1
|
Rao ER, Thaxton T, Gama E, Godfrey J, Wei C, Lin Q, Li Y, Hejazi Pastor DP, Hansel C, Du X, Gomez CM. Calcium channel-coupled transcription factors facilitate direct nuclear signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.09.637126. [PMID: 39990342 PMCID: PMC11844367 DOI: 10.1101/2025.02.09.637126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
VGCCs play crucial roles within the CNS, in maintaining cell excitability, enabling activity- dependent neuronal development, and forming long-term memory by regulating Ca 2+ influx. The intracellular carboxyl-terminal domains of VGCC α1 subunits help regulate VGCC function. Emerging evidence suggests that some VGCC C-termini have functions independent of channel gating and exist as stable proteins. Here, we demonstrate that all VGCC gene family members express bicistronic mRNA transcripts that produce functionally distinct C-terminal proteins (CTPs) in tandem with full-length VGCC α1 subunits. Two of these CTPs, α1CCT and α1ACT, cycle to and from the nucleus in a Ca 2+ - and calmodulin-dependent fashion. α1CCT, α1ACT, and α1HCT regulate chromatin accessibility and/or bind directly to genes, regulating gene networks involved in neuronal differentiation and synaptic function in a Ca 2+ -dependent manner. This study elucidates a conserved process of coordinated protein expression within the VGCC family, coupling the channel function with VGCC C-terminal transcription factors.
Collapse
|
2
|
Pedroni A, Dai YWE, Lafouasse L, Chang W, Srivastava I, Del Vecchio L, Ampatzis K. Neuroprotective gap-junction-mediated bystander transformations in the adult zebrafish spinal cord after injury. Nat Commun 2024; 15:4331. [PMID: 38773121 PMCID: PMC11109231 DOI: 10.1038/s41467-024-48729-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/07/2024] [Indexed: 05/23/2024] Open
Abstract
The adult zebrafish spinal cord displays an impressive innate ability to regenerate after traumatic insults, yet the underlying adaptive cellular mechanisms remain elusive. Here, we show that while the cellular and tissue responses after injury are largely conserved among vertebrates, the large-size fast spinal zebrafish motoneurons are remarkably resilient by remaining viable and functional. We also reveal the dynamic changes in motoneuron glutamatergic input, excitability, and calcium signaling, and we underscore the critical role of calretinin (CR) in binding and buffering the intracellular calcium after injury. Importantly, we demonstrate the presence and the dynamics of a neuron-to-neuron bystander neuroprotective biochemical cooperation mediated through gap junction channels. Our findings support a model in which the intimate and dynamic interplay between glutamate signaling, calcium buffering, gap junction channels, and intercellular cooperation upholds cell survival and promotes the initiation of regeneration.
Collapse
Affiliation(s)
- Andrea Pedroni
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Yu-Wen E Dai
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Leslie Lafouasse
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Weipang Chang
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Ipsit Srivastava
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Lisa Del Vecchio
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | | |
Collapse
|
3
|
Kimizoğlu O, Kirca ND, Kandis S, Micili SC, Harzadin NU, Kocturk S. Daily Consumption of High-Polyphenol Olive Oil Enhances Hippocampal Neurogenesis in Old Female Rats. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2023; 42:668-677. [PMID: 36416641 DOI: 10.1080/27697061.2022.2144540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/03/2022] [Accepted: 10/31/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVE The aim of this study is to evaluate the effect of daily consumption of high-polyphenol (HP) olive oil on neurogenesis by investigating neuronal cell proliferation and maturation in the hippocampus of old rats, and to evaluate the relationship between neurogenesis, spatial memory, and anxiety-like behavior. METHODS A total of 34 female, 20-22-month-old Sprague Dawley rats were divided into three groups: control group, low-polyphenol (LP) group, and high-polyphenol (HP) group. The animals were fed distilled water, LP olive oil and HP-extra virgin olive oil, respectively for 6 weeks using an oral gavage. At 43 days, animals were tested using the Morris Water Maze to evaluate spatial memory, and the Open-field test to evaluate anxiety-like behavior. Neural cell proliferation in the dentate gyrus (DG) was determined by BrdU labeling and Nestin protein expression. Neuronal maturation was determined by NeuN labeling. Synaptic density in the hippocampus and prefrontal cortex was examined by measuring Synaptophysin (SYN) levels. Hippocampal Calbindin levels were measured to assess cellular calcium metabolism. RESULTS Daily consumption of HP olive oil significantly improved cell proliferation and neuronal maturation in the DG of old rats. HP-olive oil significantly increased SYN levels in the prefrontal cortex, and nestin and calbindin levels in the hippocampus (p < 0.05). LP olive oil diet has shown no effect on any parameter (p > 0.05). We also did not find any statistically significant difference between the groups in terms of spatial memory and anxiety-like behavior (p > 0.05). CONCLUSION Our study is first to show that daily consumption of HP-olive oil enhances hippocampal neurogenesis in old rats, which has been confirmed by proliferation and maturation biomarkers. In addition, increased SYN and calbindin levels showed that the generated cells were also functionally developed in the HP group. We suggest that daily consumption of HP olive oil may have beneficial effects on brain aging by triggering neurogenesis.
Collapse
Affiliation(s)
- Ozgun Kimizoğlu
- Institute of Health Sciences, Department of Neurosciences, Dokuz Eylul University, Izmir, Turkey
| | - N Deniz Kirca
- Institute of Health Sciences, Department of Neurosciences, Dokuz Eylul University, Izmir, Turkey
| | - Sevim Kandis
- Faculty of Medicine, Department of Physiology, Dokuz Eylul University, Izmir, Turkey
| | - Serap Cilaker Micili
- Faculty of Medicine, Department of Histology and Embryology, Dokuz Eylul University, Izmir, Turkey
| | - Nazan Uysal Harzadin
- Faculty of Medicine, Department of Physiology, Dokuz Eylul University, Izmir, Turkey
| | - Semra Kocturk
- Institute of Health Sciences, Department of Neurosciences, Dokuz Eylul University, Izmir, Turkey
- Faculty of Medicine, Department of Medical Biochemistry, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
4
|
Zhang H, Zhou H, Guo X, Zhang G, Xiao M, Wu S, Jin C, Yang J, Lu X. Cigarette smoke triggers calcium overload in mouse hippocampal neurons via the ΔFOSB-CACNA2D1 axis to impair cognitive performance. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 258:114996. [PMID: 37167740 DOI: 10.1016/j.ecoenv.2023.114996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/24/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023]
Abstract
A growing body of evidence shows that cigarette smoking impairs cognitive performance. The 'Calcium Hypothesis' theory of neuronopathies reveals a critical role of aberrant calcium signaling in compromised cognitive functions. However, the underlying implications of abnormalities in calcium signaling in the neurotoxicity induced by cigarette smoke (CS) have not yet been identified. CACNA2D1, an important auxiliary subunit involved in the composition of voltage-gated calcium channels (VGCCs), was reported to affect the calcium signaling in neurons by facilitating VGCCs-mediated Ca2+ influx. ΔFOSB, an alternatively-spliced product of the Fosb gene, is an activity-dependent transcription factor induced robustly in the brain in response to environmental stimuli such as CS. Interestingly, our preliminary bioinformatics analysis revealed a significant co-expression between ΔFOSB and CACNA2D1 in brain tissues of patients with neurodegenerative diseases characterized by progressive cognitive decline. Therefore, we hypothesized that the activation of the ΔFOSB-CACNA2D1 axis in response to CS exposure might cause dysregulation of calcium homeostasis in hippocampal neurons via VGCCs-mediated Ca2+ influx, thereby contributing to cognitive deficits. To this end, the present study established a CS-induced mouse model of hippocampus-dependent cognitive impairment, in which the activation of the ΔFOSB-CACNA2D1 axis accompanied by severe calcium overload was observed in the mouse hippocampal tissues. More importantly, ΔFOSB knockdown-/overexpression-mediated inactivation/activation of the ΔFOSB-CACNA2D1 axis interdicted/mimicked CS-induced dysregulation of calcium homeostasis followed by severe cellular damage in HT22 mouse hippocampal neurons. Mechanistically speaking, a further ChIP-qPCR assay confirmed the physical interaction between transcription factor ΔFOSB and the Cacna2d1 gene promoter, suggesting a direct transcriptional regulation of the Cacna2d1 gene by ΔFOSB. Overall, our current work aims to deliver a unique insight into the neurotoxic mechanisms induced by CS to explore potential targets for intervention.
Collapse
Affiliation(s)
- Hongchao Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, People's Republic of China
| | - Huabin Zhou
- Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, People's Republic of China
| | - Xianhe Guo
- Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, People's Republic of China
| | - Guopei Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, People's Republic of China
| | - Mingyang Xiao
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, People's Republic of China
| | - Shengwen Wu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, People's Republic of China
| | - Cuihong Jin
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, People's Republic of China
| | - Jinghua Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, People's Republic of China
| | - Xiaobo Lu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang 110122, People's Republic of China.
| |
Collapse
|
5
|
Takla M, Saadeh K, Tse G, Huang CLH, Jeevaratnam K. Ageing and the Autonomic Nervous System. Subcell Biochem 2023; 103:201-252. [PMID: 37120470 DOI: 10.1007/978-3-031-26576-1_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
The vertebrate nervous system is divided into central (CNS) and peripheral (PNS) components. In turn, the PNS is divided into the autonomic (ANS) and enteric (ENS) nervous systems. Ageing implicates time-related changes to anatomy and physiology in reducing organismal fitness. In the case of the CNS, there exists substantial experimental evidence of the effects of age on individual neuronal and glial function. Although many such changes have yet to be experimentally observed in the PNS, there is considerable evidence of the role of ageing in the decline of ANS function over time. As such, this chapter will argue that the ANS constitutes a paradigm for the physiological consequences of ageing, as well as for their clinical implications.
Collapse
Affiliation(s)
| | | | - Gary Tse
- Kent and Medway Medical School, Canterbury, UK
- University of Surrey, Guildford, UK
| | | | | |
Collapse
|
6
|
Calderazzo S, Covert M, Alba DD, Bowley BE, Pessina MA, Rosene DL, Buller B, Medalla M, Moore TL. Neural recovery after cortical injury: Effects of MSC derived extracellular vesicles on motor circuit remodeling in rhesus monkeys. IBRO Neurosci Rep 2022; 13:243-254. [PMID: 36590089 PMCID: PMC9795302 DOI: 10.1016/j.ibneur.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 07/01/2022] [Accepted: 08/07/2022] [Indexed: 01/04/2023] Open
Abstract
Reorganization of motor circuits in the cortex and corticospinal tract are thought to underlie functional recovery after cortical injury, but the mechanisms of neural plasticity that could be therapeutic targets remain unclear. Recent work from our group have shown that systemic treatment with mesenchymal stem cell derived (MSCd) extracellular vesicles (EVs) administered after cortical damage to the primary motor cortex (M1) of rhesus monkeys resulted in a robust recovery of fine motor function and reduced chronic inflammation. Here, we used immunohistochemistry for cfos, an activity-dependent intermediate early gene, to label task-related neurons in the surviving primary motor and premotor cortices, and markers of axonal and synaptic plasticity in the spinal cord. Compared to vehicle, EV treatment was associated with a greater density of cfos+ pyramidal neurons in the deep layers of M1, greater density of cfos+ inhibitory interneurons in premotor areas, and lower density of synapses on MAP2+ lower motor neurons in the cervical spinal cord. These data suggest that the anti-inflammatory effects of EVs may reduce injury-related upper motor neuron damage and hyperexcitability, as well as aberrant compensatory re-organization in the cervical spinal cord to improve motor function.
Collapse
Key Words
- CB, Calbindin
- CR, Calretinin
- CSC, Cervical Spinal Cord
- Circuit Remodeling
- Cortical Injury
- DH, Dorsal Horn
- EVs, Extracellular Vesicles
- Extracellular Vesicles
- Ischemia
- LCST, Lateral Corticospinal Tract
- M1, Primary Motor Cortex
- MAP2, Microtubule Associated Protein 2
- MSCd, Mesenchymal Stem Cell derived
- Motor Cortex
- NHP, Non-Human Primate
- PV, Parvalbumin
- Plasticity
- ROS, Reactive Oxygen Species
- SYN, Synaptophysin
- Stem Cell-Based Treatments
- VH, Ventral Horn
- dPMC, dorsal Premotor Cortex
- miRNA, Micro RNA
- periM1, Perilesional Primary Motor Cortex
Collapse
Affiliation(s)
| | | | | | | | | | - Douglas L. Rosene
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| | | | - Maria Medalla
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| | - Tara L. Moore
- Anatomy and Neurobiology Dept, BUSM, USA
- Center for Systems Neuroscience, BU, USA
| |
Collapse
|
7
|
Bazwinsky-Wutschke I, Dehghani F. Impact of cochlear ablation on calbindin and synaptophysin in the gerbil medial nucleus of the trapezoid body before hearing onset. J Chem Neuroanat 2021; 118:102023. [PMID: 34481914 DOI: 10.1016/j.jchemneu.2021.102023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 11/19/2022]
Abstract
Spontaneous bursting activity is already generated in the cochlea before hearing onset and represents an important condition of the functional and anatomical organization of auditory brainstem nuclei. In the present study, cochlea ablation induced changes were characterized in auditory brainstem nuclei indirectly innervated by auditory nerve fibers before hearing onset. In Meriones unguiculatus immunohistochemical labeling of calbindin-D28k (CB) and synaptophysin (SYN) were performed. The influence of cochlea-ablation on CB or SYN was analyzed by considering their differential immunoreaction during development. During the normal postnatal development, CB was first detected in somata of the medial nucleus of the trapezoid body (MNTB) at postnatal day (P)4. The immunoreaction increased gradually in parallel to the appearance of CB-immunoreactive terminal fields in distinct superior olivary complex (SOC) nuclei. Cochlear removal at P5 or P9 in animals with 24 and 48 h survival times resulted in an increase in somatic CB-labeling in the lesioned MNTB including terminal fields compared to the non-lesioned MNTB. SYN-immunolabeling was first detected at P0 and began to strongly encircle the MNTB neurons at P4. A further progression was observed with age. Cochlear ablation resulted in a significant reduction of SYN-labeled MNTB areas of P5-cochlea-ablated gerbils after 48 h post-lesion. In P9 cochlea-ablated gerbils, a redistribution of SYN-positive terminals was seen after 24 and 48 h. Taken together, the destruction of cochlea differentially influences CB- and SYN-labeling in the MNTB, which should be considered in association with different critical periods before hearing onset.
Collapse
Affiliation(s)
- Ivonne Bazwinsky-Wutschke
- Department of Biology, University of Leipzig, Talstrasse 33, D-04103, Leipzig, Germany; Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, D-06108, Halle (Saale), Germany.
| | - Faramarz Dehghani
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, D-06108, Halle (Saale), Germany
| |
Collapse
|
8
|
Geula C, Dunlop SR, Ayala I, Kawles AS, Flanagan ME, Gefen T, Mesulam MM. Basal forebrain cholinergic system in the dementias: Vulnerability, resilience, and resistance. J Neurochem 2021; 158:1394-1411. [PMID: 34272732 PMCID: PMC8458251 DOI: 10.1111/jnc.15471] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 01/15/2023]
Abstract
The basal forebrain cholinergic neurons (BFCN) provide the primary source of cholinergic innervation of the human cerebral cortex. They are involved in the cognitive processes of learning, memory, and attention. These neurons are differentially vulnerable in various neuropathologic entities that cause dementia. This review summarizes the relevance to BFCN of neuropathologic markers associated with dementias, including the plaques and tangles of Alzheimer's disease (AD), the Lewy bodies of diffuse Lewy body disease, the tauopathy of frontotemporal lobar degeneration (FTLD-TAU) and the TDP-43 proteinopathy of FTLD-TDP. Each of these proteinopathies has a different relationship to BFCN and their corticofugal axons. Available evidence points to early and substantial degeneration of the BFCN in AD and diffuse Lewy body disease. In AD, the major neurodegenerative correlate is accumulation of phosphotau in neurofibrillary tangles. However, these neurons are less vulnerable to the tauopathy of FTLD. An intriguing finding is that the intracellular tau of AD causes destruction of the BFCN, whereas that of FTLD does not. This observation has profound implications for exploring the impact of different species of tauopathy on neuronal survival. The proteinopathy of FTLD-TDP shows virtually no abnormal inclusions within the BFCN. Thus, the BFCN are highly vulnerable to the neurodegenerative effects of tauopathy in AD, resilient to the neurodegenerative effect of tauopathy in FTLD and apparently resistant to the emergence of proteinopathy in FTLD-TDP and perhaps also in Pick's disease. Investigations are beginning to shed light on the potential mechanisms of this differential vulnerability and their implications for therapeutic intervention.
Collapse
Affiliation(s)
- Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Sara R Dunlop
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Ivan Ayala
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Allegra S Kawles
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Marek-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
9
|
Influence of Nitric Oxide-Cyclic GMP and Oxidative STRESS on Amyloid-β Peptide Induced Decrease of Na,K-ATPase Activity in Rat Hippocampal Slices. J Membr Biol 2021; 254:463-473. [PMID: 34327545 DOI: 10.1007/s00232-021-00196-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/19/2021] [Indexed: 12/11/2022]
Abstract
Amyloid-β peptide (Aβ) has been shown to cause synaptic dysfunction and can render neurons vulnerable to excitotoxicity and oxidative stress. Na,K-ATPase plays an important role to maintain cell ionic equilibrium and it can be modulated by N-methyl-D-aspartate (NMDA)-nitric oxide (NO)-cyclic GMP pathway. Disruption of NO synthase (NOS) activity and reactive oxygen species (ROS) production could lead to changes in Na,K-ATPase isoforms' activities that may be detrimental to the cells. Our aim was to evaluate the signaling pathways of Aβ in relation to NMDA-NOS-cyclic GMP versus oxidative stress on α1-/α2,3-Na,K-ATPase activities in rat hippocampal slices. Aβ1-40 induced a concentration-dependent increase of NOS activity and increased cyclic guanosine monophosphate (cGMP), TBARS (thiobarbituric acid reactive substances), and 3-Nitrotyrosine (3-NT)-modified protein levels in rat hippocampal slices. The increase in NOS activity and cyclic GMP levels induced by Aβ1-40 was completely blocked by MK-801 (inhibitor of NMDA receptor) and L-NAME (inhibitor of NOS) pre-treatment but changes in TBARS levels were only partially blocked by both compounds. The Aβ treatment also decreased Na,K-ATPase activity which was reverted by N-nitro-L-arginine methyl ester hydrochloride (L-NAME) but not by MK-801 pre-treatment. The decrease in enzyme activity induced by Aβ was isoform-specific since only α1-Na,K-ATPase was affected. These findings suggest that the activation of NMDA-NOS signaling cascade linked to α2,3-Na,K-ATPase activity may mediate an adaptive, neuroprotective response to Aβ in rat hippocampus.
Collapse
|
10
|
Hamrick MW, Stranahan AM. Metabolic regulation of aging and age-related disease. Ageing Res Rev 2020; 64:101175. [PMID: 32971259 DOI: 10.1016/j.arr.2020.101175] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 08/19/2020] [Accepted: 09/03/2020] [Indexed: 12/23/2022]
Abstract
Inquiry into relationships between energy metabolism and brain function requires a uniquely interdisciplinary mindset, and implementation of anti-aging lifestyle strategies based on this work also involves consistent mental and physical discipline. Dr. Mark P. Mattson embodies both of these qualities, based on the breadth and depth of his work on neurobiological responses to energetic stress, and on his own diligent practice of regular exercise and caloric restriction. Dr. Mattson created a neurotrophic niche in his own laboratory, allowing trainees to grow their skills, form new connections, and eventually migrate, forming their own labs while remaining part of the extended lab family. In this historical review, we highlight Dr. Mattson's many contributions to understanding neurobiological responses to physical exercise and dietary restriction, with an emphasis on the mechanisms that may underlie neuroprotection in ageing and age-related disease. On the occasion of Dr. Mattson's retirement from the National Institute on Aging, we highlight his foundational work on metabolism and neuroplasticity by reviewing the context for these findings and considering their impact on future research on the neuroscience of aging.
Collapse
|
11
|
Posada-Duque RA, Cardona-Gómez GP. CDK5 Targeting as a Therapy for Recovering Neurovascular Unit Integrity in Alzheimer's Disease. J Alzheimers Dis 2020; 82:S141-S161. [PMID: 33016916 DOI: 10.3233/jad-200730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The neurovascular unit (NVU) is responsible for synchronizing the energetic demand, vasodynamic changes, and neurochemical and electrical function of the brain through a closed and interdependent interaction of cell components conforming to brain tissue. In this review, we will focus on cyclin-dependent kinase 5 (CDK5) as a molecular pivot, which plays a crucial role in the healthy function of neurons, astrocytes, and the endothelium and is implicated in the cross-talk of cellular adhesion signaling, ion transmission, and cytoskeletal remodeling, thus allowing the individual and interconnected homeostasis of cerebral parenchyma. Then, we discuss how CDK5 overactivation affects the integrity of the NVU in Alzheimer's disease (AD) and cognitive impairment; we emphasize how CDK5 is involved in the excitotoxicity spreading of glutamate and Ca2+ imbalance under acute and chronic injury. Additionally, we present pharmacological and gene therapy strategies for producing partial depletion of CDK5 activity on neurons, astrocytes, or endothelium to recover neuroplasticity and neurotransmission, suggesting that the NVU should be the targeted tissue unit in protective strategies. Finally, we conclude that CDK5 could be effective due to its intervention on astrocytes by its end feet on the endothelium and neurons, acting as an intermediary cell between systemic and central communication in the brain. This review provides integrated guidance regarding the pathogenesis of and potential repair strategies for AD.
Collapse
Affiliation(s)
- Rafael Andrés Posada-Duque
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, SIU, University of Antioquia, Medellín, Colombia.,Institute of Biology, Faculty of Exact and Natural Sciences, University of Antioquia, Medellín, Colombia
| | - Gloria Patricia Cardona-Gómez
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, SIU, University of Antioquia, Medellín, Colombia
| |
Collapse
|
12
|
Involvement of GABAergic interneuron dysfunction and neuronal network hyperexcitability in Alzheimer's disease: Amelioration by metabolic switching. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:191-205. [DOI: 10.1016/bs.irn.2020.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
13
|
Kumar D, Sharma A, Sharma L. A Comprehensive Review of Alzheimer's Association with Related Proteins: Pathological Role and Therapeutic Significance. Curr Neuropharmacol 2020; 18:674-695. [PMID: 32172687 PMCID: PMC7536827 DOI: 10.2174/1570159x18666200203101828] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/26/2019] [Accepted: 01/31/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's is an insidious, progressive, chronic neurodegenerative disease which causes the devastation of neurons. Alzheimer's possesses complex pathologies of heterogeneous nature counting proteins as one major factor along with enzymes and mutated genes. Proteins such as amyloid precursor protein (APP), apolipoprotein E (ApoE), presenilin, mortalin, calbindin-D28K, creactive protein, heat shock proteins (HSPs), and prion protein are some of the chief elements in the foremost hypotheses of AD like amyloid-beta (Aβ) cascade hypothesis, tau hypothesis, cholinergic neuron damage, etc. Disturbed expression of these proteins results in synaptic dysfunction, cognitive impairment, memory loss, and neuronal degradation. On the therapeutic ground, attempts of developing anti-amyloid, anti-inflammatory, anti-tau therapies are on peak, having APP and tau as putative targets. Some proteins, e.g., HSPs, which ameliorate oxidative stress, calpains, which help in regulating synaptic plasticity, and calmodulin-like skin protein (CLSP) with its neuroprotective role are few promising future targets for developing anti-AD therapies. On diagnostic grounds of AD C-reactive protein, pentraxins, collapsin response mediator protein-2, and growth-associated protein-43 represent the future of new possible biomarkers for diagnosing AD. The last few decades were concentrated over identifying and studying protein targets of AD. Here, we reviewed the physiological/pathological roles and therapeutic significance of nearly all the proteins associated with AD that addresses putative as well as probable targets for developing effective anti-AD therapies.
Collapse
Affiliation(s)
- Deepak Kumar
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P. India
| | - Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P. India
| | - Lalit Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P. India
| |
Collapse
|
14
|
Fischer N, Johnson Chacko L, Majerus A, Potrusil T, Riechelmann H, Schmutzhard J, Schrott-Fischer A, Glueckert R. Age-Dependent Calcium-Binding Protein Expression in the Spiral Ganglion and Hearing Performance of C57BL/6J and 129/SvJ Mice. ORL J Otorhinolaryngol Relat Spec 2019; 81:138-154. [PMID: 31170714 DOI: 10.1159/000499472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/08/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Calcium-binding proteins in neurons buffer intracellular free Ca2+ ions, which interact with proteins controlling enzymatic and ion channel activity. The heterogeneous distribution of calretinin, calbindin, and parvalbumin influences calcium homeostasis, and calcium-related neuronal processes play an important role in neuronal aging and degeneration. This study evaluated age-related changes in calretinin, calbindin, and parvalbumin immune reactivity in spiral ganglion cells. METHODS A total of 16 C57BL/6J and 16 129/SvJ mice at different ages (2, 4, 7, and 12 months) were included in the study. Hearing thresholds were assessed using auditory brainstem response before inner ears were excised for further evaluation. Semiquantitative immunohistochemistry for the aforementioned calcium-binding proteins was performed at the cellular level. RESULTS The hearing thresholds of C57BL/6J and 129/SvJ mice increased significantly by 7 months of age. The average immune reactivity of calbin-din as well as the relative number of positive cells increased significantly with aging, but no significant alterations in calretinin or parvalbumin were observed. CONCLUSIONS Upregulation of calbindin could serve as a protection to compensate for functional deficits that occur with aging. Expression of both calretinin and parvalbumin seem to be stabilizing factors in murine inner ears up to the age of 12 months in C57BL/6J and 129/SvJ mice.
Collapse
Affiliation(s)
- Natalie Fischer
- Department of Otorhinolaryngology, Medical University Innsbruck, Innsbruck, Austria
| | - Lejo Johnson Chacko
- Department of Otorhinolaryngology, Medical University Innsbruck, Innsbruck, Austria
| | - Alexandra Majerus
- Department of Otorhinolaryngology, Medical University Innsbruck, Innsbruck, Austria
| | - Thomas Potrusil
- Department of Otorhinolaryngology, Medical University Innsbruck, Innsbruck, Austria
| | - Herbert Riechelmann
- Department of Otorhinolaryngology, Medical University Innsbruck, Innsbruck, Austria
| | - Joachim Schmutzhard
- Department of Otorhinolaryngology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Rudolf Glueckert
- Department of Otorhinolaryngology, Medical University Innsbruck, Innsbruck, Austria.,Department of Otorhinolaryngology, Tirol Kliniken, University Clinics of Innsbruck, Innsbruck, Austria
| |
Collapse
|
15
|
Constantinof A, Moisiadis VG, Kostaki A, Szyf M, Matthews SG. Antenatal Glucocorticoid Exposure Results in Sex-Specific and Transgenerational Changes in Prefrontal Cortex Gene Transcription that Relate to Behavioural Outcomes. Sci Rep 2019; 9:764. [PMID: 30679753 PMCID: PMC6346022 DOI: 10.1038/s41598-018-37088-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 11/28/2018] [Indexed: 11/17/2022] Open
Abstract
Synthetic glucocorticoids (sGC) are administered to women at risk for pre-term delivery to reduce respiratory distress syndrome in the newborn. The prefrontal cortex (PFC) is important in regulating stress responses and related behaviours and expresses high levels of glucocorticoid receptors (GR). Further, antenatal exposure to sGC results in a hyperactive phenotype in first generation (F1) juvenile male and female offspring, as well as F2 and F3 juvenile females from the paternal lineage. We hypothesized that multiple courses of antenatal sGC modify gene expression in the PFC, that these effects are sex-specific and maintained across multiple generations, and that the gene sets affected relate to modified locomotor activity. We performed RNA sequencing on PFC of F1 juvenile males and females, as well as F2 and F3 juvenile females from the paternal lineage and used regression modelling to relate gene expression and behavior. Antenatal sGC resulted in sex-specific and generation-specific changes in gene expression. Further, the expression of 4 genes (C9orf116, Calb1, Glra3, and Gpr52) explained 20–29% of the observed variability in locomotor activity. Antenatal exposure to sGC profoundly influences the developing PFC; effects are evident across multiple generations and may drive altered behavioural phenotypes.
Collapse
Affiliation(s)
- Andrea Constantinof
- Departments of Physiology, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Vasilis G Moisiadis
- Departments of Physiology, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Alisa Kostaki
- Departments of Physiology, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Moshe Szyf
- Departments of Pharmacology & Therapeutics, Sackler Program for Epigenetics & Psychobiology, McGill University, Montreal, QC, H3G1Y6, Canada
| | - Stephen G Matthews
- Departments of Physiology, University of Toronto, Toronto, ON, M5S1A8, Canada. .,Departments of Obstetrics and Gynecology, University of Toronto, Toronto, ON, M5S1A8, Canada. .,Departments of Medicine, University of Toronto, Toronto, ON, M5S1A8, Canada. .,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G1X5, Canada.
| |
Collapse
|
16
|
Chandran R, Kumar M, Kesavan L, Jacob RS, Gunasekaran S, Lakshmi S, Sadasivan C, Omkumar R. Cellular calcium signaling in the aging brain. J Chem Neuroanat 2019; 95:95-114. [DOI: 10.1016/j.jchemneu.2017.11.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/03/2017] [Accepted: 11/07/2017] [Indexed: 12/21/2022]
|
17
|
Voltage-Dependent Calcium Channels, Calcium Binding Proteins, and Their Interaction in the Pathological Process of Epilepsy. Int J Mol Sci 2018; 19:ijms19092735. [PMID: 30213136 PMCID: PMC6164075 DOI: 10.3390/ijms19092735] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 01/08/2023] Open
Abstract
As an important second messenger, the calcium ion (Ca2+) plays a vital role in normal brain function and in the pathophysiological process of different neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), and epilepsy. Ca2+ takes part in the regulation of neuronal excitability, and the imbalance of intracellular Ca2+ is a trigger factor for the occurrence of epilepsy. Several anti-epileptic drugs target voltage-dependent calcium channels (VDCCs). Intracellular Ca2+ levels are mainly controlled by VDCCs located in the plasma membrane, the calcium-binding proteins (CBPs) inside the cytoplasm, calcium channels located on the intracellular calcium store (particular the endoplasmic reticulum/sarcoplasmic reticulum), and the Ca2+-pumps located in the plasma membrane and intracellular calcium store. So far, while many studies have established the relationship between calcium control factors and epilepsy, the mechanism of various Ca2+ regulatory factors in epileptogenesis is still unknown. In this paper, we reviewed the function, distribution, and alteration of VDCCs and CBPs in the central nervous system in the pathological process of epilepsy. The interaction of VDCCs with CBPs in the pathological process of epilepsy was also summarized. We hope this review can provide some clues for better understanding the mechanism of epileptogenesis, and for the development of new anti-epileptic drugs targeting on VDCCs and CBPs.
Collapse
|
18
|
Inoue KI, Miyachi S, Nishi K, Okado H, Nagai Y, Minamimoto T, Nambu A, Takada M. Recruitment of calbindin into nigral dopamine neurons protects against MPTP-Induced parkinsonism. Mov Disord 2018; 34:200-209. [PMID: 30161282 DOI: 10.1002/mds.107] [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: 02/15/2018] [Revised: 06/06/2018] [Accepted: 06/29/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Parkinson's disease is caused by dopamine deficiency in the striatum, which is a result of loss of dopamine neurons from the substantia nigra pars compacta. There is a consensus that a subpopulation of nigral dopamine neurons that expresses the calcium-binding protein calbindin is selectively invulnerable to parkinsonian insults. The objective of the present study was to test the hypothesis that dopamine neuron degeneration might be prevented by viral vector-mediated gene delivery of calbindin into the dopamine neurons that do not normally contain it. METHODS A calbindin-expressing adenoviral vector was injected into the striatum of macaque monkeys to be conveyed to cell bodies of nigral dopamine neurons through retrograde axonal transport, or the calbindin-expressing lentiviral vector was injected into the nigra directly because of its predominant uptake from cell bodies and dendrites. The animals in which calbindin was successfully recruited into nigral dopamine neurons were administered systemically with MPTP. RESULTS In the monkeys that had received unilateral vector injections, parkinsonian motor deficits, such as muscular rigidity and akinesia/bradykinesia, appeared predominantly in the limbs corresponding to the non-calbindin-recruited hemisphere after MPTP administration. Data obtained from tyrosine hydroxylase immunostaining and PET imaging for the dopamine transporter revealed that the nigrostriatal dopamine system was preserved better on the calbindin-recruited side. Conversely, on the non-calbindin-recruited control side, many more dopamine neurons expressed α-synuclein. CONCLUSIONS The present results indicate that calbindin recruitment into nigral dopamine neurons protects against the onset of parkinsonian insults, thus providing a novel approach to PD prevention. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ken-Ichi Inoue
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan.,Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Shigehiro Miyachi
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,Cognitive Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan
| | - Katsunori Nishi
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan
| | - Haruo Okado
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Yuji Nagai
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takafumi Minamimoto
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Atsushi Nambu
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan.,Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan
| |
Collapse
|
19
|
Supraphysiological Levels of Oxygen Exposure During the Neonatal Period Impairs Signaling Pathways Required for Learning and Memory. Sci Rep 2018; 8:9914. [PMID: 29967535 PMCID: PMC6028393 DOI: 10.1038/s41598-018-28220-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/04/2018] [Indexed: 12/31/2022] Open
Abstract
Preterm infants often require prolonged oxygen supplementation and are at high risk of neurodevelopmental impairment. We recently reported that adult mice exposed to neonatal hyperoxia (postnatal day [P] 2 to 14) had spatial navigation memory deficits associated with hippocampal shrinkage. The mechanisms by which early oxidative stress impair neurodevelopment are not known. Our objective was to identify early hyperoxia-induced alterations in hippocampal receptors and signaling pathways necessary for memory formation. We evaluated C57BL/6 mouse pups at P14, exposed to either 85% oxygen or air from P2 to 14. We performed targeted analysis of hippocampal ligand-gated ion channels and proteins necessary for memory formation, and global bioinformatic analysis of differentially expressed hippocampal genes and proteins. Hyperoxia decreased hippocampal mGLU7, TrkB, AKT, ERK2, mTORC1, RPS6, and EIF4E and increased α3, α5, and ɤ2 subunits of GABAA receptor and PTEN proteins, although changes in gene expression were not always concordant. Bioinformatic analysis indicated dysfunction in mitochondria and global protein synthesis and translational processes. In conclusion, supraphysiological oxygen exposure reduced proteins necessary for hippocampus-dependent memory formation and may adversely impact hippocampal mitochondrial function and global protein synthesis. These early hippocampal changes may account for memory deficits seen in preterm survivors following prolonged oxygen supplementation.
Collapse
|
20
|
Mattson MP, Arumugam TV. Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States. Cell Metab 2018; 27:1176-1199. [PMID: 29874566 PMCID: PMC6039826 DOI: 10.1016/j.cmet.2018.05.011] [Citation(s) in RCA: 705] [Impact Index Per Article: 100.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/02/2018] [Accepted: 05/15/2018] [Indexed: 02/06/2023]
Abstract
During aging, the cellular milieu of the brain exhibits tell-tale signs of compromised bioenergetics, impaired adaptive neuroplasticity and resilience, aberrant neuronal network activity, dysregulation of neuronal Ca2+ homeostasis, the accrual of oxidatively modified molecules and organelles, and inflammation. These alterations render the aging brain vulnerable to Alzheimer's and Parkinson's diseases and stroke. Emerging findings are revealing mechanisms by which sedentary overindulgent lifestyles accelerate brain aging, whereas lifestyles that include intermittent bioenergetic challenges (exercise, fasting, and intellectual challenges) foster healthy brain aging. Here we provide an overview of the cellular and molecular biology of brain aging, how those processes interface with disease-specific neurodegenerative pathways, and how metabolic states influence brain health.
Collapse
Affiliation(s)
- Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
21
|
Berg EM, Bertuzzi M, Ampatzis K. Complementary expression of calcium binding proteins delineates the functional organization of the locomotor network. Brain Struct Funct 2018; 223:2181-2196. [PMID: 29423637 PMCID: PMC5968073 DOI: 10.1007/s00429-018-1622-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/30/2018] [Indexed: 12/18/2022]
Abstract
Neuronal networks in the spinal cord generate and execute all locomotor-related movements by transforming descending signals from supraspinal areas into appropriate rhythmic activity patterns. In these spinal networks, neurons that arise from the same progenitor domain share similar distribution patterns, neurotransmitter phenotypes, morphological and electrophysiological features. However, subgroups of them participate in different functionally distinct microcircuits to produce locomotion at different speeds and of different modalities. To better understand the nature of this network complexity, here we characterized the distribution of parvalbumin (PV), calbindin D-28 k (CB) and calretinin (CR) which are regulators of intracellular calcium levels and can serve as anatomical markers for morphologically and potential functionally distinct neuronal subpopulations. We observed wide expression of CBPs in the adult zebrafish, in several spinal and reticulospinal neuronal populations with a diverse neurotransmitter phenotype. We also found that several spinal motoneurons express CR and PV. However, only the motoneuron pools that are responsible for generation of fast locomotion were CR-positive. CR can thus be used as a marker for fast motoneurons and might potentially label the fast locomotor module. Moreover, CB was mainly observed in the neuronal progenitor cells that are distributed around the central canal. Thus, our results suggest that during development the spinal neurons utilize CB and as the neurons mature and establish a neurotransmitter phenotype they use CR or/and PV. The detailed characterization of CBPs expression, in the spinal cord and brainstem neurons, is a crucial step toward a better understanding of the development and functionality of neuronal locomotor networks.
Collapse
Affiliation(s)
- Eva M Berg
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Maria Bertuzzi
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | | |
Collapse
|
22
|
Palus K, Bulc M, Czajkowska M, Miciński B, Całka J. Neurochemical characteristics of calbindin-like immunoreactive coeliac-cranial mesenteric ganglion complex (CCMG) neurons supplying the pre-pyloric region of the porcine stomach. Tissue Cell 2018; 50:8-14. [DOI: 10.1016/j.tice.2017.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 01/29/2023]
|
23
|
Roque C, Baltazar G. Impact of Astrocytes on the Injury Induced by In Vitro Ischemia. Cell Mol Neurobiol 2017; 37:1521-1528. [PMID: 28315110 PMCID: PMC11482236 DOI: 10.1007/s10571-017-0483-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/12/2017] [Indexed: 12/29/2022]
Abstract
Cell cultures are characterized by their simplicity, controllability, and ability to provide detailed basic information on how a particular cell population responds to specific stimuli or insult. These characteristics led to their extensive application in the study of molecular interactions and represent a valuable tool in the study of different pathologies. However, due to the lack of interactions between the different components that form an in vivo system, the results obtained in pure cell cultures not always translate what occurs in vivo. In this context, the use of co-cultures has the advantage of allowing the study of interactions between different types of cells present in a tissue, which in many situations are determinant for the effects obtained. The present study aimed to characterize cortical neuron-glia and neuron-enriched primary cultures and evaluate their response to an ischemic insult. Cell viability was assessed by the MTT assay and cell number/phenotype was analyzed by immunocytochemistry in control cultures and in cells subjected to 4 h of oxygen and glucose deprivation. The results obtained demonstrate that astrocytes have a substantial impact on the injury induced by an ischemic insult, thus suggesting that the crosstalk between glia and neurons is crucial to the neuronal protection in conditions of ischemia.
Collapse
Affiliation(s)
- Cláudio Roque
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
- Instituto Politécnico de Castelo Branco, Escola Superior de Saúde Dr. Lopes Dias, IPCB-ESALD, Castelo Branco, Portugal
| | - Graça Baltazar
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal.
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
24
|
Epigenetic suppression of hippocampal calbindin-D28k by ΔFosB drives seizure-related cognitive deficits. Nat Med 2017; 23:1377-1383. [PMID: 29035369 PMCID: PMC5747956 DOI: 10.1038/nm.4413] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 08/29/2017] [Indexed: 12/13/2022]
Abstract
The calcium-binding protein calbindin-D28k is critical for hippocampal function and cognition1-3, but its expression is markedly decreased in various neurological disorders associated with epileptiform activity and seizures4-7. In Alzheimer's disease (AD) and epilepsy, both of which are accompanied by recurrent seizures8, the severity of cognitive deficits reflects the degree of calbindin reduction in the hippocampal dentate gyrus (DG)4,9,10. However, despite the importance of calbindin in both neuronal physiology and pathology, the regulatory mechanisms that control its expression in the hippocampus are poorly understood. Here we report an epigenetic mechanism by which seizures chronically suppress hippocampal calbindin expression and impair cognition. We demonstrate that ΔFosB, a highly stable transcription factor, is induced in the hippocampus of mouse models of AD and seizures, where it binds and triggers histone deacetylation at the calbindin gene (Calb1) promoter, and downregulates Calb1 transcription. Notably, increasing DG calbindin levels, either by direct virus-mediated expression or inhibition of ΔFosB signaling, improves spatial memory in a mouse model of AD. Moreover, levels of ΔFosB and calbindin expression are inversely related in DG of patients with temporal lobe epilepsy (TLE) or AD, and correlate with performance on the Mini-Mental State Examination (MMSE). We propose that chronic suppression of calbindin by ΔFosB is one mechanism by which intermittent seizures drive persistent cognitive deficits in conditions accompanied by recurrent seizures.
Collapse
|
25
|
Lee JC, Cho JH, Lee TK, Kim IH, Won MH, Cho GS, Shin BN, Hwang IK, Park JH, Ahn JH, Kang IJ, Lee YJ, Kim YH. Effect of hyperthermia on calbindin-D 28k immunoreactivity in the hippocampal formation following transient global cerebral ischemia in gerbils. Neural Regen Res 2017; 12:1458-1464. [PMID: 29089991 PMCID: PMC5649466 DOI: 10.4103/1673-5374.215256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Calbindin D-28K (CB), a Ca2+-binding protein, maintains Ca2+ homeostasis and protects neurons against various insults. Hyperthermia can exacerbate brain damage produced by ischemic insults. However, little is reported about the role of CB in the brain under hyperthermic condition during ischemic insults. We investigated the effects of transient global cerebral ischemia on CB immunoreactivity as well as neuronal damage in the hippocampal formation under hyperthermic condition using immunohistochemistry for neuronal nuclei (NeuN) and CB, and Fluoro-Jade B histofluorescence staining in gerbils. Hyperthermia (39.5 ± 0.2°C) was induced for 30 minutes before and during transient ischemia. Hyperthermic ischemia resulted in neuronal damage/death in the pyramidal layer of CA1–3 area and in the polymorphic layer of the dentate gyrus at 1, 2, 5 days after ischemia. In addition, hyperthermic ischemia significantly decreaced CB immunoreactivity in damaged or dying neurons at 1, 2, 5 days after ischemia. In brief, hyperthermic condition produced more extensive and severer neuronal damage/death, and reduced CB immunoreactivity in the hippocampus following transient global cerebral ischemia. Present findings indicate that the degree of reduced CB immunoreactivity might be related with various neuronal damage/death overtime and corresponding areas after ischemic insults.
Collapse
Affiliation(s)
- Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jeong-Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Geum-Sil Cho
- Department of Pharmacology & Toxicology, Shinpoong Pharmaceutical Co., Ltd., Ansan, South Korea
| | - Bich-Na Shin
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, South Korea
| | - Young Joo Lee
- Department of Emergency Medicine, Seoul Hospital, College of Medicine, Sooncheonhyang University, Seoul, South Korea
| | - Yang Hee Kim
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
26
|
Wächter C, Eiden LE, Naumann N, Depboylu C, Weihe E. Loss of cerebellar neurons in the progression of lentiviral disease: effects of CNS-permeant antiretroviral therapy. J Neuroinflammation 2016; 13:272. [PMID: 27737697 PMCID: PMC5064958 DOI: 10.1186/s12974-016-0726-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/20/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The majority of investigations on HIV-associated neurocognitive disorders (HAND) neglect the cerebellum in spite of emerging evidence for its role in higher cognitive functions and dysfunctions in common neurodegenerative diseases. METHODS We systematically investigated the molecular and cellular responses of the cerebellum as contributors to lentiviral infection-induced neurodegeneration, in the simian immunodeficiency virus (SIV)-infected rhesus macaque model for HIV infection and HAND. Four cohorts of animals were studied: non-infected controls, SIV-infected asymptomatic animals, and SIV-infected AIDS-diseased animals with and without brain-permeant antiretroviral treatment. The antiretroviral utilized was 6-chloro-2',3'-dideoxyguanosine (6-Cl-ddG), a CNS-permeable nucleoside reverse transcriptase inhibitor. Quantitation of granule cells and Purkinje cells, of an established biomarker of SIV infection (gp41), of microglial/monocyte/macrophage markers (IBA-1, CD68, CD163), and of the astroglial marker (GFAP) were used to reveal cell-specific cerebellar responses to lentiviral infection and antiretroviral therapy (ART). The macromolecular integrity of the blood brain barrier was tested by albumin immunohistochemistry. RESULTS Productive CNS infection was observed in the symptomatic stage of disease, and correlated with extensive microglial/macrophage and astrocyte activation, and widespread macromolecular blood brain barrier defects. Signs of productive infection, and inflammation, were reversed upon treatment with 6-Cl-ddG, except for a residual low-grade activation of microglial cells and astrocytes. There was an extensive loss of granule cells in the SIV-infected asymptomatic cohort, which was further increased in the symptomatic stage of the disease and persisted after 6-Cl-ddG (administered after the onset of symptoms of AIDS). In the symptomatic stage, Purkinje cell density was reduced. Purkinje cell loss was likewise unaffected by 6-Cl-ddG treatment at this time. CONCLUSIONS Our findings suggest that neurodegenerative mechanisms are triggered by SIV infection early in the disease process, i. e., preceding large-scale cerebellar productive infection and marked neuroinflammation. These affect primarily granule cells early in disease, with later involvement of Purkinje cells, indicating differential vulnerability of the two neuronal populations. The results presented here indicate a role for the cerebellum in neuro-AIDS. They also support the conclusion that, in order to attenuate the development of motor and cognitive dysfunctions in HIV-positive individuals, CNS-permeant antiretroviral therapy combined with anti-inflammatory and neuroprotective treatment is indicated even before overt signs of CNS inflammation occur.
Collapse
Affiliation(s)
- Christian Wächter
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany
| | - Lee E Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Nedye Naumann
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany
| | - Candan Depboylu
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.,Experimental Neurology, Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Eberhard Weihe
- Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps University Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.
| |
Collapse
|
27
|
Yang X, Zhao Q, An R, Zhou H, Lin Z, Xu Y. SNP rs1805874 of the Calbindin1 Gene Is Associated with Parkinson's Disease in Han Chinese. Genet Test Mol Biomarkers 2016; 20:753-757. [PMID: 27611799 DOI: 10.1089/gtmb.2016.0149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
AIMS The single-nucleotide polymorphism, rs1805874, in the Calbindin1 gene has been associated with Parkinson's disease among the Japanese people, but not among Europeans or Americans. To help clarify these contrasting results, we conducted a case-control study to explore whether such an association exists among the Han Chinese. METHODS We used the ligase detection reaction to genotype the rs1805874 SNP in 514 Han Chinese with Parkinson's disease as well as in 475 healthy Han Chinese controls. RESULTS We identified a significant association between the A allele of rs1805874 and the risk of Parkinson's disease (OR 1.257, 95% CI 1.036-1.524, p = 0.020). Subgroup analysis revealed a significant association of the A allele with male gender (OR 1.306, 95% CI 1.009-1.691, p = 0.042) and late onset of disease (OR 1.247, 95% CI 1.010-1.540, p = 0.040). CONCLUSION Our findings suggest that the A allele of rs1805874 is associated with risk of Parkinson's disease among the Han Chinese. Our results, combined with previous studies, suggest that rs1805874 is associated with Parkinson's disease in East Asians, but not Caucasians.
Collapse
Affiliation(s)
- Xinglong Yang
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| | - Quanzhen Zhao
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| | - Ran An
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| | - Huayong Zhou
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| | - Zhenfang Lin
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| | - Yanming Xu
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| |
Collapse
|
28
|
Bradbury A, Bagel J, Sampson M, Farhat N, Ding W, Swain G, Prociuk M, O'Donnell P, Drobatz K, Gurda B, Wassif C, Remaley A, Porter F, Vite C. Cerebrospinal Fluid Calbindin D Concentration as a Biomarker of Cerebellar Disease Progression in Niemann-Pick Type C1 Disease. J Pharmacol Exp Ther 2016; 358:254-61. [PMID: 27307499 PMCID: PMC4959104 DOI: 10.1124/jpet.116.232975] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/06/2016] [Indexed: 01/29/2023] Open
Abstract
Niemann-Pick type C (NPC) 1 disease is a rare, inherited, neurodegenerative disease. Clear evidence of the therapeutic efficacy of 2-hydroxypropyl-β-cyclodextrin (HPβCD) in animal models resulted in the initiation of a phase I/IIa clinical trial in 2013 and a phase IIb/III trial in 2015. With clinical trials ongoing, validation of a biomarker to track disease progression and serve as a supporting outcome measure of therapeutic efficacy has become compulsory. In this study, we evaluated calcium-binding protein calbindin D-28K (calbindin) concentrations in the cerebrospinal fluid (CSF) as a biomarker of NPC1 disease. In the naturally occurring feline model, CSF calbindin was significantly elevated at 3 weeks of age, prior to the onset of cerebellar dysfunction, and steadily increased to >10-fold over normal at end-stage disease. Biweekly intrathecal administration of HPβCD initiated prior to the onset of neurologic dysfunction completely normalized CSF calbindin in NPC1 cats at all time points analyzed when followed up to 78 weeks of age. Initiation of HPβCD after the onset of clinical signs (16 weeks of age) resulted in a delayed reduction of calbindin levels in the CSF. Evaluation of CSF from patients with NPC1 revealed that calbindin concentrations were significantly elevated compared with CSF samples collected from unaffected patients. Off-label treatment of patients with NPC1 with miglustat, an inhibitor of glycosphingolipid biosynthesis, significantly decreased CSF calbindin compared with pretreatment concentrations. These data suggest that the CSF calbindin concentration is a sensitive biomarker of NPC1 disease that could be instrumental as an outcome measure of therapeutic efficacy in ongoing clinical trials.
Collapse
Affiliation(s)
- Allison Bradbury
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Jessica Bagel
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Maureen Sampson
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Nicole Farhat
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Wenge Ding
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Gary Swain
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Maria Prociuk
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Patricia O'Donnell
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Kenneth Drobatz
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Brittney Gurda
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Christopher Wassif
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Alan Remaley
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Forbes Porter
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Charles Vite
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| |
Collapse
|
29
|
Gonadal hormone modulation of intracellular calcium as a mechanism of neuroprotection. Front Neuroendocrinol 2016; 42:40-52. [PMID: 26930421 DOI: 10.1016/j.yfrne.2016.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/22/2016] [Accepted: 02/26/2016] [Indexed: 12/28/2022]
Abstract
Hormones have wide-ranging effects throughout the nervous system, including the ability interact with and modulate many aspects of intracellular calcium regulation and calcium signaling. Indeed, these interactions specifically may help to explain the often opposing or paradoxical effects of hormones, such as their ability to both promote and prevent neuronal cell death during development, as well as reduce or exacerbate damage following an insult or injury in adulthood. Here, we review the basic mechanisms underlying intracellular calcium regulation-perhaps the most dynamic and flexible of all signaling molecules-and discuss how gonadal hormones might manipulate these mechanisms to coordinate diverse cellular responses and achieve disparate outcomes. Additional future research that specifically addresses questions of sex and hormone effects on calcium signaling at different ages will be critical to understanding hormone-mediated neuroprotection.
Collapse
|
30
|
Bharti AR, Woods SP, Ellis RJ, Cherner M, Rosario D, Potter M, Heaton RK, Everall IP, Masliah E, Grant I, Letendre SL. Fibroblast growth factors 1 and 2 in cerebrospinal fluid are associated with HIV disease, methamphetamine use, and neurocognitive functioning. HIV AIDS-RESEARCH AND PALLIATIVE CARE 2016; 8:93-9. [PMID: 27199571 PMCID: PMC4857802 DOI: 10.2147/hiv.s93306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Human immunodeficiency virus (HIV) and methamphetamine use commonly affect neurocognitive (NC) functioning. We evaluated the relationships between NC functioning and two fibroblast growth factors (FGFs) in volunteers who differed in HIV serostatus and methamphetamine dependence (MAD). Methods A total of 100 volunteers were categorized into four groups based on HIV serostatus and MAD in the prior year. FGF-1 and FGF-2 were measured in cerebrospinal fluid by enzyme-linked immunosorbent assays along with two reference biomarkers (monocyte chemotactic protein [MCP]-1 and neopterin). Comprehensive NC testing was summarized by global and domain impairment ratings. Results Sixty-three volunteers were HIV+ and 59 had a history of MAD. FGF-1, FGF-2, and both reference biomarkers differed by HIV and MAD status. For example, FGF-1 levels were lower in subjects who had either HIV or MAD than in HIV− and MAD− controls (P=0.003). Multivariable regression identified that global NC impairment was associated with an interaction between FGF-1 and FGF-2 (model R2=0.09, P=0.01): higher FGF-2 levels were only associated with neurocognitive impairment among subjects who had lower FGF-1 levels. Including other covariates in the model (including antidepressant use) strengthened the model (model R2=0.18, P=0.004) but did not weaken the association with FGF-1 and FGF-2. Lower FGF-1 levels were associated with impairment in five of seven cognitive domains, more than FGF-2, MCP-1, or neopterin. Conclusion These findings provide in vivo support that HIV and MAD alter expression of FGFs, which may contribute to the NC abnormalities associated with these conditions. These cross-sectional findings cannot establish causality and the therapeutic benefits of recombinant FGF-1 need to be investigated.
Collapse
Affiliation(s)
- Ajay R Bharti
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Steven Paul Woods
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Ronald J Ellis
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Mariana Cherner
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Debra Rosario
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Michael Potter
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Robert K Heaton
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Ian P Everall
- Department of Psychiatry, University of Melbourne, Victoria, Australia
| | - Eliezer Masliah
- Department of Pathology, University of Californa San Diego, San Diego, CA, USA
| | - Igor Grant
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Scott L Letendre
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
31
|
Ray S, Brecht M. Structural development and dorsoventral maturation of the medial entorhinal cortex. eLife 2016; 5:e13343. [PMID: 27036175 PMCID: PMC4876644 DOI: 10.7554/elife.13343] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/27/2016] [Indexed: 12/11/2022] Open
Abstract
We investigated the structural development of superficial-layers of medial entorhinal cortex and parasubiculum in rats. The grid-layout and cholinergic-innervation of calbindin-positive pyramidal-cells in layer-2 emerged around birth while reelin-positive stellate-cells were scattered throughout development. Layer-3 and parasubiculum neurons had a transient calbindin-expression, which declined with age. Early postnatally, layer-2 pyramidal but not stellate-cells co-localized with doublecortin - a marker of immature neurons - suggesting delayed functional-maturation of pyramidal-cells. Three observations indicated a dorsal-to-ventral maturation of entorhinal cortex and parasubiculum: (i) calbindin-expression in layer-3 neurons decreased progressively from dorsal-to-ventral, (ii) doublecortin in layer-2 calbindin-positive-patches disappeared dorsally before ventrally, and (iii) wolframin-expression emerged earlier in dorsal than ventral parasubiculum. The early appearance of calbindin-pyramidal-grid-organization in layer-2 suggests that this pattern is instructed by genetic information rather than experience. Superficial-layer-microcircuits mature earlier in dorsal entorhinal cortex, where small spatial-scales are represented. Maturation of ventral-entorhinal-microcircuits - representing larger spatial-scales - follows later around the onset of exploratory behavior.
Collapse
Affiliation(s)
- Saikat Ray
- Bernstein Center for Computational
Neuroscience, Humboldt University of
Berlin, Berlin, Germany
| | - Michael Brecht
- Bernstein Center for Computational
Neuroscience, Humboldt University of
Berlin, Berlin, Germany
| |
Collapse
|
32
|
Dieni S, Nestel S, Sibbe M, Frotscher M, Hellwig S. Distinct synaptic and neurochemical changes to the granule cell-CA3 projection in Bassoon mutant mice. Front Synaptic Neurosci 2015; 7:18. [PMID: 26557085 PMCID: PMC4615824 DOI: 10.3389/fnsyn.2015.00018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/05/2015] [Indexed: 01/19/2023] Open
Abstract
Proper synaptic function depends on a finely-tuned balance between events such as protein synthesis and structural organization. In particular, the functional loss of just one synaptic-related protein can have a profound impact on overall neuronal network function. To this end, we used a mutant mouse model harboring a mutated form of the presynaptic scaffolding protein Bassoon (Bsn), which is phenotypically characterized by: (i) spontaneous generalized epileptic seizure activity, representing a chronically-imbalanced neuronal network; and (ii) a dramatic increase in hippocampal brain-derived neurotrophic factor (BDNF) protein concentration, a key player in synaptic plasticity. Detailed morphological and neurochemical analyses revealed that the increased BDNF levels are associated with: (i) modified neuropeptide distribution; (ii) perturbed expression of selected markers of synaptic activation or plasticity; (iii) subtle changes to microglial structure; and (iv) morphological alterations to the mossy fiber (MF) synapse. These findings emphasize the important contribution of Bassoon protein to normal hippocampal function, and further characterize the Bsn-mutant as a useful model for studying the effects of chronic changes to network activity.
Collapse
Affiliation(s)
- Sandra Dieni
- Neurochemistry Laboratory, Department of Molecular Psychiatry, University Hospital Freiburg Freiburg, Germany
| | - Sigrun Nestel
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, University of Freiburg Freiburg, Germany
| | - Mirjam Sibbe
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, University of Freiburg Freiburg, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg Hamburg, Germany
| | - Sabine Hellwig
- Neurochemistry Laboratory, Department of Molecular Psychiatry, University Hospital Freiburg Freiburg, Germany
| |
Collapse
|
33
|
Ahmadian SS, Rezvanian A, Peterson M, Weintraub S, Bigio EH, Mesulam MM, Geula C. Loss of calbindin-D28K is associated with the full range of tangle pathology within basal forebrain cholinergic neurons in Alzheimer's disease. Neurobiol Aging 2015; 36:3163-3170. [PMID: 26417681 DOI: 10.1016/j.neurobiolaging.2015.09.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 01/21/2023]
Abstract
Basal forebrain cholinergic neurons (BFCN) are selectively vulnerable in Alzheimer's disease (AD). We have shown that most of the BFCN in the human brain contain the calcium-binding protein calbindin-D28K (CB), a large proportion lose their CB in the course of normal aging, and the BFCN which degenerate in AD lack CB. Here, we investigated the relationship between CB in the BFCN and the process of tangle formation in AD using antibodies to tau epitopes that appear early, intermediate or late in the process of tangle formation. Very small percentages (0%-3.7%) of CB-positive BFCN contained pretangles and/or tangles, and very small percentages (0%-5%) of the total BFCN pretangles and/or tangles were in CB-immunoreactive neurons. The number of CB-positive BFCN which contained tau immunoreactivity was highest for the early epitope and lower for intermediate epitopes. A late appearing epitope was absent from CB-positive BFCN. Age-related loss of CB appears to coincide with tangle formation in the BFCN and is associated with the full range of tau pathology, including late appearing epitopes.
Collapse
Affiliation(s)
- Saman S Ahmadian
- Laboratory for Cognitive and Molecular Morphometry, Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| | - Aras Rezvanian
- Laboratory for Cognitive and Molecular Morphometry, Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| | - Melanie Peterson
- Laboratory for Cognitive and Molecular Morphometry, Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| | - Sandra Weintraub
- Laboratory for Cognitive and Molecular Morphometry, Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| | - Eileen H Bigio
- Laboratory for Cognitive and Molecular Morphometry, Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| | - Marek-Marsel Mesulam
- Laboratory for Cognitive and Molecular Morphometry, Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago IL, USA
| | - Changiz Geula
- Laboratory for Cognitive and Molecular Morphometry, Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago IL, USA.
| |
Collapse
|
34
|
Norvin D, Kim G, Baker-Nigh A, Geula C. Accumulation and age-related elevation of amyloid-β within basal forebrain cholinergic neurons in the rhesus monkey. Neuroscience 2015; 298:102-11. [DOI: 10.1016/j.neuroscience.2015.04.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 03/31/2015] [Accepted: 04/06/2015] [Indexed: 11/29/2022]
|
35
|
Freire MAM, Faber J, Lemos NAM, Santos JR, Cavalcanti PF, Lima RH, Morya E. Distribution and Morphology of Calcium-Binding Proteins Immunoreactive Neurons following Chronic Tungsten Multielectrode Implants. PLoS One 2015; 10:e0130354. [PMID: 26098896 PMCID: PMC4476592 DOI: 10.1371/journal.pone.0130354] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 05/19/2015] [Indexed: 12/25/2022] Open
Abstract
The development of therapeutic approaches to improve the life quality of people suffering from different types of body paralysis is a current major medical challenge. Brain-machine interface (BMI) can potentially help reestablishing lost sensory and motor functions, allowing patients to use their own brain activity to restore sensorimotor control of paralyzed body parts. Chronic implants of multielectrodes, employed to record neural activity directly from the brain parenchyma, constitute the fundamental component of a BMI. However, before this technique may be effectively available to human clinical trials, it is essential to characterize its long-term impact on the nervous tissue in animal models. In the present study we evaluated how chronic implanted tungsten microelectrode arrays impact the distribution and morphology of interneurons reactive to calcium-binding proteins calbindin (CB), calretinin (CR) and parvalbumin (PV) across the rat’s motor cortex. Our results revealed that chronic microelectrode arrays were well tolerated by the nervous tissue, with recordings remaining viable for up to 6 months after implantation. Furthermore, neither the morphology nor the distribution of inhibitory neurons were broadly impacted. Moreover, restricted microglial activation was observed on the implanted sites. On the whole, our results confirm and expand the notion that tungsten multielectrodes can be deemed as a feasible candidate to future human BMI studies.
Collapse
Affiliation(s)
- Marco Aurelio M. Freire
- Edmond and Lily Safra International Institute of Neurosciences (ELS-IIN), Santos Dumont Institute, Macaiba, RN, Brazil
| | - Jean Faber
- Laboratory of Neuroengineering, Department of Science and Technology, Federal University of São Paulo (UNIFESP), Sao Jose dos Campos, SP, Brazil
| | - Nelson Alessandretti M. Lemos
- Edmond and Lily Safra International Institute of Neurosciences (ELS-IIN), Santos Dumont Institute, Macaiba, RN, Brazil
| | - Jose Ronaldo Santos
- Laboratory of Neuroscience, Department of Biosciences, Federal University of Sergipe (UFS), Itabaiana, SE, Brazil
| | - Pedro França Cavalcanti
- Edmond and Lily Safra International Institute of Neurosciences (ELS-IIN), Santos Dumont Institute, Macaiba, RN, Brazil
| | - Ramon Hypolito Lima
- Memory Studies Laboratory, Department of Physiology, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Edgard Morya
- Edmond and Lily Safra International Institute of Neurosciences (ELS-IIN), Santos Dumont Institute, Macaiba, RN, Brazil
- Associação Alberto Santos Dumont para Apoio a Pesquisa, Sirio-Libanes Hospital, São Paulo, SP, Brazil
- * E-mail:
| |
Collapse
|
36
|
Spruill MM, Kuncl RW. Calbindin-D28K is increased in the ventral horn of spinal cord by neuroprotective factors for motor neurons. J Neurosci Res 2015; 93:1184-91. [PMID: 25914366 DOI: 10.1002/jnr.23562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 11/27/2014] [Accepted: 01/01/2015] [Indexed: 02/06/2023]
Abstract
Slow glutamate-mediated neuronal degeneration is implicated in the pathophysiology of motor neuron diseases such as amyotrophic lateral sclerosis (ALS). The calcium-binding proteins calbindin-D28K and parvalbumin have been reported to protect neurons against excitotoxic insults. Expression of calbindin-D28K is low in adult human motor neurons, and vulnerable motor neurons additionally may lack parvalbumin. Thus, it has been speculated that the lack of calcium-binding proteins may, in part, be responsible for early degeneration of the population of motor neurons most vulnerable in ALS. Using a rat organotypic spinal cord slice system, we examined whether the most potent neuroprotective factors for motor neurons can increase the expression of calbindin-D28K or parvalbumin proteins in the postnatal spinal cord. After 4 weeks of incubation of spinal cord slices with 1) glial cell line-derived neurotrophic factor (GDNF), 2) neurturin, 3) insulin-like growth factor I (IGF-I), or 4) pigment epithelium-derived factor (PEDF), the number of calbindin-D28K -immunopositive large neurons (>20 μm) in the ventral horn was higher under the first three conditions, but not after PEDF, compared with untreated controls. Under the same conditions, parvalbumin was not upregulated by any neuroprotective factor. The same calbindin increase was true of IGF-I and GDNF in a parallel glutamate toxicity model of motor neuron degeneration. Taken together with our previous reports from the same model, which showed that all these neurotrophic factors can potently protect motor neurons from slow glutamate injury, the data here suggest that upregulation of calbindin-D28K by some of these factors may be one mechanism by which motor neurons can be protected from glutamate-induced, calcium-mediated excitotoxicity.
Collapse
Affiliation(s)
- Maria M Spruill
- Department of Neurology and Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ralph W Kuncl
- Department of Neurology and Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
37
|
Baker-Nigh A, Vahedi S, Davis EG, Weintraub S, Bigio EH, Klein WL, Geula C. Neuronal amyloid-β accumulation within cholinergic basal forebrain in ageing and Alzheimer's disease. Brain 2015; 138:1722-37. [PMID: 25732182 DOI: 10.1093/brain/awv024] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/14/2014] [Indexed: 11/14/2022] Open
Abstract
The mechanisms that contribute to selective vulnerability of the magnocellular basal forebrain cholinergic neurons in neurodegenerative diseases, such as Alzheimer's disease, are not fully understood. Because age is the primary risk factor for Alzheimer's disease, mechanisms of interest must include age-related alterations in protein expression, cell type-specific markers and pathology. The present study explored the extent and characteristics of intraneuronal amyloid-β accumulation, particularly of the fibrillogenic 42-amino acid isoform, within basal forebrain cholinergic neurons in normal young, normal aged and Alzheimer's disease brains as a potential contributor to the selective vulnerability of these neurons using immunohistochemistry and western blot analysis. Amyloid-β1-42 immunoreactivity was observed in the entire cholinergic neuronal population regardless of age or Alzheimer's disease diagnosis. The magnitude of this accumulation as revealed by optical density measures was significantly greater than that in cortical pyramidal neurons, and magnocellular neurons in the globus pallidus did not demonstrate a similar extent of amyloid immunoreactivity. Immunoblot analysis with a panel of amyloid-β antibodies confirmed accumulation of high concentration of amyloid-β in basal forebrain early in adult life. There was no age- or Alzheimer-related alteration in total amyloid-β content within this region. In contrast, an increase in the large molecular weight soluble oligomer species was observed with a highly oligomer-specific antibody in aged and Alzheimer brains when compared with the young. Similarly, intermediate molecular weight oligomeric species displayed an increase in aged and Alzheimer brains when compared with the young using two amyloid-β42 antibodies. Compared to cortical homogenates, small molecular weight oligomeric species were lower and intermediate species were enriched in basal forebrain in ageing and Alzheimer's disease. Regional and age-related differences in accumulation were not the result of alterations in expression of the amyloid precursor protein, as confirmed by both immunostaining and western blot. Our results demonstrate that intraneuronal amyloid-β accumulation is a relatively selective trait of basal forebrain cholinergic neurons early in adult life, and increases in the prevalence of intermediate and large oligomeric assembly states are associated with both ageing and Alzheimer's disease. Selective intraneuronal amyloid-β accumulation in adult life and oligomerization during the ageing process are potential contributors to the degeneration of basal forebrain cholinergic neurons in Alzheimer's disease.
Collapse
Affiliation(s)
- Alaina Baker-Nigh
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Shahrooz Vahedi
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Elena Goetz Davis
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sandra Weintraub
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Eileen H Bigio
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - William L Klein
- 2 Neurobiology, Northwestern University, Evanston, IL 60201, USA
| | - Changiz Geula
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| |
Collapse
|
38
|
Murayama N, Noshita T, Ogino R, Masuda T, Kadoshima T, Oka T, Ueno N, Takemoto N, Toba T, Ueno S, Schulze W, Igawa Y, Morita Y, Inoue T. SUN11602-induced hyperexpression of calbindin D-28k is pivotal for the survival of hippocampal neurons under neurotoxic conditions. Brain Res 2015; 1594:71-81. [DOI: 10.1016/j.brainres.2014.10.066] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 09/12/2014] [Accepted: 10/29/2014] [Indexed: 02/03/2023]
|
39
|
Torres LB, Araujo BHS, Marruaz KS, de Souza JS, Sousa BS, Gomes da Silva S, Cabral FR, Cavalheiro EA. Parvalbumin expression and distribution in the hippocampal formation of Cebus apella. Am J Primatol 2014; 77:449-61. [PMID: 25472893 DOI: 10.1002/ajp.22366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 10/30/2014] [Accepted: 11/05/2014] [Indexed: 11/06/2022]
Abstract
New World primates play an important role in biomedical research. However, the literature still lacks information on many structural features of the brain in these species, particularly structures of the hippocampal formation that are related to long-term memory storage. This study was designed to provide information, for the first time, about the distribution and number of neurons expressing parvalbumin-immunoreactivity (PV-I) in the subregions of the hippocampal formation in Cebus apella, a New World primate species commonly used in biomedical research. Our results revealed that for several morphometric variables, PV-I cells differ significantly among the subregions CA1, CA2, CA3, and the hilus. Based upon our findings and those of other studies, we hypothesize that the proportional increase from monkeys to humans in PV-I cell density within CA1 is a factor contributing to the evolution of increased memory formation and storage.
Collapse
Affiliation(s)
- Laila Brito Torres
- Universidade Federal de São Paulo, Unifesp/EPM, Departamento de Neurologia e Neurocirurgia, Laboratório de Neurociências, São Paulo, Brazil; Hospital Israelita Albert Einstein, Instituto do Cérebro, São Paulo, Brazil; Instituto Evandro Chagas (IEC), Centro Nacional de Primatas (CENP), Ananindeua, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Siddharthan V, Wang H, Davies CJ, Hall JO, Morrey JD. Inhibition of West Nile virus by calbindin-D28k. PLoS One 2014; 9:e106535. [PMID: 25180779 PMCID: PMC4152291 DOI: 10.1371/journal.pone.0106535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 08/08/2014] [Indexed: 11/19/2022] Open
Abstract
Evidence indicates that West Nile virus (WNV) employs Ca2+ influx for its replication. Moreover, calcium buffer proteins, such as calbindin D28k (CB-D28k), may play an important role mitigating cellular destruction due to disease processes, and more specifically, in some neurological diseases. We addressed the hypothesis that CB-D28k inhibits WNV replication in cell culture and infected rodents. WNV envelope immunoreactivity (ir) was not readily co-localized with CB-D28k ir in WNV-infected Vero 76 or motor neuron-like NSC34 cells that were either stably or transiently transfected with plasmids coding for CB-D28k gene. This was confirmed in cultured cells fixed on glass coverslips and by flow cytometry. Moreover, WNV infectious titers were reduced in CB-D28k-transfected cells. As in cell culture studies, WNV env ir was not co-localized with CB-D28k ir in the cortex of an infected WNV hamster, or in the hippocampus of an infected mouse. Motor neurons in the spinal cord typically do not express CB-D28k and are susceptible to WNV infection. Yet, CB-D28k was detected in the surviving motor neurons after the initial phase of WNV infection in hamsters. These data suggested that induction of CB-D28k elicit a neuroprotective response to WNV infection.
Collapse
Affiliation(s)
- Venkatraman Siddharthan
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
| | - Hong Wang
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
| | - Christopher J. Davies
- Center for Integrated BioSystems, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
| | - Jeffery O. Hall
- Utah Veterinary Diagnostic Laboratory, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
| | - John D. Morrey
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
- * E-mail:
| |
Collapse
|
41
|
Foo KS, Hellysaz A, Broberger C. Expression and colocalization patterns of calbindin-D28k, calretinin and parvalbumin in the rat hypothalamic arcuate nucleus. J Chem Neuroanat 2014; 61-62:20-32. [PMID: 25014433 DOI: 10.1016/j.jchemneu.2014.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/26/2014] [Accepted: 06/26/2014] [Indexed: 11/16/2022]
Abstract
Calcium binding proteins (CaBPs) form a diverse group of molecules that function as signal transducers or as intracellular buffers of Ca(2+) concentration. They have been extensively used to histochemically categorize cell types throughout the brain. One region which has not yet been characterized with regard to CaBP expression is the hypothalamic arcuate nucleus, which plays a vital role in neuroendocrine control and the central regulation of energy metabolism. Using in situ hybridization and immunofluorescence, we have investigated the cellular distribution of the three CaBPs, calbindin-D28k (CB), calretinin (CR) and parvalbumin (PV) in the rat arcuate nucleus. Both mRNA and immunoreactivity was detected in the arcuate nucleus for CB - located in the medial aspects - and CR - located ventrolaterally. No PV mRNA was detected in the arcuate nucleus. Immunofluorescence results for PV were ambiguous; while one antibody detected a group of cell somata, a different antibody failed to visualize any arcuate nucleus cell profiles. Using double-labeling, neither of the examined CaBPs were observed in cells immunoreactive for the signaling molecules agouti gene-related protein, tyrosine hydroxylase, neurotensin, growth hormone-releasing hormone, somatostatin, enkephalin, dynorphin or galanin. We did, however, observe CB- and CR-immunoreactivity, in two distinct populations of neurons immunoreactive for the melanocortin peptide α-melanocyte-stimulating hormone. These data identify distinct subpopulations of arcuate neurons defined by their expression of CaBPs and provide further support for differentiation between subpopulations of anorexigenic melanocortin neurons.
Collapse
Affiliation(s)
- Kylie S Foo
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Arash Hellysaz
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
42
|
Abstract
Progressive neurodegenerative diseases are among the most frequently occurring aging-associated human pathologies. In a screen for Caenorhabditis elegans mutant animals that lack their normal complement of dopaminergic neurons, we identified two strains with progressive loss of dopaminergic neurons during postembryonic life. Through whole-genome sequencing we show that both strains harbor dominant (d), gain-of-function mutations in the Transient Receptor Potential (TRP) mechanosensory channel trp-4, a member of the invertebrate and vertebrate TRPN-type of the TRP family channels. Gain-of-function mutations in TRP channels have not been previously implicated in dopaminergic neuronal degeneration. We show that trp-4(d) induces cell death in dopamine neurons through a defined, calcium-related downstream pathway.
Collapse
|
43
|
Levi I, Eskira Y, Eisenstein M, Gilon C, Hoffman A, Tal-Gan Y, Fanous J, Bersudsky Y, Belmaker RH, Agam G, Almog O, Almog O. Inhibition of inositol monophosphatase (IMPase) at the calbindin-D28k binding site: molecular and behavioral aspects. Eur Neuropsychopharmacol 2013; 23:1806-15. [PMID: 23619164 DOI: 10.1016/j.euroneuro.2013.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 01/01/2013] [Accepted: 02/08/2013] [Indexed: 01/13/2023]
Abstract
Bipolar-disorder (manic-depressive illness) is a severe chronic illness affecting ∼1% of the adult population. It is treated with mood-stabilizers, the prototypic one being lithium-salts (lithium), but it has life threatening side-effects and a significant number of patients fail to respond. The lithium-inhibitable enzyme inositol-monophosphatase (IMPase) is one of the viable targets for lithium's mechanism of action. Calbindin-D28k (calbindin) up-regulates IMPase activity. The IMPase-calbindincomplex was modeled using the program MolFit. The in-silico model indicated that the 55-66 amino-acid segment of IMPase anchors calbindin via Lys59 and Lys61 with a glutamate in between (Lys-Glu-Lys motif) and that the motif interacts with residues Asp24 and Asp26 of calbindin. We found that differently from wildtype calbindin, IMPase was not activated by mutated calbindin in which Asp24 and Asp26 were replaced by alanine. Calbindin's effect was significantly reduced by a linear peptide with the sequence of amino acids 58-63 of IMPase (peptide 1) and by six amino-acid linear peptides including at least part of the Lys-Glu-Lys motif. The three amino-acid peptide Lys-Glu-Lys or five amino-acid linear peptides containing this motif were ineffective. Mice administered peptide 1 intracerebroventricularly exhibited a significant anti-depressant-like reduced immobility in the forced-swim test. Based on the sequence of peptide 1, and to potentially increase the peptide's stability, cyclic and linear pre-cyclic analog peptides were synthesized. One cyclic peptide and one linear pre-cyclic analog peptide inhibited calbindin-activated brain IMPase activity in-vitro. Our findings may lead to the development of molecules capable of inhibiting IMPase activity at an alternative site than that of lithium.
Collapse
Affiliation(s)
- Itzhak Levi
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Psychiatry Research Unit, Mental Health Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Pretreatment with apoaequorin protects hippocampal CA1 neurons from oxygen-glucose deprivation. PLoS One 2013; 8:e79002. [PMID: 24244400 PMCID: PMC3823939 DOI: 10.1371/journal.pone.0079002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 09/26/2013] [Indexed: 12/26/2022] Open
Abstract
Ischemic stroke affects ∼795,000 people each year in the U.S., which results in an estimated annual cost of $73.7 billion. Calcium is pivotal in a variety of neuronal signaling cascades, however, during ischemia, excess calcium influx can trigger excitotoxic cell death. Calcium binding proteins help neurons regulate/buffer intracellular calcium levels during ischemia. Aequorin is a calcium binding protein isolated from the jellyfish Aequorea victoria, and has been used for years as a calcium indicator, but little is known about its neuroprotective properties. The present study used an in vitro rat brain slice preparation to test the hypothesis that an intra-hippocampal infusion of apoaequorin (the calcium binding component of aequorin) protects neurons from ischemic cell death. Bilaterally cannulated rats received an apoaequorin infusion in one hemisphere and vehicle control in the other. Hippocampal slices were then prepared and subjected to 5 minutes of oxygen-glucose deprivation (OGD), and cell death was assayed by trypan blue exclusion. Apoaequorin dose-dependently protected neurons from OGD--doses of 1% and 4% (but not 0.4%) significantly decreased the number of trypan blue-labeled neurons. This effect was also time dependent, lasting up to 48 hours. This time dependent effect was paralleled by changes in cytokine and chemokine expression, indicating that apoaequorin may protect neurons via a neuroimmunomodulatory mechanism. These data support the hypothesis that pretreatment with apoaequorin protects neurons against ischemic cell death, and may be an effective neurotherapeutic.
Collapse
|
45
|
Celio MR. Perineuronal nets of extracellular matrix around parvalbumin‐containing neurons of the hippocampus. Hippocampus 2013. [DOI: 10.1002/hipo.1993.4500030709] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Marco R. Celio
- Institute of Histology and General Embryology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
46
|
Mattson MP, Barger SW. Roles for calcium signaling in structural plasticity and pathology in the hippocampal system. Hippocampus 2013. [DOI: 10.1002/hipo.1993.4500030711] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Mark P. Mattson
- Sanders‐Brown Center on Aging and Department of Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky, U.S.A
| | - Steven W. Barger
- Sanders‐Brown Center on Aging and Department of Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky, U.S.A
| |
Collapse
|
47
|
Fazeli AS, Nasrabadi D, Pouya A, Mirshavaladi S, Sanati MH, Baharvand H, Salekdeh GH. Proteome analysis of post-transplantation recovery mechanisms of an EAE model of multiple sclerosis treated with embryonic stem cell-derived neural precursors. J Proteomics 2013; 94:437-50. [PMID: 23791935 DOI: 10.1016/j.jprot.2013.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Revised: 06/02/2013] [Accepted: 06/09/2013] [Indexed: 12/21/2022]
Abstract
UNLABELLED Multiple sclerosis (MS) is a chronic inflammatory and progressive disorder of the central nervous system (CNS), which ultimately causes demyelination and subsequent axonal injury. Experimental autoimmune encephalomyelitis (EAE) is a well-characterized animal model to study the etiology and pathogenesis of MS. This model can also be used to investigate various therapeutic approaches for MS. Herein; we have treated a score 3 EAE mouse model with an embryonic stem cell-derived neural precursor. Clinical analysis showed recovery of the EAE model of MS following transplantation. We analyzed the proteome of spinal cords of healthy and EAE samples before and after transplantation. Proteome analysis revealed that expressions of 86 spinal cord protein spots changed in the EAE or transplanted mouse compared to controls. Mass spectrometry resulted in identification of 72 proteins. Of these, the amounts of 27 differentially expressed proteins in EAE samples returned to sham levels after transplantation, suggesting a possible correlation between changes at the proteome level and clinical signs of EAE in transplanted mice. The recovered proteins belonged to various functional groups that included disturbances in ionic and neurotransmitter release, apoptosis, iron hemostasis, and signal transduction. Our results provided a proteomic view of the molecular mechanisms of EAE recovery after stem cell transplantation. BIOLOGICAL SIGNIFICANCE In this study, we applied proteomics to analyze the changes in proteome pattern of EAE mouse model after embryonic stem cell-derived neural precursor transplantation. Our proteome results clearly showed that the expression levels of several differentially expressed proteins in EAE samples returned to sham levels after transplantation, which suggested a possible correlation between changes at the proteome level and decreased clinical signs of EAE in transplanted mice. These results will serve as a basis to address new questions and design new experiments to elucidate the biology of EAE and recovery after transplantation. A thorough understanding of stem cell-mediated therapeutic mechanisms might result in the development of more efficacious therapies for MS than are currently available.
Collapse
Affiliation(s)
- Abolhassan Shahzadeh Fazeli
- Department of Molecular Systems Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Genetics at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
48
|
The spatial relationship between neurons and astrocytes in HIV-associated dementia. J Neurovirol 2013; 19:123-30. [PMID: 23430713 DOI: 10.1007/s13365-013-0149-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 11/05/2012] [Indexed: 12/30/2022]
Abstract
Specific neuronal spatial distributional patterns have previously been correlated with increasing severity of HIV-associated dementia (HAD). As astrocytes are also a putative site of neurotoxicity, we investigated the spatial relationships of astrocytes with pyramidal and interneurons in the superior frontal gyrus from 29 patients who died from acquired immunodeficiency syndrome. Frontal cortical brain tissue was taken from diseased HIV patients who had been assessed for the presence and severity of HAD using the Memorial Sloan-Kettering Scale. No correlation was found between neuronal density and severity of dementia. However, the pattern of astrocytes became more clustered as dementia progressed. Bivariate spatial pattern analysis of neuronal populations with astrocytes revealed that, with increasing dementia severity, astrocytes and large pyramidal neurons increasingly "repelled" each other, while astrocytes and interneurons evidenced increasing "attraction." This implies that astrocytes may be more likely to be situated in the vicinity of surviving interneurons but less likely to be situated near surviving large pyramidal neurons in the setting of progressing HAD.
Collapse
|
49
|
Murayama N, Kadoshima T, Takemoto N, Kodama S, Toba T, Ogino R, Noshita T, Oka T, Ueno S, Kuroda M, Shimmyo Y, Morita Y, Inoue T. SUN11602, a novel aniline compound, mimics the neuroprotective mechanisms of basic fibroblast growth factor. ACS Chem Neurosci 2013; 4:266-76. [PMID: 23421678 DOI: 10.1021/cn300183k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Basic fibroblast growth factor (bFGF) offers some measure of protection against excitotoxic neuronal injuries by upregulating the expression of the calcium-binding protein calbindin-D28k (Calb). The newly synthesized small molecule 4-({4-[[(4-amino-2,3,5,6-tetramethylanilino)acetyl](methyl)amino]-1-piperidinyl}methyl)benzamide (SUN11602) mimics the neuroprotective effects of bFGF, and thus, we examined how SUN11602 exerts its actions on neurons in toxic conditions of glutamate. In primary cultures of rat cerebrocortical neurons, SUN11602 and bFGF prevented glutamate-induced neuronal death. This neuroprotection, which occurred in association with the augmented phosphorylation of the bFGF receptor-1 (FGFR-1) and the extracellular signal-regulated kinase-1/2 (ERK-1/2), was abolished by pretreatment with PD166866 (a FGFR-1 tyrosine kinase-specific inhibitor) and PD98059 (a mitogen-activated protein kinase [MAPK]/[ERK-1/2] kinase [MEK] inhibitor). In addition, SUN11602 and bFGF increased the levels of CALB1 gene expression in cerebrocortical neurons. Whether this neuroprotection was linked to Calb was investigated with primary cultures of cerebrocortical neurons from homozygous knockout (Calb(-/-)) and wild-type (WT) mice. In WT mice, SUN11602 and bFGF increased the levels of newly synthesized Calb in cerebrocortical neurons and suppressed the glutamate-induced rise in intracellular Ca(2+). This Ca(2+)-capturing ability of Calb allowed the neurons to survive severe toxic conditions of glutamate. In contrast, Calb levels remained unchanged in Calb(-/-) mice after exposure to SUN11602 or bFGF, and due to a loss of function of the gene, these neurons were no longer resistant to toxic conditions of glutamate. These findings indicated that SUN11602 activated a number of cellular molecules (FGFR-1, MEK/ERK intermediates, and Calb) and consequently contributed to intracellular Ca(2+) homeostasis as observed in the case of bFGF.
Collapse
Affiliation(s)
| | | | | | | | | | - Ryoko Ogino
- Asubio Pharma Co., Ltd., Kobe 650-0047, Japan
| | | | | | - Shinya Ueno
- Asubio Pharma Co., Ltd., Kobe 650-0047, Japan
| | | | | | - Yasuhiro Morita
- Faculty of Pharmacy, Laboratory
of Physiology and Morphology, Yasuda Women’s University, Hiroshima 731-0153, Japan
| | | |
Collapse
|
50
|
Magloire H, Couble ML, Romeas A, Bleicher F. Odontoblast primary cilia: facts and hypotheses. Cell Biol Int 2013; 28:93-9. [PMID: 14984754 DOI: 10.1016/j.cellbi.2003.11.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2003] [Accepted: 11/04/2003] [Indexed: 11/22/2022]
Abstract
Odontoblasts, the cells responsible for the dentine formation, are organized as a single layer of highly polarized and differentiated post-mitotic cells along the interface between the dental pulp and the mineralized tubules. They lay down the physiological secondary dentine throughout the life of the teeth. Odontoblasts play a central role in the transportation of calcium to the dentine and they possibly mediate early stages of sensory processing in teeth. A primary cilium, 9+0 configuration, have been regularly identified in a supra nuclear location. Calbindin D28k has been detected at the base of the cilium membrane. The cilium structure was positive with detyrosinated alpha tubulin antibodies in vivo and in cultured human odontoblasts. Transcripts of tektin, a protein involved in ciliogenesis, were expressed in vitro. The putative role of the primary cilium constituting a critical link between external teeth stimuli and odontoblast responses is extensively discussed.
Collapse
Affiliation(s)
- Henry Magloire
- Laboratoire du Développement des tissus dentaires, EA 1892, IFR 62, Faculté d'Odontologie, rue G. Paradin, 69372 Lyon Cedex 08, France.
| | | | | | | |
Collapse
|