1
|
Meejang J, Busboom MT, Baker SE, Arif Y, Kastner O, Wilson TW, Kurz MJ. Adults with down syndrome exhibit altered somatosensory cortical inhibition. Neuroimage Clin 2025; 46:103797. [PMID: 40347551 DOI: 10.1016/j.nicl.2025.103797] [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: 12/17/2024] [Revised: 03/25/2025] [Accepted: 05/02/2025] [Indexed: 05/14/2025]
Abstract
Down syndrome (DS) is a developmental genetic disorder that is associated with an accelerated aging profile and high probability of early incidence Alzheimer's disease like symptoms. It is well established that there are morphological differences in the brains of adults with DS, but the net impact of the genetic disruption on cortical function remains poorly understood. To address this knowledge gap, we used magnetoencephalographic (MEG) brain imaging to assess the somatosensory cortical activity elicited by a paired-pulse electrical stimulation of the right median nerve of adults with DS (N = 19; Age = 28.05 ± 7.9 yrs.) and neurotypical controls (NT) (N = 21; Age = 30.81 ± 8.2 yrs.). sLORETA was used to image neural responses to the somatosensory stimulation, which were centered on the left central sulcus posterior to the motor hand knob region. Our results revealed that adults with DS had weaker somatosensory cortical activity after the second electrical stimulation in the paired-pulse paradigm (DS = 594.1 ± 194.22 AU; NT = 750.48 ± 256.6; P = 0.038) and a pronounced hyper-gating response (DS = 78.9 ± 6.8 %; NT = 87.4 ± 9.9 %; P = 0.003). Together, these results suggest that adults with DS may have an imbalance in the excitatory/inhibitory ratio. These novel data enhance our understanding of the neurophysiological aberrations associated with DS and may hold promise in understanding the origins of Alzheimer's disease like symptoms in this population. Future studies should examine whether these inhibitory alterations are restricted to the sensorimotor cortices or extend across the brain.
Collapse
Affiliation(s)
- Jiraros Meejang
- Institute for Human Neuroscience, Boys Town National Research Hospital, Omaha, NE, USA
| | - Morgan T Busboom
- Institute for Human Neuroscience, Boys Town National Research Hospital, Omaha, NE, USA
| | - Sarah E Baker
- Institute for Human Neuroscience, Boys Town National Research Hospital, Omaha, NE, USA
| | - Yasra Arif
- Institute for Human Neuroscience, Boys Town National Research Hospital, Omaha, NE, USA
| | - Olyvia Kastner
- Institute for Human Neuroscience, Boys Town National Research Hospital, Omaha, NE, USA
| | - Tony W Wilson
- Institute for Human Neuroscience, Boys Town National Research Hospital, Omaha, NE, USA; Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE, USA
| | - Max J Kurz
- Institute for Human Neuroscience, Boys Town National Research Hospital, Omaha, NE, USA; Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE, USA.
| |
Collapse
|
2
|
Piriform cortex alterations in the Ts65Dn model for down syndrome. Brain Res 2020; 1747:147031. [PMID: 32726601 DOI: 10.1016/j.brainres.2020.147031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/02/2020] [Accepted: 07/22/2020] [Indexed: 11/22/2022]
Abstract
The piriform cortex is involved in olfactory information processing, that is altered in Down Syndrome. Moreover, piriform cortex has a crucial involvement in epilepsy generation and is one of the first regions affected in Alzheimer's Disease, both maladies being prevalent among Down Syndrome individuals. In this work, we studied the alterations in neuronal morphology, synaptology and structural plasticity in the piriform cortex of the Ts65Dn mouse model, which is the most used model for the study of this syndrome and mimics some of their alterations. We have observed that Ts65Dn piriform cortex displays: a reduction in dendritic arborisation, a higher density of inhibitory synapses (GAD67), a lower density of excitatory synapses (vGLUT1) and a higher density of inhibitory postsynaptic puncta (gephyrin). Under electron microscopy the excitatory presynaptic and postsynaptic elements were larger in trisomic mice than in controls. Similar results were obtained using confocal microscopy. There were less immature neurons in piriform cortex layer II in addition to a reduction in the expression of PSA-NCAM in the neuropil that subsequently can reflect impairment in structural plasticity. These data support the idea of an impaired environment with altered ratio of inhibition and excitation that involves a reduction in plasticity and dendritic atrophy, providing a possible substrate for the olfactory processing impairment observed in DS individuals.
Collapse
|
3
|
Rueda N, Vidal V, García-Cerro S, Puente A, Campa V, Lantigua S, Narcís O, Bartesaghi R, Martínez-Cué C. Prenatal, but not Postnatal, Curcumin Administration Rescues Neuromorphological and Cognitive Alterations in Ts65Dn Down Syndrome Mice. J Nutr 2020; 150:2478-2489. [PMID: 32729926 DOI: 10.1093/jn/nxaa207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/27/2020] [Accepted: 06/26/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The cognitive dysfunction in Down syndrome (DS) is partially caused by deficient neurogenesis during fetal stages. Curcumin enhances neurogenesis and learning and memory. OBJECTIVES We aimed to test the ability of curcumin to rescue the neuromorphological and cognitive alterations of the Ts65Dn (TS) mouse model of DS when administered prenatally or during early postnatal stages, and to evaluate whether these effects were maintained several weeks after the treatment. METHODS To evaluate the effects of prenatal curcumin administration, 65 pregnant TS females were subcutaneously treated with curcumin (300 mg/kg) or vehicle from ED (Embryonic Day) 10 to PD (Postnatal Day) 2. All the analyses were performed on their TS and Control (CO) male and female progeny. At PD2, the changes in neurogenesis, cellularity, and brain weight were analyzed in 30 TS and CO pups. The long-term effects of prenatal curcumin were evaluated in another cohort of 44 TS and CO mice between PD30 and PD45. The neuromorphological effects of the early postnatal administration of curcumin were assessed on PD15 in 30 male and female TS and CO pups treated with curcumin (300 mg/kg) or vehicle from PD2 to PD15. The long-term neuromorphological and cognitive effects were assessed from PD60 to PD90 in 45 mice. Data was compared by ANOVAs. RESULTS Prenatal administration of curcumin increased the brain weight (+45%, P < 0.001), the density of BrdU (bromodeoxyuridine)- (+150%, P < 0.001) and DAPI (4',6-diamidino-2-phenylindole)- (+38%, P = 0.005) positive cells, and produced a long-term improvement of cognition in TS (+35%, P = 0.007) mice with respect to vehicle-treated mice. Postnatal administration of curcumin did not rescue any of the short- or long-term altered phenotypes of TS mice. CONCLUSION The beneficial effects of prenatal curcumin administration to TS mice suggest that it could be a therapeutic strategy to treat DS cognitive disabilities.
Collapse
Affiliation(s)
- Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Verónica Vidal
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Susana García-Cerro
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Alba Puente
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Víctor Campa
- Institute of Molecular Biology and Biomedicine, Santander, Cantabria, Spain
| | - Sara Lantigua
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Oriol Narcís
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| |
Collapse
|
4
|
Illouz T, Madar R, Biragyn A, Okun E. Restoring microglial and astroglial homeostasis using DNA immunization in a Down Syndrome mouse model. Brain Behav Immun 2019; 75:163-180. [PMID: 30389461 PMCID: PMC6358279 DOI: 10.1016/j.bbi.2018.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/22/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022] Open
Abstract
Down Syndrome (DS), the most common cause of genetic intellectual disability, is characterized by over-expression of the APP and DYRK1A genes, located on the triplicated chromosome 21. This chromosomal abnormality leads to a cognitive decline mediated by Amyloid-β (Aβ) overproduction and tau hyper-phosphorylation as early as the age of 40. In this study, we used the Ts65Dn mouse model of DS to evaluate the beneficial effect of a DNA vaccination against the Aβ1-11 fragment, in ameliorating Aβ-related neuropathology and rescue of cognitive and behavioral abilities. Anti-Aβ1-11 vaccination induced antibody production and facilitated clearance of soluble oligomers and small extracellular inclusions of Aβ from the hippocampus and cortex of Ts65Dn mice. This was correlated with reduced neurodegeneration and restoration of the homeostatic phenotype of microglial and astroglial cells. Vaccinated Ts65Dn mice performed better in spatial-learning tasks, exhibited reduced motor hyperactivity typical for this strain, and restored short-term memory abilities. Our findings support the hypothesis that DS individuals may benefit from active immunotherapy against Aβ from a young age by slowing the progression of dementia.
Collapse
Affiliation(s)
- Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Ravit Madar
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Arya Biragyn
- Laboratory of Molecular Biology and Immunology, NIA, NIH, MD 21224, USA
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel.
| |
Collapse
|
5
|
Zhao X, Bhattacharyya A. Human Models Are Needed for Studying Human Neurodevelopmental Disorders. Am J Hum Genet 2018; 103:829-857. [PMID: 30526865 DOI: 10.1016/j.ajhg.2018.10.009] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 10/09/2018] [Indexed: 12/19/2022] Open
Abstract
The analysis of animal models of neurological disease has been instrumental in furthering our understanding of neurodevelopment and brain diseases. However, animal models are limited in revealing some of the most fundamental aspects of development, genetics, pathology, and disease mechanisms that are unique to humans. These shortcomings are exaggerated in disorders that affect the brain, where the most significant differences between humans and animal models exist, and could underscore failures in targeted therapeutic interventions in affected individuals. Human pluripotent stem cells have emerged as a much-needed model system for investigating human-specific biology and disease mechanisms. However, questions remain regarding whether these cell-culture-based models are sufficient or even necessary. In this review, we summarize human-specific features of neurodevelopment and the most common neurodevelopmental disorders, present discrepancies between animal models and human diseases, demonstrate how human stem cell models can provide meaningful information, and discuss the challenges that exist in our pursuit to understand distinctively human aspects of neurodevelopment and brain disease. This information argues for a more thoughtful approach to disease modeling through consideration of the valuable features and limitations of each model system, be they human or animal, to mimic disease characteristics.
Collapse
Affiliation(s)
- Xinyu Zhao
- Waisman Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison WI 53705, USA.
| | - Anita Bhattacharyya
- Waisman Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison WI 53705, USA; Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison WI 53705, USA.
| |
Collapse
|
6
|
Giacomini A, Stagni F, Emili M, Guidi S, Salvalai ME, Grilli M, Vidal-Sanchez V, Martinez-Cué C, Bartesaghi R. Treatment with corn oil improves neurogenesis and cognitive performance in the Ts65Dn mouse model of Down syndrome. Brain Res Bull 2018; 140:378-391. [PMID: 29935232 DOI: 10.1016/j.brainresbull.2018.06.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/07/2018] [Accepted: 06/18/2018] [Indexed: 12/12/2022]
Abstract
Individuals with Down syndrome (DS), a genetic condition due to triplication of Chromosome 21, are characterized by intellectual disability that worsens with age. Since impairment of neurogenesis and dendritic maturation are very likely key determinants of intellectual disability in DS, interventions targeted to these defects may translate into a behavioral benefit. While most of the neurogenesis enhancers tested so far in DS mouse models may pose some caveats due to possible side effects, substances naturally present in the human diet may be regarded as therapeutic tools with a high translational impact. Linoleic acid and oleic acid are major constituents of corn oil that positively affect neurogenesis and neuron maturation. Based on these premises, the goal of the current study was to establish whether treatment with corn oil improves hippocampal neurogenesis and hippocampus-dependent memory in the Ts65Dn model of DS. Four-month-old Ts65Dn and euploid mice were treated with saline or corn oil for 30 days. Evaluation of behavior at the end of treatment showed that Ts65Dn mice treated with corn oil underwent a large improvement in hippocampus-dependent learning and memory. Evaluation of neurogenesis and dendritogenesis showed that in treated Ts65Dn mice the number of new granule cells of the hippocampal dentate gyrus and their dendritic pattern became similar to those of euploid mice. In addition, treated Ts65Dn mice underwent an increase in body and brain weight. This study shows for the first time that fatty acids have a positive impact on the brain of the Ts65Dn mouse model of DS. These results suggest that a diet that is rich in fatty acids may exert beneficial effects on cognitive performance in individuals with DS without causing adverse effects.
Collapse
Affiliation(s)
- Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fiorenza Stagni
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Maria Elisa Salvalai
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Veronica Vidal-Sanchez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria, Santander, Spain
| | - Carmen Martinez-Cué
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria, Santander, Spain
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
7
|
Dang T, Duan WY, Yu B, Tong DL, Cheng C, Zhang YF, Wu W, Ye K, Zhang WX, Wu M, Wu BB, An Y, Qiu ZL, Wu BL. Autism-associated Dyrk1a truncation mutants impair neuronal dendritic and spine growth and interfere with postnatal cortical development. Mol Psychiatry 2018; 23:747-758. [PMID: 28167836 PMCID: PMC5822466 DOI: 10.1038/mp.2016.253] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 10/07/2016] [Accepted: 10/17/2016] [Indexed: 11/30/2022]
Abstract
Autism is a prevailing neurodevelopmental disorder with a large genetic/genomic component. Recently, the dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 1 A (DYRK1A) gene was implicated as a risk factor for autism spectrum disorder (ASD). We identified five DYRK1A variants in ASD patients and found that the dose of DYRK1A protein has a crucial role in various aspects of postnatal neural development. Dyrk1a loss of function and gain of function led to defects in dendritic growth, dendritic spine development and radial migration during cortical development. Importantly, two autism-associated truncations, R205X and E239X, were shown to be Dyrk1a loss-of-function mutants. Studies of the truncated Dyrk1a mutants may provide new insights into the role of Dyrk1a in brain development, as well as the role of Dyrk1a loss of function in the pathophysiology of autism.
Collapse
Affiliation(s)
- T Dang
- Children’s Hospital of Fudan University and Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, China
| | - W Y Duan
- Exome Sequencing Collaboration at Boston Children’s Hospital and Institute of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - B Yu
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - D L Tong
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - C Cheng
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Y F Zhang
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - W Wu
- Exome Sequencing Collaboration at Boston Children’s Hospital and Institute of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - K Ye
- Exome Sequencing Collaboration at Boston Children’s Hospital and Institute of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - W X Zhang
- Exome Sequencing Collaboration at Boston Children’s Hospital and Institute of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - M Wu
- Children’s Hospital of Fudan University and Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, China
- Exome Sequencing Collaboration at Boston Children’s Hospital and Institute of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - B B Wu
- Children’s Hospital of Fudan University and Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, China
- Exome Sequencing Collaboration at Boston Children’s Hospital and Institute of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Y An
- Children’s Hospital of Fudan University and Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, China
- Exome Sequencing Collaboration at Boston Children’s Hospital and Institute of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Z L Qiu
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - B L Wu
- Children’s Hospital of Fudan University and Institutes of Biomedical Sciences of Shanghai Medical College, Fudan University, Shanghai, China
- Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Coskun P, Helguera P, Nemati Z, Bohannan RC, Thomas J, Samuel SE, Argueta J, Doran E, Wallace DC, Lott IT, Busciglio J. Metabolic and Growth Rate Alterations in Lymphoblastic Cell Lines Discriminate Between Down Syndrome and Alzheimer's Disease. J Alzheimers Dis 2018; 55:737-748. [PMID: 27802222 DOI: 10.3233/jad-160278] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Deficits in mitochondrial function and oxidative stress play pivotal roles in Down syndrome (DS) and Alzheimer's disease (AD) and these alterations in mitochondria occur systemically in both conditions. OBJECTIVE We hypothesized that peripheral cells of elder subjects with DS exhibit disease-specific and dementia-specific metabolic features. To test this, we performed a comprehensive analysis of energy metabolism in lymphoblastic-cell-lines (LCLs) derived from subjects belonging to four groups: DS-with-dementia (DSAD), DS-without-dementia (DS), sporadic AD, and age-matched controls. METHODS LCLs were studied under regular or minimal feeding regimes with galactose or glucose as primary carbohydrate sources. We assessed metabolism under glycolysis or oxidative phosphorylation by quantifying cell viability, oxidative stress, ATP levels, mitochondrial membrane potential (MMP), mitochondrial calcium uptake, and autophagy. RESULTS DS and DSAD LCLs showed slower growth rates under minimal feeding. DS LCLs mainly dependent on mitochondrial respiration exhibited significantly slower growth and higher levels of oxidative stress compared to other groups. While ATP levels (under mitochondrial inhibitors) and mitochondrial calcium uptake were significantly reduced in DSAD and AD cells, MMP was decreased in DS, DSAD, and AD LCLs. Finally, DS LCLs showed markedly reduced levels of the autophagy marker LC3-II, underscoring the close association between metabolic dysfunction and impaired autophagy in DS. CONCLUSION There are significant mitochondrial functional changes in LCLs derived from DS, DSAD, and AD patients. Several parameters analyzed were consistently different between DS, DSAD, and AD lines suggesting that metabolic indicators between LCL groups may be utilized as biomarkers of disease progression and/or treatment outcomes.
Collapse
Affiliation(s)
- Pinar Coskun
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| | - Pablo Helguera
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA.,Instituto de Investigación Médica Mercedes y Martin Ferreyra, Córdoba, Argentina, USA
| | - Zahra Nemati
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| | - Ryan C Bohannan
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| | - Jean Thomas
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| | - Schriner E Samuel
- Department of Pharmaceutical Science, University of California, Irvine, CA, USA
| | - Jocelyn Argueta
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| | - Eric Doran
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine (CMEM), Children's Hospital of Philadelphia, and Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ira T Lott
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Jorge Busciglio
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| |
Collapse
|
9
|
Block A, Ahmed M, Rueda N, Hernandez MC, Martinez-Cué C, Gardiner K. The GABA A α5-selective Modulator, RO4938581, Rescues Protein Anomalies in the Ts65Dn Mouse Model of Down Syndrome. Neuroscience 2018; 372:192-212. [DOI: 10.1016/j.neuroscience.2017.12.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 12/21/2022]
|
10
|
Zorrilla de San Martin J, Delabar JM, Bacci A, Potier MC. GABAergic over-inhibition, a promising hypothesis for cognitive deficits in Down syndrome. Free Radic Biol Med 2018; 114:33-39. [PMID: 28993272 DOI: 10.1016/j.freeradbiomed.2017.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/01/2017] [Accepted: 10/04/2017] [Indexed: 12/31/2022]
Abstract
Down syndrome (DS), also known as trisomy 21, is the most common genetic cause of intellectual disability. It is also a model human disease for exploring consequences of gene dosage imbalance on complex phenotypes. Learning and memory impairments linked to intellectual disabilities in DS could result from synaptic plasticity deficits and excitatory-inhibitory alterations leading to changes in neuronal circuitry in the brain of affected individuals. Increasing number of studies in mouse and cellular models converge towards the assumption that excitatory-inhibitory imbalance occurs in DS, likely early during development. Thus increased inhibition appears to be a common trend that could explain synaptic and circuit disorganization. Interestingly using several potent pharmacological tools, preclinical studies strongly demonstrated that cognitive deficits could be restored in mouse models of DS. Clinical trials have not yet provided robust data for therapeutic application and additional studies are needed. Here we review the literature and our own published work emphasizing the over-inhibition hypothesis in DS and their links with gene dosage imbalance paving the way for future basic and clinical research.
Collapse
Affiliation(s)
- Javier Zorrilla de San Martin
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Jean-Maurice Delabar
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Alberto Bacci
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Marie-Claude Potier
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.
| |
Collapse
|
11
|
Tramutola A, Pupo G, Di Domenico F, Barone E, Arena A, Lanzillotta C, Brokeaart D, Blarzino C, Head E, Butterfield DA, Perluigi M. Activation of p53 in Down Syndrome and in the Ts65Dn Mouse Brain is Associated with a Pro-Apoptotic Phenotype. J Alzheimers Dis 2017; 52:359-371. [PMID: 26967221 DOI: 10.3233/jad-151105] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Down syndrome (DS) is the most common genetic cause of intellectual disability, resulting from trisomy of chromosome 21. The main feature of DS neuropathology includes early onset of Alzheimer's disease (AD), with deposition of senile plaques and tangles. We hypothesized that apoptosis may be activated in the presence of AD neuropathology in DS, thus we measured proteins associated with upstream and downstream pathways of p53 in the frontal cortex from DS cases with and without AD pathology and from Ts65Dn mice, at different ages. We observed increased acetylation and phosphorylation of p53, coupled to reduced MDM2/p53 complex level and lower levels of SIRT1. Activation of p53 was associated with a number of targets (BAX, PARP1, caspase-3, p21, heat shock proteins, and PGC1α) that were modulated in both DS and DS/AD compared with age-matched controls. In particular, the most relevant changes (increased p-p53 and acetyl-p53 and reduced formation of MDM2/p53 complex) were found to be modified only in the presence of AD pathology in DS. In addition, a similar pattern of alterations in the p53 pathway was found in Ts65Dn mice. These results suggest that p53 may integrate different signals, which can result in a pro-apoptotic-phenotype contributing to AD neuropathology in people with DS.
Collapse
Affiliation(s)
| | - Gilda Pupo
- Department of Biochemical Sciences, Sapienza University of Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences, Sapienza University of Rome, Italy.,Universidad Autónoma de Chile, Instituto de Ciencias Biomédicas, Facultad de Salud, Providencia, Santiago, Chile
| | - Andrea Arena
- Department of Biochemical Sciences, Sapienza University of Rome, Italy
| | | | | | - Carla Blarzino
- Department of Biochemical Sciences, Sapienza University of Rome, Italy
| | - Elizabeth Head
- Sanders-Brown Center of Aging, University of Kentucky, Lexington KY, USA
| | - D Allan Butterfield
- Sanders-Brown Center of Aging, University of Kentucky, Lexington KY, USA.,Department of Chemistry, University of Kentucky, Lexington KY, USA
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Italy
| |
Collapse
|
12
|
López-Hidalgo R, Ballestín R, Vega J, Blasco-Ibáñez JM, Crespo C, Gilabert-Juan J, Nácher J, Varea E. Hypocellularity in the Murine Model for Down Syndrome Ts65Dn Is Not Affected by Adult Neurogenesis. Front Neurosci 2016; 10:75. [PMID: 26973453 PMCID: PMC4773601 DOI: 10.3389/fnins.2016.00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/17/2016] [Indexed: 01/08/2023] Open
Abstract
Down syndrome (DS) is caused by the presence of an extra copy of the chromosome 21 and it is the most common aneuploidy producing intellectual disability. Neural mechanisms underlying this alteration may include defects in the formation of neuronal networks, information processing and brain plasticity. The murine model for DS, Ts65Dn, presents reduced adult neurogenesis. This reduction has been suggested to underlie the hypocellularity of the hippocampus as well as the deficit in olfactory learning in the Ts65Dn mice. Similar alterations have also been observed in individuals with DS. To determine whether the impairment in adult neurogenesis is, in fact, responsible for the hypocellularity in the hippocampus and physiology of the olfactory bulb, we have analyzed cell proliferation and neuronal maturation in the two major adult neurogenic niches in the Ts656Dn mice: the subgranular zone (SGZ) of the hippocampus and the subventricular zone (SVZ). Additionally, we carried out a study to determine the survival rate and phenotypic fate of newly generated cells in both regions, injecting 5'BrdU and sacrificing the mice 21 days later, and analyzing the number and phenotype of the remaining 5'BrdU-positive cells. We observed a reduction in the number of proliferating (Ki67 positive) cells and immature (doublecortin positive) neurons in the subgranular and SVZ of Ts65Dn mice, but we did not observe changes in the number of surviving cells or in their phenotype. These data correlated with a lower number of apoptotic cells (cleaved caspase 3 positive) in Ts65Dn. We conclude that although adult Ts65Dn mice have a lower number of proliferating cells, it is compensated by a lower level of cell death. This higher survival rate in Ts65Dn produces a final number of mature cells similar to controls. Therefore, the reduction of adult neurogenesis cannot be held responsible for the neuronal hypocellularity in the hippocampus or for the olfactory learning deficit of Ts65Dn mice.
Collapse
Affiliation(s)
- Rosa López-Hidalgo
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Raul Ballestín
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Jessica Vega
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| | - José M. Blasco-Ibáñez
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Carlos Crespo
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| | - Javier Gilabert-Juan
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVAValència, Spain
- CIBERSAM, Spanish National Network for Research in Mental HealthValència, Spain
- Genetics Department, CIBERSAM, Universitat de ValènciaValència, Spain
| | - Juan Nácher
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVAValència, Spain
- CIBERSAM, Spanish National Network for Research in Mental HealthValència, Spain
- Genetics Department, CIBERSAM, Universitat de ValènciaValència, Spain
| | - Emilio Varea
- Neurobiology Unit and Program in Basic and Applied Neurosciences, Cell Biology Department, Universitat de ValènciaValència, Spain
- Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (BIOTECMED), Universitat de ValènciaValència, Spain
| |
Collapse
|
13
|
Deidda G, Bozarth IF, Cancedda L. Modulation of GABAergic transmission in development and neurodevelopmental disorders: investigating physiology and pathology to gain therapeutic perspectives. Front Cell Neurosci 2014; 8:119. [PMID: 24904277 PMCID: PMC4033255 DOI: 10.3389/fncel.2014.00119] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/14/2014] [Indexed: 01/30/2023] Open
Abstract
During mammalian ontogenesis, the neurotransmitter GABA is a fundamental regulator of neuronal networks. In neuronal development, GABAergic signaling regulates neural proliferation, migration, differentiation, and neuronal-network wiring. In the adult, GABA orchestrates the activity of different neuronal cell-types largely interconnected, by powerfully modulating synaptic activity. GABA exerts these functions by binding to chloride-permeable ionotropic GABAA receptors and metabotropic GABAB receptors. According to its functional importance during development, GABA is implicated in a number of neurodevelopmental disorders such as autism, Fragile X, Rett syndrome, Down syndrome, schizophrenia, Tourette's syndrome and neurofibromatosis. The strength and polarity of GABAergic transmission is continuously modulated during physiological, but also pathological conditions. For GABAergic transmission through GABAA receptors, strength regulation is achieved by different mechanisms such as modulation of GABAA receptors themselves, variation of intracellular chloride concentration, and alteration in GABA metabolism. In the never-ending effort to find possible treatments for GABA-related neurological diseases, of great importance would be modulating GABAergic transmission in a safe and possibly physiological way, without the dangers of either silencing network activity or causing epileptic seizures. In this review, we will discuss the different ways to modulate GABAergic transmission normally at work both during physiological and pathological conditions. Our aim is to highlight new research perspectives for therapeutic treatments that reinstate natural and physiological brain functions in neuro-pathological conditions.
Collapse
Affiliation(s)
- Gabriele Deidda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genova, Italy
| | - Ignacio F Bozarth
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genova, Italy
| | - Laura Cancedda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genova, Italy
| |
Collapse
|
14
|
Edgin JO, Spanò G, Kawa K, Nadel L. Remembering things without context: development matters. Child Dev 2014; 85:1491-502. [PMID: 24597709 DOI: 10.1111/cdev.12232] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Spatial context supports memory retrieval in adults. To understand the development of these effects, context effects on object recognition were tested in neurotypical children ages 3 years to adulthood (n 3-6 years = 34, n 10-16 years = 32, n college age = 22) and individuals with Down syndrome (DS) ages 10-29 years (n = 21). Participants engaged in an object recognition task; objects were presented in scenes and either remained in that same scene or were removed at test. In some groups (< 4.5 years and with DS) context effects were present even though object recognition was poor. After 4.5 years, children demonstrated memory flexibility, while later in adolescence context effects reemerged, showing nonlinearity in the development of these effects.
Collapse
|
15
|
Martínez-Cué C, Delatour B, Potier MC. Treating enhanced GABAergic inhibition in Down syndrome: use of GABA α5-selective inverse agonists. Neurosci Biobehav Rev 2014; 46 Pt 2:218-27. [PMID: 24412222 DOI: 10.1016/j.neubiorev.2013.12.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/27/2013] [Accepted: 12/16/2013] [Indexed: 11/27/2022]
Abstract
Excess inhibition in the brain of individuals carrying an extra copy of chromosome 21 could be responsible for cognitive deficits observed throughout their lives. A change in the excitatory/inhibitory balance in adulthood would alter synaptic plasticity, potentially triggering learning and memory deficits. γ-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the mature central nervous system and binds to GABAA receptors, opens a chloride channel, and reduces neuronal excitability. In this review we discuss methods to alleviate neuronal inhibition in a mouse model of Down syndrome, the Ts65Dn mouse, using either an antagonist (pentylenetetrazol) or two different inverse agonists selective for the α5-subunit containing receptor. Both inverse agonists, which reduce inhibitory GABAergic transmission, could rescue learning and memory deficits in Ts65Dn mice. We also discuss safety issues since modulation of the excitatory-inhibitory balance to improve cognition without inducing seizures remains particularly difficult when using GABA antagonists.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Benoît Delatour
- Institut du Cerveau et de Moelle Epinière, CNRS UMR7225, INSERM U1127, UPMC, IHUA-ICM, Hôpital Pitié-Salpêtrière, Paris, France
| | - Marie-Claude Potier
- Institut du Cerveau et de Moelle Epinière, CNRS UMR7225, INSERM U1127, UPMC, IHUA-ICM, Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
16
|
Corrales A, Vidal R, García S, Vidal V, Martínez P, García E, Flórez J, Sanchez-Barceló EJ, Martínez-Cué C, Rueda N. Chronic melatonin treatment rescues electrophysiological and neuromorphological deficits in a mouse model of Down syndrome. J Pineal Res 2014; 56:51-61. [PMID: 24147912 DOI: 10.1111/jpi.12097] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 09/20/2013] [Indexed: 12/22/2022]
Abstract
The Ts65Dn mouse (TS), the most commonly used model of Down syndrome (DS), exhibits several key phenotypic characteristics of this condition. In particular, these animals present hypocellularity in different areas of their CNS due to impaired neurogenesis and have alterations in synaptic plasticity that compromise their cognitive performance. In addition, increases in oxidative stress during adulthood contribute to the age-related progression of cognitive and neuronal deterioration. We have previously demonstrated that chronic melatonin treatment improves learning and memory and reduces cholinergic neurodegeneration in TS mice. However, the molecular and physiological mechanisms that mediate these beneficial cognitive effects are not yet fully understood. In this study, we analyzed the effects of chronic melatonin treatment on different mechanisms that have been proposed to underlie the cognitive impairments observed in TS mice: reduced neurogenesis, altered synaptic plasticity, enhanced synaptic inhibition and oxidative damage. Chronic melatonin treatment rescued both impaired adult neurogenesis and the decreased density of hippocampal granule cells in trisomic mice. In addition, melatonin administration reduced synaptic inhibition in TS mice by increasing the density and/or activity of glutamatergic synapses in the hippocampus. These effects were accompanied by a full recovery of hippocampal LTP in trisomic animals. Finally, melatonin treatment decreased the levels of lipid peroxidation in the hippocampus of TS mice. These results indicate that the cognitive-enhancing effects of melatonin in adult TS mice could be mediated by the normalization of their electrophysiological and neuromorphological abnormalities and suggest that melatonin represents an effective treatment in retarding the progression of DS neuropathology.
Collapse
Affiliation(s)
- Andrea Corrales
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria, Santander, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Reducing GABAA α5 receptor-mediated inhibition rescues functional and neuromorphological deficits in a mouse model of down syndrome. J Neurosci 2013; 33:3953-66. [PMID: 23447605 DOI: 10.1523/jneurosci.1203-12.2013] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Down syndrome (DS) is associated with neurological complications, including cognitive deficits that lead to impairment in intellectual functioning. Increased GABA-mediated inhibition has been proposed as a mechanism underlying deficient cognition in the Ts65Dn (TS) mouse model of DS. We show that chronic treatment of these mice with RO4938581 (3-bromo-10-(difluoromethyl)-9H-benzo[f]imidazo[1,5-a][1,2,4]triazolo[1,5-d][1,4]diazepine), a selective GABA(A) α5 negative allosteric modulator (NAM), rescued their deficits in spatial learning and memory, hippocampal synaptic plasticity, and adult neurogenesis. We also show that RO4938581 normalized the high density of GABAergic synapse markers in the molecular layer of the hippocampus of TS mice. In addition, RO4938581 treatment suppressed the hyperactivity observed in TS mice without inducing anxiety or altering their motor abilities. These data demonstrate that reducing GABAergic inhibition with RO4938581 can reverse functional and neuromorphological deficits of TS mice by facilitating brain plasticity and support the potential therapeutic use of selective GABA(A) α5 NAMs to treat cognitive dysfunction in DS.
Collapse
|
18
|
Functional implications of hippocampal adult neurogenesis in intellectual disabilities. Amino Acids 2013; 45:113-31. [DOI: 10.1007/s00726-013-1489-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 03/15/2013] [Indexed: 12/19/2022]
|
19
|
Modulating cognitive deficits and tau accumulation in a mouse model of aging Down syndrome through neonatal implantation of neural progenitor cells. Exp Gerontol 2012; 47:723-33. [DOI: 10.1016/j.exger.2012.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 06/27/2012] [Accepted: 06/28/2012] [Indexed: 01/04/2023]
|
20
|
Molecular and cellular alterations in Down syndrome: toward the identification of targets for therapeutics. Neural Plast 2012; 2012:171639. [PMID: 22848846 PMCID: PMC3403492 DOI: 10.1155/2012/171639] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/18/2012] [Accepted: 04/19/2012] [Indexed: 12/25/2022] Open
Abstract
Down syndrome is a complex disease that has challenged molecular and cellular research for more than 50 years. Understanding the molecular bases of morphological, cellular, and functional alterations resulting from the presence of an additional complete chromosome 21 would aid in targeting specific genes and pathways for rescuing some phenotypes. Recently, progress has been made by characterization of brain alterations in mouse models of Down syndrome. This review will highlight the main molecular and cellular findings recently described for these models, particularly with respect to their relationship to Down syndrome phenotypes.
Collapse
|
21
|
Mouse models of Down syndrome as a tool to unravel the causes of mental disabilities. Neural Plast 2012; 2012:584071. [PMID: 22685678 PMCID: PMC3364589 DOI: 10.1155/2012/584071] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 03/23/2012] [Accepted: 03/24/2012] [Indexed: 12/16/2022] Open
Abstract
Down syndrome (DS) is the most common genetic cause of mental disability. Based on the homology of Hsa21 and the murine chromosomes Mmu16, Mmu17 and Mmu10, several mouse models of DS have been developed. The most commonly used model, the Ts65Dn mouse, has been widely used to investigate the neural mechanisms underlying the mental disabilities seen in DS individuals. A wide array of neuromorphological alterations appears to compromise cognitive performance in trisomic mice. Enhanced inhibition due to alterations in GABA(A)-mediated transmission and disturbances in the glutamatergic, noradrenergic and cholinergic systems, among others, has also been demonstrated. DS cognitive dysfunction caused by neurodevelopmental alterations is worsened in later life stages by neurodegenerative processes. A number of pharmacological therapies have been shown to partially restore morphological anomalies concomitantly with cognition in these mice. In conclusion, the use of mouse models is enormously effective in the study of the neurobiological substrates of mental disabilities in DS and in the testing of therapies that rescue these alterations. These studies provide the basis for developing clinical trials in DS individuals and sustain the hope that some of these drugs will be useful in rescuing mental disabilities in DS individuals.
Collapse
|
22
|
Mazur-Kolecka B, Golabek A, Kida E, Rabe A, Hwang YW, Adayev T, Wegiel J, Flory M, Kaczmarski W, Marchi E, Frackowiak J. Effect of DYRK1A activity inhibition on development of neuronal progenitors isolated from Ts65Dn mice. J Neurosci Res 2012; 90:999-1010. [PMID: 22252917 DOI: 10.1002/jnr.23007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 11/14/2011] [Accepted: 11/17/2011] [Indexed: 01/09/2023]
Abstract
Overexpression of dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 1A (DYRK1A), encoded by a gene located in the Down syndrome (DS) critical region, is considered a major contributor to developmental abnormalities in DS. DYRK1A regulates numerous genes involved in neuronal commitment, differentiation, maturation, and apoptosis. Because alterations of neurogenesis could lead to impaired brain development and mental retardation in individuals with DS, pharmacological normalization of DYRK1A activity has been postulated as DS therapy. We tested the effect of harmine, a specific DYRK1A inhibitor, on the development of neuronal progenitor cells (NPCs) isolated from the periventricular zone of newborn mice with segmental trisomy 16 (Ts65Dn mice), a mouse model for DS that overexpresses Dyrk1A by 1.5-fold. Trisomy did not affect the ability of NPCs to expand in culture. Twenty-four hours after stimulation of migration and neuronal differentiation, NPCs showed increased expression of Dyrk1A, particularly in the trisomic cultures. After 7 days, NPCs developed into a heterogeneous population of differentiating neurons and astrocytes that expressed Dyrk1A in the nuclei. In comparison with disomic cells, NPCs with trisomy showed premature neuronal differentiation and enhanced γ-aminobutyric acid (GABA)-ergic differentiation, but astrocyte development was unchanged. Harmine prevented premature neuronal maturation of trisomic NPCs but not acceleration of GABA-ergic development. In control NPCs, harmine treatment caused altered neuronal development of NPCs, similar to that in trisomic NPCs with Dyrk1A overexpression. This study suggests that pharmacological normalization of DYRK1A activity may have a potential role in DS therapy.
Collapse
Affiliation(s)
- Bozena Mazur-Kolecka
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Rachubinski AL, Crowley SK, Sladek JR, Maclean KN, Bjugstad KB. Effects of neonatal neural progenitor cell implantation on adult neuroanatomy and cognition in the Ts65Dn model of Down syndrome. PLoS One 2012; 7:e36082. [PMID: 22558337 PMCID: PMC3338504 DOI: 10.1371/journal.pone.0036082] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 03/26/2012] [Indexed: 12/13/2022] Open
Abstract
As much of the aberrant neural development in Down syndrome (DS) occurs postnatally, an early opportunity exists to intervene and influence life-long cognitive development. Recent success using neural progenitor cells (NPC) in models of adult neurodegeneration indicate such therapy may be a viable option in diseases such as DS. Murine NPC (mNPC, C17.2 cell line) or saline were implanted bilaterally into the dorsal hippocampus of postnatal day 2 (PND 2) Ts65Dn pups to explore the feasibility of early postnatal treatment in this mouse model of DS. Disomic littermates provided karyotype controls for trisomic pups. Pups were monitored for developmental milestone achievement, and then underwent adult behavior testing at 14 weeks of age. We found that implanted mNPC survived into adulthood and migrated beyond the implant site in both karyotypes. The implantation of mNPC resulted in a significant increase in the density of dentate granule cells. However, mNPC implantation did not elicit cognitive changes in trisomic mice either neonatally or in adulthood. To the best of our knowledge, these results constitute the first assessment of mNPC as an early intervention on cognitive ability in a DS model.
Collapse
Affiliation(s)
- Angela L. Rachubinski
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Shannon K. Crowley
- Departments of Exercise Science, and Neuropsychiatry and Behavioral Science, University of South Carolina, Columbia, South Carolina, United States of America
| | - John R. Sladek
- Department of Neurology and Center for Neuroscience, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Kenneth N. Maclean
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Colorado Intellectual and Developmental Disabilities Research Center (IDDRC), University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Kimberly B. Bjugstad
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Colorado Intellectual and Developmental Disabilities Research Center (IDDRC), University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| |
Collapse
|
24
|
Martin KR, Corlett A, Dubach D, Mustafa T, Coleman HA, Parkington HC, Merson TD, Bourne JA, Porta S, Arbonés ML, Finkelstein DI, Pritchard MA. Over-expression of RCAN1 causes Down syndrome-like hippocampal deficits that alter learning and memory. Hum Mol Genet 2012; 21:3025-41. [PMID: 22511596 DOI: 10.1093/hmg/dds134] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
People with Down syndrome (DS) exhibit abnormal brain structure. Alterations affecting neurotransmission and signalling pathways that govern brain function are also evident. A large number of genes are simultaneously expressed at abnormal levels in DS; therefore, it is a challenge to determine which gene(s) contribute to specific abnormalities, and then identify the key molecular pathways involved. We generated RCAN1-TG mice to study the consequences of RCAN1 over-expression and investigate the contribution of RCAN1 to the brain phenotype of DS. RCAN1-TG mice exhibit structural brain abnormalities in those areas affected in DS. The volume and number of neurons within the hippocampus is reduced and this correlates with a defect in adult neurogenesis. The density of dendritic spines on RCAN1-TG hippocampal pyramidal neurons is also reduced. Deficits in hippocampal-dependent learning and short- and long-term memory are accompanied by a failure to maintain long-term potentiation (LTP) in hippocampal slices. In response to LTP induction, we observed diminished calcium transients and decreased phosphorylation of CaMKII and ERK1/2-proteins that are essential for the maintenance of LTP and formation of memory. Our data strongly suggest that RCAN1 plays an important role in normal brain development and function and its up-regulation likely contributes to the neural deficits associated with DS.
Collapse
Affiliation(s)
- Katherine R Martin
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3168 Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Gotti S, Caricati E, Panzica G. Alterations of brain circuits in Down syndrome murine models. J Chem Neuroanat 2011; 42:317-26. [DOI: 10.1016/j.jchemneu.2011.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 09/04/2011] [Accepted: 09/06/2011] [Indexed: 10/17/2022]
|
26
|
Kida E, Walus M, Jarząbek K, Palminiello S, Albertini G, Rabe A, Hwang YW, Golabek AA. Form of dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 1A nonphosphorylated at tyrosine 145 and 147 is enriched in the nuclei of astroglial cells, adult hippocampal progenitors, and some cholinergic axon terminals. Neuroscience 2011; 195:112-27. [PMID: 21878370 DOI: 10.1016/j.neuroscience.2011.08.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 08/10/2011] [Accepted: 08/12/2011] [Indexed: 01/01/2023]
Abstract
Compelling lines of evidence indicate that overexpression of dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A (DYRK1A) in subjects with trisomy 21 (Down syndrome[DS]) contributes to the abnormal structure and function of the DS brain. In the present study, we used a novel, phospho-dependent antibody recognizing DYRK1A only with nonphosphorylated tyrosine 145 and 147 (DYRK1A Tyr-145/147P(-)), to investigate the expression pattern of this DYRK1A species in trisomic and disomic human and mouse brains. Immunoblotting and dephosphorylation experiments demonstrated higher levels of DYRK1A Tyr-145/147P(-) in postnatal trisomic brains in comparison with controls (by ∼40%) than those of the DYRK1A visualized by three other N- and C-terminally directed antibodies to DYRK1A. By immunofluorescence, the immunoreactivity to DYRK1A Tyr-145/147P(-) was the strongest in the nuclei of astroglial cells, which contrasted with the predominantly neuronal localization of DYRK1A visualized by the three other antibodies to DYRK1A we used. In addition, DYRK1A Tyr-145/147P(-) was enriched in the nuclei of neuronal progenitors and newly born neurons in the adult hippocampal proliferative zone and also occurred in some cholinergic axonal terminals. Our data show a distinctive expression pattern of DYRK1A forms nonphosphorylated at Tyr-145 and Tyr-147 in the brain tissue and suggest that DS subjects may exhibit not only upregulation of total DYRK1A, but also more subtle differences in phosphorylation levels of this kinase in comparison with control individuals.
Collapse
Affiliation(s)
- E Kida
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Rueda N, Flórez J, Martínez-Cué C. The Ts65Dn mouse model of Down syndrome shows reduced expression of the Bcl-X(L) antiapoptotic protein in the hippocampus not accompanied by changes in molecular or cellular markers of cell death. Int J Dev Neurosci 2011; 29:711-6. [PMID: 21684326 DOI: 10.1016/j.ijdevneu.2011.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 05/20/2011] [Accepted: 06/02/2011] [Indexed: 11/19/2022] Open
Abstract
The Ts65Dn (TS) mouse, the most widely used model of Down syndrome (DS), has a partial trisomy of a segment of chromosome 16 that is homologous to the distal part of human chromosome 21. This mouse shares many phenotypic characteristics with people with DS including neuromorphological, neurochemical, and cognitive disturbances. Both TS and DS brains show earlier aging and neurodegeneration. Since fibroblast cultures from TS mice and human DS hippocampal regions show increased apoptotic cell death it has been suggested that alterations in cerebral apoptosis might be implicated in the cognitive deficits found in TS mice and in people with DS. In the present study we have evaluated brain expression levels of several proapoptotic and antiapoptotic proteins from the mitochondrial (Bcl-2, Bcl-X(L), Bax and Bad) and the extrinsic (Fas-R and Fas-L) apoptotic pathways as well as the final executioner caspase-3, in the cortex and hippocampus of TS mice. No significant alterations in the expression levels of the proapoptotic Bad and Bax or the antiapoptotic Bcl-2 proteins in the cortex or hippocampus were found in TS mice. However, TS mice showed downregulation of Bcl-X(L) in the hippocampus. In the extrinsic pathway we found unchanged levels of Fas-L in both structures and also in the expression levels of Fas-R in the hippocampus. Although Bcl-X(L) downregulation suggests that the hippocampus of TS mice is less protected against programmed cell death, we did not find any evidence for increased apoptosis in TS mice since neither TUNEL-positive cells nor active caspase-3 expression were found in cortex or hippocampus of TS or CO mice.
Collapse
Affiliation(s)
- Noemí Rueda
- Department of Physiology and Pharmacology, University of Cantabria, Spain
| | | | | |
Collapse
|
28
|
Guidi S, Ciani E, Bonasoni P, Santini D, Bartesaghi R. Widespread proliferation impairment and hypocellularity in the cerebellum of fetuses with down syndrome. Brain Pathol 2010; 21:361-73. [PMID: 21040072 DOI: 10.1111/j.1750-3639.2010.00459.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Evidence in mouse models for Down syndrome (DS) and human fetuses with DS clearly shows severe neurogenesis impairment in various telencephalic regions, suggesting that this defect may underlie the cognitive abnormalities of DS. As cerebellar hypotrophy and motor disturbances are part of the clinical features of DS, the goal of our study was to establish whether these defects may be related to neurogenesis impairment during cerebellar development. We found that in fetuses with DS (17-21 weeks of gestation) the cerebellum had an immature pattern, a reduced volume and notably fewer cells (-25%/-50%) in all cerebellar layers. Immunohistochemistry for Ki-67, a marker of cycling cells, showed impaired proliferation (-17%/-50%) of precursors from both cerebellar neurogenic regions (external granular layer and ventricular zone). No differences in apoptotic cell death were found in DS vs. control fetuses. The current study provides novel evidence that in the cerebellum of DS fetuses there is a generalized hypocellularity and that this defect is due to proliferation impairment, rather than to an increased cell death. The reduced proliferation potency found in the DS fetal cerebellum, in conjunction with previous evidence, strengthens the idea that the trisomic brain is characterized by widespread neurogenesis disruption.
Collapse
Affiliation(s)
- Sandra Guidi
- Dipartimento di Fisiologia Umana e Generale, Università di Bologna, Bologna, Italy
| | | | | | | | | |
Collapse
|
29
|
Llorens-Martín MV, Rueda N, Tejeda GS, Flórez J, Trejo JL, Martínez-Cué C. Effects of voluntary physical exercise on adult hippocampal neurogenesis and behavior of Ts65Dn mice, a model of Down syndrome. Neuroscience 2010; 171:1228-40. [PMID: 20875841 DOI: 10.1016/j.neuroscience.2010.09.043] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 08/18/2010] [Accepted: 09/22/2010] [Indexed: 11/15/2022]
Abstract
The Ts65Dn (TS) mouse is the most widely used model of Down syndrome (DS). This mouse shares many phenotypic characteristics with the human condition including cognitive and neuromorphological alterations. In this study the effects of physical exercise on hippocampal neurogenesis and behavior in TS mice were assessed. 10-12 month-old male TS and control (CO) mice were submitted to voluntary physical exercise for 7 weeks and the effects of this protocol on hippocampal morphology, neurogenesis and apoptosis were evaluated. Physical exercise improved performance in the acquisition sessions of the Morris water maze in TS but not in CO mice. Conversely, it did not have any effect on anxiety or depressive behavior in TS mice but it did reduce the cognitive components of anxiety in CO mice. TS mice presented a reduced dentate gyrus (DG) volume, subgranular zone area and number of granule neurons. Hippocampal neurogenesis was reduced in TS mice as shown by the reduced number of 5-bromo-2-deoxyuridine (BrdU) positive cells. Voluntary physical exercise did not rescue these alterations in TS mice but it did increase the number of doublecortin (DCX)-and phospho histone 3 (PH3)-positive neurons in CO mice. It is concluded that physical exercise produced a modest anxiolytic effect in CO mice and that this was accompanied by an increased number of immature cells in the hippocampal DG. On the other hand, voluntary physical exercise exerted a positive effect on TS mice learning of the platform position in the Morris water maze that seems to be mediated by a neurogenesis-independent mechanism.
Collapse
Affiliation(s)
- M V Llorens-Martín
- Cajal Institute, Consejo Superior de Investigaciones Científicas, Av. Doctor Arce, 37. 28002, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
30
|
Hewitt CA, Ling KH, Merson TD, Simpson KM, Ritchie ME, King SL, Pritchard MA, Smyth GK, Thomas T, Scott HS, Voss AK. Gene network disruptions and neurogenesis defects in the adult Ts1Cje mouse model of Down syndrome. PLoS One 2010; 5:e11561. [PMID: 20661276 PMCID: PMC2905390 DOI: 10.1371/journal.pone.0011561] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2010] [Accepted: 05/31/2010] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Down syndrome (DS) individuals suffer mental retardation with further cognitive decline and early onset Alzheimer's disease. METHODOLOGY/PRINCIPAL FINDINGS To understand how trisomy 21 causes these neurological abnormalities we investigated changes in gene expression networks combined with a systematic cell lineage analysis of adult neurogenesis using the Ts1Cje mouse model of DS. We demonstrated down regulation of a number of key genes involved in proliferation and cell cycle progression including Mcm7, Brca2, Prim1, Cenpo and Aurka in trisomic neurospheres. We found that trisomy did not affect the number of adult neural stem cells but resulted in reduced numbers of neural progenitors and neuroblasts. Analysis of differentiating adult Ts1Cje neural progenitors showed a severe reduction in numbers of neurons produced with a tendency for less elaborate neurites, whilst the numbers of astrocytes was increased. CONCLUSIONS/SIGNIFICANCE We have shown that trisomy affects a number of elements of adult neurogenesis likely to result in a progressive pathogenesis and consequently providing the potential for the development of therapies to slow progression of, or even ameliorate the neuronal deficits suffered by DS individuals.
Collapse
Affiliation(s)
- Chelsee A. Hewitt
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Pathology, The Peter MacCallum Cancer Centre, Melbourne, Australia
| | - King-Hwa Ling
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Molecular Pathology, The Centre for Cancer Biology, The Institute of Medical and Veterinary Science and The Hanson Institute, SA Pathology, and The Adelaide Cancer Research Institute, School of Medicine, University of Adelaide, Adelaide, Australia
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Tobias D. Merson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Ken M. Simpson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Matthew E. Ritchie
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Sarah L. King
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Melanie A. Pritchard
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Gordon K. Smyth
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Tim Thomas
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Hamish S. Scott
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Molecular Pathology, The Centre for Cancer Biology, The Institute of Medical and Veterinary Science and The Hanson Institute, SA Pathology, and The Adelaide Cancer Research Institute, School of Medicine, University of Adelaide, Adelaide, Australia
| | - Anne K. Voss
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
31
|
Gardiner KJ. Molecular basis of pharmacotherapies for cognition in Down syndrome. Trends Pharmacol Sci 2010; 31:66-73. [PMID: 19963286 PMCID: PMC2815198 DOI: 10.1016/j.tips.2009.10.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 10/27/2009] [Accepted: 10/30/2009] [Indexed: 12/18/2022]
Abstract
Intellectual disability in Down syndrome (DS) ranges from low normal to severely impaired and has a significant impact on the quality-of-life of the individuals affected and their families. Because the incidence of DS remains at approximately 1 in 700 live births and the lifespan is now >50 years, development of pharmacotherapies for cognitive deficits is an important goal. DS is due to an extra copy of human chromosome 21 and has often been considered too complex a genetic abnormality to be amenable to intervention. However, recent successes in rescuing learning/memory impairments in a mouse model of DS suggest that this negative outlook may not be justified. In this contribution, we first review the DS phenotype, chromosome 21 gene content and mouse models. We then discuss recent successes and the remaining challenges in the identification of targets for and preclinical evaluation of potential therapeutics.
Collapse
Affiliation(s)
- Katheleen J Gardiner
- Department of Pediatrics, Intellectual and Developmental Disability Research Center, Human Medical Genetics and Neuroscience Programs, University of Colorado Denver, 12800 E 19(th) Avenue, Aurora, Colorado 80045, USA.
| |
Collapse
|
32
|
Communication breaks-Down: from neurodevelopment defects to cognitive disabilities in Down syndrome. Prog Neurobiol 2010; 91:1-22. [PMID: 20097253 DOI: 10.1016/j.pneurobio.2010.01.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 12/10/2009] [Accepted: 01/14/2010] [Indexed: 12/31/2022]
Abstract
Down syndrome (DS) is the leading cause of genetically-defined intellectual disability and congenital birth defects. Despite being one of the first genetic diseases identified, only recently, thanks to the phenotypic analysis of DS mouse genetic models, we have begun to understand how trisomy may impact cognitive function. Cognitive disabilities in DS appear to result mainly from two pathological processes: neurogenesis impairment and Alzheimer-like degeneration. In DS brain, suboptimal network architecture and altered synaptic communication arising from neurodevelopmental impairment are key determinants of cognitive defects. Hypocellularity and hypoplasia start at early developmental stages and likely depend upon impaired proliferation of neuronal precursors, resulting in reduction of numbers of neurons and synaptic contacts. The impairment of neuronal precursor proliferation extends to adult neurogenesis and may affect learning and memory. Neurodegenerative mechanisms also contribute to DS cognitive impairment. Early onset Alzheimer disease occurs with extremely high incidence in DS patients and is causally-related to overexpression of beta-amyloid precursor protein (betaAPP), which is one of the triplicated genes in DS. In this review, we will survey the available findings on neurodevelopmental and neurodegenerative changes occurring in DS throughout life. Moreover, we will discuss the potential mechanisms by which defects in neurogenesis and neurodegenerative processes lead to altered formation of neural circuits and impair cognitive function, in connection with findings on pharmacological treatments of potential benefit for DS.
Collapse
|
33
|
Baroncelli L, Braschi C, Spolidoro M, Begenisic T, Sale A, Maffei L. Nurturing brain plasticity: impact of environmental enrichment. Cell Death Differ 2009; 17:1092-103. [PMID: 20019745 DOI: 10.1038/cdd.2009.193] [Citation(s) in RCA: 200] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Environmental enrichment (EE) is known to profoundly affect the central nervous system (CNS) at the functional, anatomical and molecular level, both during the critical period and during adulthood. Recent studies focusing on the visual system have shown that these effects are associated with the recruitment of previously unsuspected neural plasticity processes. At early stages of brain development, EE triggers a marked acceleration in the maturation of the visual system, with maternal behaviour acting as a fundamental mediator of the enriched experience in both the foetus and the newborn. In adult brain, EE enhances plasticity in the cerebral cortex, allowing the recovery of visual functions in amblyopic animals. The molecular substrate of the effects of EE on brain plasticity is multi-factorial, with reduced intracerebral inhibition, enhanced neurotrophin expression and epigenetic changes at the level of chromatin structure. These findings shed new light on the potential of EE as a non-invasive strategy to ameliorate deficits in the development of the CNS and to treat neurological disorders.
Collapse
Affiliation(s)
- L Baroncelli
- Laboratory of Neurobiology, Scuola Normale Superiore, Pisa I-56100, Italy.
| | | | | | | | | | | |
Collapse
|
34
|
Moldrich RX, Dauphinot L, Laffaire J, Vitalis T, Hérault Y, Beart PM, Rossier J, Vivien D, Gehrig C, Antonarakis SE, Lyle R, Potier MC. Proliferation deficits and gene expression dysregulation in Down's syndrome (Ts1Cje) neural progenitor cells cultured from neurospheres. J Neurosci Res 2009; 87:3143-52. [PMID: 19472221 DOI: 10.1002/jnr.22131] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Down's syndrome neurophenotypes are characterized by mental retardation and a decreased brain volume. To identify whether deficits in proliferation could be responsible for this phenotype, neural progenitor cells were isolated from the developing E14 neocortex of Down's syndrome partial trisomy Ts1Cje mice and euploid (WT) littermates and grown as neurospheres. Ts1Cje neural progenitors proliferated at a slower rate, because of a longer cell cycle, and a greater number of cells were positive for glial fibrillary acidic protein. An increase in cell death was also noted. Gene expression profiles of neural progenitor cells from Ts1Cje and WT showed that 54% of triploid genes had expression ratios (Ts1Cje/WT) significantly greater than the expected diploid gene ratio of 1.0. Some diploid genes associated with proliferation, differentiation, and glial function were dysregulated. Interestingly, proliferation and gene expression dysregulation detected in the Ts1Cje mice did not require overexpression of the chromosome 21 genes amyloid precursor protein (App) and soluble superoxide dismutase 1 (Sod1).
Collapse
Affiliation(s)
- Randal X Moldrich
- Laboratoire de Neurobiologie et Diversité Cellulaire, CNRS UMR7637, ESPCI, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Iwamoto T, Yamada A, Yuasa K, Fukumoto E, Nakamura T, Fujiwara T, Fukumoto S. Influences of interferon-gamma on cell proliferation and interleukin-6 production in Down syndrome derived fibroblasts. Arch Oral Biol 2009; 54:963-9. [PMID: 19700144 DOI: 10.1016/j.archoralbio.2009.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Accepted: 07/24/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Down syndrome, a frequently encountered genetic disorder, is usually associated with medical problems related to infectious disease, such as periodontal diseases and prolonged wound healing. Although affected individuals are considered to have clinical problems related to high interferon (IFN) sensitivity, the molecular mechanisms of IFN activities are not completely understood. DESIGN Down syndrome derived fibroblasts, Detroit 539 (D1) and Hs 52.Sk (D2) cells, were used. To analyse the expressions of interferon (IFN) receptors and downstream of IFN-gamma, western blotting was performed. Cell proliferation was determined by counting cells following trypan blue staining. Media levels of IL-1beta, TNF-alpha, and IL-6 were quantified using ELISA. RESULTS IFN-gamma receptor 2 and IFN-alpha receptor 1, but not IFN-gamma receptor 1, were highly expressed in D1 and D2 cells, as compared to the control fibroblast cells. Cell proliferation by D1 and D2 cells was lower than that by the control fibroblasts, further, IFN-gamma had a greater effect to inhibit cell proliferation by D1 and D2 cells. In addition, IFN-gamma treatment increased the phosphorylation of STAT1 and MAPK in D1 cells as compared to normal fibroblasts. Also, the presence of exogenous IFN-gamma in the growth medium significantly induced IL-6, but not IL-1beta or TNF-alpha, in D1 and D2 cells. CONCLUSION Taken together, our results are consistent with hypersensitive reactions to IFN-gamma seen in patients with Down syndrome and may provide useful information to elucidate the mechanisms of IFN-gamma activities in those individuals.
Collapse
Affiliation(s)
- Tsutomu Iwamoto
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Aoba-ku, Sendai 980-8575, Japan.
| | | | | | | | | | | | | |
Collapse
|
36
|
Dierssen M, Herault Y, Estivill X. Aneuploidy: from a physiological mechanism of variance to Down syndrome. Physiol Rev 2009; 89:887-920. [PMID: 19584316 DOI: 10.1152/physrev.00032.2007] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Quantitative differences in gene expression emerge as a significant source of variation in natural populations, representing an important substrate for evolution and accounting for a considerable fraction of phenotypic diversity. However, perturbation of gene expression is also the main factor in determining the molecular pathogenesis of numerous aneuploid disorders. In this review, we focus on Down syndrome (DS) as the prototype of "genomic disorder" induced by copy number change. The understanding of the pathogenicity of the extra genomic material in trisomy 21 has accelerated in the last years due to the recent advances in genome sequencing, comparative genome analysis, functional genome exploration, and the use of model organisms. We present recent data on the role of genome-altering processes in the generation of diversity in DS neural phenotypes focusing on the impact of trisomy on brain structure and mental retardation and on biological pathways and cell types in target brain regions (including prefrontal cortex, hippocampus, cerebellum, and basal ganglia). We also review the potential that genetically engineered mouse models of DS bring into the understanding of the molecular biology of human learning disorders.
Collapse
Affiliation(s)
- Mara Dierssen
- Genes and Disease Program, Genomic Regulation Center-CRG, Pompeu Fabra University, Barcelona Biomedical Research Park, Dr Aiguader 88, PRBB building E, Barcelona 08003, Catalonia, Spain.
| | | | | |
Collapse
|
37
|
Laffaire J, Rivals I, Dauphinot L, Pasteau F, Wehrle R, Larrat B, Vitalis T, Moldrich RX, Rossier J, Sinkus R, Herault Y, Dusart I, Potier MC. Gene expression signature of cerebellar hypoplasia in a mouse model of Down syndrome during postnatal development. BMC Genomics 2009; 10:138. [PMID: 19331679 PMCID: PMC2678156 DOI: 10.1186/1471-2164-10-138] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 03/30/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Down syndrome is a chromosomal disorder caused by the presence of three copies of chromosome 21. The mechanisms by which this aneuploidy produces the complex and variable phenotype observed in people with Down syndrome are still under discussion. Recent studies have demonstrated an increased transcript level of the three-copy genes with some dosage compensation or amplification for a subset of them. The impact of this gene dosage effect on the whole transcriptome is still debated and longitudinal studies assessing the variability among samples, tissues and developmental stages are needed. RESULTS We thus designed a large scale gene expression study in mice (the Ts1Cje Down syndrome mouse model) in which we could measure the effects of trisomy 21 on a large number of samples (74 in total) in a tissue that is affected in Down syndrome (the cerebellum) and where we could quantify the defect during postnatal development in order to correlate gene expression changes to the phenotype observed. Statistical analysis of microarray data revealed a major gene dosage effect: for the three-copy genes as well as for a 2 Mb segment from mouse chromosome 12 that we show for the first time as being deleted in the Ts1Cje mice. This gene dosage effect impacts moderately on the expression of euploid genes (2.4 to 7.5% differentially expressed). Only 13 genes were significantly dysregulated in Ts1Cje mice at all four postnatal development stages studied from birth to 10 days after birth, and among them are 6 three-copy genes. The decrease in granule cell proliferation demonstrated in newborn Ts1Cje cerebellum was correlated with a major gene dosage effect on the transcriptome in dissected cerebellar external granule cell layer. CONCLUSION High throughput gene expression analysis in the cerebellum of a large number of samples of Ts1Cje and euploid mice has revealed a prevailing gene dosage effect on triplicated genes. Moreover using an enriched cell population that is thought responsible for the cerebellar hypoplasia in Down syndrome, a global destabilization of gene expression was not detected. Altogether these results strongly suggest that the three-copy genes are directly responsible for the phenotype present in cerebellum. We provide here a short list of candidate genes.
Collapse
Affiliation(s)
- Julien Laffaire
- Laboratoire de Neurobiologie, CNRS UMR7637, ESPCI, Paris, France
- CRICM, CNRS UMR7225, INSERM UMR975, UPMC, CHU Pitie-Salpetriere, Paris, France
| | | | - Luce Dauphinot
- Laboratoire de Neurobiologie, CNRS UMR7637, ESPCI, Paris, France
- CRICM, CNRS UMR7225, INSERM UMR975, UPMC, CHU Pitie-Salpetriere, Paris, France
| | - Fabien Pasteau
- Laboratoire de Neurobiologie, CNRS UMR7637, ESPCI, Paris, France
| | - Rosine Wehrle
- Neurobiologie des Processus Adaptatifs, CNRS UMR7102, Paris, France
- UPMC, Paris, France
| | - Benoit Larrat
- Laboratoire Ondes et Accoustique, UMR7587, ESPCI, Paris, France
| | - Tania Vitalis
- Laboratoire de Neurobiologie, CNRS UMR7637, ESPCI, Paris, France
| | - Randal X Moldrich
- Laboratoire de Neurobiologie, CNRS UMR7637, ESPCI, Paris, France
- The Queensland Brain Institute, St Lucia, Australia
| | - Jean Rossier
- Laboratoire de Neurobiologie, CNRS UMR7637, ESPCI, Paris, France
| | - Ralph Sinkus
- Laboratoire Ondes et Accoustique, UMR7587, ESPCI, Paris, France
| | | | - Isabelle Dusart
- Neurobiologie des Processus Adaptatifs, CNRS UMR7102, Paris, France
- UPMC, Paris, France
| | - Marie-Claude Potier
- Laboratoire de Neurobiologie, CNRS UMR7637, ESPCI, Paris, France
- CRICM, CNRS UMR7225, INSERM UMR975, UPMC, CHU Pitie-Salpetriere, Paris, France
| |
Collapse
|
38
|
Contestabile A, Fila T, Bartesaghi R, Ciani E. Cell cycle elongation impairs proliferation of cerebellar granule cell precursors in the Ts65Dn mouse, an animal model for Down syndrome. Brain Pathol 2008; 19:224-37. [PMID: 18482164 DOI: 10.1111/j.1750-3639.2008.00168.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mental retardation, the hallmark of Down syndrome (DS), has been attributed to the reduced number of neurons populating the DS brain. The Ts65Dn mouse model of DS displays several anomalies analogous to those in individuals with DS, including neurogenesis impairment. The goal of the current study was to determine whether cell cycle alterations underlie neurogenesis impairment in the cerebellum of the Ts65Dn mouse and to identify the molecular mechanisms responsible for this defect. In neonatal (2-day old) Ts65Dn mice, cerebellar granule cell precursors exhibited a reduced proliferation rate (-40%) and a notable elongation (+45%) of the cell cycle. Alteration of cell cycle rate was due to elongation of the G(2) and G(1) phases. Microarray screening of cell cycle regulatory genes showed that Ts65Dn mice had a decreased expression of Cyclin B1 and Skp2, two key regulators of G(2)/M and G(1)/S transition. Results point at cell cycle elongation as major determinant of neurogenesis reduction in the cerebellum of Ts65Dn mice and suggest that this defect is specifically linked to an altered expression of two cell-cycle regulatory genes, Cyclin B1 and Skp2. These findings may establish the basis for a therapeutic approach aimed at restoring neurogenesis in the DS brain.
Collapse
Affiliation(s)
- Andrea Contestabile
- Department of Human and General Physiology, University of Bologna, Piazza di Porta San Donato 2, Bologna (BO), Italy
| | | | | | | |
Collapse
|
39
|
Guidi S, Bonasoni P, Ceccarelli C, Santini D, Gualtieri F, Ciani E, Bartesaghi R. Neurogenesis impairment and increased cell death reduce total neuron number in the hippocampal region of fetuses with Down syndrome. Brain Pathol 2007; 18:180-97. [PMID: 18093248 DOI: 10.1111/j.1750-3639.2007.00113.x] [Citation(s) in RCA: 206] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
We previously obtained evidence for reduced cell proliferation in the dentate gyrus (DG) of fetuses with Down syndrome (DS), suggesting that the hippocampal hypoplasia seen in adulthood may be caused by defective early neuron production. The goal of this study was to establish whether DS fetuses (17-21 weeks of gestation) exhibit reduction in total cell number in the DG, hippocampus and parahippocampal gyrus (PHG). Volumes of the cellular layers and cell number were estimated with Cavalieri's principle and the optical fractionator method, respectively. We found that in DS fetuses all investigated structures had a reduced volume and cell number. Analysis of cell phenotype showed that DS fetuses had a higher percentage of cells with astrocytic phenotype but a smaller percentage of cells with neuronal phenotype. Immunohistochemistry for Ki-67, a marker of cycling cells, showed that DS fetuses had less proliferating cells in the germinal zones of the hippocampus and PHG. We additionally found that in the hippocampal region of DS fetuses there was a higher incidence of apoptotic cell death. Results show reduced neuron number in the DS hippocampal region and suggest that this defect is caused by disruption of neurogenesis and apoptosis, two fundamental processes underlying brain building.
Collapse
Affiliation(s)
- Sandra Guidi
- Dipartimento di Fisiologia Umana e Generale, Università di Bologna, Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
40
|
Rueda N, Flórez J, Martínez-Cué C. Effects of chronic administration of SGS-111 during adulthood and during the pre- and post-natal periods on the cognitive deficits of Ts65Dn mice, a model of Down syndrome. Behav Brain Res 2007; 188:355-67. [PMID: 18178265 DOI: 10.1016/j.bbr.2007.11.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Revised: 11/20/2007] [Accepted: 11/24/2007] [Indexed: 11/24/2022]
Abstract
The Ts65Dn mouse is the most commonly used model of Down syndrome. This mouse shows many phenotypic characteristics present in people with Down syndrome, including behavioral and cognitive deficits. SGS-111 is a novel analogue of the nootropic piracetam, which prevents oxidative damage and apoptosis in both normal and Down syndrome human cortical neurons. In this work we tested the ability of chronic administration of SGS-111 to adult Ts65Dn mice to reverse the cognitive deficit found in these mice. Moreover, since oxidative stress has been reported as early as the fetal stage, SGS-111 was also administered to pregnant Ts65Dn females from the day of conception throughout the pregnancy and to Ts65Dn pups during their entire life (5 months), from birth to the end of the behavioral testing period. A characterization of the effects of SGS-111 treatment on Ts65Dn and control mice sensorimotor abilities, motor coordination, spontaneous activity, activity in the open field, exploration, anxiety and spatial and non-spatial short- and long-term learning and memory was performed. The behavioral characterization showed that chronic administration of the antioxidant SGS-111 reduced the hyperactivity shown by Ts65Dn mice in their home cage, in the open field and in the hole board test. SGS-111 administration during adulthood improved performance in the first session in the Morris water maze in control mice, and when administered during the pre- and post-natal periods, improved spatial learning in the control mice but not in Ts65Dn mice. Chronic SGS-111 administration failed to affect behavior and cognition in Ts65Dn mice.
Collapse
Affiliation(s)
- Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | | | | |
Collapse
|
41
|
Contestabile A, Fila T, Ceccarelli C, Bonasoni P, Bonapace L, Santini D, Bartesaghi R, Ciani E. Cell cycle alteration and decreased cell proliferation in the hippocampal dentate gyrus and in the neocortical germinal matrix of fetuses with Down syndrome and in Ts65Dn mice. Hippocampus 2007; 17:665-78. [PMID: 17546680 DOI: 10.1002/hipo.20308] [Citation(s) in RCA: 215] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Down syndrome (DS), the leading genetic cause of mental retardation, is characterized by reduced number of cortical neurons and brain size. The occurrence of these defects starting from early life stages points at altered developmental neurogenesis as their major determinant. The goal of our study was to obtain comparative evidence for impaired neurogenesis in the hippocampal dentate gyrus (DG) of DS fetuses and Ts65Dn mice, an animal model for DS. Cell proliferation in human fetuses was evaluated with Ki-67 (a marker of cells in S + G(2) + M phases of cell cycle) and cyclin A (a marker of cells in S phase) immunohistochemistry. We found that in the DG of DS fetuses the number of proliferating cells was notably reduced when compared with controls. A similar reduction was observed in the germinal matrix of the lateral ventricle. In both structures, DS fetuses showed a reduced ratio between cyclin A- and Ki-67-positive cells when compared with controls, indicating that they had a reduced number of cycling cells in S phase. In the DG of P2 Ts65Dn mice cell proliferation, assessed 2 h after an injection of bromodeoxyuridine (BrdU), was notably reduced, similarly to DS fetuses. After 28 days, Ts65Dn mice had still less BrdU-positive cells than controls. Phenotypic analysis of the surviving cells showed that Ts65Dn mice had a percent number of cells with astrocytic phenotype larger than controls. Using phospho-histone H3 immunohistochemistry we found that both DS fetuses and P2 Ts65Dn mice had a higher number of proliferating cells in G(2) and a smaller number of cells in M phase of cell cycle. Results provide novel evidence for proliferation impairment in the hippocampal DG of the DS fetal brain, comparable to that of the P2 mouse model, and suggest that cell cycle alterations may be critical determinants of the reduced proliferation potency.
Collapse
Affiliation(s)
- Andrea Contestabile
- Department of Human and General Physiology, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Chakrabarti L, Galdzicki Z, Haydar TF. Defects in embryonic neurogenesis and initial synapse formation in the forebrain of the Ts65Dn mouse model of Down syndrome. J Neurosci 2007; 27:11483-95. [PMID: 17959791 PMCID: PMC6673208 DOI: 10.1523/jneurosci.3406-07.2007] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 08/30/2007] [Accepted: 09/03/2007] [Indexed: 11/21/2022] Open
Abstract
Trisomy 21, one of the most prevalent congenital birth defects, results in a constellation of phenotypes collectively termed Down syndrome (DS). Mental retardation and motor and sensory deficits are among the many debilitating symptoms of DS. Alterations in brain growth and synaptic development are thought to underlie the cognitive impairments in DS, but the role of early brain development has not been studied because of the lack of embryonic human tissue and because of breeding difficulties in mouse models of DS. We generated a breeding colony of the Ts65Dn mouse model of DS to test the hypothesis that early defects in embryonic brain development are a component of brain dysfunction in DS. We found substantial delays in prenatal growth of the Ts65Dn cerebral cortex and hippocampus because of longer cell cycle duration and reduced neurogenesis from the ventricular zone neural precursor population. In addition, the Ts65Dn neocortex remains hypocellular after birth and there is a lasting decrease in synaptic development beginning in the first postnatal week. These results demonstrate that specific abnormalities in embryonic forebrain precursor cells precede early deficits in synaptogenesis and may underlie the postnatal disabilities in Ts65Dn and DS. The early prenatal period is therefore an important new window for possible therapeutic amelioration of the cognitive symptoms in DS.
Collapse
Affiliation(s)
- Lina Chakrabarti
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC 20010, and
| | - Zygmunt Galdzicki
- Department of Anatomy, Physiology, and Genetics, Neuroscience Program, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | - Tarik F. Haydar
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC 20010, and
| |
Collapse
|
43
|
Moore CS, Roper RJ. The power of comparative and developmental studies for mouse models of Down syndrome. Mamm Genome 2007; 18:431-43. [PMID: 17653795 PMCID: PMC1998891 DOI: 10.1007/s00335-007-9030-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Accepted: 04/12/2007] [Indexed: 11/15/2022]
Abstract
Since the genetic basis for Down syndrome (DS) was described, understanding the causative relationship between genes at dosage imbalance and phenotypes associated with DS has been a principal goal of researchers studying trisomy 21 (Ts21). Though inferences to the gene-phenotype relationship in humans have been made, evidence linking a specific gene or region to a particular congenital phenotype has been limited. To further understand the genetic basis for DS phenotypes, mouse models with three copies of human chromosome 21 (Hsa21) orthologs have been developed. Mouse models offer access to every tissue at each stage of development, opportunity to manipulate genetic content, and ability to precisely quantify phenotypes. Numerous approaches to recreate trisomic composition and analyze phenotypes similar to DS have resulted in diverse trisomic mouse models. A murine intraspecies comparative analysis of different genetic models of Ts21 and specific DS phenotypes reveals the complexity of trisomy and important considerations to understand the etiology of and strategies for amelioration or prevention of trisomic phenotypes. By analyzing individual phenotypes in different mouse models throughout development, such as neurologic, craniofacial, and cardiovascular abnormalities, greater insight into the gene-phenotype relationship has been demonstrated. In this review we discuss how phenotype-based comparisons between DS mouse models have been useful in analyzing the relationship of trisomy and DS phenotypes.
Collapse
Affiliation(s)
- Clara S. Moore
- Department of Biology, Franklin and Marshall College, Lancaster, Pennsylvania 17604 USA
| | - Randall J. Roper
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 W. Michigan Street, SL 306, Indianapolis, Indiana 46202 USA
| |
Collapse
|
44
|
Llorens-Martín MV, Rueda N, Martínez-Cué C, Torres-Alemán I, Flórez J, Trejo JL. Both increases in immature dentate neuron number and decreases of immobility time in the forced swim test occurred in parallel after environmental enrichment of mice. Neuroscience 2007; 147:631-8. [PMID: 17570601 DOI: 10.1016/j.neuroscience.2007.04.054] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Revised: 04/23/2007] [Accepted: 04/24/2007] [Indexed: 10/23/2022]
Abstract
A direct relation between the rate of adult hippocampal neurogenesis in mice and the immobility time in a forced swim test after living in an enriched environment has been suggested previously. In the present work, young adult mice living in an enriched environment for 2 months developed considerably more immature differentiating neurons (doublecortin-positive, DCX(+)) than control, non-enriched animals. Furthermore, we found that the more DCX(+) cells they possessed, the lower the immobility time they scored in the forced swim test. This DCX(+) subpopulation is composed of mostly differentiating dentate neurons independently of the birthdates of every individual cell. However, variations found in this subpopulation were not the result of a general effect on the survival of any newborn neuron in the granule cell layer, as 5-bromo-2-deoxyuridine (BrdU)-labeled cells born during a narrow time window included in the longer lifetime period of DCX(+) cells, were not significantly modified after enrichment. In contrast, the survival of the mature population of neurons in the granule cell layer of the dentate gyrus in enriched animals increased, although this did not influence their performance in the Porsolt test, nor did it influence the dentate gyrus volume or granule neuronal nuclei size. These results indicate that the population of immature, differentiating neurons in the adult hippocampus is one factor directly related to the protective effect of an enriched environment against a highly stressful event.
Collapse
Affiliation(s)
- M V Llorens-Martín
- Cajal Institute, Consejo Superior Investigaciones Cientificas, Doctor Arce 37, 28002 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
45
|
Fernandez F, Garner CC. Object recognition memory is conserved in Ts1Cje, a mouse model of Down syndrome. Neurosci Lett 2007; 421:137-41. [PMID: 17566652 DOI: 10.1016/j.neulet.2007.04.075] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2007] [Revised: 04/06/2007] [Accepted: 04/06/2007] [Indexed: 12/15/2022]
Abstract
Ts1Cje and Ts65Dn are genetic mouse models of Down syndrome (DS). Like individuals with DS, these mice exhibit various hallmarks of hippocampal pathology, and deficits in hippocampal-based, declarative learning and memory tasks. Both spatial navigation and novel object recognition, two prototypical domains of declarative memory function, have been strongly characterized in the Ts65Dn DS model. Indeed, Ts65Dn mice show navigation problems in the Morris water maze, impaired alternation in a T-maze, and deficient working and reference memory in the radial arm maze task. They, likewise, show an inability to detect object novelty over time. In contrast to the Ts65Dn model, hippocampal-dependent cognition has been less well characterized in Ts1Cje. Although Ts1Cje mice have been found to exhibit spatial difficulties in the Morris water maze and reduced spontaneous alternation, their ability to process object-based information has never been examined. Here, we report that Ts1Cje mice perform normally in short-term and long-term novel object recognition tasks. The ability of Ts1Cje mice to detect object novelty, unlike Ts65Dn, may point to differences in the extent of hippocampal pathology in the two DS mouse models.
Collapse
Affiliation(s)
- Fabian Fernandez
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University, 1201 Welch Rd., Palo Alto, CA 94304-5485, USA
| | | |
Collapse
|
46
|
Sahir N, Brenneman DE, Hill JM. Neonatal mice of the Down syndrome model, Ts65Dn, exhibit upregulated VIP measures and reduced responsiveness of cortical astrocytes to VIP stimulation. J Mol Neurosci 2007; 30:329-40. [PMID: 17401158 DOI: 10.1385/jmn:30:3:329] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/11/2022]
Abstract
The Ts65Dn segmental mouse model of Down syndrome (DS) possesses a triplication of the section of chromosome 16 that is most homologous to the human chromosome 21 that is trisomic in DS. This model exhibits many of the characteristics of DS including small size, developmental delays, and a decline of cholinergic systems and cognitive function with age. Recent studies have shown that vasoactive intestinal peptide (VIP) systems are upregulated in aged Ts65Dn mice and that VIP dysregulation during embryogenesis is followed by the hypotonia and developmental delays as seen in both DS and in Ts65Dn mice. Additionally, astrocytes from aged Ts65Dn brains do not respond to VIP stimulation to release survival-promoting substances. To determine if VIP dysregulation is age-related in Ts65Dn mice, the current study examined VIP and VIP receptors (VPAC-1 and VPAC-2) in postnatal day 8 Ts65Dn mice. VIP and VPAC-1 expression was significantly increased in the brains of trisomic mice compared with wild-type mice. VIP-binding sites were also significantly increased in several brain areas of young Ts65Dn mice, especially in the cortex, caudate/putamen, and hippocampus. Further, in vitro treatment of normal neurons with conditioned medium from VIP-stimulated Ts65Dn astrocytes from neonatal mice did not enhance neuronal survival. This study indicates that VIP anomalies are present in neonatal Ts65Dn mice, a defect occurs in the signal transduction mechanism of the VPAC-1 VIP receptor, cortical astrocytes from neonatal brains are dysfunctional, and further, that VIP dysregulation may play a significant role in DS.
Collapse
Affiliation(s)
- Nadia Sahir
- Section on Developmental and Molecular Pharmacology, NICHD/NIH, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
47
|
Gardiner K, Costa ACS. The proteins of human chromosome 21. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2006; 142C:196-205. [PMID: 17048356 PMCID: PMC3299406 DOI: 10.1002/ajmg.c.30098] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Recent genomic sequence annotation suggests that the long arm of human chromosome 21 encodes more than 400 genes. Because there is no evidence to exclude any significant segment of 21 q from containing genes relevant to the Down syndrome (DS) cognitive phenotype, all genes in this entire set must be considered as candidates. Only a subset, however, is likely to make critical contributions. Determining which these are is both a major focus in biology and a critical step in efficient development of therapeutics. The subtle molecular abnormality in DS, the 50% increase in chromosome 21 gene expression, presents significant challenges for researchers in detection and quantitation. Another challenge is the current limitation in understanding gene functions and in interpreting biological characteristics. Here, we review information on chromosome 21-encoded proteins compiled from the literature and from genomics and proteomics databases. For each protein, we summarize their evolutionary conservation, the complexity of their known protein interactions and their level of expression in brain, and discuss the implications and limitations of these data. For a subset, we discuss neurologically relevant phenotypes of mouse models that include knockouts, mutations, or overexpression. Lastly, we highlight a small number of genes for which recent evidence suggests a function in biochemical/cellular pathways that are relevant to cognition. Until knowledge deficits are overcome, we suggest that effective development of gene-phenotype correlations in DS requires a serious and continuous effort to assimilate broad categories of information on chromosome 21 genes, plus the creation of more versatile mouse models.
Collapse
Affiliation(s)
- Katheleen Gardiner
- Eleanor Roosevelt Institute at the University of Denver, 1899 Gaylord Street, Denver, Colorado 80206
- Department of Biochemistry and Molecular Genetics, University of Colorado at Denver and Health Science Center, Denver, CO
| | - Alberto C. S. Costa
- Eleanor Roosevelt Institute at the University of Denver, 1899 Gaylord Street, Denver, Colorado 80206
- Department of Psychiatry, University of Colorado at Denver and Health Science Center, Denver, CO
| |
Collapse
|
48
|
Lorenzi HA, Reeves RH. Hippocampal hypocellularity in the Ts65Dn mouse originates early in development. Brain Res 2006; 1104:153-9. [PMID: 16828061 DOI: 10.1016/j.brainres.2006.05.022] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 05/03/2006] [Accepted: 05/08/2006] [Indexed: 10/24/2022]
Abstract
Ts65Dn, a well-characterized animal model for Down syndrome, has three copies of the distal end of mouse chromosome 16 and therefore has segmental trisomy for orthologs for nearly half of the genes located on human chromosome 21. Ts65Dn mice have learning and memory impairments, especially in tasks involving the hippocampus. Previous studies have shown that older adult Ts65Dn mice have structural abnormalities in the hippocampus including fewer granule cells in dentate gyrus and more pyramidal cells in the CA3 subfield of cornus ammonis. However, it is not clear whether those changes are secondary to the age-related neurodegeneration of the basal forebrain cholinergic neurons that project to the hippocampus or if they originate earlier during hippocampal development. To address this question, we performed a quantitative study of the hippocampal volume and the numbers of granule cell and pyramidal neurons in young (postnatal day 6, P6) and adult (3-month-old) mice using the optical fractionator method. At P6, Ts65Dn mice had 20% fewer granule cells in dentate gyrus than did euploid littermates. Similarly, compared to euploid, P6 trisomic mice showed an 18% reduction in mitotic cells in the granule cell layer and the hilus, where granule cell precursors divide to generate the internal granule cell layer. Granule cell hypocellularity persists in 3-month-old Ts65Dn mice before the onset of cholinergic atrophy. The hypocellularity seen in the trisomic adult hippocampus originates early in development and may contribute to specific cognitive deficits in these mice.
Collapse
Affiliation(s)
- Hernan A Lorenzi
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2185, USA
| | | |
Collapse
|
49
|
Clark S, Schwalbe J, Stasko MR, Yarowsky PJ, Costa ACS. Fluoxetine rescues deficient neurogenesis in hippocampus of the Ts65Dn mouse model for Down syndrome. Exp Neurol 2006; 200:256-61. [PMID: 16624293 DOI: 10.1016/j.expneurol.2006.02.005] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2005] [Revised: 02/02/2006] [Accepted: 02/06/2006] [Indexed: 11/29/2022]
Abstract
The Ts65Dn mouse, an adult model of Down syndrome displays behavioral deficits consistent with a dysfunctional hippocampus, similar to that seen with DS. In looking for mechanisms underlying these performance deficits, we have assessed adult neurogenesis in the dentate gyrus of Ts65Dn. Under untreated conditions, Ts65Dn mice (2-5 months old) showed markedly fewer BrdU-labeled cells than euploid animals. Chronic antidepressant treatment for over 3 weeks with the serotonin selective reuptake inhibitor, fluoxetine, increased neurogenesis in the Ts65Dn to comparable levels seen in the euploid by augmenting both proliferation and survival of BrdU-labeled cells in the subgranular layer and granule cell layer of the hippocampus, respectively.
Collapse
Affiliation(s)
- Sarah Clark
- Department of Pharmacology and Exp. Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1509, USA
| | | | | | | | | |
Collapse
|
50
|
Roper RJ, Baxter LL, Saran NG, Klinedinst DK, Beachy PA, Reeves RH. Defective cerebellar response to mitogenic Hedgehog signaling in Down [corrected] syndrome mice. Proc Natl Acad Sci U S A 2006; 103:1452-6. [PMID: 16432181 PMCID: PMC1360600 DOI: 10.1073/pnas.0510750103] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Trisomy 21 is the cause of Down [corrected] syndrome (DS) which is characterized by a number of phenotypes, including a brain which is small and hypocellular compared to that of euploid individuals. The cerebellum is disproportionately reduced. Ts65Dn mice are trisomic for orthologs of about half of the genes on human chromosome 21 and provide a genetic model for DS. These mice display a number of developmental anomalies analogous to those in DS, including a small cerebellum with a significantly decreased number of both granule and Purkinje cell neurons. Here we trace the origin of the granule cell deficit to precursors in early postnatal development, which show a substantially reduced mitogenic response to Hedgehog protein signaling. Purified cultures of trisomic granule cell precursors show a reduced but dose-dependent response to the Sonic hedgehog protein signal in vitro, demonstrating that this is a cell-autonomous deficit. Systemic treatment of newborn trisomic mice with a small molecule agonist of Hedgehog pathway activity increases mitosis and restores granule cell precursor populations in vivo. These results demonstrate a basis for and a potential therapeutic approach to a fundamental aspect of CNS pathology in DS.
Collapse
Affiliation(s)
- Randall J Roper
- Department of Physiology and McKusick-Nathans Institute for Genetic Medicine, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|