Ca2+/calmodulin-dependent protein kinase II

Last updated
Calcium/calmodulin dependent protein kinase II association domain
PDB 1hkx EBI.jpg
Crystal structure of calcium/calmodulin-dependent protein kinase
Identifiers
SymbolCaMKII_AD
Pfam PF08332
Pfam clan CL0051
InterPro IPR013543
Available protein structures:
Pfam   structures / ECOD  
PDB RCSB PDB; PDBe; PDBj
PDBsum structure summary
CaMKII gamma holoenzyme in its (A) closed and the (B) open conformations Camkii open close.jpg
CaMKII gamma holoenzyme in its (A) closed and the (B) open conformations

Ca2+
/calmodulin-dependent protein kinase II
(CaM kinase II or CaMKII) is a serine/threonine-specific protein kinase that is regulated by the Ca2+
/calmodulin complex. CaMKII is involved in many signaling cascades and is thought to be an important mediator of learning and memory. [1] CaMKII is also necessary for Ca2+
homeostasis and reuptake in cardiomyocytes, [2] chloride transport in epithelia, [3] positive T-cell selection, [4] and CD8 T-cell activation. [5]

Contents

Misregulation of CaMKII is linked to Alzheimer's disease, Angelman syndrome, and heart arrhythmia. [6]

Types

There are two types of CaM kinase:

The structure of the association domain of CaMKII gamma rendered by pymol from PDB 2ux0 (left) space fill holoenzyme (center) cartoon holoemzyme (right) cartoon monome Camkii association domain.jpg
The structure of the association domain of CaMKII gamma rendered by pymol from PDB 2ux0 (left) space fill holoenzyme (center) cartoon holoemzyme (right) cartoon monome
The structure of the kinase domain of CaMKII (gamma) rendered by pymol from PDB 2v7O, green sticks = nucleotide Camkii kinase domain.jpg
The structure of the kinase domain of CaMKII (gamma) rendered by pymol from PDB 2v7O, green sticks = nucleotide

Structure, function, and autoregulation

Activation and autoregulation of CaMKII Psy161ST redrawn.svg
Activation and autoregulation of CaMKII

CaMKII accounts for 1–2% of all proteins in the brain, [7] [8] and has 28 different isoforms. The isoforms derive from the alpha, beta, gamma, and delta genes.

Structural domain

All of the isoforms of CaMKII have: a catalytic domain, an autoinhibitory domain, a variable segment, and a self-association domain. [9]

The catalytic domain has several binding sites for ATP and other substrate anchor proteins. It is responsible for the transfer of phosphate from ATP to Ser or Thr residues in substrates. The autoinhibitory domain features a pseudosubstrate site, which binds to the catalytic domain and blocks its ability to phosphorylate proteins. [10]

The structural feature that governs this autoinhibition is the Threonine 286 residue. Phosphorylation of this site will permanently activate the CaMKII enzyme. Once the Threonine 286 residue has been phosphorylated, the inhibitory domain is blocked from the pseudosubstrate site. This effectively blocks autoinhibition, allowing for permanent activation of the CaMKII enzyme. This enables CamKII to be active, even in the absence of calcium and calmodulin. [11]

The other two domains in CaMKII are the variable and self-association domains. Differences in these domains contribute to the various CaMKII isoforms. [12]

The self-association domain (CaMKII AD) is found at the C terminus, the function of this domain is the assembly of the single proteins into large (8 to 14 subunits) multimers [13]

Calcium and calmodulin dependence

The sensitivity of the CaMKII enzyme to calcium and calmodulin is governed by the variable and self-associative domains. This sensitivity level of CaMKII will also modulate the different states of activation for the enzyme. Initially, the enzyme is activated; however, autophosphorylation does not occur because there is not enough calcium or calmodulin present to bind to neighboring subunits. As greater amounts of calcium and calmodulin accumulate, autophosphorylation occurs leading to persistent activation of the CaMKII enzyme for a short period of time. However, the Threonine 286 residue eventually becomes dephosphorylated, leading to inactivation of CaMKII. [14] [15]

Autophosphorylation

Autophosphorylation is the process in which a kinase attaches a phosphate group to itself. When CaMKII autophosphorylates, it becomes persistently active. Phosphorylation of the Threonine 286 site allows for the activation of the catalytic domain. Autophosphorylation is enhanced by the structure of the holoenzyme because it is present in two stacked rings. The close proximity of these adjacent rings increases the probability of phosphorylation of neighboring CaMKII enzymes, furthering autophosphorylation. [16] A mechanism that promotes autophosphorylation features inhibition of the PP1 (protein phosphatase I). This enables CaMKII to be constantly active by increasing the likelihood of autophosphorylation. [17]

Long-term potentiation

Calcium/ calmodulin dependent protein kinase II is also heavily implicated in long-term potentiation (LTP) – the molecular process of strengthening active synapses that is thought to underlie the processes of memory. It is involved in many aspects of this process. LTP is initiated when the NMDA receptors are in a local environment with a voltage potential high enough to displace the positively-charged Mg2+ ion from the channel pore. As a result of the channel being unblocked, Ca2+ ions are able to enter into the postsynaptic neuron through the NMDA receptor channel. This Ca2+ influx activates CaMKII. It has been shown that there is an increase in CaMKII activity directly in the post synaptic density of dendrites after LTP induction, suggesting that activation is a direct result of stimulation. [18] [19]

In LTP

When alpha-CaMKII is knocked out in mice, LTP is reduced by 50%. This can be explained by the fact that beta-CaMKII is responsible for approximately 65% of CaMKII activity. [20] [21] LTP can be completely blocked if CaMKII is modified so that it cannot remain active. [2] [22] After LTP induction, CaMKII moves to the postsynaptic density (PSD). However, if the stimulation does not induce LTP, the translocation is quickly reversible. Binding to the PSD changes CaMKII so that it is less likely to become dephosphorylated. CaMKII transforms from a substrate for Protein Phosphatase 2A (PP2A), which is responsible for dephosphorylating CaMKII, to that of Protein Phosphatase 1. Strack, S. (1997) [18] demonstrated this phenomenon by chemically stimulating hippocampal slices. This experiment illustrates that CaMKII contributes to the enhancement of synaptic strength. Sanhueza et al. [23] found that persistent activation of CaMKII is necessary for the maintenance of LTP. She induced LTP in hippocampal slices and experimentally applied an antagonist (CaMKIINtide) to prevent CaMKII from remaining active. The slices that were applied with CaMKIINtide showed a decrease in Normalized EPSP slope after the drug infusion, meaning that the induced LTP reversed itself. The Normalized EPSP slope remained constant in the control; CaMKII continues to be involved in the LTP maintenance process even after LTP establishment. CaMKII is activated by calcium/calmodulin, but it is maintained by autophosphorylation. CaMKII is activated by the NMDA-receptor-mediated Calcium elevation that occurs during LTP induction. Activation is accompanied by phosphorylation of both the alpha and beta-subunits and Thr286/287.

Independent induction of LTP

LTP can be induced by artificially injecting CaMKII. When CaMKII is infused in postsynaptically in the hippocampal slices and intracellular perfusion or viral expression, there is a two- to threefold increase in the response of the synapse to glutamate and other chemical signals. [24] [25]

Mechanistic role in LTP

There is strong evidence that after activation of CaMKII, CaMKII plays a role in the trafficking of AMPA receptors into the membrane and then the PSD of the dendrite. Movement of AMPA receptors increases postsynaptic response to presynaptic depolarization through strengthening the synapses. This produces LTP.

Mechanistically, CaMKII phosphorylates AMPA receptors at the P2 serine 831 site. This increases channel conductance of GluA1 subunits of AMPA receptors, [26] which allows AMPA receptors to be more sensitive than normal during LTP. Increased AMPA receptor sensitivity leads to increased synaptic strength.

In addition to increasing the channel conductance of GluA1 subunits, CaMKII has also been shown to aid in the process of AMPA receptor exocytosis. Reserve AMPA receptors are embedded in endosomes within the cell. CaMKII can stimulate the endosomes to move to the outer membrane and activate the embedded AMPA receptors. [27] Exocytosis of endosomes enlarges and increases the number of AMPA receptors in the synapse. The greater number of AMPA receptors increases the sensitivity of the synapse to presynaptic depolarization, and generates LTP.

Maintenance of LTP

Along with helping to establish LTP, CaMKII has been shown to be crucial in maintaining LTP. Its ability to autophosphorylate is thought to play an important role in this maintenance. Administration of certain CaMKII blockers has been shown not only to block LTP but also to reverse it in a time-dependent manner. [28]

Behavioral memory

As LTP is thought to underlie the processes of learning and memory, CaMKII is also crucial to memory formation. Behavioral studies involving genetically engineered mice have demonstrated the importance of CaMKII.

Preventing autophosphorylation

Deficit in spatial learning

In 1998, Giese and colleagues studied knockout mice that have been genetically engineered to prevent CaMKII autophosphorylation. They observed that mice had trouble finding the hidden platform in the Morris water maze task. The Morris water maze task is often used to represent hippocampus-dependent spatial learning. The mice's inability to find the hidden platform implies deficits in spatial learning. [17]

However, these results were not entirely conclusive because memory formation deficit could also be associated with sensory motor impairment resulting from genetic alteration. [29]

Deficit in fear memories

Irvine and colleagues in 2006 showed that preventing autophosphorylation of CaMKII cause mice to have impaired initial learning of fear conditioning. However, after repeated trials, the impaired mice exhibited similar fear memory formation as the control mice. CaMKII may play a role in rapid fear memory, but does not completely prevent fear memory in the long run. [30]

In 2004, Rodrigues and colleagues found that fear conditioning increased phosphorylated CaMKII in lateral amygdala synapses and dendritic spines, indicating that fear conditioning could be responsible for regulating and activating the kinase. They also discovered a drug, KN-62, that inhibited CaMKII and prevented acquisition of fear conditioning and LTP. [31]

Deficit in consolidation of memory traces

α-CaMKII heterozygous mice express half the normal protein level as the wild-type level. These mice showed normal memory storage in the hippocampus, but deficits in consolidation of memory in the cortex. [32]

Overexpression

Mayford and colleagues engineered transgenic mice that express CaMKII with a point mutation of Thr-286 to aspartate, which mimics autophosphorylation and increases kinase activity. These mice failed to show LTP response to weak stimuli, and failed to perform hippocampus-dependent spatial learning that depended on visual or olfactory cues. [33]

Researchers speculate these results could be due to lack of stable hippocampal place cells in these animals. [34]

However, because genetic modifications might cause unintentional developmental changes, viral vector delivery allows the mice's genetic material to be modified at specific stages of development. It is possible with viral vector delivery to inject a specific gene of choice into a particular region of the brain in an already developed animal. This, in fact, has been done by Tonegawa group in early 1990s and by Poulsen and colleagues in 2007. Both groups used this method to inject CaMKII into the hippocampus. They found that overexpression of CaMKII resulted in slight enhancement of acquisition of new memories. [35] [36]

Addiction

Drug-induced changes in CaMKII function have been implicated in addiction.

Different forms

CaMK2A

CaMKIIA is one of the major forms of CamKII. It has been found to play a critical role in sustaining activation of CamKII at the postsynaptic density. Studies have found that knockout mice without CaMKIIA demonstrate a low frequency of LTP. Additionally, these mice do not form persistent, stable place cells in the hippocampus. [37]

CaMK2B

CaMK2B has an autophosphorylation site at Thr287. It functions as a targeting or docking module. Reverse transcription-polymerase chain reaction and sequencing analysis identified at least five alternative splicing variants of beta CaMKII (beta, beta6, betae, beta'e, and beta7) in brain and two of them (beta6 and beta7) were first detected in any species. [38]

CaMK2D

CaMK2D appears in both neuronal and non-neuronal cell types. It is characterized particularly in many tumor cells, such as a variety of pancreatic, leukemic, breast and other tumor cells. [39] found that CaMK2D is downregulated in human tumor cells.

CaMK2G

CaMK2G has been shown to be a crucial extracellular signal-regulated kinase in differentiated smooth muscle cells. [40]

Genes

See also

Related Research Articles

<span class="mw-page-title-main">Calmodulin</span> Messenger protein

Calmodulin (CaM) (an abbreviation for calcium-modulated protein) is a multifunctional intermediate calcium-binding messenger protein expressed in all eukaryotic cells. It is an intracellular target of the secondary messenger Ca2+, and the binding of Ca2+ is required for the activation of calmodulin. Once bound to Ca2+, calmodulin acts as part of a calcium signal transduction pathway by modifying its interactions with various target proteins such as kinases or phosphatases.

<span class="mw-page-title-main">Dendritic spine</span> Small protrusion on a dendrite that receives input from a single axon

A dendritic spine is a small membranous protrusion from a neuron's dendrite that typically receives input from a single axon at the synapse. Dendritic spines serve as a storage site for synaptic strength and help transmit electrical signals to the neuron's cell body. Most spines have a bulbous head, and a thin neck that connects the head of the spine to the shaft of the dendrite. The dendrites of a single neuron can contain hundreds to thousands of spines. In addition to spines providing an anatomical substrate for memory storage and synaptic transmission, they may also serve to increase the number of possible contacts between neurons. It has also been suggested that changes in the activity of neurons have a positive effect on spine morphology.

<span class="mw-page-title-main">Long-term potentiation</span> Persistent strengthening of synapses based on recent patterns of activity

In neuroscience, long-term potentiation (LTP) is a persistent strengthening of synapses based on recent patterns of activity. These are patterns of synaptic activity that produce a long-lasting increase in signal transmission between two neurons. The opposite of LTP is long-term depression, which produces a long-lasting decrease in synaptic strength.

<span class="mw-page-title-main">AMPA receptor</span> Transmembrane protein family

The α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (also known as AMPA receptor, AMPAR, or quisqualate receptor) is an ionotropic transmembrane receptor for glutamate (iGluR) and predominantly Na+ ion channel that mediates fast synaptic transmission in the central nervous system (CNS). It has been traditionally classified as a non-NMDA-type receptor, along with the kainate receptor. Its name is derived from its ability to be activated by the artificial glutamate analog AMPA. The receptor was first named the "quisqualate receptor" by Watkins and colleagues after a naturally occurring agonist quisqualate and was only later given the label "AMPA receptor" after the selective agonist developed by Tage Honore and colleagues at the Royal Danish School of Pharmacy in Copenhagen. The GRIA2-encoded AMPA receptor ligand binding core (GluA2 LBD) was the first glutamate receptor ion channel domain to be crystallized.

In neuroscience, synaptic plasticity is the ability of synapses to strengthen or weaken over time, in response to increases or decreases in their activity. Since memories are postulated to be represented by vastly interconnected neural circuits in the brain, synaptic plasticity is one of the important neurochemical foundations of learning and memory.

In neurophysiology, long-term depression (LTD) is an activity-dependent reduction in the efficacy of neuronal synapses lasting hours or longer following a long patterned stimulus. LTD occurs in many areas of the CNS with varying mechanisms depending upon brain region and developmental progress.

In neuroscience, a silent synapse is an excitatory glutamatergic synapse whose postsynaptic membrane contains NMDA-type glutamate receptors but no AMPA-type glutamate receptors. These synapses are named "silent" because normal AMPA receptor-mediated signaling is not present, rendering the synapse inactive under typical conditions. Silent synapses are typically considered to be immature glutamatergic synapses. As the brain matures, the relative number of silent synapses decreases. However, recent research on hippocampal silent synapses shows that while they may indeed be a developmental landmark in the formation of a synapse, that synapses can be "silenced" by activity, even once they have acquired AMPA receptors. Thus, silence may be a state that synapses can visit many times during their lifetimes.

<span class="mw-page-title-main">Brain-derived neurotrophic factor</span> Protein found in humans

Brain-derived neurotrophic factor (BDNF), or abrineurin, is a protein that, in humans, is encoded by the BDNF gene. BDNF is a member of the neurotrophin family of growth factors, which are related to the canonical nerve growth factor (NGF), a family which also includes NT-3 and NT-4/NT-5. Neurotrophic factors are found in the brain and the periphery. BDNF was first isolated from a pig brain in 1982 by Yves-Alain Barde and Hans Thoenen.

Schaffer collaterals are axon collaterals given off by CA3 pyramidal cells in the hippocampus. These collaterals project to area CA1 of the hippocampus and are an integral part of memory formation and the emotional network of the Papez circuit, and of the hippocampal trisynaptic loop. It is one of the most studied synapses in the world and named after the Hungarian anatomist-neurologist Károly Schaffer.

Metaplasticity is a term originally coined by W.C. Abraham and M.F. Bear to refer to the plasticity of synaptic plasticity. Until that time synaptic plasticity had referred to the plastic nature of individual synapses. However this new form referred to the plasticity of the plasticity itself, thus the term meta-plasticity. The idea is that the synapse's previous history of activity determines its current plasticity. This may play a role in some of the underlying mechanisms thought to be important in memory and learning such as long-term potentiation (LTP), long-term depression (LTD) and so forth. These mechanisms depend on current synaptic "state", as set by ongoing extrinsic influences such as the level of synaptic inhibition, the activity of modulatory afferents such as catecholamines, and the pool of hormones affecting the synapses under study. Recently, it has become clear that the prior history of synaptic activity is an additional variable that influences the synaptic state, and thereby the degree, of LTP or LTD produced by a given experimental protocol. In a sense, then, synaptic plasticity is governed by an activity-dependent plasticity of the synaptic state; such plasticity of synaptic plasticity has been termed metaplasticity. There is little known about metaplasticity, and there is much research currently underway on the subject, despite its difficulty of study, because of its theoretical importance in brain and cognitive science. Most research of this type is done via cultured hippocampus cells or hippocampal slices.

<span class="mw-page-title-main">Synapse</span> Structure connecting neurons in the nervous system

In the nervous system, a synapse is a structure that permits a neuron to pass an electrical or chemical signal to another neuron or to the target effector cell.

<span class="mw-page-title-main">Calcium/calmodulin-dependent protein kinase type II subunit alpha</span> Protein-coding gene in the species Homo sapiens

Calcium/calmodulin-dependent protein kinase type II subunit alpha (CAMKIIα), a.k.a.Ca2+/calmodulin-dependent protein kinase II alpha, is one subunit of CamKII, a protein kinase (i.e., an enzyme which phosphorylates proteins) that in humans is encoded by the CAMK2A gene.

Coincidence detection is a neuronal process in which a neural circuit encodes information by detecting the occurrence of temporally close but spatially distributed input signals. Coincidence detectors influence neuronal information processing by reducing temporal jitter and spontaneous activity, allowing the creation of variable associations between separate neural events in memory. The study of coincidence detectors has been crucial in neuroscience with regards to understanding the formation of computational maps in the brain.

<span class="mw-page-title-main">CAMK2D</span> Protein-coding gene in the species Homo sapiens

Calcium/calmodulin-dependent protein kinase type II delta chain is an enzyme that in humans is encoded by the CAMK2D gene.

The spine apparatus (SA) is a specialized form of endoplasmic reticulum (ER) that is found in a subpopulation of dendritic spines in central neurons. It was discovered by Edward George Gray in 1959 when he applied electron microscopy to fixed cortical tissue. The SA consists of a series of stacked discs that are connected to each other and to the dendritic system of ER-tubules. The actin binding protein synaptopodin is an essential component of the SA. Mice that lack the gene for synaptopodin do not form a spine apparatus. The SA is believed to play a role in synaptic plasticity, learning and memory, but the exact function of the spine apparatus is still enigmatic.

<span class="mw-page-title-main">Activity-regulated cytoskeleton-associated protein</span> Protein-coding gene in the species Homo sapiens

Activity-regulated cytoskeleton-associated protein is a plasticity protein that in humans is encoded by the ARC gene. The gene is believed to derive from a retrotransposon. The protein is found in the neurons of tetrapods and other animals where it can form virus-like capsids that transport RNA between neurons.

Synaptic tagging, or the synaptic tagging hypothesis, was first proposed in 1997 by Julietta U. Frey and Richard G. Morris; it seeks to explain how neural signaling at a particular synapse creates a target for subsequent plasticity-related product (PRP) trafficking essential for sustained LTP and LTD. Although the molecular identity of the tags remains unknown, it has been established that they form as a result of high or low frequency stimulation, interact with incoming PRPs, and have a limited lifespan.

<span class="mw-page-title-main">Homosynaptic plasticity</span> Type of synaptic plasticity.

Homosynaptic plasticity is one type of synaptic plasticity. Homosynaptic plasticity is input-specific, meaning changes in synapse strength occur only at post-synaptic targets specifically stimulated by a pre-synaptic target. Therefore, the spread of the signal from the pre-synaptic cell is localized.

<span class="mw-page-title-main">Mary B. Kennedy</span> American biochemist and neuroscientist

Mary Bernadette Kennedy is an American biochemist and neuroscientist. She is a member of the American Academy of Arts and Sciences, and is the Allen and Lenabelle Davis Professor of Biology at the California Institute of Technology, where she has been a member of the faculty since 1981. Her research focuses on the molecular mechanisms of synaptic plasticity, the process underlying formation of memory in the central nervous system. Her lab uses biochemical and molecular biological methods to study the protein machinery within a structure called the postsynaptic density. Kennedy has published over 100 papers with over 20,000 total citations.

Early long-term potentiation (E-LTP) is the first phase of long-term potentiation (LTP), a well-studied form of synaptic plasticity, and consists of an increase in synaptic strength. LTP could be produced by repetitive stimulation of the presynaptic terminals, and it is believed to play a role in memory function in the hippocampus, amygdala and other cortical brain structures in mammals.

References

  1. Yamauchi, Takashi (2005). "Neuronal Ca2+/Calmodulin-Dependent Protein Kinase II—Discovery, Progress in a Quarter of a Century, and Perspective: Implication for Learning and Memory". Biological & Pharmaceutical Bulletin. 28 (8): 1342–54. doi: 10.1248/bpb.28.1342 . PMID   16079472.
  2. 1 2 Anderson, M (2005). "Calmodulin kinase signaling in heart: an intriguing candidate target for therapy of myocardial dysfunction and arrhythmias". Pharmacology & Therapeutics. 106 (1): 39–55. doi:10.1016/j.pharmthera.2004.11.002. PMID   15781121.
  3. Fährmann, Michael; Kaufhold, Marc-André (2006). "Functional partitioning of epithelial protein kinase CaMKII in signal transduction". Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 1763 (1): 101–9. doi:10.1016/j.bbamcr.2005.11.012. PMID   16406114.
  4. McGargill, Maureen A.; Sharp, Leslie L.; Bui, Jack D.; Hedrick, Stephen M.; Calbo, Sébastien (July 2005). "Active Ca2+
    /calmodulin-dependent protein kinase II gamma B impairs positive selection of T cells by modulating TCR signaling"
    . The Journal of Immunology. 175 (2): 656–64. doi: 10.4049/jimmunol.175.2.656 . PMID   16002660. S2CID   35436952.
  5. Lin, Meei Yun; Zal, Tomasz; Ch'en, Irene L.; Gascoigne, Nicholas R. J.; Hedrick, Stephen M. (May 2005). "A pivotal role for the multifunctional calcium/calmodulin-dependent protein kinase II in T cells: from activation to unresponsiveness". The Journal of Immunology. 174 (9): 5583–92. doi: 10.4049/jimmunol.174.9.5583 . PMID   15843557. S2CID   21614214.
  6. Yamauchi, Takashi (August 2005). "Neuronal Ca2+
    /calmodulin-dependent protein kinase II—discovery, progress in a quarter of a century, and perspective: implication for learning and memory"
    . Biological & Pharmaceutical Bulletin. 28 (8): 1342–54. doi: 10.1248/bpb.28.1342 . PMID   16079472.
  7. Bennett, M.K., Erondu, N.E., and Kennedy, M.B. (1983). Purification and characterization of a calmodulin-dependent protein kinase that is highly concentrated in brain. J Biol Chem 258, 12735-12744.
  8. Erondu, N.E., and Kennedy, M.B. (1985). Regional distribution of type II Ca2+/calmodulin-dependent protein kinase in rat brain. J Neurosci 5, 3270-3277.
  9. Hudmon, Andy; Schulman, Howard (2002). "Neuronal Ca2+/Calmodulin-Dependent Protein Kinase II: The Role of Structure and Autoregulation in Cellular Function". Annual Review of Biochemistry. 71: 473–510. doi:10.1146/annurev.biochem.71.110601.135410. PMID   12045104.
  10. Kanaseki, T; Ikeuchi, Y; Sugiura, H; Yamauchi, T (1991). "Structural features of Ca2+/calmodulin-dependent protein kinase II revealed by electron microscopy". The Journal of Cell Biology. 115 (4): 1049–60. doi:10.1083/jcb.115.4.1049. PMC   2289961 . PMID   1659571.
  11. Yang, E; Schulman, H (1999). "Structural examination of autoregulation of multifunctional calcium/calmodulin-dependent protein kinase II". The Journal of Biological Chemistry. 274 (37): 26199–208. doi: 10.1074/jbc.274.37.26199 . PMID   10473573. S2CID   16106663.
  12. Giese, K. P. (1998). "Autophosphorylation at Thr286 of the  Calcium-Calmodulin Kinase II in LTP and Learning". Science. 279 (5352): 26199–208. doi:10.1126/science.279.5352.870. PMID   9452388.
  13. Griffith LC, Lu CS, Sun XX (October 2003). "CaMKII, an enzyme on the move: regulation of temporospatial localization". Mol. Interv. 3 (7): 386–403. doi:10.1124/mi.3.7.386. PMID   14993460.
  14. Miller, S.G.; Kennedy, M.B. (1986). "Regulation of brain type II Ca2+/calmodulin-dependent protein kinase by autophosphorylation: a Ca2+-triggered molecular switch". Cell . 44 (6): 861–870. doi:10.1016/0092-8674(86)90008-5. PMID   3006921. S2CID   491812.
  15. Lisman, J (1994). "The CaM kinase II hypothesis for the storage of synaptic memory". Trends in Neurosciences. 17 (10): 406–12. doi:10.1016/0166-2236(94)90014-0. PMID   7530878. S2CID   33109273.
  16. Blitzer, Robert D.; Wong, Tony; Nouranifar, Rabin; Iyengar, Ravi; Landau, Emmanuel M. (1995). "Postsynaptic CAMP pathway gates early LTP in hippocampal CA1 region". Neuron. 15 (6): 1403–14. doi: 10.1016/0896-6273(95)90018-7 . PMID   8845163. S2CID   8220445.
  17. 1 2 Giese, K. P.; Fedorov, NB; Filipkowski, RK; Silva, AJ (1998). "Autophosphorylation at Thr286 of the  Calcium-Calmodulin Kinase II in LTP and Learning". Science. 279 (5352): 870–3. doi:10.1126/science.279.5352.870. PMID   9452388.
  18. 1 2 Strack, S.; Choi, S; Lovinger, DM; Colbran, RJ (1997). "Translocation of Autophosphorylated Calcium/Calmodulin-dependent Protein Kinase II to the Postsynaptic Density". Journal of Biological Chemistry. 272 (21): 13467–70. doi: 10.1074/jbc.272.21.13467 . PMID   9153188. S2CID   37467211.
  19. Gardoni, F; Schrama, LH; Kamal, A; Gispen, WH; Cattabeni, F; Di Luca, M (2001). "Hippocampal synaptic plasticity involves competition between Ca2+/calmodulin-dependent protein kinase II and postsynaptic density 95 for binding to the NR2A subunit of the NMDA receptor". The Journal of Neuroscience. 21 (5): 1501–9. doi:10.1523/JNEUROSCI.21-05-01501.2001. hdl:1874/3794. PMC   6762931 . PMID   11222640.
  20. Silva, A.; Stevens, C.; Tonegawa, S; Wang, Y (1992). "Deficient hippocampal long-term potentiation in alpha-calcium-calmodulin kinase II mutant mice". Science. 257 (5067): 201–6. Bibcode:1992Sci...257..201S. doi:10.1126/science.1378648. PMID   1378648.
  21. Hinds H. L.; Tonegawa, S.; Malinow, R. (1998). "CA1 Long-Term Potentiation Is Diminished but Present in Hippocampal Slices from α-CaMKII Mutant Mice". Learning & Memory. 5 (4): 344–354. doi: 10.1101/lm.5.4.344 . S2CID   9166287.
  22. Hrabetova, S; Sacktor, TC (1996). "Bidirectional regulation of protein kinase M zeta in the maintenance of long-term potentiation and long-term depression". The Journal of Neuroscience. 16 (17): 5324–33. doi:10.1523/JNEUROSCI.16-17-05324.1996. PMC   6578881 . PMID   8757245.
  23. Sanhueza, M; McIntyre, CC; Lisman, JE (2007). "Reversal of synaptic memory by Ca2+/calmodulin-dependent protein kinase II inhibitor". The Journal of Neuroscience. 27 (19): 5190–9. doi:10.1523/JNEUROSCI.5049-06.2007. PMC   6672374 . PMID   17494705.
  24. Davies, SN; Lester, RA; Reymann, KG; Collingridge, GL (1989). "Temporally distinct pre- and post-synaptic mechanisms maintain long-term potentiation". Nature. 338 (6215): 500–3. Bibcode:1989Natur.338..500D. doi:10.1038/338500a0. PMID   2564640. S2CID   4339539.
  25. Montgomery, JM; Pavlidis, P; Madison, DV (2001). "Pair recordings reveal all-silent synaptic connections and the postsynaptic expression of long-term potentiation". Neuron. 29 (3): 691–701. doi: 10.1016/S0896-6273(01)00244-6 . PMID   11301028. S2CID   2441189.
  26. Collingridge, Graham L.; Benke, Tim A.; Lüthi, Andreas; Isaac, John T. R. (1998). "Modulation of AMPA receptor unitary conductance by synaptic activity". Nature. 393 (6687): 793–7. Bibcode:1998Natur.393..793B. doi:10.1038/31709. PMID   9655394. S2CID   47246118.
  27. Lisman, John; Schulman, Howard; Cline, Hollis (2002). "The Molecular Basis of CaMKII Function in Synaptic and Behavioural Memory". Nature Reviews Neuroscience. 3 (3): 175–90. doi:10.1038/nrn753. PMID   11994750. S2CID   5844720.
  28. Yang, H.-W.; Hu, XD; Zhang, HM; Xin, WJ; Li, MT; Zhang, T; Zhou, LJ; Liu, XG (2003). "Roles of CaMKII, PKA, and PKC in the Induction and Maintenance of LTP of C-Fiber-Evoked Field Potentials in Rat Spinal Dorsal Horn". Journal of Neurophysiology. 91 (3): 1122–33. doi:10.1152/jn.00735.2003. PMID   14586032.
  29. Rudy, Jerry W. (2004). The Neurobiology of Learning and Memory. Snauer. ISBN   978-0-87893-669-4.[ page needed ]
  30. Irvine, Elaine E.; Von Hertzen, Laura S. J.; Plattner, Florian; Giese, Karl Peter (2006). "αCaMKII autophosphorylation: a fast track to memory". Trends in Neurosciences. 29 (8): 459–65. doi:10.1016/j.tins.2006.06.009. PMID   16806507. S2CID   53151434.
  31. Rodrigues, S. M.; Farb, CR; Bauer, EP; Ledoux, JE; Schafe, GE (2004). "Pavlovian Fear Conditioning Regulates Thr286 Autophosphorylation of Ca2+/Calmodulin-Dependent Protein Kinase II at Lateral Amygdala Synapses". Journal of Neuroscience. 24 (13): 3281–8. doi:10.1523/JNEUROSCI.5303-03.2004. PMC   6730013 . PMID   15056707.
  32. Frankland, Paul W.; O'Brien, Cara; Ohno, Masuo; Kirkwood, Alfredo; Silva, Alcino J. (2001). "Alpha-CaMKII-dependent plasticity in the cortex is required for permanent memory". Nature. 411 (6835): 309–13. Bibcode:2001Natur.411..309F. doi:10.1038/35077089. PMID   11357133. S2CID   4384100.
  33. Mayford, Mark; Wang, Jian; Kandel, Eric R; O'Dell, Thomas J (1995). "CaMKII regulates the frequency-response function of hippocampal synapses for the production of both LTD and LTP". Cell. 81 (6): 891–904. doi: 10.1016/0092-8674(95)90009-8 . PMID   7781066. S2CID   17934142.
  34. Rotenberg, Alexander; Mayford, Mark; Hawkins, Robert D; Kandel, Eric R; Muller, Robert U (1996). "Mice Expressing Activated CaMKII Lack Low Frequency LTP and Do Not Form Stable Place Cells in the CA1 Region of the Hippocampus". Cell. 87 (7): 1351–61. doi: 10.1016/S0092-8674(00)81829-2 . PMID   8980240. S2CID   16704390.
  35. Tonegawa S (1994). "Gene targeting: a new approach for the analysis of mammalian memory and learning". Molecular Neurobiology: Mechanisms Common to Brain, Skin and Immune System. Series: Progress in Clinical and Biological Research. Willey-Liss, Inc. 390: 5–18. PMID   7724650.
  36. Poulsen, D.J.; Standing, D.; Bullshields, K.; Spencer, K.; Micevych, P.E.; Babcock, A.M. (2007). "Overexpression of hippocampal Ca2+/calmodulin-dependent protein kinase II improves spatial memory". Journal of Neuroscience Research. 85 (4): 735–9. doi:10.1002/jnr.21163. PMID   17171706. S2CID   45751857.
  37. Soderling, T (2000). "CaM-kinases: modulators of synaptic plasticity". Current Opinion in Neurobiology. 10 (3): 375–80. doi:10.1016/S0959-4388(00)00090-8. PMID   10851169. S2CID   31122499.
  38. Wang, P; Wu, YL; Zhou, TH; Sun, Y; Pei, G (2000). "Identification of alternative splicing variants of the β subunit of human Ca2+/calmodulin-dependent protein kinase II with different activities". FEBS Letters. 475 (2): 107–10. doi: 10.1016/S0014-5793(00)01634-3 . PMID   10858498. S2CID   39732332.
  39. Wang, P; Wu, YL; Zhou, TH; Sun, Y; Pei, G (2000). "Identification of alternative splicing variants of the β subunit of human Ca2+/calmodulin-dependent protein kinase II with different activities". FEBS Letters. 475 (2): 1–11. doi: 10.1016/S0014-5793(00)01634-3 . PMID   10858498. S2CID   39732332.
  40. Marganski, W. A.; Gangopadhyay, SS; Je, HD; Gallant, C; Morgan, KG (2005). "Targeting of a Novel Ca+2/Calmodulin-Dependent Protein Kinase II Is Essential for Extracellular Signal-Regulated Kinase-Mediated Signaling in Differentiated Smooth Muscle Cells". Circulation Research. 97 (6): 541–549. doi: 10.1161/01.RES.0000182630.29093.0d . PMID   16109919. S2CID   10316848.
This article incorporates text from the public domain Pfam and InterPro: IPR013543