Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nature ; 621(7977): 146-153, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37648853

ABSTRACT

Learning and memory are thought to require hippocampal long-term potentiation (LTP), and one of the few central dogmas of molecular neuroscience that has stood undisputed for more than three decades is that LTP induction requires enzymatic activity of the Ca2+/calmodulin-dependent protein kinase II (CaMKII)1-3. However, as we delineate here, the experimental evidence is surprisingly far from conclusive. All previous interventions inhibiting enzymatic CaMKII activity and LTP4-8 also interfere with structural CaMKII roles, in particular binding to the NMDA-type glutamate receptor subunit GluN2B9-14. Thus, we here characterized and utilized complementary sets of new opto-/pharmaco-genetic tools to distinguish between enzymatic and structural CaMKII functions. Several independent lines of evidence demonstrated LTP induction by a structural function of CaMKII rather than by its enzymatic activity. The sole contribution of kinase activity was autoregulation of this structural role via T286 autophosphorylation, which explains why this distinction has been elusive for decades. Directly initiating the structural function in a manner that circumvented this T286 role was sufficient to elicit robust LTP, even when enzymatic CaMKII activity was blocked.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Long-Term Potentiation , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Glutamic Acid/metabolism , Hippocampus/physiology , Learning/physiology , Long-Term Potentiation/physiology , Optogenetics , Phosphorylation , Protein Binding
2.
Sci Signal ; 16(795): eade5892, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37490545

ABSTRACT

CaMKII has molecular memory functions because transient calcium ion stimuli can induce long-lasting increases in its synaptic localization and calcium ion-independent (autonomous) activity, thereby leaving memory traces of calcium ion stimuli beyond their duration. The synaptic effects of two mechanisms that induce CaMKII autonomy are well studied: autophosphorylation at threonine-286 and binding to GluN2B. Here, we examined the neuronal functions of additional autonomy mechanisms: nitrosylation and oxidation of the CaMKII regulatory domain. We generated a knock-in mouse line with mutations that render the CaMKII regulatory domain nitrosylation/oxidation-incompetent, CaMKIIΔSNO, and found that it had deficits in memory and synaptic plasticity that were similar to those in aged wild-type mice. In addition, similar to aged wild-type mice, in which CaMKII was hyponitrosylated, but unlike mice with impairments of other CaMKII autonomy mechanisms, CaMKIIΔSNO mice showed reduced long-term potentiation (LTP) when induced by theta-burst stimulation but not high-frequency stimulation (HFS). As in aged wild-type mice, the HFS-LTP in the young adult CaMKIIΔSNO mice required L-type voltage-gated calcium ion channels. The effects in aged mice were likely caused by the loss of nitrosylation because no decline in CaMKII oxidation was detected. In hippocampal neurons, nitrosylation of CaMKII induced its accumulation at synapses under basal conditions in a manner mediated by GluN2B binding, like after LTP stimuli. However, LTP-induced synaptic CaMKII accumulation did not require nitrosylation. Thus, an aging-associated decrease in CaMKII nitrosylation may cause impairments by chronic synaptic effects, such as the decrease in basal synaptic CaMKII.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium , Animals , Mice , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/metabolism , Long-Term Potentiation/physiology , Neuronal Plasticity , Phosphorylation , Synapses/metabolism
3.
J Biol Chem ; 299(5): 104693, 2023 05.
Article in English | MEDLINE | ID: mdl-37037305

ABSTRACT

The Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a central regulator of learning and memory, which poses a problem for targeting it therapeutically. Indeed, our study supports prior conclusions that long-term interference with CaMKII signaling can erase pre-formed memories. By contrast, short-term pharmacological CaMKII inhibition with the neuroprotective peptide tatCN19o interfered with learning in mice only mildly and transiently (for less than 1 h) and did not at all reverse pre-formed memories. These results were obtained with ≥500-fold of the dose that protected hippocampal neurons from cell death after a highly clinically relevant pig model of transient global cerebral ischemia: ventricular fibrillation followed by advanced life support and electrical defibrillation to induce the return of spontaneous circulation. Of additional importance for therapy development, our preliminary cardiovascular safety studies in mice and pig did not indicate any concerns with acute tatCN19o injection. Taken together, although prolonged interference with CaMKII signaling can erase memory, acute short-term CaMKII inhibition with tatCN19o did not cause such retrograde amnesia that would pose a contraindication for therapy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Memory , Animals , Mice , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/metabolism , Memory/drug effects , Memory/physiology , Neurons/metabolism , Phosphorylation/physiology , Swine , Peptides/pharmacology
4.
bioRxiv ; 2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36747773

ABSTRACT

The Ca 2+ /calmodulin-dependent protein kinase II (CaMKII) is a central regulator of learning and memory, which poses a problem for targeting it therapeutically. Indeed, our study supports prior conclusions that long-term interference with CaMKII signaling can erase pre-formed memories. By contrast, short-term pharmacological CaMKII inhibition with tatCN19o interfered with learning in mice only mildly and transiently (for less than 1 h) and did not at all reverse pre-formed memories. This was at ≥500fold of the dose that protected hippocampal neurons from cell death after a highly clinically relevant pig model of transient global cerebral ischemia: ventricular fibrillation followed by advanced life support and electrical defibrillation to induce return of spontaneous circulation. Of additional importance for therapeutic development, cardiovascular safety studies in mice and pig did not indicate any concerns with acute tatCN19o injection. Taken together, even though prolonged interference with CaMKII signaling can erase memory, acute short-term CaMKII inhibition with tatCN19o did not cause such retrograde amnesia that would pose a contraindication for therapy.

5.
J Biol Chem ; 298(9): 102299, 2022 09.
Article in English | MEDLINE | ID: mdl-35872016

ABSTRACT

The Ca2+/calmodulin-dependent protein kinase II (CaMKII) mediates long-term potentiation or depression (LTP or LTD) after distinct stimuli of hippocampal NMDA-type glutamate receptors (NMDARs). NMDAR-dependent LTD prevails in juvenile mice, but a mechanistically different form of LTD can be readily induced in adults by instead stimulating metabotropic glutamate receptors (mGluRs). However, the role that CaMKII plays in the mGluR-dependent form of LTD is not clear. Here we show that mGluR-dependent LTD also requires CaMKII and its T286 autophosphorylation (pT286), which induces Ca2+-independent autonomous kinase activity. In addition, we compared the role of pT286 among three forms of long-term plasticity (NMDAR-dependent LTP and LTD, and mGluR-dependent LTD) using simultaneous live imaging of endogenous CaMKII together with synaptic marker proteins. We determined that after LTP stimuli, pT286 autophosphorylation accelerated CaMKII movement to excitatory synapses. After NMDAR-LTD stimuli, pT286 was strictly required for any movement to inhibitory synapses. Similar to NMDAR-LTD, we found the mGluR-LTD stimuli did not induce CaMKII movement to excitatory synapses. However, in contrast to NMDAR-LTD, we demonstrate that the mGluR-LTD did not involve CaMKII movement to inhibitory synapses and did not require additional T305/306 autophosphorylation. Thus, despite its prominent role in LTP, we conclude that CaMKII T286 autophosphorylation is also required for both major forms of hippocampal LTD, albeit with differential requirements for the heterosynaptic communication of excitatory signals to inhibitory synapses.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Hippocampus , Long-Term Synaptic Depression , Receptors, Metabotropic Glutamate , Synapses , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/metabolism , Hippocampus/physiology , Long-Term Synaptic Depression/physiology , Mice , N-Methylaspartate/metabolism , Phosphorylation , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Synapses/physiology
6.
iScience ; 25(6): 104368, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35620430

ABSTRACT

Aß bears homology to the CaMKII regulatory domain, and peptides derived from this domain can bind and disrupt the CaMKII holoenzyme, suggesting that Aß could have a similar effect. Notably, Aß impairs the synaptic CaMKII accumulation that is mediated by GluN2B binding, which requires CaMKII assembly into holoenzymes. Furthermore, this Aß-induced impairment is prevented by CaMKII inhibitors that should also inhibit the putative direct Aß binding. However, our study did not find any evidence for direct effects of Aß on CaMKII: Aß did not directly disrupt CaMKII holoenzymes, GluN2B binding, T286 autophosphorylation, or kinase activity in vitro. Most importantly, in neurons, the Aß-induced impairment of CaMKII synaptic accumulation was prevented by an ATP-competitive CaMKII inhibitor that would not interfere with the putative direct Aß binding. Together, our results indicate that synaptic Aß effects are not mediated by direct binding to CaMKII, but instead require CaMKII activation via indirect signaling events.

7.
Brain Res ; 1773: 147699, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34687697

ABSTRACT

CaMKIIα plays a dual role in synaptic plasticity, as it can mediate synaptic changes in opposing directions. We hypothesized that CaMKIIα plays a similar dual role also in neuronal cell death and survival. Indeed, the CaMKII inhibitor tatCN21 is neuroprotective when added during or after excitotoxic/ischemic insults, but was described to cause sensitization when applied long-term prior to such insult. However, when comparing long-term CaMKII inhibition by several different inhibitors in neuronal cultures, we did not detect any sensitization. Likewise, in a mouse in vivo model of global cerebral ischemia (cardiac arrest followed by cardiopulmonary resuscitation), complete knockout of the neuronal CaMKIIα isoform did not cause sensitization but instead significant neuroprotection.


Subject(s)
Brain Ischemia/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Death/physiology , Heart Arrest/metabolism , Animals , Brain Ischemia/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cerebral Cortex/metabolism , Disease Models, Animal , Heart Arrest/genetics , Mice , Mice, Knockout , Neurons/metabolism , Phosphorylation
8.
PLoS One ; 15(7): e0236478, 2020.
Article in English | MEDLINE | ID: mdl-32716967

ABSTRACT

CaMKII is an important mediator of forms of synaptic plasticity that are thought to underly learning and memory. The CaMKII mutants K42M and K42R have been used interchangeably as research tools, although some reported phenotypic differences suggest that they may differ in the extent to which they impair ATP binding. Here, we directly compared the two mutations at the high ATP concentrations that exist within cells (~4 mM). We found that both mutations equally blocked GluA1 phosphorylation in vitro and GluN2B binding within cells. Both mutations also reduced but did not completely abolish CaMKII T286 autophosphorylation in vitro or CaMKII movement to excitatory synapses in neurons. Thus, despite previously suggested differences, both mutations appear to interfere with ATP binding to the same extent.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Mutation/genetics , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Cells, Cultured , Female , Glutamic Acid/pharmacology , HEK293 Cells , Hippocampus/cytology , Humans , Male , Movement , Phosphorylation , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism
9.
Cell Rep ; 30(1): 1-8.e4, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31914378

ABSTRACT

DAPK1 binding to GluN2B was prominently reported to mediate ischemic cell death in vivo. DAPK1 and CaMKII bind to the same GluN2B region, and their binding is mutually exclusive. Here, we show that mutating the binding region on GluN2B (L1298A/R1300Q) protected against neuronal cell death induced by cardiac arrest followed by resuscitation. Importantly, the GluN2B mutation selectively abolished only CaMKII, but not DAPK1, binding. During ischemic or excitotoxic insults, CaMKII further accumulated at excitatory synapses, and this accumulation was mediated by GluN2B binding. Interestingly, extra-synaptic GluN2B decreased after ischemia, but its relative association with DAPK1 increased. Thus, ischemic neuronal death requires CaMKII binding to synaptic GluN2B, whereas any potential role for DAPK1 binding is restricted to a different, likely extra-synaptic population of GluN2B.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Death-Associated Protein Kinases/metabolism , Heart Arrest/metabolism , Heart Arrest/pathology , Ischemia/metabolism , Ischemia/pathology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cell Death/drug effects , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Female , Glutamic Acid/toxicity , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Protein Binding/drug effects , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Resuscitation
10.
Stem Cell Reports ; 10(6): 1705-1720, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29805108

ABSTRACT

Neural stem and precursor cell (NSPC) proliferation in the rodent adult hippocampus is essential to maintain stem cell populations and produce new neurons. Retinoic acid (RA) signaling is implicated in regulation of adult hippocampal neurogenesis, but its exact role in control of NSPC behavior has not been examined. We show RA signaling in all hippocampal NSPC subtypes and that inhibition of RA synthesis or signaling significantly decreases NSPC proliferation via abrogation of cell-cycle kinetics and cell-cycle regulators. RA signaling controls NSPC proliferation through hypoxia inducible factor-1α (HIF1α), where stabilization of HIF1α concurrent with disruption of RA signaling can prevent NSPC defects. These studies demonstrate a cell-autonomous role for RA signaling in hippocampal NSPCs that substantially broadens RA's function beyond its well-described role in neuronal differentiation.


Subject(s)
Cell Differentiation/drug effects , Hippocampus/cytology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Tretinoin/pharmacology , Age Factors , Animals , Biomarkers , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Hippocampus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Tretinoin/metabolism , Vascular Endothelial Growth Factor A/metabolism
11.
Eur J Neurosci ; 45(3): 342-357, 2017 02.
Article in English | MEDLINE | ID: mdl-27763700

ABSTRACT

Manipulating gut microbes may improve mental health. Prebiotics are indigestible compounds that increase the growth and activity of health-promoting microorganisms, yet few studies have examined how prebiotics affect CNS function. Using an acute inescapable stressor known to produce learned helplessness behaviours such as failure to escape and exaggerated fear, we tested whether early life supplementation of a blend of two prebiotics, galactooligosaccharide (GOS) and polydextrose (PDX), and the glycoprotein lactoferrin (LAC) would attenuate behavioural and biological responses to stress later in life. Juvenile, male F344 rats were fed diets containing either GOS and PDX alone, LAC alone, or GOS, PDX and LAC. All diets altered gut bacteria, while diets containing GOS and PDX increased Lactobacillus spp. After 4 weeks, rats were exposed to inescapable stress, and either immediately killed for blood and tissues, or assessed for learned helplessness 24 h later. Diets did not attenuate stress effects on spleen weight, corticosterone and blood glucose; however, all diets differentially attenuated stress-induced learned helplessness. Notably, in situ hybridization revealed that all diets reduced stress-evoked cfos mRNA in the dorsal raphe nucleus (DRN), a structure important for learned helplessness behaviours. In addition, GOS, PDX and LAC diet attenuated stress-evoked decreases in mRNA for the 5-HT1A autoreceptor in the DRN and increased basal BDNF mRNA within the prefrontal cortex. These data suggest early life diets containing prebiotics and/or LAC promote behavioural stress resistance and uniquely modulate gene expression in corresponding circuits.


Subject(s)
Diet , Helplessness, Learned , Lactoferrin/therapeutic use , Prebiotics , Stress, Psychological/diet therapy , Animals , Brain-Derived Neurotrophic Factor/metabolism , Lactoferrin/pharmacology , Male , Prefrontal Cortex/drug effects , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Raphe Nuclei/drug effects , Raphe Nuclei/growth & development , Raphe Nuclei/metabolism , Rats , Rats, Inbred F344 , Receptor, Serotonin, 5-HT1A/metabolism , Stress, Psychological/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...