1
|
Yao J, Chen SRW. RyR2-dependent modulation of neuronal hyperactivity: A potential therapeutic target for treating Alzheimer's disease. J Physiol 2024; 602:1509-1518. [PMID: 36866974 DOI: 10.1113/jp283824] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/27/2023] [Indexed: 03/04/2023] Open
Abstract
Increasing evidence suggests that simply reducing β-amyloid (Aβ) plaques may not significantly affect the progression of Alzheimer's disease (AD). There is also increasing evidence indicating that AD progression is driven by a vicious cycle of soluble Aβ-induced neuronal hyperactivity. In support of this, it has recently been shown that genetically and pharmacologically limiting ryanodine receptor 2 (RyR2) open time prevents neuronal hyperactivity, memory impairment, dendritic spine loss and neuronal cell death in AD mouse models. By contrast, increased RyR2 open probability (Po) exacerbates the onset of familial AD-associated neuronal dysfunction and induces AD-like defects in the absence of AD-causing gene mutations. Thus, RyR2-dependent modulation of neuronal hyperactivity represents a promising new target for combating AD.
Collapse
Affiliation(s)
- Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
2
|
Yao J, Chen SRW. R-carvedilol, a potential new therapy for Alzheimer's disease. Front Pharmacol 2022; 13:1062495. [PMID: 36532759 PMCID: PMC9756136 DOI: 10.3389/fphar.2022.1062495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
Abstract
For decades, the amyloid cascade hypothesis has been the leading hypothesis in studying Alzheimer's disease (AD) pathology and drug development. However, a growing body of evidence indicates that simply removing amyloid plaques may not significantly affect AD progression. Alternatively, it has been proposed that AD progression is driven by increased neuronal excitability. Consistent with this alternative hypothesis, recent studies showed that pharmacologically limiting ryanodine receptor 2 (RyR2) open time with the R-carvedilol enantiomer prevented and reversed neuronal hyperactivity, memory impairment, and neuron loss in AD mouse models without affecting the accumulation of ß-amyloid (Aβ). These data indicate that R-carvedilol could be a potential new therapy for AD.
Collapse
Affiliation(s)
- Jinjing Yao
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada,Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,*Correspondence: Jinjing Yao, ; S. R. Wayne Chen,
| | - S. R. Wayne Chen
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada,Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,*Correspondence: Jinjing Yao, ; S. R. Wayne Chen,
| |
Collapse
|
3
|
Yao J, Liu Y, Sun B, Zhan X, Estillore JP, Turner RW, Chen SRW. Increased RyR2 open probability induces neuronal hyperactivity and memory loss with or without Alzheimer's disease-causing gene mutations. Alzheimers Dement 2022; 18:2088-2098. [PMID: 34985200 DOI: 10.1002/alz.12543] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/01/2021] [Accepted: 10/25/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Neuronal hyperactivity is an early neuronal defect commonly observed in familial and sporadic Alzheimer's disease (AD), but the underlying mechanisms are unclear. METHODS We employed a ryanodine receptor 2 (RyR2) mutant mouse model harboring the R4496C+/- mutation that markedly increases the channel's open probability (Po) to determine the impact of increased RyR2 activity in neuronal function without AD gene mutations. RESULTS Genetically increasing RyR2 Po induced neuronal hyperactivity in vivo in anesthetized and awake mice. Increased RyR2 Po induced hyperactive behaviors, impaired learning and memory, defective dendritic spines, and neuronal cell death. Increased RyR2 Po exacerbated the onset of neuronal hyperexcitability and learning and memory impairments in 5xFAD mice. DISCUSSION Increased RyR2 Po exacerbates the onset of familial AD-associated neuronal dysfunction, and induces AD-like defects in the absence of AD-causing gene mutations, suggesting that RyR2-associated neuronal hyperactivity represents a common target for combating AD with or without AD gene mutations.
Collapse
Affiliation(s)
- Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yajing Liu
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Sun
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Medical School, Kunming University of Science and Technology, Kunming, China
| | - Xiaoqin Zhan
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - John Paul Estillore
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ray W Turner
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
4
|
Bidoli C, Miccoli A, Buonocore F, Fausto AM, Gerdol M, Picchietti S, Scapigliati G. Transcriptome Analysis Reveals Early Hemocyte Responses upon In Vivo Stimulation with LPS in the Stick Insect Bacillus rossius (Rossi, 1788). INSECTS 2022; 13:insects13070645. [PMID: 35886821 PMCID: PMC9316843 DOI: 10.3390/insects13070645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023]
Abstract
Simple Summary Non-model insect species such as B. rossius suffer from a profound gap of knowledge regarding the temporal progression of physiological responses following the challenge with bacterial pathogens or cell wall components thereof. The reason for this mostly lies in the lack of genomic/transcriptomic resources, which would provide an unparalleled in-depth capacity in the analysis of molecular, biochemical, and metabolic mechanisms. We present a high-quality transcriptome obtained from high-coverage sequencing of hemocytes harvested from adult stick insect specimens both pre- and post-LPS stimulation. Such a resource served as the basis for a stringent differential gene expression and functional enrichment analyses, the results of which were characterized and discussed in depth. Selected transcripts encoding for C-type lectins and ML-domain containing proteins were further investigated from a phylogenetic perspective. Overall, these findings shed light on the physiological responses driven by a short-term LPS stimulation in the European stick insect. Abstract Despite a growing number of non-model insect species is being investigated in recent years, a greater understanding of their physiology is prevented by the lack of genomic resources. This is the case of the common European stick insect Bacillus rossius (Rossi, 1788): in this species, some knowledge is available on hemocyte-related defenses, but little is known about the physiological changes occurring in response to natural or experimental challenges. Here, the transcriptional signatures of adult B. rossius hemocytes were investigated after a short-term (2 h) LPS stimulation in vivo: a total of 2191 differentially expressed genes, mostly involved in proteolysis and carbohydrate and lipid metabolic processes, were identified in the de novo assembled transcriptome and in-depth discussed. Overall, the significant modulation of immune signals—such as C-type lectins, ML domain-containing proteins, serpins, as well as Toll signaling-related molecules—provide novel information on the early progression of LPS-induced responses in B. rossius.
Collapse
Affiliation(s)
- Carlotta Bidoli
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (C.B.); (M.G.)
| | - Andrea Miccoli
- Department for Innovation in Biological, Agro-Food and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (F.B.); (A.M.F.); (S.P.); (G.S.)
- Correspondence:
| | - Francesco Buonocore
- Department for Innovation in Biological, Agro-Food and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (F.B.); (A.M.F.); (S.P.); (G.S.)
| | - Anna Maria Fausto
- Department for Innovation in Biological, Agro-Food and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (F.B.); (A.M.F.); (S.P.); (G.S.)
| | - Marco Gerdol
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (C.B.); (M.G.)
| | - Simona Picchietti
- Department for Innovation in Biological, Agro-Food and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (F.B.); (A.M.F.); (S.P.); (G.S.)
| | - Giuseppe Scapigliati
- Department for Innovation in Biological, Agro-Food and Forest Systems, University of Tuscia, 01100 Viterbo, Italy; (F.B.); (A.M.F.); (S.P.); (G.S.)
| |
Collapse
|
5
|
Hiess F, Yao J, Song Z, Sun B, Zhang Z, Huang J, Chen L, Institoris A, Estillore JP, Wang R, Ter Keurs HEDJ, Stys PK, Gordon GR, Zamponi GW, Ganguly A, Chen SRW. Subcellular localization of hippocampal ryanodine receptor 2 and its role in neuronal excitability and memory. Commun Biol 2022; 5:183. [PMID: 35233070 PMCID: PMC8888588 DOI: 10.1038/s42003-022-03124-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/01/2022] [Indexed: 11/09/2022] Open
Abstract
Ryanodine receptor 2 (RyR2) is abundantly expressed in the heart and brain. Mutations in RyR2 are associated with both cardiac arrhythmias and intellectual disability. While the mechanisms of RyR2-linked arrhythmias are well characterized, little is known about the mechanism underlying RyR2-associated intellectual disability. Here, we employed a mouse model expressing a green fluorescent protein (GFP)-tagged RyR2 and a specific GFP probe to determine the subcellular localization of RyR2 in hippocampus. GFP-RyR2 was predominantly detected in the soma and dendrites, but not the dendritic spines of CA1 pyramidal neurons or dentate gyrus granular neurons. GFP-RyR2 was also detected within the mossy fibers in the stratum lucidum of CA3, but not in the presynaptic terminals of CA1 neurons. An arrhythmogenic RyR2-R4496C+/− mutation downregulated the A-type K+ current and increased membrane excitability, but had little effect on the afterhyperpolarization current or presynaptic facilitation of CA1 neurons. The RyR2-R4496C+/− mutation also impaired hippocampal long-term potentiation, learning, and memory. These data reveal the precise subcellular distribution of hippocampal RyR2 and its important role in neuronal excitability, learning, and memory. A mouse model containing a GFP-tagged ryanodine receptor 2 (RyR2) has shed light on the precise subcellular localization of hippocampal RyR2 and mechanisms underlying neuronal excitability, learning, and memory.
Collapse
Affiliation(s)
- Florian Hiess
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Zhenpeng Song
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Bo Sun
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Zizhen Zhang
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Junting Huang
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Lina Chen
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - John Paul Estillore
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Ruiwu Wang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Henk E D J Ter Keurs
- Libin Cardiovascular Institute, Department of Cardiovascular Science, Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Peter K Stys
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.,Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Grant R Gordon
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Gerald W Zamponi
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Anutosh Ganguly
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada. .,Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
6
|
Arnsten AFT, Datta D, Wang M. The genie in the bottle-magnified calcium signaling in dorsolateral prefrontal cortex. Mol Psychiatry 2021; 26:3684-3700. [PMID: 33319854 PMCID: PMC8203737 DOI: 10.1038/s41380-020-00973-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/20/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023]
Abstract
Neurons in the association cortices are particularly vulnerable in cognitive disorders such as schizophrenia and Alzheimer's disease, while those in primary visual cortex remain relatively resilient. This review proposes that the special molecular mechanisms needed for higher cognitive operations confer vulnerability to dysfunction, atrophy, and neurodegeneration when regulation is lost due to genetic and/or environmental insults. Accumulating data suggest that higher cortical circuits rely on magnified levels of calcium (from NMDAR, calcium channels, and/or internal release from the smooth endoplasmic reticulum) near the postsynaptic density to promote the persistent firing needed to maintain, manipulate, and store information without "bottom-up" sensory stimulation. For example, dendritic spines in the primate dorsolateral prefrontal cortex (dlPFC) express the molecular machinery for feedforward, cAMP-PKA-calcium signaling. PKA can drive internal calcium release and promote calcium flow through NMDAR and calcium channels, while in turn, calcium activates adenylyl cyclases to produce more cAMP-PKA signaling. Excessive levels of cAMP-calcium signaling can have a number of detrimental effects: for example, opening nearby K+ channels to weaken synaptic efficacy and reduce neuronal firing, and over a longer timeframe, driving calcium overload of mitochondria to induce inflammation and dendritic atrophy. Thus, calcium-cAMP signaling must be tightly regulated, e.g., by agents that catabolize cAMP or inhibit its production (PDE4, mGluR3), and by proteins that bind calcium in the cytosol (calbindin). Many genetic or inflammatory insults early in life weaken the regulation of calcium-cAMP signaling and are associated with increased risk of schizophrenia (e.g., GRM3). Age-related loss of regulatory proteins which result in elevated calcium-cAMP signaling over a long lifespan can additionally drive tau phosphorylation, amyloid pathology, and neurodegeneration, especially when protective calcium binding proteins are lost from the cytosol. Thus, the "genie" we need for our remarkable cognitive abilities may make us vulnerable to cognitive disorders when we lose essential regulation.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA.
| | - Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| |
Collapse
|
7
|
Elevating the Levels of Calcium Ions Exacerbate Alzheimer's Disease via Inducing the Production and Aggregation of β-Amyloid Protein and Phosphorylated Tau. Int J Mol Sci 2021; 22:ijms22115900. [PMID: 34072743 PMCID: PMC8198078 DOI: 10.3390/ijms22115900] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/08/2021] [Accepted: 05/08/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease with a high incidence rate. The main pathological features of AD are β-amyloid plaques (APs), which are formed by β-amyloid protein (Aβ) deposition, and neurofibrillary tangles (NFTs), which are formed by the excessive phosphorylation of the tau protein. Although a series of studies have shown that the accumulation of metal ions, including calcium ions (Ca2+), can promote the formation of APs and NFTs, there is no systematic review of the mechanisms by which Ca2+ affects the development and progression of AD. In view of this, the current review summarizes the mechanisms by which Ca2+ is transported into and out of cells and organelles, such as the cell, endoplasmic reticulum, mitochondrial and lysosomal membranes to affect the balance of intracellular Ca2+ levels. In addition, dyshomeostasis of Ca2+ plays an important role in modulating the pathogenesis of AD by influencing the production and aggregation of Aβ peptides and tau protein phosphorylation and the ways that disrupting the metabolic balance of Ca2+ can affect the learning ability and memory of people with AD. In addition, the effects of these mechanisms on the synaptic plasticity are also discussed. Finally, the molecular network through which Ca2+ regulates the pathogenesis of AD is introduced, providing a theoretical basis for improving the clinical treatment of AD.
Collapse
|
8
|
Guo W, Wei J, Estillore JP, Zhang L, Wang R, Sun B, Chen SRW. RyR2 disease mutations at the C-terminal domain intersubunit interface alter closed-state stability and channel activation. J Biol Chem 2021; 297:100808. [PMID: 34022226 PMCID: PMC8214192 DOI: 10.1016/j.jbc.2021.100808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 11/19/2022] Open
Abstract
Ryanodine receptors (RyRs) are ion channels that mediate the release of Ca2+ from the sarcoplasmic reticulum/endoplasmic reticulum, mutations of which are implicated in a number of human diseases. The adjacent C-terminal domains (CTDs) of cardiac RyR (RyR2) interact with each other to form a ring-like tetrameric structure with the intersubunit interface undergoing dynamic changes during channel gating. This mobile CTD intersubunit interface harbors many disease-associated mutations. However, the mechanisms of action of these mutations and the role of CTD in channel function are not well understood. Here, we assessed the impact of CTD disease-associated mutations P4902S, P4902L, E4950K, and G4955E on Ca2+− and caffeine-mediated activation of RyR2. The G4955E mutation dramatically increased both the Ca2+-independent basal activity and Ca2+-dependent activation of [3H]ryanodine binding to RyR2. The P4902S and E4950K mutations also increased Ca2+ activation but had no effect on the basal activity of RyR2. All four disease mutations increased caffeine-mediated activation of RyR2 and reduced the threshold for activation and termination of spontaneous Ca2+ release. G4955D dramatically increased the basal activity of RyR2, whereas G4955K mutation markedly suppressed channel activity. Similarly, substitution of P4902 with a negatively charged residue (P4902D), but not a positively charged residue (P4902K), also dramatically increased the basal activity of RyR2. These data suggest that electrostatic interactions are involved in stabilizing the CTD intersubunit interface and that the G4955E disease mutation disrupts this interface, and thus the stability of the closed state. Our studies shed new insights into the mechanisms of action of RyR2 CTD disease mutations.
Collapse
Affiliation(s)
- Wenting Guo
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Jinhong Wei
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - John Paul Estillore
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Lin Zhang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Ruiwu Wang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Bo Sun
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada; Medical School, Kunming University of Science and Technology, Kunming, China.
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
9
|
Yao J, Sun B, Institoris A, Zhan X, Guo W, Song Z, Liu Y, Hiess F, Boyce AKJ, Ni M, Wang R, Ter Keurs H, Back TG, Fill M, Thompson RJ, Turner RW, Gordon GR, Chen SRW. Limiting RyR2 Open Time Prevents Alzheimer's Disease-Related Neuronal Hyperactivity and Memory Loss but Not β-Amyloid Accumulation. Cell Rep 2021; 32:108169. [PMID: 32966798 PMCID: PMC7532726 DOI: 10.1016/j.celrep.2020.108169] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 07/23/2020] [Accepted: 08/27/2020] [Indexed: 12/31/2022] Open
Abstract
Neuronal hyperactivity is an early primary dysfunction in Alzheimer’s disease (AD) in humans and animal models, but effective neuronal hyperactivity-directed anti-AD therapeutic agents are lacking. Here we define a previously unknown mode of ryanodine receptor 2 (RyR2) control of neuronal hyperactivity and AD progression. We show that a single RyR2 point mutation, E4872Q, which reduces RyR2 open time, prevents hyperexcitability, hyperactivity, memory impairment, neuronal cell death, and dendritic spine loss in a severe early-onset AD mouse model (5xFAD). The RyR2-E4872Q mutation upregulates hippocampal CA1-pyramidal cell A-type K+ current, a well-known neuronal excitability control that is downregulated in AD. Pharmacologically limiting RyR2 open time with the R-carvedilol enantiomer (but not racemic carvedilol) prevents and rescues neuronal hyperactivity, memory impairment, and neuron loss even in late stages of AD. These AD-related deficits are prevented even with continued β-amyloid accumulation. Thus, limiting RyR2 open time may be a hyperactivity-directed, non-β-amyloid-targeted anti-AD strategy. Yao et al. show that genetically or pharmacologically limiting the open duration of ryanodine receptor 2 upregulates the A-type potassium current and prevents neuronal hyperexcitability and hyperactivity, memory impairment, neuronal cell death, and dendritic spine loss in a severe early-onset Alzheimer’s disease mouse model, even with continued accumulation of β-amyloid.
Collapse
Affiliation(s)
- Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Bo Sun
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; Medical School, Kunming University of Science and Technology, Kunming 650504, China
| | - Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Xiaoqin Zhan
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Wenting Guo
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Zhenpeng Song
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Yajing Liu
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Florian Hiess
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Andrew K J Boyce
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mingke Ni
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Ruiwu Wang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Henk Ter Keurs
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Thomas G Back
- Department of Chemistry, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Michael Fill
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL 60612, USA
| | - Roger J Thompson
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Ray W Turner
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Grant R Gordon
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
10
|
Zhang H. Aluminum-Induced Electrophysiological Variation, Synaptic Plasticity Impairment, and Related Mechanism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1091:161-172. [PMID: 30315454 DOI: 10.1007/978-981-13-1370-7_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Aluminum, an environmentally abundant non-redox trivalent cation, has long been reported to alter blood-brain barrier and gets deposited in different regions of the brain. Many reports strongly indicated that Al had an adverse impact on the central nervous system (CNS), particularly on cognitive ability. Until now, studies in animal models and cell cultures have revealed that Al exposure results in altered behavioral performance and memory damage. The present paper reviews the scientific literature linking aluminum and the impairment of electrophysiological variation and synaptic plasticity. The focus is on the changes of electrical excitability, voltage-operated ion channels, and synaptic plasticity induced by aluminum. A detailed mechanism of the role of aluminum in hippocampal LTP which is the most widely studied example of synaptic plasticity is highlighted. Evidence revealed that glutamate-NO-cGMP, PLC, Ca2+-CaM-CaMKII, MAPK, and Wnt pathway may be important in the mechanism underlying Al-induced long-term memory impairment. Further studies are required to establish the upstream activators and downstream effectors of these cascades and to answer how so many signaling cascades relate to the other signaling processes that might be involved in the Al-induced inhibition of synaptic plasticity.
Collapse
Affiliation(s)
- Huifang Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
11
|
Reuveni I, Barkai E. Tune it in: mechanisms and computational significance of neuron-autonomous plasticity. J Neurophysiol 2018; 120:1781-1795. [DOI: 10.1152/jn.00102.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The activity of a neural network is a result of synaptic signals that convey the communication between neurons and neuron-based intrinsic currents that determine the neuron’s input-output transfer function. Ample studies have demonstrated that cell-based excitability, and in particular intrinsic excitability, is modulated by learning and that these modifications play a key role in learning-related behavioral changes. The field of cell-based plasticity is largely growing, and it entails numerous experimental findings that demonstrate a large diversity of currents that are affected by learning. The diverse effect of learning on the neuron’s excitability emphasizes the need for a framework under which cell-based plasticity can be categorized to enable the assessment of the computational roles of the intrinsic modifications. We divide the domain of cell-based plasticity into three main categories, where the first category entails the currents that mediate the passive properties and single-spike generation, the second category entails the currents that mediate spike frequency adaptation, and the third category entails a novel learning-induced mechanism where all excitatory and inhibitory synapses double their strength. Curiously, this elementary division enables a natural categorization of the computational roles of these learning-induced plasticities. The computational roles are diverse and include modification of the neuronal mode of action, such as bursting, prolonged, and fast responsive; attention-like effect where the signal detection is improved; transfer of the network into an active state; biasing the competition for memory allocation; and transforming an environmental cue into a dominant cue and enabling a quicker formation of new memories.
Collapse
Affiliation(s)
- Iris Reuveni
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Edi Barkai
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
12
|
Pregnolato M, Damiani G, Pereira A. Patterns of calcium signaling: A link between chronic emotions and cancer. J Integr Neurosci 2018; 16:S43-S63. [PMID: 29154288 DOI: 10.3233/jin-170066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Intra and inter-cellular calcium signaling is present in all types of cells and body tissues. In the human brain, calcium currents and waves are related to mental activities, including emotions. We present a theoretical interpretation of these phenomena suggesting their involvement in chronic emotional patterns and in the pathology of cancer. Recent developments on biophysics, translational biology and psychoneuroendocrinoimmunology (PNEI) can support explanatory hypotheses about the link between emotional stresses and the origin and development of different types of tumor cells. Chronic stresses may cause perturbations of rhythms of the PNEI system, excessive activation of HPA axis and abnormal activation of calcium signals in somatic tissues, with deleterious effects on different parts of the body. The increasing of calcium signaling inside cells may lead to a deregulation of different pathways and epigenetic systems that promote the production of genomic mutations in a second phase. In particular, the hyperactivation of the transcription nuclear factor kappaB (NF-κB), if is not counterbalanced by the following activation of the nuclear factor (erythroid-derived 2)-like 2 (NFE2L2 or Nrf2), increases the production of oxidative catabolites, as the advanced glycation end products (AGE), which play a key role in the progression of different types of cancer and other degenerative diseases. Cortisol binding to glucocorticoid receptor (GR) reduces the activity of both NF-κB and Nrf2 inside the cells but inhibits the cellular immunity and the anabolic processes of tissue regeneration. The tissue atrophy and the defective anti-ageing mechanisms promotes the tumoral cells growth and their escape from the immune-surveillance.
Collapse
Affiliation(s)
| | | | - Alfredo Pereira
- Institute of Biosciences, São Paulo State University, Brasil. E-mail:
| |
Collapse
|
13
|
Castellano-Muñoz M, Schnee ME, Ricci AJ. Calcium-induced calcium release supports recruitment of synaptic vesicles in auditory hair cells. J Neurophysiol 2015; 115:226-39. [PMID: 26510758 DOI: 10.1152/jn.00559.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 10/23/2015] [Indexed: 01/31/2023] Open
Abstract
Hair cells from auditory and vestibular systems transmit continuous sound and balance information to the central nervous system through the release of synaptic vesicles at ribbon synapses. The high activity experienced by hair cells requires a unique mechanism to sustain recruitment and replenishment of synaptic vesicles for continuous release. Using pre- and postsynaptic electrophysiological recordings, we explored the potential contribution of calcium-induced calcium release (CICR) in modulating the recruitment of vesicles to auditory hair cell ribbon synapses. Pharmacological manipulation of CICR with agents targeting endoplasmic reticulum calcium stores reduced both spontaneous postsynaptic multiunit activity and the frequency of excitatory postsynaptic currents (EPSCs). Pharmacological treatments had no effect on hair cell resting potential or activation curves for calcium and potassium channels. However, these drugs exerted a reduction in vesicle release measured by dual-sine capacitance methods. In addition, calcium substitution by barium reduced release efficacy by delaying release onset and diminishing vesicle recruitment. Together these results demonstrate a role for calcium stores in hair cell ribbon synaptic transmission and suggest a novel contribution of CICR in hair cell vesicle recruitment. We hypothesize that calcium entry via calcium channels is tightly regulated to control timing of vesicle fusion at the synapse, whereas CICR is used to maintain a tonic calcium signal to modulate vesicle trafficking.
Collapse
Affiliation(s)
- Manuel Castellano-Muñoz
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, California; and
| | - Michael E Schnee
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, California; and
| | - Anthony J Ricci
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, California; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
14
|
Sun MK, Nelson TJ, Alkon DL. Towards universal therapeutics for memory disorders. Trends Pharmacol Sci 2015; 36:384-94. [DOI: 10.1016/j.tips.2015.04.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/07/2015] [Accepted: 04/08/2015] [Indexed: 12/22/2022]
|
15
|
Del Prete D, Checler F, Chami M. Ryanodine receptors: physiological function and deregulation in Alzheimer disease. Mol Neurodegener 2014; 9:21. [PMID: 24902695 PMCID: PMC4063224 DOI: 10.1186/1750-1326-9-21] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 05/18/2014] [Indexed: 12/21/2022] Open
Abstract
Perturbed Endoplasmic Reticulum (ER) calcium (Ca2+) homeostasis emerges as a central player in Alzheimer disease (AD). Accordingly, different studies have reported alterations of the expression and the function of Ryanodine Receptors (RyR) in human AD-affected brains, in cells expressing familial AD-linked mutations on the β amyloid precursor protein (βAPP) and presenilins (the catalytic core in γ-secretase complexes cleaving the βAPP, thereby generating amyloid β (Aβ) peptides), as well as in the brain of various transgenic AD mice models. Data converge to suggest that RyR expression and function alteration are associated to AD pathogenesis through the control of: i) βAPP processing and Aβ peptide production, ii) neuronal death; iii) synaptic function; and iv) memory and learning abilities. In this review, we document the network of evidences suggesting that RyR could play a complex dual "compensatory/protective versus pathogenic" role contributing to the setting of histopathological lesions and synaptic deficits that are associated with the disease stages. We also discuss the possible mechanisms underlying RyR expression and function alterations in AD. Finally, we review recent publications showing that drug-targeting blockade of RyR and genetic manipulation of RyR reduces Aβ production, stabilizes synaptic transmission, and prevents learning and memory deficits in various AD mouse models. Chemically-designed RyR "modulators" could therefore be envisioned as new therapeutic compounds able to delay or block the progression of AD.
Collapse
Affiliation(s)
| | - Frédéric Checler
- Université de Nice Sophia Antipolis, IPMC, Sophia Antipolis, Nice, F-06560 Valbonne, France.
| | | |
Collapse
|
16
|
Xu C, Liu QY, Alkon DL. PKC activators enhance GABAergic neurotransmission and paired-pulse facilitation in hippocampal CA1 pyramidal neurons. Neuroscience 2014; 268:75-86. [PMID: 24637095 DOI: 10.1016/j.neuroscience.2014.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 11/18/2022]
Abstract
Bryostatin-1, a potent agonist of protein kinase C (PKC), has recently been found to enhance spatial learning and long-term memory in rats, mice, rabbits and the nudibranch Hermissenda, and to exert profound neuroprotective effects on Alzheimer's disease (AD) in transgenic mice. However, details of the mechanistic effects of bryostatin on learning and memory remain unclear. To address this issue, whole-cell recording, a dual-recording approach and extracellular recording techniques were performed on young (2-4months) Brown-Norway rats. We found that bath-applied bryostatin-1 significantly increased the frequency and amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs). The firing rate of GABAergic interneurons significantly was also increased as recorded with a loosely-attached extracellular recording configuration. Simultaneous recordings from communicating cell pairs of interneuron and pyramidal neuron revealed unique activity-dependent properties of GABAergic synapses. Furthermore, the bryostatin-induced increase of the frequency and amplitude of IPSCs was blocked by methionine enkephalin which selectively suppressed the excitability of interneurons. Pretreatment with RO-32-0432, a relatively specific PKCα antagonist, blocked the effect of bryostatin on sIPSCs. Finally, bryostatin increased paired-pulse ratio of GABAergic synapses that lasted for at least 20min while pretreatment with RO-32-0432 significantly reduced the ratio. In addition, 8-[2-(2-pentyl-cyclopropylmethl)-cyclopropyl]-octanoic acid (DCP-LA), a selective PKCε activator, also increased the frequency and amplitude of sIPSCs. Taken together, these results suggest that bryostatin enhances GABAergic neurotransmission in pyramidal neurons by activating the PKCα & ε-dependent pathway and by a presynaptic mechanism with excitation of GABAergic interneurons. These effects of bryostatin on GABAergic transmissions and modifiability may contribute to the improvement of learning and memory previously observed to be induced by bryostatin.
Collapse
Affiliation(s)
- C Xu
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV 26506, United States of America.
| | - Q-Y Liu
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV 26506, United States of America
| | - D L Alkon
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV 26506, United States of America
| |
Collapse
|
17
|
Takigami S, Sunada H, Lukowiak K, Kuzirian AM, Alkon DL, Sakakibara M. Protein kinase C mediates memory consolidation of taste avoidance conditioning in Lymnaea stagnalis. Neurobiol Learn Mem 2014; 111:9-18. [PMID: 24613854 DOI: 10.1016/j.nlm.2014.02.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 02/05/2014] [Accepted: 02/23/2014] [Indexed: 11/25/2022]
Abstract
In Lymnaea stagnalis, in order to obtain a 10 min short-term memory (STM) of taste avoidance conditioning (TAC) at least 10 paired presentations of a conditioned stimulus (CS), sucrose, and an unconditioned stimulus (US), tactile stimulation to the animal's head, are required. Pre-exposure of snails to the protein kinase C (PKC) α and ε activator bryostatin (Bryo) facilitated STM formation in that only 5 paired CS-US trials were required. Typically 20 paired presentations of the CS-US are required for formation of STM and LTM. However, 20 paired presentations do not result in STM or LTM if snails are pre-incubated with a PKC inhibitor, Ro-32-0432. We also found that LTM lasting longer than 48 h was acquired with Bryo incubation for 45 min even after termination of the conditioning paradigm. These data suggest that activation of the α and ε isozymes of PKC is crucially involved in the formation of LTM and provide further support for a mechanism that has been conserved across the evolution of species ranging from invertebrate molluscs to higher mammals.
Collapse
Affiliation(s)
- Satoshi Takigami
- Graduate School of Bioscience, Tokai University, 410-0321 Numazu, Shizuoka, Japan
| | - Hiroshi Sunada
- Hotchkiss Brain Institute, University of Calgary, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Ken Lukowiak
- Hotchkiss Brain Institute, University of Calgary, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Alan M Kuzirian
- Program in Sensory Physiology & Behavior, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Daniel L Alkon
- Blanchette Rockefeller Neuroscience Institute, 9601 Medical Center Drive, Rockville, MD 20850-3332, USA
| | - Manabu Sakakibara
- Graduate School of Bioscience, Tokai University, 410-0321 Numazu, Shizuoka, Japan; School of High-Technology for Human Welfare, Tokai University, 410-0321 Numazu, Shizuoka, Japan.
| |
Collapse
|
18
|
The "memory kinases": roles of PKC isoforms in signal processing and memory formation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 122:31-59. [PMID: 24484697 DOI: 10.1016/b978-0-12-420170-5.00002-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The protein kinase C (PKC) isoforms, which play an essential role in transmembrane signal conduction, can be viewed as a family of "memory kinases." Evidence is emerging that they are critically involved in memory acquisition and maintenance, in addition to their involvement in other functions of cells. Deficits in PKC signal cascades in neurons are one of the earliest abnormalities in the brains of patients suffering from Alzheimer's disease. Their dysfunction is also involved in several other types of memory impairments, including those related to emotion, mental retardation, brain injury, and vascular dementia/ischemic stroke. Inhibition of PKC activity leads to a reduced capacity of many types of learning and memory, but may have therapeutic values in treating substance abuse or aversive memories. PKC activators, on the other hand, have been shown to possess memory-enhancing and antidementia actions. PKC pharmacology may, therefore, represent an attractive area for developing effective cognitive drugs for the treatment of many types of memory disorders and dementias.
Collapse
|
19
|
Uno Y, Fujiyuki T, Morioka M, Kubo T. Mushroom body-preferential expression of proteins/genes involved in endoplasmic reticulum Ca(2+)-transport in the worker honeybee (Apis mellifera L.) brain. INSECT MOLECULAR BIOLOGY 2013; 22:52-61. [PMID: 23170949 DOI: 10.1111/imb.12002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
To identify the molecular characteristics specific to the mushroom body (MB, a higher processing centre) neurones in the honeybee brain, we previously used proteomics to identify proteins that are preferentially expressed in these MBs. Here we continued our proteomic analysis to show that reticulocalbin, which is involved in endoplasmic reticulum (ER) Ca(2+) transport, is also preferentially expressed in the MBs in the honeybee brain. Gene expression analysis revealed that reticulocalbin is preferentially expressed in the large-type Kenyon cells, which are MB-intrinsic neurones. In addition, the gene for the ryanodine receptor, which is also involved in ER Ca(2+) transport, was also preferentially expressed in the large-type Kenyon cells. In contrast, the expression of three other ER-related genes, protein disulphide isomerase, sec61 and erp60, was not enriched in the MBs. These findings further support the notion that the function of ER Ca(2+)-signalling, but not the mere intracellular density of ER, is specifically enhanced in the large-type Kenyon cells in the honeybee brain.
Collapse
Affiliation(s)
- Y Uno
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | | | | | | |
Collapse
|
20
|
Wayman GA, Bose DD, Yang D, Lesiak A, Bruun D, Impey S, Ledoux V, Pessah IN, Lein PJ. PCB-95 modulates the calcium-dependent signaling pathway responsible for activity-dependent dendritic growth. ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:1003-9. [PMID: 22534176 PMCID: PMC3404671 DOI: 10.1289/ehp.1104833] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 04/02/2012] [Indexed: 05/17/2023]
Abstract
BACKGROUND Non-dioxin-like (NDL) polychlorinated biphenyls (PCBs) promote dendritic growth in hippocampal neurons via ryanodine receptor (RyR)-dependent mechanisms; however, downstream signaling events that link enhanced RyR activity to dendritic growth are unknown. Activity-dependent dendritic growth, which is a critical determinant of neuronal connectivity in the developing brain, is mediated by calcium ion (Ca(2+))-dependent activation of Ca(2+)/calmodulin kinase-I (CaMKI), which triggers cAMP response element binding protein (CREB)-dependent Wnt2 transcription. RyRs regulate the spatiotemporal dynamics of intracellular Ca(2+) signals, but whether RyRs promote dendritic growth via modulation of this signaling pathway is not known. OBJECTIVE We tested the hypothesis that the CaMKI-CREB-Wnt2 signaling pathway couples NDL PCB-enhanced RyR activity to dendritic arborization. METHODS AND RESULTS Ca(2+) imaging of dissociated cultures of primary rat hippocampal neurons indicated that PCB-95 (2,2',3,5'6-pentachlorobiphenyl; a potent RyR potentiator), enhanced synchronized Ca(2+) oscillations in somata and dendrites that were blocked by ryanodine. As determined by Western blotting and quantitative polymerase chain reaction, PCB-95 also activated CREB and up-regulated Wnt2. Blocking CaMKK, CaMKIα/γ, MEK/ERK, CREB, or Wnt2 prevented PCB-95-induced dendritic growth. Antagonism of γ-aminobutyric acid (GABA) receptors with bicuculline (BIC) phenocopied the dendrite-promoting effects of PCB-95, and pharmacological antagonism or siRNA knockdown of RyR blocked BIC-induced dendritic growth in dissociated and slice cultures of hippocampal neurons. CONCLUSIONS RyR activity contributes to dynamic remodeling of dendritic architecture in response to NDL PCBs via CaMKI-CREB-Wnt2 signaling in rats. Our findings identify PCBs as candidate environmental risk factors for neurodevelopmental disorders, especially in children with heritable deficits in calcium signaling associated with autism.
Collapse
Affiliation(s)
- Gary A Wayman
- Program in Neuroscience, Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, Washington State University, Pullman, Washington, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kurokawa K, Mizuno K, Ohkuma S. Increase of ryanodine receptors by dopamine D1 receptors is negatively regulated by γ-aminobutyric acid type B receptors in primary cultures of mouse cerebral cortical neurons. J Neurosci Res 2012; 90:1626-38. [PMID: 22504960 DOI: 10.1002/jnr.23058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 02/23/2012] [Accepted: 03/04/2012] [Indexed: 11/06/2022]
Abstract
Although upregulation of ryanodine receptor (RyR)-1 and -2 is mediated through the activation of dopamine D1 receptors (D1DRs) in the development of psychostimulant-induced place preference, little is known about how such increased expressions of RyRs are negatively regulated. This study investigated negative regulatory mechanisms of increase of RyR-1 and -2 expression by D1DR stimulation with its full agonist, SKF82958 or A 68930, using cultures of mouse cerebral cortical neurons. Sustained exposure to SKF82958 or A 68930 of the neurons increased RyR-1 and -2 proteins in a dose- and time-dependent-manner. The SKF82958-induced increases of RyR-1 and -2 proteins were significantly suppressed by SCH23390 (a selective D1DR antagonist). In addition, the SKF82958- or A 68930-induced increases of RyR-1 and -2 proteins were completely abolished by baclofen (a selective γ-aminobutyric acid type B [GABA(B)] receptor agonist), whereas muscimol (an agonist specific to GABA(A) receptors) had no effect. SKF82958 or A 68930 significantly increased intracellular cAMP level, which was completely suppressed by baclofen. Furthermore, sustained exposure to phorbol 12,13-dibutyrate, a protein kinase C activator, did not change the expression of RyR-1 or -2 proteins. Immunohistochemical study showed colocalizaton of immunoreactivities for three types of proteins, D1DRs and GABA(B) receptor R1 and R2 subunits in the same neuronal bodies, suggesting that the neurochemical changes induced by the activation of D1DRs and GABA(B) receptors occur in the same neurons. These results indicate that RyR-1 and -2 expression facilitated by D1DR stimulation are negatively regulated by GABA(B) receptor via suppression of cAMP production.
Collapse
Affiliation(s)
- Kazuhiro Kurokawa
- Department of Pharmacology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | | | | |
Collapse
|
22
|
Sun MK, Alkon DL. Activation of protein kinase C isozymes for the treatment of dementias. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 64:273-302. [PMID: 22840750 DOI: 10.1016/b978-0-12-394816-8.00008-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Memories are much more easily impaired than improved. Dementias, a lasting impairment of memory function, occur in a variety of cognitive disorders and become more clinically dominant as the population ages. Protein kinase C is one of the "cognitive kinases," and plays an essential role in both memory acquisition and maintenance. Deficits in protein kinase C (PKC) signal cascades in neurons represent one of the earliest changes in the brains of patients with Alzheimer's disease (AD) and other types of memory impairment, including those related to cerebral ischemia and ischemic stroke. Inhibition or impairment of PKC activity results in compromised learning and memory, whereas an appropriate activation of certain PKC isozymes leads to an enhancement of learning and memory and/or antidementic effects. In preclinical studies, PKC activators have been shown to increase the expression and activity of PKC isozymes, thereby restoring PKC signaling and downstream activity, including stimulation of neurotrophic activity, synaptic/structural remodeling, and synaptogenesis in the hippocampus and related cortical areas. PKC activators also reduce the accumulation of neurotoxic amyloid and tau protein hyperphosphorylation and support anti-apoptotic processes in the brain. These observations strongly suggest that PKC pharmacology may represent an attractive area for the development of effective cognition-enhancing therapeutics for the treatment of dementias.
Collapse
Affiliation(s)
- Miao-Kun Sun
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV, USA
| | | |
Collapse
|
23
|
van Welie I, du Lac S. Bidirectional control of BK channel open probability by CAMKII and PKC in medial vestibular nucleus neurons. J Neurophysiol 2011; 105:1651-9. [PMID: 21307321 DOI: 10.1152/jn.00058.2011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Large conductance K(+) (BK) channels are a key determinant of neuronal excitability. Medial vestibular nucleus (MVN) neurons regulate eye movements to ensure image stabilization during head movement, and changes in their intrinsic excitability may play a critical role in plasticity of the vestibulo-ocular reflex. Plasticity of intrinsic excitability in MVN neurons is mediated by kinases, and BK channels influence excitability, but whether endogenous BK channels are directly modulated by kinases is unknown. Double somatic patch-clamp recordings from MVN neurons revealed large conductance potassium channel openings during spontaneous action potential firing. These channels displayed Ca(2+) and voltage dependence in excised patches, identifying them as BK channels. Recording isolated single channel currents at physiological temperature revealed a novel kinase-mediated bidirectional control in the range of voltages over which BK channels are activated. Application of activated Ca(2+)/calmodulin-dependent kinase II (CAMKII) increased BK channel open probability by shifting the voltage activation range towards more hyperpolarized potentials. An opposite shift in BK channel open probability was revealed by inhibition of phosphatases and was occluded by blockade of protein kinase C (PKC), suggesting that active PKC associated with BK channel complexes in patches was responsible for this effect. Accordingly, direct activation of endogenous PKC by PMA induced a decrease in BK open probability. BK channel activity affects excitability in MVN neurons and bidirectional control of BK channels by CAMKII, and PKC suggests that cellular signaling cascades engaged during plasticity may dynamically control excitability by regulating BK channel open probability.
Collapse
Affiliation(s)
- Ingrid van Welie
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, San Diego, CA 92037, USA
| | | |
Collapse
|
24
|
Pereira A, Furlan FA. Astrocytes and human cognition: modeling information integration and modulation of neuronal activity. Prog Neurobiol 2010; 92:405-20. [PMID: 20633599 DOI: 10.1016/j.pneurobio.2010.07.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Revised: 06/18/2010] [Accepted: 07/06/2010] [Indexed: 10/19/2022]
Abstract
Recent research focusing on the participation of astrocytes in glutamatergic tripartite synapses has revealed mechanisms that support cognitive functions common to human and other mammalian species, such as learning, perception, conscious integration, memory formation/retrieval and the control of voluntary behavior. Astrocytes can modulate neuronal activity by means of release of glutamate, d-serine, adenosine triphosphate and other signaling molecules, contributing to sustain, reinforce or depress pre- and post-synaptic membranes. We review molecular mechanisms present in tripartite synapses and model the cognitive role of astrocytes. Single protoplasmic astrocytes operate as a "Local Hub", integrating information patterns from neuronal and glial populations. Two mechanisms, here modeled as the "domino" and "carousel" effects, contribute to the formation of intercellular calcium waves. As waves propagate through gap junctions and reach other types of astrocytes (interlaminar, polarized, fibrous and varicose projection), the active astroglial network functions as a "Master Hub" that integrates results of distributed processing from several brain areas and supports conscious states. Response of this network would define the effect exerted on neuronal plasticity (membrane potentiation or depression), behavior and psychosomatic processes. Theoretical results of our modeling can contribute to the development of new experimental research programs to test cognitive functions of astrocytes.
Collapse
Affiliation(s)
- Alfredo Pereira
- Institute of Biosciences, State University of São Paulo (UNESP), Campus Rubião Jr., 18618-000 Botucatu-SP, Brazil.
| | | |
Collapse
|
25
|
Gericke GS. Common chromosomal fragile sites (CFS) may be involved in normal and traumatic cognitive stress memory consolidation and altered nervous system immunity. Med Hypotheses 2010; 74:911-8. [PMID: 20138440 DOI: 10.1016/j.mehy.2009.05.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 05/22/2009] [Accepted: 05/26/2009] [Indexed: 11/18/2022]
Abstract
Previous reports of specific patterns of increased fragility at common chromosomal fragile sites (CFS) found in association with certain neurobehavioural disorders did not attract attention at the time due to a shift towards molecular approaches to delineate neuropsychiatric disorder candidate genes. Links with miRNA, altered methylation and the origin of copy number variation indicate that CFS region characteristics may be part of chromatinomic mechanisms that are increasingly linked with neuroplasticity and memory. Current reports of large-scale double-stranded DNA breaks in differentiating neurons and evidence of ongoing DNA demethylation of specific gene promoters in adult hippocampus may shed new light on the dynamic epigenetic changes that are increasingly appreciated as contributing to long-term memory consolidation. The expression of immune recombination activating genes in key stress-induced memory regions suggests the adoption by the brain of this ancient pattern recognition and memory system to establish a structural basis for long-term memory through controlled chromosomal breakage at highly specific genomic regions. It is furthermore considered that these mechanisms for management of epigenetic information related to stress memory could be linked, in some instances, with the transfer of the somatically acquired information to the germline. Here, rearranged sequences can be subjected to further selection and possible eventual retrotranscription to become part of the more stable coding machinery if proven to be crucial for survival and reproduction. While linkage of cognitive memory with stress and fear circuitry and memory establishment through structural DNA modification is proposed as a normal process, inappropriate activation of immune-like genomic rearrangement processes through traumatic stress memory may have the potential to lead to undesirable activation of neuro-inflammatory processes. These theories could have a significant impact on the interpretation of risks posed by heredity and the environment and the search for neuropsychiatric candidate genes.
Collapse
Affiliation(s)
- G S Gericke
- Department of Biomedical Sciences, Tshwane University of Technology, Brooklyn Square, Pretoria, Gauteng, South Africa.
| |
Collapse
|
26
|
Pessah IN, Cherednichenko G, Lein PJ. Minding the calcium store: Ryanodine receptor activation as a convergent mechanism of PCB toxicity. Pharmacol Ther 2010; 125:260-85. [PMID: 19931307 PMCID: PMC2823855 DOI: 10.1016/j.pharmthera.2009.10.009] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 10/30/2009] [Indexed: 11/24/2022]
Abstract
Chronic low-level polychlorinated biphenyl (PCB) exposures remain a significant public health concern since results from epidemiological studies indicate that PCB burden is associated with immune system dysfunction, cardiovascular disease, and impairment of the developing nervous system. Of these various adverse health effects, developmental neurotoxicity has emerged as a particularly vulnerable endpoint in PCB toxicity. Arguably the most pervasive biological effects of PCBs could be mediated by their ability to alter the spatial and temporal fidelity of Ca2+ signals through one or more receptor-mediated processes. This review will focus on our current knowledge of the structure and function of ryanodine receptors (RyRs) in muscle and nerve cells and how PCBs and related non-coplanar structures alter these functions. The molecular and cellular mechanisms by which non-coplanar PCBs and related structures alter local and global Ca2+ signaling properties and the possible short and long-term consequences of these perturbations on neurodevelopment and neurodegeneration are reviewed.
Collapse
Affiliation(s)
- Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.
| | | | | |
Collapse
|
27
|
Sun MK, Alkon DL. Protein kinase C activators as synaptogenic and memory therapeutics. Arch Pharm (Weinheim) 2010; 342:689-98. [PMID: 19899099 DOI: 10.1002/ardp.200900050] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The last decade has witnessed a rapid progress in understanding of the molecular cascades that may underlie memory and memory disorders. Among the critical players, activity of protein kinase C (PKC) isoforms is essential for many types of learning and memory and their dysfunction, and is critical in memory disorders. PKC inhibition and functional deficits lead to an impairment of various types of learning and memory, consistent with the observations that neurotoxic amyloid inhibits PKC activity and that transgenic animal models with PKCbeta deficit exhibit impaired capacity in cognition. In addition, PKC isozymes play a regulatory role in amyloid production and accumulation. Restoration of the impaired PKC signal pathway pharmacologically results in an enhanced memory capacity and synaptic remodeling / repair and synaptogenesis, and, therefore, represents a potentially important strategy for the treatment of memory disorders, including Alzheimer's dementia. The PKC activators, especially those that are isozyme-specific, are a new class of drug candidates that may be developed as future memory therapeutics.
Collapse
Affiliation(s)
- Miao-Kun Sun
- Blanchette Rockefeller Neurosciences Institute, Rockville, MD 20850, USA.
| | | |
Collapse
|
28
|
Garcia L, Saraiva Garcia CH, Calábria LK, Costa Nunes da Cruz G, Sánchez Puentes A, Báo SN, Fontes W, Ricart CAO, Salmen Espindola F, Valle de Sousa M. Proteomic Analysis of Honey Bee Brain upon Ontogenetic and Behavioral Development. J Proteome Res 2009; 8:1464-73. [DOI: 10.1021/pr800823r] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Liudy Garcia
- Mass Spectrometry Group, Physics Department, CEADEN, Havana, Cuba, Brazilian Center for Protein Research, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil, Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil, Department for Proteome Analysis, CIGB, Havana, Cuba, and Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil
| | - Carlos H. Saraiva Garcia
- Mass Spectrometry Group, Physics Department, CEADEN, Havana, Cuba, Brazilian Center for Protein Research, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil, Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil, Department for Proteome Analysis, CIGB, Havana, Cuba, and Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil
| | - Luciana Karen Calábria
- Mass Spectrometry Group, Physics Department, CEADEN, Havana, Cuba, Brazilian Center for Protein Research, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil, Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil, Department for Proteome Analysis, CIGB, Havana, Cuba, and Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil
| | - Gabriel Costa Nunes da Cruz
- Mass Spectrometry Group, Physics Department, CEADEN, Havana, Cuba, Brazilian Center for Protein Research, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil, Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil, Department for Proteome Analysis, CIGB, Havana, Cuba, and Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil
| | - Aniel Sánchez Puentes
- Mass Spectrometry Group, Physics Department, CEADEN, Havana, Cuba, Brazilian Center for Protein Research, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil, Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil, Department for Proteome Analysis, CIGB, Havana, Cuba, and Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil
| | - Sonia N. Báo
- Mass Spectrometry Group, Physics Department, CEADEN, Havana, Cuba, Brazilian Center for Protein Research, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil, Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil, Department for Proteome Analysis, CIGB, Havana, Cuba, and Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil
| | - Wagner Fontes
- Mass Spectrometry Group, Physics Department, CEADEN, Havana, Cuba, Brazilian Center for Protein Research, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil, Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil, Department for Proteome Analysis, CIGB, Havana, Cuba, and Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil
| | - Carlos A. O. Ricart
- Mass Spectrometry Group, Physics Department, CEADEN, Havana, Cuba, Brazilian Center for Protein Research, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil, Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil, Department for Proteome Analysis, CIGB, Havana, Cuba, and Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil
| | - Foued Salmen Espindola
- Mass Spectrometry Group, Physics Department, CEADEN, Havana, Cuba, Brazilian Center for Protein Research, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil, Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil, Department for Proteome Analysis, CIGB, Havana, Cuba, and Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil
| | - Marcelo Valle de Sousa
- Mass Spectrometry Group, Physics Department, CEADEN, Havana, Cuba, Brazilian Center for Protein Research, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil, Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil, Department for Proteome Analysis, CIGB, Havana, Cuba, and Laboratory of Electron Microscopy, Department of Cell Biology, University of Brasília, Brasília, DF, Brazil
| |
Collapse
|
29
|
Uhl GR, Drgon T, Johnson C, Li CY, Contoreggi C, Hess J, Naiman D, Liu QR. Molecular genetics of addiction and related heritable phenotypes: genome-wide association approaches identify "connectivity constellation" and drug target genes with pleiotropic effects. Ann N Y Acad Sci 2008; 1141:318-81. [PMID: 18991966 PMCID: PMC3922196 DOI: 10.1196/annals.1441.018] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Genome-wide association (GWA) can elucidate molecular genetic bases for human individual differences in complex phenotypes that include vulnerability to addiction. Here, we review (a) evidence that supports polygenic models with (at least) modest heterogeneity for the genetic architectures of addiction and several related phenotypes; (b) technical and ethical aspects of importance for understanding GWA data, including genotyping in individual samples versus DNA pools, analytic approaches, power estimation, and ethical issues in genotyping individuals with illegal behaviors; (c) the samples and the data that shape our current understanding of the molecular genetics of individual differences in vulnerability to substance dependence and related phenotypes; (d) overlaps between GWA data sets for dependence on different substances; and (e) overlaps between GWA data for addictions versus other heritable, brain-based phenotypes that include bipolar disorder, cognitive ability, frontal lobe brain volume, the ability to successfully quit smoking, neuroticism, and Alzheimer's disease. These convergent results identify potential targets for drugs that might modify addictions and play roles in these other phenotypes. They add to evidence that individual differences in the quality and quantity of brain connections make pleiotropic contributions to individual differences in vulnerability to addictions and to related brain disorders and phenotypes. A "connectivity constellation" of brain phenotypes and disorders appears to receive substantial pathogenic contributions from individual differences in a constellation of genes whose variants provide individual differences in the specification of brain connectivities during development and in adulthood. Heritable brain differences that underlie addiction vulnerability thus lie squarely in the midst of the repertoire of heritable brain differences that underlie vulnerability to other common brain disorders and phenotypes.
Collapse
Affiliation(s)
- George R Uhl
- Molecular Neurobiology Branch, National Institutes of Health (NIH), Intramural Research Program (IRP), National Institute on Drug Abuse (NIDA), Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Nelson TJ, Sun MK, Hongpaisan J, Alkon DL. Insulin, PKC signaling pathways and synaptic remodeling during memory storage and neuronal repair. Eur J Pharmacol 2008; 585:76-87. [PMID: 18402935 DOI: 10.1016/j.ejphar.2008.01.051] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Revised: 11/29/2007] [Accepted: 01/21/2008] [Indexed: 01/04/2023]
Abstract
Protein kinase C (PKC) is involved in synaptic remodeling, induction of protein synthesis, and many other processes important in learning and memory. Activation of neuronal protein kinase C correlates with, and may be essential for, all phases of learning, including acquisition, consolidation, and reconsolidation. Protein kinase C activation is closely tied to hydrolysis of membrane lipids. Phospholipases C and A2 produce 1,2-diacylglycerol and arachidonic acid, which are direct activators of protein kinase C. Phospholipase C also produces inositol triphosphate, which releases calcium from internal stores. Protein kinase C interacts with many of the same pathways as insulin; therefore, it should not be surprising that insulin signaling and protein kinase C activation can both have powerful effects on memory storage and synaptic remodeling. However, investigating the possible roles of insulin in memory storage can be challenging, due to the powerful peripheral effects of insulin on glucose and the low concentration of insulin in the brain. Although peripheral for insulin, synthesized in the beta-cells of the pancreas, is primarily involved in regulating glucose, small amounts of insulin are also present in the brain. The functions of this brain insulin are inadequately understood. Protein kinase C may also contribute to insulin resistance by phosphorylating the insulin receptor substrates required for insulin signaling. Insulin is also responsible insulin-long term depression, a type of synaptic plasticity that is also dependent on protein kinase C. However, insulin can also activate PKC signaling pathways via PLC gamma, Erk 1/2 MAP kinase, and src stimulation. Taken together, the available evidence suggests that the major impact of protein kinase C and its interaction with insulin in the mature, fully differentiated nervous system appears to be to induce synaptogenesis, enhance memory, reduce Alzheimer's pathophysiology, and stimulate neurorepair.
Collapse
Affiliation(s)
- Thomas J Nelson
- Blanchette Rockefeller Neurosciences Institute, 9601 Medical Center Drive, Rockville, Maryland 20850 USA
| | | | | | | |
Collapse
|
31
|
Alkon DL, Sun MK, Nelson TJ. PKC signaling deficits: a mechanistic hypothesis for the origins of Alzheimer's disease. Trends Pharmacol Sci 2007; 28:51-60. [PMID: 17218018 DOI: 10.1016/j.tips.2006.12.002] [Citation(s) in RCA: 183] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Revised: 11/06/2006] [Accepted: 12/19/2006] [Indexed: 11/29/2022]
Abstract
There is strong evidence that protein kinase C (PKC) isozyme signaling pathways are causally involved in associative memory storage. Other observations have indicated that PKC signaling pathways regulate important molecular events in the neurodegenerative pathophysiology of Alzheimer's disease (AD), which is a progressive dementia that is characterized by loss of recent memory. This parallel involvement of PKC signaling in both memory and neurodegeneration indicates a common basis for the origins of both the symptoms and the pathology of AD. Here, we discuss this conceptual framework as a basis for an autopsy-validated peripheral biomarker--and for AD drug design targeting drugs (bryostatin and bryologs) that activate PKC isozymes--that has already demonstrated significant promise for treating both AD neurodegeneration and its symptomatic memory loss.
Collapse
Affiliation(s)
- Daniel L Alkon
- Blanchette Rockefeller Neurosciences Institute, 9601 Medical Center Drive, Rockville, MD 20850, USA.
| | | | | |
Collapse
|
32
|
Uno Y, Fujiyuki T, Morioka M, Takeuchi H, Kubo T. Identification of proteins whose expression is up- or down-regulated in the mushroom bodies in the honeybee brain using proteomics. FEBS Lett 2006; 581:97-101. [PMID: 17182037 DOI: 10.1016/j.febslet.2006.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Revised: 12/02/2006] [Accepted: 12/04/2006] [Indexed: 11/28/2022]
Abstract
To identify protein(s) with different expression patterns in the mushroom bodies (MBs) in the honeybee brain, we compared the protein profiles of MBs and optic lobes (OLs) using proteomics. Two-dimensional gel electrophoresis revealed that five and three spots were selectively expressed in the MBs or OLs, respectively. Liquid chromatography tandem mass spectrometry analysis identified juvenile hormone diol kinase and glyceraldehyde-3-phosphate dehydrogenase as MB- and OL-selective proteins, respectively. In situ hybridization revealed that jhdk expression was upregulated in MB neuron subsets, whereas gapdh expression was downregulated, indicating that MBs have a distinct gene and protein expression profile in the honeybee brain.
Collapse
Affiliation(s)
- Yuko Uno
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
33
|
Abstract
Bryostatin-1 is a powerful protein kinase C (PKC) agonist, activating PKC isozymes at nanomolar concentrations. Pharmacological studies of bryostatin-1 have mainly been focused on its action in preventing tumor growth. Emerging evidence suggests, however, that bryostatin-1 exhibits additional important pharmacological activities. In preclinical studies bryostatin-1 has been shown at appropriate doses to have cognitive restorative and antidepressant effects. The underlying pharmacological mechanisms may involve an activation of PKC isozymes, induction of synthesis of proteins required for long-term memory, restoration of stress-evoked inhibition of PKC activity, and reduction of neurotoxic amyloid accumulation and tau protein hyperphosphorylation. The therapeutic potential of bryostatin-1 as a CNS drug should be further explored.
Collapse
Affiliation(s)
- Miao-Kun Sun
- Blanchette Rockefeller Neurosciences Institute, Rockville, MD 20850, USA.
| | | |
Collapse
|
34
|
Kuzirian AM, Epstein HT, Gagliardi CJ, Nelson TJ, Sakakibara M, Taylor C, Scioletti AB, Alkon DL. Bryostatin enhancement of memory in Hermissenda. THE BIOLOGICAL BULLETIN 2006; 210:201-14. [PMID: 16801495 DOI: 10.2307/4134558] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Bryostatin, a potent agonist of protein kinase C (PKC), when administered to Hermissenda was found to affect acquisition of an associative learning paradigm. Low bryostatin concentrations (0.1 to 0.5 ng/ml) enhanced memory acquisition, while concentrations higher than 1.0 ng/ml down-regulated the pathway and no recall of the associative training was exhibited. The extent of enhancement depended upon the conditioning regime used and the memory stage normally fostered by that regime. The effects of two training events (TEs) with paired conditioned and unconditioned stimuli, which standardly evoked only short-term memory (STM) lasting 7 min, were--when bryostatin was added concurrently--enhanced to a long-term memory (LTM) that lasted about 20 h. The effects of both 4- and 6-paired TEs (which by themselves did not generate LTM), were also enhanced by bryostatin to induce a consolidated memory (CM) that lasted at least 5 days. The standard positive 9-TE regime typically produced a CM lasting at least 6 days. Low concentrations of bryostatin (<0.5 ng/ml) elicited no demonstrable enhancement of CM from 9-TEs. However, animals exposed to bryostatin concentrations higher than 1.0 ng/ml exhibited no behavioral learning. Sharp-electrode intracellular recordings of type-B photoreceptors in the eyes from animals conditioned in vivo with bryostatin revealed changes in input resistance and an enhanced long-lasting depolarization (LLD) in response to light. Likewise, quantitative immunocytochemical measurements using an antibody specific for the PKC-activated Ca2+/GTP-binding protein calexcitin showed enhanced antibody labeling with bryostatin. Animals exposed to the PKC inhibitor bisindolylmaleimide-XI (Ro-32-0432) administered by immersion prior to 9-TE conditioning showed no training-induced changes with or without bryostatin exposure. However, if animals received bryostatin before Ro-32, the enhanced acquisition and demonstrated recall still occurred. Therefore, pathways responsible for the enhancement effects induced by bryostatin were putatively mediated by PKC. Overall, the data indicated that PKC activation occurred and calexcitin levels were raised during the acquisition phases of associative conditioning and memory initiation, and subsequently returned to baseline levels within 24 and 48 h, respectively. Therefore, the protracted recall measured by the testing regime used was probably due to bryostatin-induced changes during the acquisition and facilitated storage of memory, and not necessarily to enhanced recall of the stored memory when tested many days after training.
Collapse
Affiliation(s)
- A M Kuzirian
- Marine Biological Laboratory, Woods Hole, Massachusetts 02543, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Gombos Z, Yap KL, Ikura M, Chakrabartty A. NMR-driven secondary and tertiary structure model of Ca2+-loaded calexcitin. Biochem Biophys Res Commun 2006; 343:520-4. [PMID: 16546129 DOI: 10.1016/j.bbrc.2006.02.182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Accepted: 02/27/2006] [Indexed: 11/17/2022]
Abstract
Calexcitin (CE) is a Ca2+-binding protein which is expressed in neuronal cells and is a member of the sarcoplasmic Ca2+-binding protein subfamily. The peptide backbone of Ca2+-CE has been assigned by NMR and it shows that CE is composed of nine alpha-helices-forming four EF-hands and an additional helix near the C-terminus. A structural model of CE suggests the presence of a putative recessed hydrophobic pocket that may be involved in Ca2+-mediated protein-ligand interactions. This feature is unique to CE and is absent in other SCPs, such as those from Branchiostoma and Nereis, and from calerythrin.
Collapse
Affiliation(s)
- Zoltan Gombos
- Ontario Cancer Institute and Department of Medical Biophysics, University of Toronto, Ont., Canada M5G 1L7.
| | | | | | | |
Collapse
|
36
|
Shin H, Hwang IK, Yoo KY, Song JH, Jung JY, Kang TC, Choi SY, Han BH, Kim JS, Won MH. Expression and changes of Ca2+-ATPase in neurons and astrocytes in the gerbil hippocampus after transient forebrain ischemia. Brain Res 2005; 1049:43-51. [PMID: 15922996 DOI: 10.1016/j.brainres.2005.04.084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Revised: 04/21/2005] [Accepted: 04/25/2005] [Indexed: 10/25/2022]
Abstract
Ca2+-ATPase is one of the most powerful modulators of intracellular calcium levels. In this study, we focused on chronological changes in the immunoreactivity and protein levels of Ca2+-ATPase in the hippocampus after 5 min of transient forebrain ischemia. Ca2+-ATPase immunoreactivity was significantly altered in the hippocampal CA1 region and in the dentate gyrus, but not in the CA2/3 region after ischemic insult. In the sham-operated group, Ca2+-ATPase immunoreactivity was detected in the hippocampus. Ca2+-ATPase immunoreactivity in the CA1 region and in the dentate gyrus, and its protein levels peaked 3 h after ischemic insult. At this time, CA1 pyramidal cells and dentate polymorphic cells showed strong Ca2+-ATPase immunoreactivity. Thereafter, Ca2+-ATPase immunoreactivity reduced in the CA1 region and in the dentate gyrus. One day after ischemic insult, Ca2+-ATPase immunoreactivity was observed in some CA1 non-pyramidal cells, and 4 days after ischemic insult, Ca2+-ATPase immunoreactivity was detected in astrocytes throughout the CA1 region, but Ca2+-ATPase immunoreactivity in the dentate gyrus had nearly disappeared. Our results suggest that Ca2+-ATPase changes may be associated with a response to ischemic damage in hippocampal CA1 pyramidal cells, and that increased Ca2+-ATPase immunoreactivity in the reactive astrocytes may be associated with the maintenance of intracellular calcium levels.
Collapse
Affiliation(s)
- Hyoseon Shin
- Department of Anatomy, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Sun MK, Alkon DL. Protein kinase C substrate activators: potential as novel antidepressants. Drug Dev Res 2005. [DOI: 10.1002/ddr.20019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
38
|
Barkai E. Dynamics of learning-induced cellular modifications in the cortex. BIOLOGICAL CYBERNETICS 2005; 92:360-6. [PMID: 15906082 DOI: 10.1007/s00422-005-0564-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2004] [Accepted: 03/18/2005] [Indexed: 05/02/2023]
Abstract
This aim of this review is to describe the dynamics of learning-induced cellular modifications in the rat piriform (olfactory) cortex after olfactory discrimination learning and to describe their functional significance to long-term memory consolidation. The first change to occur is in the intrinsic properties of the neurons. One day after learning, pyramidal neurons show enhanced neuronal excitability. This enhancement results from reduction in calcium-dependent conductance that mediates the post burst after-hyperpolarization. Such enhanced excitability lasts for 3 days and is followed by a series of synaptic modifications. Several forms of long-term enhancement in synaptic connections between layer II pyramidal neurons in the piriform cortex accompany olfactory learning. Enhanced synaptic release is indicated by reduced paired-pulse facilitation. Post-synaptic enhancement of synaptic transmission is indicated by reduced rise time of post-synaptic potentials and formation of new synaptic connections is indicated by increased spine density along dendrites of these neurons. Such modifications last for up to 5 days. Thus, olfactory discrimination rule learning is accompanied by a series of cellular modifications which occur and then disappear at different times. These modifications overlap partially, allowing the maintenance of the cortical system in a 'learning mode' in which memories for specific odors can be acquired rapidly and efficiently.
Collapse
Affiliation(s)
- Edi Barkai
- Center for Brain and Behavior, Faculty of Sciences, University of Haifa, Haifa, 39105, Israel.
| |
Collapse
|
39
|
Sun MK, Alkon DL. Dual effects of bryostatin-1 on spatial memory and depression. Eur J Pharmacol 2005; 512:43-51. [PMID: 15814089 DOI: 10.1016/j.ejphar.2005.02.028] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Accepted: 02/18/2005] [Indexed: 02/05/2023]
Abstract
Dementia and depression are clinical symptoms commonly associated in patients. Emerging evidence suggests that the two diseases share many profiles in their development and underlying neural/molecular mechanisms. Thus, interest is raised in developing new classes of antidepressant agents with activity of cognitive enhancement. Here, we show that bryostatin-1, a protein kinase C substrate activator, at bilateral intracerebroventricular doses of 0.64 or 2 pmol/site, significantly enhanced learning and memory of rats in a spatial water maze task. When applied at the doses at which it exhibits memory-enhancing activity, bryostatin-1 showed a significant antidepressant activity, as determined in an open space swim test. Both effects were not observed when a smaller dose was administered and were largely eliminated by co-administration of 1-(5-isoquinolinesulfonyl)-2-methylpiperazine (H-7), a protein kinase C inhibitor. These results support the hypothesis that memory processing and mood regulation share common neural mechanisms. Restoring impaired mood regulation with antidepressant agents that also exhibit memory-enhancing activity may represent one of the new strategies in the fight against depression associated with memory impairments.
Collapse
Affiliation(s)
- Miao-Kun Sun
- Blânchette Rockefeller Neurosciences Institute, 9601 Medical Center Drive, Johns Hopkins Academic and Research Building, Room 319, Rockville, MD 20850, USA.
| | | |
Collapse
|
40
|
Cavallaro S. DNA microarrays and animal models of learning and memory. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2005; 60:97-133. [PMID: 15474588 DOI: 10.1016/s0074-7742(04)60004-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- Sebastiano Cavallaro
- Institute of Neurological Sciences, Italian National Research Council, 95123 Catania, Italy
| |
Collapse
|
41
|
Verkhratsky A. Physiology and Pathophysiology of the Calcium Store in the Endoplasmic Reticulum of Neurons. Physiol Rev 2005; 85:201-79. [PMID: 15618481 DOI: 10.1152/physrev.00004.2004] [Citation(s) in RCA: 567] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The endoplasmic reticulum (ER) is the largest single intracellular organelle, which is present in all types of nerve cells. The ER is an interconnected, internally continuous system of tubules and cisterns, which extends from the nuclear envelope to axons and presynaptic terminals, as well as to dendrites and dendritic spines. Ca2+release channels and Ca2+pumps residing in the ER membrane provide for its excitability. Regulated ER Ca2+release controls many neuronal functions, from plasmalemmal excitability to synaptic plasticity. Enzymatic cascades dependent on the Ca2+concentration in the ER lumen integrate rapid Ca2+signaling with long-lasting adaptive responses through modifications in protein synthesis and processing. Disruptions of ER Ca2+homeostasis are critically involved in various forms of neuropathology.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester, Faculty of Biological Sciences, United Kingdom.
| |
Collapse
|
42
|
Maiorov VI. Local bicuculline disinhibition does not disrupt acquired coordination between the tactile input and the motor output in the motor cortex of the cat. ACTA ACUST UNITED AC 2004; 34:321-2. [PMID: 15341205 DOI: 10.1023/b:neab.0000018739.16608.de] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
MESH Headings
- Animals
- Bicuculline/pharmacology
- Cats
- Conditioning, Classical/drug effects
- Conditioning, Classical/physiology
- Evoked Potentials, Motor/drug effects
- Evoked Potentials, Motor/physiology
- Extinction, Psychological/drug effects
- Extinction, Psychological/physiology
- GABA Antagonists/pharmacology
- Motor Cortex/cytology
- Motor Cortex/drug effects
- Neural Inhibition/drug effects
- Neural Inhibition/physiology
- Neurons, Afferent/drug effects
- Neurons, Afferent/physiology
- Neurons, Efferent/drug effects
- Neurons, Efferent/physiology
- Touch/physiology
Collapse
Affiliation(s)
- V I Maiorov
- Department of Higher Nervous Activity, M V Lomonosov Moscow State University.
| |
Collapse
|
43
|
Abstract
Tissue plasminogen activator (tPA), a fibrin specific activator for the conversion of plasminogen to plasmin, stimulates thrombolysis and rescues ischemic brain by restoring blood flow. However, emerging data suggests that under some conditions, both tPA and plasmin, which are broad spectrum protease enzymes, are potentially neurotoxic if they reach the extracellular space. Animal models suggest that in severe ischemia with injury to the blood brain barrier (BBB) there is injury attributed to the protease effects of this exogenous tPA. Besides clot lysis per se, tPA may have pleiotropic actions in the brain, including direct vasoactivity, cleaveage of the N-methyl-D-aspartate (NMDA) NR1 subunit, amplification of intracellular Ca++ conductance, and activation of other extracellular proteases from the matrix metalloproteinase (MMP) family, e.g. MMP-9. These effects may increase excitotoxicity, further damage the BBB, and worsen edema and cerebral hemorrhage. If tPA is effective and reverses ischemia promptly, the BBB remains intact and exogenous tPA remains within the vascular space. If tPA is ineffective and ischemia is prolonged, there is the risk that exogenous tPA will injure both the neurovascular unit and the brain. Methods of neuroprotection, which prevent tPA toxicity or additional mechanical means to open cerebral vessels, are now needed.
Collapse
Affiliation(s)
- Jaspreet Kaur
- Stroke Program, Calgary Brain Institute, University of Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
44
|
Crow T. Pavlovian conditioning of Hermissenda: current cellular, molecular, and circuit perspectives. Learn Mem 2004; 11:229-38. [PMID: 15169851 DOI: 10.1101/lm.70704] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The less-complex central nervous system of many invertebrates make them attractive for not only the molecular analysis of the associative learning and memory, but also in determining how neural circuits are modified by learning to generate changes in behavior. The nudibranch mollusk Hermissenda crassicornis is a preparation that has contributed to an understanding of cellular and molecular mechanisms of Pavlovian conditioning. Identified neurons in the conditioned stimulus (CS) pathway have been studied in detail using biophysical, biochemical, and molecular techniques. These studies have resulted in the identification and characterization of specific membrane conductances contributing to enhanced excitability and synaptic facilitation in the CS pathway of conditioned animals. Second-messenger systems activated by the CS and US have been examined, and proteins that are regulated by one-trial and multi-trial Pavlovian conditioning have been identified in the CS pathway. The recent progress that has been made in the identification of the neural circuitry supporting the unconditioned response (UR) and conditioned response (CR) now provides for the opportunity to understand how Pavlovian conditioning is expressed in behavior.
Collapse
Affiliation(s)
- Terry Crow
- Department of Neurobiology and Anatomy, University of Texas Medical School, Houston, Texas 77030, USA.
| |
Collapse
|
45
|
Etcheberrigaray R, Tan M, Dewachter I, Kuipéri C, Van der Auwera I, Wera S, Qiao L, Bank B, Nelson TJ, Kozikowski AP, Van Leuven F, Alkon DL. Therapeutic effects of PKC activators in Alzheimer's disease transgenic mice. Proc Natl Acad Sci U S A 2004; 101:11141-6. [PMID: 15263077 PMCID: PMC503753 DOI: 10.1073/pnas.0403921101] [Citation(s) in RCA: 261] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease (AD) characteristically presents with early memory loss. Regulation of K(+) channels, calcium homeostasis, and protein kinase C (PKC) activation are molecular events that have been implicated during associative memory which are also altered or defective in AD. PKC is also involved in the processing of the amyloid precursor protein (APP), a central element in AD pathophysiology. In previous studies, we demonstrated that benzolactam (BL), a novel PKC activator, reversed K(+) channels defects and enhanced secretion of APP alpha in AD cells. In this study we present data showing that another PKC activator, bryostatin 1, at subnanomolar concentrations dramatically enhances the secretion of the alpha-secretase product sAPP alpha in fibroblasts from AD patients. We also show that BL significantly increased the amount of sAPP alpha and reduced A beta 40 in the brains of APP[V717I] transgenic mice. In a more recently developed AD double-transgenic mouse, bryostatin was effective in reducing both brain A beta 40 and A beta 42. In addition, bryostatin ameliorated the rate of premature death and improved behavioral outcomes. Collectively, these data corroborate PKC and its activation as a potentially important means of ameliorating AD pathophysiology and perhaps cognitive impairment, thus offering a promising target for drug development. Because bryostatin 1 is devoid of tumor-promoting activity and is undergoing numerous clinical studies for cancer treatment in humans, it might be readily tested in patients as a potential therapeutic agent for Alzheimer's disease.
Collapse
|
46
|
Nesti E, Everill B, Morielli AD. Endocytosis as a mechanism for tyrosine kinase-dependent suppression of a voltage-gated potassium channel. Mol Biol Cell 2004; 15:4073-88. [PMID: 15215309 PMCID: PMC515342 DOI: 10.1091/mbc.e03-11-0788] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The voltage-gated potassium channel Kv1.2 undergoes tyrosine phosphorylation-dependent suppression of its ionic current. However, little is known about the physical mechanism behind that process. We have found that the Kv1.2 alpha-subunit protein undergoes endocytosis in response to the same stimuli that evoke suppression of Kv1.2 ionic current. The process is tyrosine phosphorylation-dependent because the same tyrosine to phenylalanine mutation in the N-terminus of Kv1.2 that confers resistance to channel suppression (Y132F) also confers resistance to channel endocytosis. Overexpression of a dominant negative form of dynamin blocked stimulus-induced Kv1.2 endocytosis and also blocked suppression of Kv1.2 ionic current. These data indicate that endocytosis of Kv1.2 from the cell surface is a key mechanism for channel suppression by tyrosine kinases.
Collapse
Affiliation(s)
- Edmund Nesti
- The University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | | | |
Collapse
|
47
|
Sadamoto H, Sato H, Kobayashi S, Murakami J, Aonuma H, Ando H, Fujito Y, Hamano K, Awaji M, Lukowiak K, Urano A, Ito E. CREB in the pond snail Lymnaea stagnalis: cloning, gene expression, and function in identifiable neurons of the central nervous system. ACTA ACUST UNITED AC 2004; 58:455-66. [PMID: 14978723 DOI: 10.1002/neu.10296] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The pond snail Lymnaea stagnalis is an excellent model system in which to study the neuronal and molecular substrates of associative learning and its consolidation into long-term memory. Until now, the presence of cyclic AMP (cAMP)-responsive element binding protein (CREB), which is believed to be a necessary component in the process of a learned behavior that is consolidated into long-term memory, has only been assumed in Lymnaea neurons. We therefore cloned and analyzed the cDNA sequences of homologues of CREB1 and CREB2 and determined the presence of these mRNAs in identifiable neurons of the central nervous system (CNS) of L. stagnalis. The deduced amino acid sequence of Lymnaea CREB1 is homologous to transcriptional activators, mammalian CREB1 and Aplysia CREB1a, in the C-terminal DNA binding (bZIP) and phosphorylation domains, whereas the deduced amino acid sequence of Lymnaea CREB2 is homologous to transcriptional repressors, human CREB2, mouse activating transcription factor-4, and Aplysia CREB2 in the bZIP domain. In situ hybridization revealed that only a relatively few neurons showed strongly positive signals for Lymnaea CREB1 mRNA, whereas all the neurons in the CNS contained Lymnaea CREB2 mRNA. Using one of the neurons (the cerebral giant cell) containing Lymnaea CREB1 mRNA, we showed that the injection of a CRE oligonucleotide inhibited a cAMP-induced, long-lasting synaptic plasticity. We therefore conclude that CREBs are present in Lymnaea neurons and may function as necessary players in behavioral plasticity.
Collapse
Affiliation(s)
- Hisayo Sadamoto
- Division of Biological Sciences, Graduate School of Science, Hokkaido University, Sapporo 060-0810, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kim IH, Park SK, Sun W, Kang Y, Kim HT, Kim H. Spatial learning enhances the expression of inositol 1,4,5-trisphosphate 3-kinase A in the hippocampal formation of rat. ACTA ACUST UNITED AC 2004; 124:12-9. [PMID: 15093681 DOI: 10.1016/j.molbrainres.2003.12.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2003] [Indexed: 12/01/2022]
Abstract
Calcium-mediated signaling is crucial for the synaptic plasticity and long-term memory storage, which requires de novo protein synthesis. Inositol 1,4,5-trisphosphate 3-kinase A (IP(3)K-A) is an enzyme, which is involved in the maintenance of intracellular calcium homeostasis by converting inositol 1,4,5-trisphosphate (IP(3)) to inositol 1,3,4,5-tetrakisphosphate (IP(4)). Because IP(3)K-A is enriched in the dendritic spines of hippocampal neurons, it has been speculated that this enzyme is involved in the memory formation. In the present study, we demonstrated that the expression of IP(3)K-A is increased in the hippocampal formation of the rats during the Morris water maze training. Immunohistochemical analysis indicated the specific induction of IP(3)K-A protein in the hippocampal formation following 5-day water maze training. Furthermore, in situ hybridization histochemistry showed that the induction of IP(3)K-A mRNA in the hippocampal formation was observed on the first day of training, and the induced level of IP(3)K-A mRNA was maintained until the fifth day of training. These results suggest that IP(3)K-A plays a role in the processing of spatial memory, most likely by regulating the calcium signaling in the dendritic spines of hippocampal formation.
Collapse
Affiliation(s)
- Il Hwan Kim
- Department of Anatomy and Division of Brain Korea 21Biomedical Science, Korea University College of Medicine, 126-1, 5-ga, Anam-dong, Sungbuk-gu, 136-705 Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Many experiments in the past have demonstrated the requirement of de novo gene expression during the long-term retention of learning and memory. Although previous studies implicated individual genes or genetic pathways in learning and memory, the collective behaviours of the genes is mostly unknown. We have used genome-scale screening by microarray analysis to examine the hippocampal expression of more than 1200 genes relevant to neurobiology during instrumental conditioning. Training rats on a step-through passive avoidance task led to unique patterns of gene expression when compared to naïve animals or those exposed to the conditioned or the unconditioned stimulus alone. The newly identified genes afford a quantitative view of the changes which accompany conditioning at the genomic level and enable deeper insights into the molecular basis underlying learning and memory.
Collapse
Affiliation(s)
- Velia D'Agata
- Institute of Neurological Sciences, Italian National Research Council, CNR, Viale Regina Margherita 6, 95123 Catania, Italy
| | | |
Collapse
|
50
|
Eyman M, Crispino M, Kaplan BB, Giuditta A. Squid photoreceptor terminals synthesize calexcitin, a learning related protein. Neurosci Lett 2003; 347:21-4. [PMID: 12865132 DOI: 10.1016/s0304-3940(03)00593-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Nerve endings of squid photoreceptor neurons generate large synaptosomes upon homogenization of the optic lobe. Using several independent methods, these presynaptic structures have been shown to synthesize a wealth of soluble, cytoskeletal and nuclear encoded mitochondrial proteins, and to account for essentially all the translation activity of the synaptosomal fraction. We are now presenting evidence that calexcitin, a learning related, Ca(2+)-binding protein of the B photoreceptors of Hermissenda crassicornis (a mollusk), is synthesized and subjected to post-translational modifications in the squid photoreceptor terminals. In view of the essential role of presynaptic protein synthesis in long-term memory formation in Aplysia, our data suggest that a comparable role may be played by calexcitin synthesized in the squid photoreceptor terminals.
Collapse
Affiliation(s)
- Maria Eyman
- Dipartimento di Fisiologia Generale e Ambientale, Università di Napoli "Federico II", Via Mezzocannone 8, 80134, Naples, Italy
| | | | | | | |
Collapse
|