of this chemotherapeutic drug combination in TNBC cells. Moreover, minocycline appeared to prevent the reduction in doublecortin-positive neural progenitor cells observed in AC-treated mice. We posit that minocycline may be useful clinically for its reported neuroprotective activity in breast cancer patients receiving AC without loss of chemotherapeutic efficacy.
Collapse
Affiliation(s)
- Lauren E Himmel
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA; Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Maryam B Lustberg
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - A Courtney DeVries
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ming Poi
- Division of Pharmacy Practice and Science, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Ching-Shih Chen
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Samuel K Kulp
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
18
|
Root JC, Andreotti C, Tsu L, Ellmore TM, Ahles TA. Learning and memory performance in breast cancer survivors 2 to 6 years post-treatment: the role of encoding versus forgetting. J Cancer Surviv 2016; 10:593-9. [PMID: 26658913 PMCID: PMC4864094 DOI: 10.1007/s11764-015-0505-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/25/2015] [Indexed: 11/29/2022]
Abstract
PURPOSE Our previous retrospective analysis of clinically referred breast cancer survivors' performance on learning and memory measures found a primary weakness in initial encoding of information into working memory with intact retention and recall of this same information at a delay. This suggests that survivors may misinterpret cognitive lapses as being due to forgetting when, in actuality, they were not able to properly encode this information at the time of initial exposure. Our objective in this study was to replicate and extend this pattern of performance to a research sample to increase the generalizability of this finding in a sample in which subjects were not clinically referred for cognitive issues. METHODS We contrasted learning and memory performance between breast cancer survivors on endocrine therapy 2 to 6 years post-treatment with age- and education-matched healthy controls. We then stratified lower- and higher-performing breast cancer survivors to examine specific patterns of learning and memory performance. Contrasts were generated for four aggregate visual and verbal memory variables from the California Verbal Learning Test-2 (CVLT-2) and the Brown Location Test (BLT): Single-trial Learning: Trial 1 performance, Multiple-trial Learning: Trial 5 performance, Delayed Recall: Long-delay Recall performance, and Memory Errors: False-positive errors. RESULTS As predicted, breast cancer survivors' performance as a whole was significantly lower on Single-trial Learning than the healthy control group but exhibited no significant difference in Delayed Recall. In the secondary analysis contrasting lower- and higher-performing survivors on cognitive measures, the same pattern of lower Single-trial Learning performance was exhibited in both groups, with the additional finding of significantly weaker Multiple-trial Learning performance in the lower-performing breast cancer group and intact Delayed Recall performance in both groups. CONCLUSIONS As with our earlier finding of weaker initial encoding with intact recall in a cohort of clinically referred breast cancer survivors, our results indicate this same profile in a research sample of breast cancer survivors. Further, when the breast cancer group was stratified by lower and higher performance, both groups exhibited significantly lower performance on initial encoding, with more pronounced encoding weakness in the lower-performing group. As in our previous research, survivors did not lose successfully encoded information over longer delays, either in the lower- or higher-performing group, again arguing against memory decay in survivors. The finding of weaker initial encoding of information together with intact delayed recall in survivors points to specific treatment interventions in rehabilitation of cognitive dysfunction. IMPLICATIONS FOR CANCER SURVIVORS The finding of weaker initial encoding of information together with intact delayed recall in survivors points to specific treatment interventions in rehabilitation of cognitive dysfunction and is discussed.
Collapse
Affiliation(s)
- James C Root
- Neurocognitive Research Laboratory, Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, 641 Lexington Avenue, 7th Floor, New York, NY, 10022, USA.
- Weill Cornell Medical College, New York, NY, USA.
| | - Charissa Andreotti
- Neurocognitive Research Laboratory, Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, 641 Lexington Avenue, 7th Floor, New York, NY, 10022, USA
| | - Loretta Tsu
- Department of Psychology, Program in Behavioral and Cognitive Neuroscience, The Graduate Center, City University of New York, The City College of the City University of New York, New York, NY, USA
| | - Timothy M Ellmore
- Department of Psychology, Program in Behavioral and Cognitive Neuroscience, The Graduate Center, City University of New York, The City College of the City University of New York, New York, NY, USA
| | - Tim A Ahles
- Neurocognitive Research Laboratory, Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, 641 Lexington Avenue, 7th Floor, New York, NY, 10022, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
19
|
Dexras1 a unique ras-GTPase interacts with NMDA receptor activity and provides a novel dissociation between anxiety, working memory and sensory gating. Neuroscience 2016; 322:408-15. [PMID: 26946266 DOI: 10.1016/j.neuroscience.2016.02.063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 11/21/2022]
Abstract
Dexras1 is a novel GTPase that acts at a confluence of signaling mechanisms associated with psychiatric and neurological disease including NMDA receptors, NOS1AP and nNOS. Recent work has shown that Dexras1 mediates iron trafficking and NMDA-dependent neurodegeneration but a role for Dexras1 in normal brain function or psychiatric disease has not been studied. To test for such a role, mice with germline knockout (KO) of Dexras1 were assayed for behavioral abnormalities as well as changes in NMDA receptor subunit protein expression. Because Dexras1 is up-regulated during stress or by dexamethasone treatment, we included measures associated with emotion including anxiety and depression. Baseline anxiety-like measures (open field and zero maze) were not altered, nor were depression-like behavior (tail suspension). Measures of memory function yielded mixed results, with no changes in episodic memory (novel object recognition) but a significant decrement on working memory (T-maze). Alternatively, there was an increase in pre-pulse inhibition (PPI), without concomitant changes in either startle amplitude or locomotor activity. PPI data are consistent with the direction of change seen following exposure to dopamine D2 antagonists. An examination of NMDA subunit expression levels revealed an increased expression of the NR2A subunit, contrary to previous studies demonstrating down-regulation of the receptor following antipsychotic exposure (Schmitt et al., 2003) and up-regulation after exposure to isolation rearing (Turnock-Jones et al., 2009). These findings suggest a potential role for Dexras1 in modulating a selective subset of psychiatric symptoms, possibly via its interaction with NMDARs and/or other disease-related binding-partners. Furthermore, data suggest that modulating Dexras1 activity has contrasting effects on emotional, sensory and cognitive domains.
Collapse
|
20
|
Seigers R, Loos M, Van Tellingen O, Boogerd W, Smit AB, Schagen SB. Neurobiological changes by cytotoxic agents in mice. Behav Brain Res 2015; 299:19-26. [PMID: 26602283 DOI: 10.1016/j.bbr.2015.10.057] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 11/15/2022]
Abstract
Cognitive deficit is a frequently reported side-effect of adjuvant chemotherapy. A large number of animal studies has been performed to examine the neurobiological mechanisms underlying this phenomenon, however, definite conclusions from these studies are restricted due to differences in experimental set-up. We systematically investigated the effects of 6 cytotoxic agents on various neurobiological parameters. C57Bl/6J mice were treated with cyclophosphamide, docetaxel, doxorubicin, 5-fluorouracil, methotrexate, or topotecan. The animals were sacrificed 3 or 15 weeks after treatment and the effect on neurogenesis, blood vessel density, and neuroinflammation was analyzed using immunohistochemistry. None of the cytostatic agents tested affected neurogenesis (cell survival or cell proliferation). Blood vessel density was increased in the hippocampus and prefrontal cortex 3 weeks after treatment with docetaxel and doxorubicin compared with control animals. A decrease in the number of microglial cells was observed in the prefrontal cortex after treatment with cyclophosphamide, docetaxel, 5-FU, and topotecan compared with control mice. The observed decrease in microglia cells is indicative of inflammation that occurred after treatment. Overall, the magnitude of the effects was relatively modest. Therefore, we conducted a similar study with topotecan in Abcg2;Abcb1a/b knock out and wildtype FVB mice. Animals were sacrificed 3 weeks after treatment and no notable effect was seen in hippocampal cell differentiation (DCX), microglia activation, or blood vessel density. Perhaps the FVB strain is more resistant to the neurotoxic effects of topotecan which makes this not the correct model to study the mechanism of chemotherapy-induced cognitive impairment.
Collapse
Affiliation(s)
- R Seigers
- Department of Psychosocial Research and Epidemiology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M Loos
- Sylics (Synaptologics BV), Amsterdam, the Netherlands
| | - O Van Tellingen
- Division of Molecular Biology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - W Boogerd
- Department of Neuro-Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, the Netherlands
| | - S B Schagen
- Department of Psychosocial Research and Epidemiology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
21
|
Assessment of disease-related cognitive impairments using the novel object recognition (NOR) task in rodents. Behav Brain Res 2015; 285:176-93. [DOI: 10.1016/j.bbr.2014.10.025] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/16/2014] [Accepted: 10/19/2014] [Indexed: 12/11/2022]
|
22
|
Fardell JE, Vardy J, Monds LA, Johnston IN. The long-term impact of oxaliplatin chemotherapy on rodent cognition and peripheral neuropathy. Behav Brain Res 2015; 291:80-88. [PMID: 25934489 DOI: 10.1016/j.bbr.2015.04.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/18/2015] [Accepted: 04/23/2015] [Indexed: 10/23/2022]
Abstract
Chemotherapy treatment is associated with cognitive dysfunction in cancer survivors after treatment completion. The duration of these impairments is unclear. Therefore this paper aims to evaluate the lasting impact of varying doses of the chemotherapy oxaliplatin (OX) on cognition and peripheral neuropathy. In Experiment 1 rats were treated once a week for 3 weeks with either physiological saline (control) or 6 mg/kg OX i.p. and were assessed for peripheral neuropathy, using von Frey filaments, and cognitive function, using novel object and location recognition, up to 2 weeks after treatment completion. For Experiment 2 rats received 3 weekly i.p. injections of either physiological saline (control), 0.6 mg/kg, 2mg/kg or 6 mg/kg OX and assessed for peripheral neuropathy and cognitive function up to 11 months after treatment completion. Systemic OX treatment induced lasting effects on cognitive function at 11 months after treatment, and peripheral neuropathy at 1 month after treatment and these were dose dependent; higher doses of OX resulted in worse cognitive outcomes and more severe peripheral neuropathy.
Collapse
Affiliation(s)
| | - Janette Vardy
- Concord Cancer Centre, Concord General Repatriation Hospital, Sydney Medical School, The University of Sydney.
| | - Lauren A Monds
- Discipline of Addiction Medicine, Central Clinical School, Sydney Medical School, The University of Sydney.
| | - Ian N Johnston
- School of Psychology, The University of Sydney, Australia.
| |
Collapse
|
23
|
Featherstone RE, McMullen MF, Ward KR, Bang J, Xiao J, Siegel SJ. EEG biomarkers of target engagement, therapeutic effect, and disease process. Ann N Y Acad Sci 2015; 1344:12-26. [DOI: 10.1111/nyas.12745] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Robert E. Featherstone
- Translational Neuroscience Program; Department of Psychiatry; University of Pennsylvania; Philadelphia Pennsylvania
| | - Mary F. McMullen
- Translational Neuroscience Program; Department of Psychiatry; University of Pennsylvania; Philadelphia Pennsylvania
| | - Katelyn R. Ward
- Translational Neuroscience Program; Department of Psychiatry; University of Pennsylvania; Philadelphia Pennsylvania
| | - Jakyung Bang
- Translational Neuroscience Program; Department of Psychiatry; University of Pennsylvania; Philadelphia Pennsylvania
| | - Jane Xiao
- Translational Neuroscience Program; Department of Psychiatry; University of Pennsylvania; Philadelphia Pennsylvania
| | - Steven J. Siegel
- Translational Neuroscience Program; Department of Psychiatry; University of Pennsylvania; Philadelphia Pennsylvania
| |
Collapse
|
24
|
Pyramidal cell selective ablation of N-methyl-D-aspartate receptor 1 causes increase in cellular and network excitability. Biol Psychiatry 2015; 77:556-68. [PMID: 25156700 PMCID: PMC4297754 DOI: 10.1016/j.biopsych.2014.06.026] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/05/2014] [Accepted: 06/22/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND Neuronal activity at gamma frequency is impaired in schizophrenia (SZ) and is considered critical for cognitive performance. Such impairments are thought to be due to reduced N-methyl-D-aspartate receptor (NMDAR)-mediated inhibition from parvalbumin interneurons, rather than a direct role of impaired NMDAR signaling on pyramidal neurons. However, recent studies suggest a direct role of pyramidal neurons in regulating gamma oscillations. In particular, a computational model has been proposed in which phasic currents from pyramidal cells could drive synchronized feedback inhibition from interneurons. As such, impairments in pyramidal neuron activity could lead to abnormal gamma oscillations. However, this computational model has not been tested experimentally and the molecular mechanisms underlying pyramidal neuron dysfunction in SZ remain unclear. METHODS In the present study, we tested the hypothesis that SZ-related phenotypes could arise from reduced NMDAR signaling in pyramidal neurons using forebrain pyramidal neuron specific NMDA receptor 1 knockout mice. RESULTS The mice displayed increased baseline gamma power, as well as sociocognitive impairments. These phenotypes were associated with increased pyramidal cell excitability due to changes in inherent membrane properties. Interestingly, mutant mice showed decreased expression of GIRK2 channels, which has been linked to increased neuronal excitability. CONCLUSIONS Our data demonstrate for the first time that NMDAR hypofunction in pyramidal cells is sufficient to cause electrophysiological, molecular, neuropathological, and behavioral changes related to SZ.
Collapse
|
25
|
Fonck C, Easter A, Pietras MR, Bialecki RA. CNS Adverse Effects: From Functional Observation Battery/Irwin Tests to Electrophysiology. Handb Exp Pharmacol 2015; 229:83-113. [PMID: 26091637 DOI: 10.1007/978-3-662-46943-9_4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This chapter describes various approaches for the preclinical assessment of drug-induced central nervous system (CNS) adverse effects. Traditionally, methods to evaluate CNS effects have consisted of observing and scoring behavioral responses of animals after drug is administered. Among several behavioral testing paradigms, the Irwin and the functional observational battery (FOB) are the most commonly used assays for the assessment of CNS effects. The Irwin and FOB are considered good first-tier assays to satisfy the ICH S7A guidance for the preclinical evaluation of new chemical entities (NCE) intended for humans. However, experts have expressed concern about the subjectivity and lack of quantitation that is derived from behavioral testing. More importantly, it is difficult to gain insight into potential mechanisms of toxicity by assessing behavioral outcomes. As a complement to behavioral testing, we propose using electrophysiology-based assays, both in vivo and in vitro, such as electroencephalograms and brain slice field-potential recordings. To better illustrate these approaches, we discuss the implementation of electrophysiology-based techniques in drug-induced assessment of seizure risk, sleep disruption, and cognitive impairment.
Collapse
Affiliation(s)
- Carlos Fonck
- Global Safety Pharmacology, AstraZeneca Pharmaceuticals LP, 35 Gatehouse Drive, Waltham, MA, 02451, USA
| | | | | | | |
Collapse
|
26
|
Seigers R, Loos M, Van Tellingen O, Boogerd W, Smit AB, Schagen SB. Cognitive impact of cytotoxic agents in mice. Psychopharmacology (Berl) 2015; 232:17-37. [PMID: 24894481 DOI: 10.1007/s00213-014-3636-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/05/2014] [Indexed: 01/12/2023]
Abstract
RATIONALE AND OBJECTIVES Adjuvant chemotherapy is associated with changes in cognition in a subgroup of cancer patients. Chemotherapy is generally given as a combination of cytotoxic agents, which makes it hard to define the agent responsible for these observed changes. Literature on animal experiments has been difficult to interpret due to variance in experimental setup. METHODS We examined the effects of cytotoxic agents administered separately on various cognitive measures in a standardized animal model. Male C57Bl/6 mice received cyclophosphamide, docetaxel, doxorubicin, 5-fluorouracil, methotrexate, or topotecan. These agents represent different compound classes based on their working mechanism and are frequently prescribed in the clinic. A control group received saline. Behavioral testing started 2 or 15 weeks after treatment and included testing general measures of behavior and cognitive task performance: spontaneous behavior in an automated home cage, open field, novel location recognition (NLR), novel object recognition (NOR), Barnes maze, contextual fear conditioning, and a simple choice reaction time task (SCRTT). RESULTS Cyclophosphamide, docetaxel, and doxorubicin administration affected spontaneous activity in the automated home cage. All cytotoxic agents affected memory (NLR and/or NOR). Spatial memory measured in the Barnes maze was affected after administration with doxorubicin, 5-fluorouracil, and topotecan. Decreased inhibition in the SCRTT was observed after treatment with cyclophosphamide, docetaxel, and topotecan. CONCLUSIONS Our data show that, in mice, a single treatment with a cytotoxic agent causes cognitive impairment. Not all cytotoxic agents affected the same cognitive domains, which might be explained by differences in working mechanisms of the various agents.
Collapse
Affiliation(s)
- R Seigers
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
27
|
Bösche K, Weissenborn K, Christians U, Witzke O, Engler H, Schedlowski M, Hadamitzky M. Neurobehavioral consequences of small molecule-drug immunosuppression. Neuropharmacology 2014; 96:83-93. [PMID: 25529273 DOI: 10.1016/j.neuropharm.2014.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/05/2014] [Accepted: 12/08/2014] [Indexed: 12/29/2022]
Abstract
60 years after the first successful kidney transplantation in humans, transplant patients have decent survival rates owing to a broad spectrum of immunosuppressive medication available today. Not only transplant patients, but also patients with inflammatory autoimmune diseases or cancer benefit from these life-saving immunosuppressive and anti-proliferative medications. However, this success is gained with the disadvantage of neuropsychological disturbances and mental health problems such as depression, anxiety and impaired quality of life after long-term treatment with immunosuppressive drugs. So far, surprisingly little is known about unwanted neuropsychological side effects of immunosuppressants and anti-proliferative drugs from the group of so called small molecule-drugs. This is partly due to the fact that it is difficult to disentangle whether and to what extent the observed neuropsychiatric disturbances are a direct result of the patient's medical history or of the immunosuppressive treatment. Thus, here we summarize experimental as well as clinical data of mammalian and human studies, with the focus on selected small-molecule drugs that are frequently employed in solid organ transplantation, autoimmune disorders or cancer therapy and their effects on neuropsychological functions, mood, and behavior. These data reveal the necessity to develop immunosuppressive and anti-proliferative drugs inducing fewer or no unwanted neuropsychological side effects, thereby increasing the quality of life in patients requiring long term immunosuppressive treatment. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Katharina Bösche
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital, Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany.
| | - Karin Weissenborn
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Uwe Christians
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Oliver Witzke
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Harald Engler
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital, Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital, Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Martin Hadamitzky
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital, Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany.
| |
Collapse
|
28
|
Evaluation of the impact of the cancer therapy everolimus on the central nervous system in mice. PLoS One 2014; 9:e113533. [PMID: 25436776 PMCID: PMC4250083 DOI: 10.1371/journal.pone.0113533] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 10/24/2014] [Indexed: 01/13/2023] Open
Abstract
Cancer and treatments may induce cognitive impairments in cancer patients, and the causal link between chemotherapy and cognitive dysfunctions was recently validated in animal models. New cancer targeted therapies have become widely used, and their impact on brain functions and quality of life needs to be explored. We evaluated the impact of everolimus, an anticancer agent targeting the mTOR pathway, on cognitive functions, cerebral metabolism, and hippocampal cell proliferation/vascular density in mice. Adult mice received everolimus daily for 2 weeks, and behavioral tests were performed from 1 week after the last treatment. Everolimus-treated mice displayed a marked reduction in weight gain from the last day of the treatment period. Ex vivo analysis showed altered cytochrome oxidase activity in selective cerebral regions involved in energy balance, food intake, reward, learning and memory modulation, sleep/wake cycle regulation, and arousal. Like chemotherapy, everolimus did not alter emotional reactivity, learning and memory performances, but in contrast to chemotherapy, did not affect behavioral flexibility or reactivity to novelty. In vivo hippocampal neural cell proliferation and vascular density were also unchanged after everolimus treatments. In conclusion, two weeks daily everolimus treatment at the clinical dose did not evoke alteration of cognitive performances evaluated in hippocampal- and prefrontal cortex-dependent tasks that would persist at one to four weeks after the end of the treatment completion. However, acute everolimus treatment caused selective CO modifications without altering the mTOR effector P70S6 kinase in cerebral regions involved in feeding behavior and/or the sleep/wake cycle, at least in part under control of the solitary nucleus and the parasubthalamic region of the hypothalamus. Thus, this area may represent a key target for everolimus-mediating peripheral modifications, which has been previously associated with symptoms such as weight loss and fatigue.
Collapse
|
29
|
Yang M, Moon C. Neurotoxicity of cancer chemotherapy. Neural Regen Res 2014; 8:1606-14. [PMID: 25206457 PMCID: PMC4145960 DOI: 10.3969/j.issn.1673-5374.2013.17.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 01/15/2013] [Indexed: 12/18/2022] Open
Abstract
There is accumulating clinical evidence that chemotherapeutic agents induce neurological side effects, including memory deficits and mood disorders, in cancer patients who have undergone chemotherapeutic treatments. This review focuses on chemotherapy-induced neurodegeneration and hippocampal dysfunctions and related mechanisms as measured by in vivo and in vitro approaches. These investigations are helpful in determining how best to further explore the causal mechanisms of chemotherapy-induced neurological side effects and in providing direction for the future development of novel optimized chemotherapeutic agents.
Collapse
Affiliation(s)
- Miyoung Yang
- Department of Veterinary Anatomy, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea ; Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | - Changjong Moon
- Department of Veterinary Anatomy, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea
| |
Collapse
|
30
|
Chemotherapy-related cognitive dysfunction: current animal studies and future directions. Brain Imaging Behav 2014; 7:453-9. [PMID: 23949877 DOI: 10.1007/s11682-013-9250-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Cognitive impairment is a potential long-term side effect of adjuvant chemotherapy that can have a major impact on the quality of life of cancer survivors. There is a growing number of preclinical studies addressing this issue, thereby extending our knowledge of the mechanisms underlying chemotherapy-induced neurotoxicity. In this review, we will summarize the recent advances and important findings presented in these studies. Emerging challenges, such as the development of neuroprotective strategies, and the role of the blood-brain barrier on cognitive impairment will be described and future directions in this field of investigation will be outlined.
Collapse
|
31
|
Parvalbumin cell ablation of NMDA-R1 causes increased resting network excitability with associated social and self-care deficits. Neuropsychopharmacology 2014; 39:1603-13. [PMID: 24525709 PMCID: PMC4023157 DOI: 10.1038/npp.2014.7] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 10/28/2013] [Accepted: 10/28/2013] [Indexed: 02/04/2023]
Abstract
NMDA-receptor (NMDAR) hypofunction is strongly implicated in the pathophysiology of schizophrenia. Several convergent lines of evidence suggest that net excitation propagated by impaired NMDAR signaling on GABAergic interneurons may be of particular interest in mediating several aspects of schizophrenia. However, it is unclear which behavioral domains are governed by a net increase of excitation and whether modulating downstream GABAergic signaling can reverse neural and thus behavioral deficits. The current study determines the selective contributions of NMDAR dysfunction on PV-containing interneurons to electrophysiological, cognitive, and negative-symptom-related behavioral phenotypes of schizophrenia using mice with a PVcre-NR1flox-driven ablation of NR1 on PV-containing interneurons. In addition, we assessed the efficacy of one agent that directly modulates GABAergic signaling (baclofen) and one agent that indirectly modifies NMDAR-mediated signaling through antagonism of mGluR5 receptors (2-methyl-6-(phenylethynyl) pyridine (MPEP)). The data indicate that loss of NMDAR function on PV interneurons impairs self-care and sociability while increasing N1 latency and baseline gamma power, and reducing induction and maintenance of long-term potentiation. Baclofen normalized baseline gamma power without corresponding effects on behavior. MPEP further increased N1 latency and reduced social behavior in PVcre/NR1+/+ mice. These two indices were negatively correlated before and following MPEP such that as N1 latency increases, sociability decreases. This finding suggests a predictive role for N1 latency with respect to social function. Although previous data suggest that MPEP may be beneficial for core features of autism spectrum disorders, current data suggest that such effects require intact function of NMDAR on PV interneurons.
Collapse
|
32
|
Dubois M, Lapinte N, Villier V, Lecointre C, Roy V, Tonon MC, Gandolfo P, Joly F, Hilber P, Castel H. Chemotherapy-induced long-term alteration of executive functions and hippocampal cell proliferation: Role of glucose as adjuvant. Neuropharmacology 2014; 79:234-48. [DOI: 10.1016/j.neuropharm.2013.11.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 11/18/2013] [Accepted: 11/19/2013] [Indexed: 11/28/2022]
|
33
|
Abstract
Psychosis is an abnormal mental state characterized by disorganization, delusions and hallucinations. Animal models have become an increasingly important research tool in the effort to understand both the underlying pathophysiology and treatment of psychosis. There are multiple animal models for psychosis, with each formed by the coupling of a manipulation and a measurement. In this manuscript we do not address the diseases of which psychosis is a prominent comorbidity. Instead, we summarize the current state of affairs and future directions for animal models of psychosis. To accomplish this, our manuscript will first discuss relevant behavioral and electrophysiological measurements. We then provide an overview of the different manipulations that are combined with these measurements to produce animal models. The strengths and limitations of each model will be addressed in order to evaluate its cross-species comparability.
Collapse
|
34
|
Kaiser J, Bledowski C, Dietrich J. Neural correlates of chemotherapy-related cognitive impairment. Cortex 2014; 54:33-50. [PMID: 24632463 DOI: 10.1016/j.cortex.2014.01.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 01/07/2014] [Accepted: 01/16/2014] [Indexed: 11/28/2022]
Abstract
Cancer survivors frequently experience cognitive deficits following chemotherapy. The most commonly affected functions include memory, attention and executive control. The present paper reviews animal research and clinical studies including event-related potential (ERP) and neuroimaging investigations of chemotherapy-related changes of brain structure and function. In rodents, chemotherapeutic substances have been shown to damage neural precursor cells and white matter tracts and are associated with impairments of learning and memory. Structural and functional changes associated with chemotherapy have also been observed in humans. Structural imaging has revealed gray and white matter volume reductions and altered white matter microstructure. Functional studies using either ERPs or hemodynamic imaging have shown that chemotherapy alters the activation patterns of cortical networks involved in higher cognitive functions. Collectively, these findings support the existence of the "chemobrain" phenomenon beyond the patients' subjective reports. However, the rather small number of studies and methodological limitations of some of the pioneering investigations call for further research of high methodological quality, including larger numbers of subjects with appropriate controls to delineate the temporal and spatial pattern of chemotherapy-associated central nervous system (CNS) toxicity. Brain activation studies in humans might systematically vary task difficulty levels to distinguish between compensatory hyper-activations on the one hand and deficient recruitment of resources on the other hand. Integrative functions could be tested by connectivity analyses using both electrophysiological and hemodynamic measures. The ultimate goal should be the development of cognitive-behavioral and pharmacological interventions to reduce the cognitive side effects of the medically indispensable but neurotoxic chemotherapeutic treatments.
Collapse
Affiliation(s)
- Jochen Kaiser
- Institute of Medical Psychology, Medical Faculty, Goethe University, Frankfurt am Main, Germany.
| | - Christoph Bledowski
- Institute of Medical Psychology, Medical Faculty, Goethe University, Frankfurt am Main, Germany
| | - Jörg Dietrich
- Department of Neurology, Massachusetts General Hospital, MGH Cancer Center and Center for Regenerative Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
35
|
Gandal MJ, Anderson RL, Billingslea EN, Carlson GC, Roberts TPL, Siegel SJ. Mice with reduced NMDA receptor expression: more consistent with autism than schizophrenia? GENES BRAIN AND BEHAVIOR 2013; 11:740-50. [PMID: 22726567 DOI: 10.1111/j.1601-183x.2012.00816.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Reduced NMDA-receptor (NMDAR) function has been implicated in the pathophysiology of neuropsychiatric disease, most strongly in schizophrenia but also recently in autism spectrum disorders (ASD). To determine the direct contribution of NMDAR dysfunction to disease phenotypes, a mouse model with constitutively reduced expression of the obligatory NR1 subunit has been developed and extensively investigated. Adult NR1(neo-/-) mice show multiple abnormal behaviors, including reduced social interactions, locomotor hyperactivity, self-injury, deficits in prepulse inhibition (PPI) and sensory hypersensitivity, among others. Whereas such phenotypes have largely been interpreted in the context of schizophrenia, these behavioral abnormalities are rather non-specific and are frequently present across models of diseases characterized by negative symptom domains. This study investigated auditory electrophysiological and behavioral paradigms relevant to autism, to determine whether NMDAR hypofunction may be more consistent with adult ASD-like phenotypes. Indeed, transgenic mice showed behavioral deficits relevant to all core ASD symptoms, including decreased social interactions, altered ultrasonic vocalizations and increased repetitive behaviors. NMDAR disruption recapitulated clinical endophenotypes including reduced PPI, auditory-evoked response N1 latency delay and reduced gamma synchrony. Auditory electrophysiological abnormalities more closely resembled those seen in clinical studies of autism than schizophrenia. These results suggest that NMDAR hypofunction may be associated with a continuum of neuropsychiatric diseases, including schizophrenia and autism. Neural synchrony abnormalities suggest an imbalance of glutamatergic and GABAergic coupling and may provide a target, along with behavioral phenotypes, for preclinical screening of novel therapeutics.
Collapse
Affiliation(s)
- M J Gandal
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Saunders JA, Tatard-Leitman VM, Suh J, Billingslea EN, Roberts TP, Siegel SJ. Knockout of NMDA receptors in parvalbumin interneurons recreates autism-like phenotypes. Autism Res 2013; 6:69-77. [PMID: 23441094 DOI: 10.1002/aur.1264] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 10/22/2012] [Indexed: 11/11/2022]
Abstract
Autism is a disabling neurodevelopmental disorder characterized by social deficits, language impairment, and repetitive behaviors with few effective treatments. New evidence suggests that autism has reliable electrophysiological endophenotypes and that these measures may be caused by n-methyl-d-aspartic acid receptor (NMDAR) disruption on parvalbumin (PV)-containing interneurons. These findings could be used to create new translational biomarkers. Recent developments have allowed for cell-type selective knockout of NMDARs in order to examine the perturbations caused by disrupting specific circuits. This study examines several electrophysiological and behavioral measures disrupted in autism using a PV-selective reduction in NMDA R1 subunit. Mouse electroencephalograph (EEG) was recorded in response to auditory stimuli. Event-related potential (ERP) component amplitude and latency analysis, social testing, and premating ultrasonic vocalizations (USVs) recordings were performed. Correlations were examined between the ERP latency and behavioral measures. The N1 ERP latency was delayed, sociability was reduced, and mating USVs were impaired in PV-selective NMDA Receptor 1 Knockout (NR1 KO) as compared with wild-type mice. There was a significant correlation between N1 latency and sociability but not between N1 latency and premating USV power or T-maze performance. The increases in N1 latency, impaired sociability, and reduced vocalizations in PV-selective NR1 KO mice mimic similar changes found in autism. Electrophysiological changes correlate to reduced sociability, indicating that the local circuit mechanisms controlling N1 latency may be utilized in social function. Therefore, we propose that behavioral and electrophysiological alterations in PV-selective NR1 KO mice may serve as a useful model for therapeutic development in autism. Autism Res 2013, 6: 69-77. © 2013 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- John A Saunders
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
37
|
Wigmore P. The effect of systemic chemotherapy on neurogenesis, plasticity and memory. Curr Top Behav Neurosci 2013; 15:211-240. [PMID: 23239468 DOI: 10.1007/7854_2012_235] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Chemotherapy has been enormously successful in treating many forms of cancer and improving patient survival rates. With the increasing numbers of survivors, a number of cognitive side effects have become apparent. These have been called "chemobrain" or "chemofog" among patient groups, who describe the symptoms as a decline in memory, concentration and executive functions. Changes which, although subtle, can cause significant distress among patients and prevent a return to the quality of life experienced before treatment. This cognitive side effect of chemotherapy was not anticipated as it had been assumed that chemotherapy agents, administered systematically, could not cross the blood-brain barrier and that the brain was therefore protected from their action. It is now realised that low concentrations of many chemotherapy agents cross the blood-brain barrier and even those that are completely prevented from doing so, can induce the production of inflammatory cytokines in peripheral tissues which in turn can cross the blood-brain barrier and impact on the brain. A large number of patient studies have shown that cognitive decline is found in a proportion of patients treated with a variety of chemotherapy agents for different types of cancer. The deficits experienced by these patients can last for up to several years and have a deleterious effect on educational attainment and ability to return to work. Imaging studies of patients after systemic chemotherapy show that this treatment produces structural and functional changes in the brain some of which seem to persist even when the cognitive deficits have ceased. This suggests that, with time, brain plasticity may be able to compensate for the deleterious effects of chemotherapy treatment. A number of mechanisms have been suggested for the changes in brain structure and function found after chemotherapy. These include both central and peripheral inflammatory changes, demyelination of white matter tracts, a reduction in stem cell proliferation in both the hippocampal neurogenic region and by oligodendrocyte precursors as well as changes in hormonal or growth factor levels. A number of possible treatments have been suggested which range from pharmacological interventions to cognitive behavioural therapies. Some of these have only been tested in animal models while others have produced varying degrees of improvement in patient populations. Currently, there is no recognised treatment and a greater understanding of the causes of the cognitive decline experienced after chemotherapy will be key to finding ways of preventing or treating the effects of chemobrain.
Collapse
Affiliation(s)
- Peter Wigmore
- School of Biomedical Sciences, Queen's Medical Centre, Nottingham, NG7 2UH, UK,
| |
Collapse
|
38
|
Saunders JA, Gandal MJ, Roberts TP, Siegel SJ. NMDA antagonist MK801 recreates auditory electrophysiology disruption present in autism and other neurodevelopmental disorders. Behav Brain Res 2012; 234:233-7. [PMID: 22771812 DOI: 10.1016/j.bbr.2012.06.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 06/21/2012] [Accepted: 06/25/2012] [Indexed: 11/19/2022]
Abstract
Autism is a highly disabling neurodevelopmental disorder characterized by social deficits, language impairment, and repetitive behaviors. There are few effective biological treatments for this disorder, partly due to the lack of translational biomarkers. However, recent data suggest that autism has reliable electrophysiological endophenotypes, along with evidence that some deficits may be caused by NMDA receptor (NMDAR) dysfunction. Similarly, the NMDAR antagonist MK801 has been used in behavioral animal models of autism. Since MK801 has also been used as a model of schizophrenia, this paper examines the independent and overlapping ways in which MK801 recreates the electrophysiogical changes present in both diseases. Mouse EEG was recorded in response to auditory stimuli after either vehicle or MK801 and the dose-response relationship for each measure was determined. ERP component amplitude and latency analysis was performed along with time-frequency analysis of gamma frequency inter-trial coherence and evoked power. Evoked gamma power and ITC were decreased by MK801 at the highest dose. P1, N1 latency and gamma baseline power were increased in dose dependent fashion following MK801. There were no amplitude changes in P1 or N1. MK801 caused alterations in evoked gamma activity, gamma ITC, gamma baseline power, P1 and N1 latency similar to findings in autism. These data provide evidence indicating that NMDAR dysfunction may contribute to deficits specific to autism and some that overlap with other disorders such as schizophrenia. Such observations could be important for developing novel therapeutics, as electrophysiological endophenotypes associate with functional measures and may provide early biomarkers for efficacy in clinical trials.
Collapse
Affiliation(s)
- John A Saunders
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, United States
| | | | | | | |
Collapse
|
39
|
Gandal MJ, Sisti J, Klook K, Ortinski PI, Leitman V, Liang Y, Thieu T, Anderson R, Pierce RC, Jonak G, Gur RE, Carlson G, Siegel SJ. GABAB-mediated rescue of altered excitatory-inhibitory balance, gamma synchrony and behavioral deficits following constitutive NMDAR-hypofunction. Transl Psychiatry 2012; 2:e142. [PMID: 22806213 PMCID: PMC3410621 DOI: 10.1038/tp.2012.69] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Reduced N-methyl-D-aspartate-receptor (NMDAR) signaling has been associated with schizophrenia, autism and intellectual disability. NMDAR-hypofunction is thought to contribute to social, cognitive and gamma (30-80 Hz) oscillatory abnormalities, phenotypes common to these disorders. However, circuit-level mechanisms underlying such deficits remain unclear. This study investigated the relationship between gamma synchrony, excitatory-inhibitory (E/I) signaling, and behavioral phenotypes in NMDA-NR1(neo-/-) mice, which have constitutively reduced expression of the obligate NR1 subunit to model disrupted developmental NMDAR function. Constitutive NMDAR-hypofunction caused a loss of E/I balance, with an increase in intrinsic pyramidal cell excitability and a selective disruption of parvalbumin-expressing interneurons. Disrupted E/I coupling was associated with deficits in auditory-evoked gamma signal-to-noise ratio (SNR). Gamma-band abnormalities predicted deficits in spatial working memory and social preference, linking cellular changes in E/I signaling to target behaviors. The GABA(B)-receptor agonist baclofen improved E/I balance, gamma-SNR and broadly reversed behavioral deficits. These data demonstrate a clinically relevant, highly translatable neural-activity-based biomarker for preclinical screening and therapeutic development across a broad range of disorders that share common endophenotypes and disrupted NMDA-receptor signaling.
Collapse
Affiliation(s)
- M J Gandal
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - J Sisti
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - K Klook
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA,Department of Psychiatry and Psychotherapy, RWTH Aachen University, Aachen, Germany
| | - P I Ortinski
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - V Leitman
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Y Liang
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - T Thieu
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - R Anderson
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - R C Pierce
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - G Jonak
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - R E Gur
- Neuropsychiatry Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - G Carlson
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - S J Siegel
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA,Director, Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Translational Research Laboratories, 125 S. 31st Street, Philadelphia, PA 19104, USA. E-mail: or
| |
Collapse
|
40
|
Abstract
Cancer therapy makes patients sick. The therapies that are available to clinicians allow them to successfully control nausea, emesis and pain. However, this is not the case for a number of other symptoms that include fatigue, distractibility, poor memory, and diminished interest in previously pleasurable activities. These symptoms cluster during the course of cancer therapy and impair patient quality of life, limit therapy options and do not always resolve at the cessation of treatment. It is possible to describe the intensity and temporal features of symptoms and assess their relationship with the inflammatory response that is associated with cancer and cancer therapy. At the preclinical level, sophisticated animal models still need to be deployed to study the causal role of inflammation in specific components of cancer-related symptoms. Various approaches can be optimally combined in a translational symptom research pathway to provide a framework for assessing in a systematic manner the neurobehavioral toxicity of existing and newly developed cancer therapies. Ultimately, this knowledge will allow derivation of mechanism-based interventions to prevent or alleviate cancer-related symptoms.
Collapse
|
41
|
Cognitive impairments caused by oxaliplatin and 5-fluorouracil chemotherapy are ameliorated by physical activity. Psychopharmacology (Berl) 2012; 220:183-93. [PMID: 21894483 DOI: 10.1007/s00213-011-2466-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 08/22/2011] [Indexed: 12/18/2022]
Abstract
RATIONALE Studies in women with breast cancer, and in animal models, have demonstrated that chemotherapy can have a negative impact on cognitive function. Which chemotherapy agents cause problems with cognition and the aetiology of the impairment is unknown. Furthermore, there is no proven treatment. OBJECTIVES This study aimed to evaluate the effects of 5-fluorouracil (5FU) and oxaliplatin (OX) chemotherapy agents commonly used to treat colorectal cancer on cognition in laboratory rodents. Furthermore, we assessed physical activity as a potential remedy for the observed chemotherapy-induced cognitive deficits. RESULTS In rodents, treatment with 5FU and OX alone impairs memory as measured by novel object recognition. But combined treatment appears to have greater detrimental effects on hippocampal-dependent tasks, contextual fear recall and spatial reference memory (water maze), yet had no effect on cued fear recall, a non-hippocampal task. These impairments were prevented by 4 weeks of wheel running overnight after 5FU/OX treatment. We found a significant interaction between chemotherapy and exercise: rats receiving both 5FU/OX and exercise had improved cognition relative to non-exercising 5FU/OX rats on novel object recognition and spatial reference memory. CONCLUSIONS The combination 5FU/OX had a significant impact on cognition. However, rats treated with 5FU/OX that exercised post chemotherapy had improved cognition relative to non-exercising rats. This suggests that physical activity may prove useful in ameliorating the cognitive impairments induced by 5FU/OX.
Collapse
|
42
|
The chemotherapy agent oxaliplatin impairs the renewal of fear to an extinguished conditioned stimulus in rats. Behav Brain Res 2012; 227:295-9. [DOI: 10.1016/j.bbr.2011.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 11/03/2011] [Accepted: 11/06/2011] [Indexed: 01/11/2023]
|
43
|
Lyons L, ELBeltagy M, Bennett G, Wigmore P. Fluoxetine counteracts the cognitive and cellular effects of 5-fluorouracil in the rat hippocampus by a mechanism of prevention rather than recovery. PLoS One 2012; 7:e30010. [PMID: 22272269 PMCID: PMC3260195 DOI: 10.1371/journal.pone.0030010] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 12/09/2011] [Indexed: 12/18/2022] Open
Abstract
5-Fluorouracil (5-FU) is a cytostatic drug associated with chemotherapy-induced cognitive impairments that many cancer patients experience after treatment. Previous work in rodents has shown that 5-FU reduces hippocampal cell proliferation, a possible mechanism for the observed cognitive impairment, and that both effects can be reversed by co-administration of the antidepressant, fluoxetine. In the present study we investigate the optimum time for administration of fluoxetine to reverse or prevent the cognitive and cellular effects of 5-FU. Male Lister-hooded rats received 5 injections of 5-FU (25 mg/kg, i.p.) over 2 weeks. Some rats were co-administered with fluoxetine (10 mg/kg/day, in drinking water) for 3 weeks before and during (preventative) or after (recovery) 5-FU treatment or both time periods (throughout). Spatial memory was tested using the novel location recognition (NLR) test and proliferation and survival of hippocampal cells was quantified using immunohistochemistry. 5-FU-treated rats showed cognitive impairment in the NLR task and a reduction in cell proliferation and survival in the subgranular zone of the dentate gyrus, compared to saline treated controls. These impairments were still seen for rats administered fluoxetine after 5-FU treatment, but were not present when fluoxetine was administered both before and during 5-FU treatment. The results demonstrate that fluoxetine is able to prevent but not reverse the cognitive and cellular effects of 5-FU. This provides information on the mechanism by which fluoxetine acts to protect against 5-FU and indicates when it would be beneficial to administer the antidepressant to cancer patients.
Collapse
Affiliation(s)
- Laura Lyons
- School of Biomedical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Maha ELBeltagy
- Department of Anatomy, Menoufiya University, Shibin el Kom, Egypt
| | - Geoffrey Bennett
- School of Biomedical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Peter Wigmore
- School of Biomedical Sciences, University of Nottingham, Nottingham, United Kingdom
- * E-mail:
| |
Collapse
|
44
|
Long JM, Lee GD, Kelley-Bell B, Spangler EL, Perez EJ, Longo DL, de Cabo R, Zou S, Rapp PR. Preserved learning and memory following 5-fluorouracil and cyclophosphamide treatment in rats. Pharmacol Biochem Behav 2011; 100:205-11. [PMID: 21875615 PMCID: PMC3183356 DOI: 10.1016/j.pbb.2011.08.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 07/26/2011] [Accepted: 08/12/2011] [Indexed: 11/25/2022]
Abstract
Some patients experience enduring cognitive impairment after cancer treatment, a condition termed "chemofog". Animal models allow assessment of chemotherapy effects on learning and memory per se, independent of changes due to cancer itself or associated health consequences such as depression. The present study examined the long-term learning and memory effects of a chemotherapy cocktail used widely in the treatment of breast cancer, consisting of 5-fluorouracil (5FU) and cyclophosphamide (CYP). Eighty 5-month old male F344 rats received contextual and cued fear conditioning before treatment with saline, or a low or high dose drug cocktail (50mg/kg CYP and 75 mg/kg 5FU, or 75 mg/kg CYP and 120 mg/kg 5FU, i.p., respectively) every 30 days for 2 months. After a 2-month, no-drug recovery, both long-term retention and new task acquisition in the water maze and 14-unit T-maze were assessed. Neither dose of the CYP/5FU cocktail impaired retrograde fear memory despite marked toxicity documented by enduring weight loss and 50% mortality at the higher dose. Acquisition in the water maze and Stone maze was also normal relative to controls in rats treated with CYP/5FU. The results contribute to a growing literature suggesting that learning and memory mediated by the hippocampus can be relatively resistant to chemotherapy. Future investigation may need to focus on assessments of processing speed, executive function and attention, and the possible interactive contribution of cancer itself and aging to the post-treatment development of cognitive impairment.
Collapse
Affiliation(s)
- Jeffrey M Long
- Laboratory of Experimental Gerontology, Intramural Research Program, National Institute on Aging, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Recent and remote spatial memory in mice treated with cytosine arabinoside. Pharmacol Biochem Behav 2011; 100:451-7. [PMID: 22020055 DOI: 10.1016/j.pbb.2011.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 09/16/2011] [Accepted: 10/06/2011] [Indexed: 11/21/2022]
Abstract
Clinical studies suggest that chemotherapy is associated with long-term cognitive impairment in some patients. A number of underlying mechanisms have been proposed, however, the etiology of chemotherapy-related cognitive dysfunction remains relatively unknown. As part of a multifaceted approach, animal models of chemotherapy-induced cognitive impairment are being developed. Thus far, the majority of animal studies have utilized a rat model, however, mice may prove particularly beneficial in studying genetic risk factors for developing chemotherapy-induced cognitive impairment. Various chemotherapy agents, including cytosine arabinoside (Ara-C), have been found to impair remote spatial memory in rats in the Morris water maze. The present study evaluated the effects of Ara-C on remote (30 d) spatial memory in mice. In addition, the possibility that time relative to chemotherapy treatment may modulate the effect of chemotherapy on spatial learning and/or recent (1 d) memory was explored. Male C57BL/6J mice received either Ara-C (275 mg/kg i.p. daily for 5 days) or saline. Spatial learning and memory was assessed using the Morris water maze. Half the mice performed a remote (30 d) memory version of the task and the other half performed a recent (1 d) memory version of the task. The experiment was designed such that the probe trial for the recent memory version occurred on the same day relative to chemotherapy treatment as the remote memory version. Despite significant toxic effects as assessed by weight loss, Ara-C treated mice performed as well as control mice during acquisition, recent memory, and remote memory portions of the task. As are some humans, C57BL/6J mice may be resistant to at least some aspects of chemotherapy induced cognitive decline.
Collapse
|
46
|
Walker EA, Foley JJ, Clark-Vetri R, Raffa RB. Effects of repeated administration of chemotherapeutic agents tamoxifen, methotrexate, and 5-fluorouracil on the acquisition and retention of a learned response in mice. Psychopharmacology (Berl) 2011; 217:539-48. [PMID: 21537942 PMCID: PMC3249240 DOI: 10.1007/s00213-011-2310-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 04/10/2011] [Indexed: 11/28/2022]
Abstract
RATIONALE A number of cancer chemotherapeutic agents have been associated with a loss of memory in breast cancer patients although little is known of the causality of this effect. OBJECTIVES To assess the potential cognitive effects of repeated exposure to chemotherapeutic agents, we administered the selective estrogen receptor modulator tamoxifen or the antimetabolite chemotherapy, methotrexate, and 5-fluorouracil, alone and in combination to mice and tested them in a learning and memory assay. METHODS Swiss-Webster male mice were injected with saline, 32 mg/kg tamoxifen, 3.2 or 32 mg/kg methotrexate, 75 mg/kg 5-fluorouracil, 3.2 or 32 mg/kg methotrexate in combination with 75 mg/kg 5-fluorouracil once per week for 3 weeks. On days 23 and 24, mice were tested for acquisition and retention of a nose-poke response in a learning procedure called autoshaping. In addition, the acute effects of tamoxifen were assessed in additional mice in a similar procedure. RESULTS The chemotherapeutic agents alone and in combination reduced body weight relative to saline treatment over the course of 4 weeks. Repeated treatment with tamoxifen produced both acquisition and retention effects relative to the saline-treated group although acute tamoxifen was without effect except at a behaviorally toxic dose. Repeated treatment with methotrexate in combination with 5-fluorouracil produced effects on retention, but the magnitude of these changes depended on the methotrexate dose. CONCLUSIONS These data demonstrate that repeated administration of tamoxifen or certain combination of methotrexate and 5-fluorouracil may produce deficits in the acquisition or retention of learned responses which suggest potential strategies for prevention or remediation might be considered in vulnerable patient populations.
Collapse
Affiliation(s)
- Ellen A. Walker
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - John J. Foley
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Rachel Clark-Vetri
- Department of Pharmacy Practice, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Robert B. Raffa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
47
|
Adriamycin-related anxiety-like behavior, brain oxidative stress and myelotoxicity in male Wistar rats. Pharmacol Biochem Behav 2011; 99:639-47. [DOI: 10.1016/j.pbb.2011.06.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 06/08/2011] [Accepted: 06/10/2011] [Indexed: 11/20/2022]
|
48
|
Fremouw T, Fessler CL, Ferguson RJ, Burguete Y. Preserved learning and memory in mice following chemotherapy: 5-Fluorouracil and doxorubicin single agent treatment, doxorubicin-cyclophosphamide combination treatment. Behav Brain Res 2011; 226:154-62. [PMID: 21930159 DOI: 10.1016/j.bbr.2011.09.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 09/02/2011] [Accepted: 09/07/2011] [Indexed: 02/07/2023]
Abstract
Clinical studies suggest that chemotherapy is associated with long-term cognitive impairment in some patients. A number of underlying mechanisms have been proposed, however, the etiology of chemotherapy-related cognitive dysfunction remains relatively unknown. As part of a multifaceted approach, animal models of chemotherapy induced cognitive impairment are being developed. Thus far, the majority of animal studies have utilized rats, however, mice may prove particularly beneficial in studying genetic risk factors for developing chemotherapy induced cognitive impairment. Thus, C57BL/6J mice were treated once a week for three weeks with saline, doxorubicin and cyclophosphamide (D&C), doxorubicin (Dox), or 5-fluorouracil (5-FU). Recent and remote contextual fear conditioning and novel object recognition (NOR) was assessed. Despite significant toxic effects as assessed by weight loss, the chemotherapy treated mice performed as well as control mice on all task. As are some humans, C57BL/6J mice may be resistant to at least some aspects of chemotherapy induced cognitive decline.
Collapse
Affiliation(s)
- Thane Fremouw
- Department of Psychology, University of Maine, Orono, ME 04469-5742, United States.
| | | | | | | |
Collapse
|
49
|
Hamed SA, Selim ZI, Elattar AM, Elserogy YM, Ahmed EA, Mohamed HO. Assessment of biocorrelates for brain involvement in female patients with rheumatoid arthritis. Clin Rheumatol 2011; 31:123-32. [PMID: 21695659 DOI: 10.1007/s10067-011-1795-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 06/01/2011] [Accepted: 06/05/2011] [Indexed: 12/22/2022]
Abstract
Central nervous system (CNS) abnormalities are rare in patients with rheumatoid arthritis (RA). Direct studies done to investigate brain involvement in RA are few or even absent. We hypothesized that CNS is not excluded from the inflammatory disease process in RA. Thus we systematically investigated markers of brain involvement in 55 females with RA. We examined patients' cognition using battery of sensitive psychometric testing [Mini-Mental State Examination, Stanford-Binet test (fourth edition) and Wechsler Memory Scale-Revised] and by recording P300 component of event-related potentials, a neurophysiological analogue. We also measured the serum levels of S100B and neuron-specific enolase (NSE), markers of glial and neuronal cells. Compared to control subjects, lower scores in cognitive testing were reported in 71% of the patients (n=39) and abnormal P300 latency and amplitude (P<0.001, 0.050). Patients had higher levels of S100B (P<0.029) and higher levels of S100B were correlated with lower total scores of cognitive functions (P<0.01), P300 latency (P<0.05), and NSE concentrations (P<0.01). However, cognitive scores did not correlate with disease activity or severity. Although depression scores were significant in patients with RA (P<0.001), but they did not correlate with cognitive scores. Seven patients had white matter hyperintensities in MRI brain suggesting vasculitis, ischemic brain lesions and dots of demyelination, and all had higher levels of S100B. Results of this study directly indicate that the disease process (inflammation and demyelination) is associated with cognitive deficits observed with RA.
Collapse
Affiliation(s)
- Sherifa A Hamed
- Department of Neurology and Psychiatry, Assiut University Hospital, P.O. Box 71516, Assiut, Egypt.
| | | | | | | | | | | |
Collapse
|
50
|
Lyons L, Elbeltagy M, Bennett G, Wigmore P. The effects of cyclophosphamide on hippocampal cell proliferation and spatial working memory in rat. PLoS One 2011; 6:e21445. [PMID: 21731752 PMCID: PMC3120875 DOI: 10.1371/journal.pone.0021445] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 05/27/2011] [Indexed: 12/25/2022] Open
Abstract
Cyclophosphamide (CP) is a chemotherapy used in combinations that are associated with cognitive impairment. In the present study male Lister-hooded rats (n = 12) were used to investigate the effects of chronic administration of CP (30mg/kg, 7 i.v. doses, or an equivalent volume of saline) on performance in the novel location recognition (NLR) task and on the proliferation and survival of hippocampal cells. The survival of hippocampal cells dividing at the beginning of treatment was significantly reduced by CP. However, no difference was seen between CP treated and control groups for the number of cells proliferating 7 days after the final injection and both groups performed equally well in the NLR task. These results indicate that the given dose of CP acutely reduces the survival of newly born hippocampal cells. However, it does not have a longer term effect on spatial working memory or hippocampal proliferation, suggesting that CP is less neurotoxic than other chemotherapies with which it is used in combination.
Collapse
Affiliation(s)
- Laura Lyons
- School of Biomedical Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom.
| | | | | | | |
Collapse
|