Next Article in Journal
The Effects of the Pragmatic Intervention Programme in Children with Autism Spectrum Disorder and Developmental Language Disorder
Previous Article in Journal
Towards Conceptual Clarification of Proactive Inhibitory Control: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CaMKIIα as a Promising Drug Target for Ischemic Grey Matter

by
Nane Griem-Krey
1,
Andrew N. Clarkson
2 and
Petrine Wellendorph
1,*
1
Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
2
Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin 9016, New Zealand
*
Author to whom correspondence should be addressed.
Brain Sci. 2022, 12(12), 1639; https://doi.org/10.3390/brainsci12121639
Submission received: 21 October 2022 / Revised: 26 November 2022 / Accepted: 28 November 2022 / Published: 29 November 2022
(This article belongs to the Section Molecular and Cellular Neuroscience)

Abstract

:
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a major mediator of Ca2+-dependent signaling pathways in various cell types throughout the body. Its neuronal isoform CaMKIIα (alpha) centrally integrates physiological but also pathological glutamate signals directly downstream of glutamate receptors and has thus emerged as a target for ischemic stroke. Previous studies provided evidence for the involvement of CaMKII activity in ischemic cell death by showing that CaMKII inhibition affords substantial neuroprotection. However, broad inhibition of this central kinase is challenging because various essential physiological processes like synaptic plasticity rely on intact CaMKII regulation. Thus, specific strategies for targeting CaMKII after ischemia are warranted which would ideally only interfere with pathological activity of CaMKII. This review highlights recent advances in the understanding of how ischemia affects CaMKII and how pathospecific pharmacological targeting of CaMKII signaling could be achieved. Specifically, we discuss direct targeting of CaMKII kinase activity with peptide inhibitors versus indirect targeting of the association (hub) domain of CaMKIIα with analogues of γ-hydroxybutyrate (GHB) as a potential way to achieve more specific pharmacological modulation of CaMKII activity after ischemia.

1. Introduction

Stroke is the third-leading cause of death and disability worldwide and its burden is expected to increase with an aging population [1]. Today, there are 6.5 million deaths from strokes every year, and over 50% of the surviving stroke patients suffer from permanent disabilities, causing an enormous public health burden [1,2]. There are two main types of strokes, ischemic and hemorrhagic, whereof ischemic stroke is the most prevalent one [1,3]. An ischemic stroke happens when the blood flow to a certain brain region is compromised by an obstruction of a blood vessel. The reduction in blood supply leads to energy failure of cells within the affected area as well as the induction of various signaling cascades and inflammation processes, ultimately resulting in cell death and infarct formation [4,5]. In particular, signaling anomalies of the main excitatory neurotransmitter glutamate (excitotoxicity) play a substantial role in mediating ischemic cell death [6,7].
Despite the heavy disease burden of ischemic stroke, treatment options are limited. To stop the progression of irreversible damage to neighboring tissue over time, timely restoration of blood flow has proven as an effective therapy capable of salvaging ischemic brain tissue [8]. This can be achieved by pharmacological thrombolysis with recombinant human tissue-type plasminogen activator (tPA, alteplase) or by mechanical thrombectomy [9,10]. However, there is a high number of patients who do not meet the criteria for recanalization therapy, and even the majority of patients receiving the therapy still experience permanent disabilities [11,12,13]. In fact, the narrow time window of 4.5 h after symptom onset and the risk of hemorrhagic transformation results in as little as ~5% of patients actually receiving tPA [11,14]. Collectively, this highlights the critical need for novel therapies improving the long-term outcome for patients. Tremendous efforts have been made to develop therapies targeting underlying cell death mechanisms to save dying tissue acutely after the initial injury (neuroprotectants), or therapies aiming to augment the function of surviving tissue and promote functional recovery after stroke [15,16]. Failures in the design of both preclinical and clinical studies have, however, among other reasons, hampered translation into the clinic [17]. Yet, a recent phase III trial (ESCAPE-NA1) on the peptide nerinetide has recently brought new hope to the field by demonstrating that neuroprotection could be a feasible treatment option [18]. Nerinetide inhibits the protein-protein interaction of N-methyl-D-aspartate (NMDA)-type glutamate receptors and the scaffold protein post-synaptic density protein 95 (PSD-95), thereby uncoupling NMDA receptors from excitotoxic signaling pathways [18,19]. Although the study observed drug-drug interactions of nerinetide with tPA, it did demonstrate that targeting a key signaling pathway upstream of excitotoxic cell death can result in clinically beneficial outcomes in the patient group not receiving tPA [18].
This review will focus on another potential target for neuroprotection, which also directly interacts with NMDA receptors, namely Ca2+/calmodulin-dependent protein kinase II alpha (CaMKIIα). CaMKIIα is a major mediator of both physiological and pathological glutamate signaling [20,21,22,23], and post-insult CaMKII inhibition has been shown to substantially reduce cell death in vitro and in vivo [24,25,26]. However, as different subtypes of CaMKII are expressed in most cell types, fulfilling critical signaling tasks, targeting CaMKIIα after ischemia requires strategies that are both subtype-selective as well as specific for pathological regulatory pathways governed by this important kinase. Here, we briefly introduce CaMKII and its regulation with specific focus on emerging functional roles of its hub domain. Next, we address the role of CaMKII in physiological glutamate signaling and present how ischemia induces dysregulation of CaMKIIα. Furthermore, we discuss recent advances in pharmacological intervention with CaMKIIα. In particular, we address the differential outcomes by interfering directly with CaMKII kinase activity versus indirect modulation of CaMKII function via targeting of its hub domain.

2. CaMKII

The CaMKII protein family is encoded by four separate genes yielding four different variants (CaMKIIα, β, γ and δ). Albeit expressed throughout the whole body, CaMKII is most known for its neuronal and cardiac functions. Specifically, CaMKIIδ is highly expressed in cardiomyocytes contributing to cardiac placemaking [23] and calcium handling [27], whereas CaMKIIα and CaMKIIβ are predominantly expressed in neurons accounting for a remarkable 1–2% of total brain protein [28]. The latter emphasizes the importance of CaMKIIα/β as a signaling module, which was shown to mediate synaptic plasticity underlying learning and memory [20,29,30,31]. Of note, this review will focus on neuronal CaMKIIα and CaMKIIβ, and the term CaMKII will thus refer to CaMKIIα/β if not otherwise specified.

2.1. CaMKII Structure and Activity Regulation

Each CaMKII protein comprises four domains, i.e., a N-terminal Ser/Thr-specific kinase domain connected to a regulatory segment and a variable linker region, which is followed by a C-terminal association (hub) domain responsible for giving the enzyme a unique architecture (Figure 1A, reviewed in [32]). In brief, CaMKII monomers can assemble into large oligomeric structures (holoenzyme), typically made up of 12 or 14 subunits, where the hub domains form two donut-shaped rings stacked on top of each other, arranging the kinase domains around it (Figure 1A) [32,33,34]. The activity of CaMKII is controlled by intracellular Ca2+ levels. Under basal conditions, the regulatory segment of each subunit binds to its own kinase domain and thereby blocks the central substrate binding site and thus suppresses its catalytic activity to a minimum (autoinhibition). Increases in intracellular Ca2+ levels activate the enzyme by Ca2+/calmodulin (CaM) binding to the regulatory segment. Since the binding site for Ca2+/CaM overlaps with the region responsible for autoinhibition, Ca2+/CaM binding relieves the inhibition and exposes the access to the substrate site (stimulated activity, Figure 1B) [35,36]. Holoenzyme activity is further regulated by multiple threonine residues within the regulatory segment. As such, Ca2+/CaM binding and subsequent release of autoinhibition exposes Thr286 (CaMKIIα numbering; used throughout the text if not otherwise stated; Thr287 in other subtypes), which is centrally located within the autoinhibitory region of the regulatory segment. Thr286 can be phosphorylated by a neighboring activated subunits (trans autophosphorylation), which prevents the interaction of the regulatory segment with the kinase domain and thus keeps the kinase active also beyond the initial Ca2+ signal when Ca2+ levels return to basal [37]. This state is referred to as autonomous activity or Ca2+-independent activity [36,38,39]. CaMKII requires Ca2+/CaM binding and autophosphorylated Thr286 (pThr286) for full catalytic activity. In contrast, autonomous activity after Ca2+/CaM dissociation amounts to only ~15–40% of its maximum activity [40,41]. Thr286 autophosphorylation enhances the affinity of Ca2+/CaM for CaMKII (CaM trapping), thus reducing the rate of Ca2+/CaM dissociation [42]. Moreover, when Ca2+/CaM is not bound, CaMKII can be autophosphorylated at two sites located within the CaM binding domain, i.e., Thr305/306. Phosphorylation at these sites can occur in an intra-subunit reaction (cis) and inhibits Ca2+/CaM binding, making CaMKII insensitive to intracellular Ca2+ signals (Figure 1B) [32,43]. Furthermore, other autophosphorylation sites on CaMKII have been described [44,45], yet their functional consequence are less well characterized. For instance, it has been shown that CaMKII can be autophosphorylated at Thr253 in vitro and in vivo, which, in contrast to Thr286 and Thr305/6, does not directly influence kinase activity, but instead influences the interaction of CaMKII with specific binding proteins, thereby affecting the subcellular targeting of the holoenzyme [45]. Of note, CaMKII is further regulated by phosphatase activity, and distinct autophosphorylation sites have been shown to undergo dephosphorylation at different rates [46,47].
The aforementioned holoenzyme structure and activation properties are conserved among all four CaMKII proteins. In fact, they share ~95% sequence identity in the kinase and regulatory domain as well as ~80% in the hub domain [32]. Notably, CaMKII is also subjected to alternative splicing generating additional diversity with ∼40 variants [48,49,50]. The four CaMKII subtypes and its splice variants differ the most in sequence and length of the linker domain [32], which is suggested to contribute to subtype-specific functional characteristics. For example, the identity of the linker influences the activation properties of CaMKII [34,46], which was demonstrated by showing that a CaMKIIα variant without its linker domain is less sensitive to activation by Ca2+/CaM compared to a CaMKIIα variant with 30-residue linker [34]. In addition, linker identity was shown to affect subcellular targeting of the holoenzyme. As such, the linker from CaMKIIβ, but not CaMKIIα, contains an F-actin binding domain [51]. As CaMKII subtypes can assemble into homomers, but also mixed heteromers [52,53], subunit composition might determine the activation and functional properties of the CaMKII holoenzyme.

2.2. Emerging Functional Roles of the CaMKII Hub Domain

The hub domain is a unique feature of CaMKII, carrying a well-established role in CaMKII oligomerization thus securing overall kinase fidelity [54]. Hence, in addition to providing a scaffold for the arrangement of the kinase domains, the unique holoenzyme architecture also gives rise to functional properties, including its response to Ca2+/CaM [34,55]. Specifically, oligomerization provides a framework for inter-subunit autophosphorylation of Thr286 by bringing the kinase domains in close proximity and increasing the effective local concentration of subunits [56]. Thr286 autophosphorylation concomitant with CaM trapping likely contributes to cooperative activation of the holoenzyme by Ca2+/CaM and allows CaMKII to act as a frequency decoder by translating the frequency of cellular Ca2+ signals into specific amounts of kinase activity under physiological conditions [34,41,55]. The functional importance of the CaMKII holoenzyme structure is further highlighted by human patients with neurodevelopmental defects, carrying a mutation in the CaMKIIα hub domain (p.His477Tyr) shown to cause impaired oligomerization [57].
Moreover, the hub assembly was suggested to be endowed with a high degree of structural flexibility [32,33,58]. As such, in vitro experiments suggest that CaMKII activation triggers the exchange of subunits between activated/autophosphorylated but also non-activated holoenzymes. Intriguingly, it has been speculated to enable the spread of activity via autophosphorylation [58,59,60]. The proposed underlying molecular mechanism involves the interconversion of dodecameric or tetradecameric CaMKII by the release and addition of vertical dimeric subunits, suggesting the tetradecameric oligomer as an intermediate state. It has been shown that the regulatory segment can bind to the hub domain, which is believed to initiate a conformational change resulting in hub destabilization and dimer release, which can then integrate into nearby holoenzymes [58,59,60]. Although this clearly demonstrates remarkable dynamics of the holoenzyme assembly in vitro, the function of subunit exchange and relevance in vivo remains elusive [61].
Finally, kinase and hub domains have been shown to interact and to functionally influence each other, thus adding further complexity to the regulation of CaMKII activation [34,49,62]. Specifically, Sloutsky and colleagues provided structural evidence that kinase domains can form direct physical contact with the hub domain, which was suggested to contribute to CaMKII autoinhibition and/or to affect Ca2+/CaM sensitivity of the holoenzyme [49]. They also demonstrated that the Ca2+/CaM sensitivity of a holoenzyme depends on the identity of its hub domain by showing that the CaMKIIα kinase domain is more sensitive to Ca2+/CaM when fused to the CaMKIIβ hub domain with no linker in comparison to its combination with the CaMKIIα hub domain without linker. Importantly, these experiments show that the hub domain is capable of allosteric modulation of kinase activity [49]. Intriguingly, it was recently demonstrated that pharmacological interaction with the hub domain can functionally modulate the CaMKII holoenzyme, e.g., by affording neuroprotection (see Section 4.3 and Section 4.4) [25].

2.3. CaMKII in Physiological Glutamate Signaling

CaMKIIα and CaMKIIβ are reported to be expressed in a 3:1 ratio in forebrain regions, and their concentration is especially high in the dendritic spines of cortical and hippocampal neurons [28,63]. Neuronal CaMKII is most known for its crucial role in the mediation of glutamate signaling underlying learning and memory processes [41]. Specifically, upon Ca2+ influx through NMDA receptors CaMKII gets activated by Ca2+/CaM and subsequent Thr286 autophosphorylation. Autophosphorylation not only infers autonomous activity, but also affects the subcellular targeting of CaMKII [64,65]. As such, increased pThr286 levels are associated with holoenzyme translocation from the center of the dendritic spine to the post-synaptic density (PSD) [66,67]. At the PSD, CaMKII can interact with several synaptic proteins, such as the NMDA receptor subunit GluN2B [30,68,69]. Interestingly, GluN2B binding renders CaMKII autonomously active because the binding site for GluN2B on CaMKII overlaps with the binding site for its regulatory domain, thus preventing autoinhibition [64]. The co-localization with GluN2B strategically locates CaMKII in close proximity of sites of Ca2+ entry into spines as well as several substrates centrally involved in strengthening of synaptic connections, i.e., long-term potentiation (LTP) [36,41]. Prominent substrates of CaMKII in LTP are α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors, whose phosphorylation by CaMKII results in both augmented channel conductance and increased number of synaptic receptors [36,70,71]. The critical involvement of both CaMKIIα and CaMKIIβ in synaptic plasticity has been demonstrated by genetic knockdown of either of them leading to reduction in LTP and learning in mice [20,72]. Other genetic studies have further demonstrated that both CaMKIIα activation and GluN2B co-localization are essential for normal synaptic plasticity by showing that knock-in mice carrying mutations that impair GluN2B-CaMKIIα binding (GluN2B L1298A/R1300Q) or Thr286 autophosphorylation (CaMKIIα T286A) also show deficits in LTP [29,73]. In contrast, the activity of CaMKIIβ is not required for LTP, but CaMKIIβ instead fulfils important structural roles by promoting the targeting of CaMKIIα to the synapse which is mediated by the ability of CaMKIIβ to bind to F-actin via its linker domain [67,72]. Consequently, it has been suggested that the CaMKIIα/β heteromer contributes to structural plasticity as a consequence of synaptic activity, and it has been shown that CaMKII is implicated in the formation of dendritic spines and synapses [74,75].
Interestingly, CaMKII and Thr286 are also critically involved in long-term depression (LTD), leading to weakening of excitatory glutamatergic synapses and potentiation of inhibitory synapses [76,77]. The mediation of this apparent opposing effect on plasticity is enabled by finetuning its activity and subcellular location via inhibitory Thr305/306 autophosphorylation [78]. As such, pThr305/306 prevents GluN2B binding, reduces CaMKII targeting to the PSD and directs CaMKII towards LTD-related substrates [66,73]. This highlights a dual role of CaMKII in the mediation of LTP and LTD as well as the complexity of CaMKII signaling.

3. Disturbances in CaMKIIα Signaling after Ischemic Stroke

Excessive release of glutamate accompanied by overstimulation of glutamate receptors, especially NMDA receptors, is a hallmark of the pathology of ischemic stroke [6]. Yet, extensive efforts to directly target the NMDA receptor for the treatment of acute ischemic stroke could not be translated to the clinic [17,79,80]. To be able to differentiate cell death from cell survival pathways or pathological from physiological functions, it has been suggested to target key signaling pathways downstream of NMDA receptors instead [16] - a concept that has recently delivered encouraging results in a phase III clinical trial (ESCAPE-NA1) [18]. CaMKII undertakes such a key signaling role downstream of the NMDA receptor by integrating physiological but also pathological Ca2+ signals, and CaMKII has thus emerged as a target in ischemic stroke [21]. Here, we will briefly describe relevant pathological signaling pathways after ischemia and we will discuss the involvement of CaMKII in ischemic cell death. For a detailed description of the pathology of ischemic stroke the reader is referred to [4,5,7]. Of note, this review focuses on neuronal CaMKII signaling in grey matter pathology. In contrast, the entire role of CaMKII as a target in white matter remains to be investigated.

3.1. Pathophysiology of Ischemic Stroke

The pathophysiology of ischemic stroke is complex and involves the activation of several interconnected signaling cascades cumulating in cell death and tissue damage (Figure 2) [4]. The area close to the occluded artery and most affected by hypoperfusion is called the ischemic core, which usually experiences irreversible tissue damage within minutes after injury [4]. The blood flow in the tissue surrounding the infarct core is less severely affected, mostly due to residual perfusion from collateral blood vessels. Here, structural and metabolic integrity is maintained, but the tissue is characterized by functional impairments. This area is called the ischemic penumbra [81], and it is the target for therapeutic interventions, like restoration of blood flow or experimental neuroprotective therapies [17]. Yet, if no treatment is initiated, potentially viable penumbral tissue gradually gets integrated into the infarct core [82].
Deprivation from glucose and oxygen initially results in energy failure of neurons in the ischemic core. Specifically, neurons are unable to produce adenosine 5′-triphosphate (ATP), which leads to the impairment of energy-depended processes and disruption of ion homeostasis (Figure 2). As a consequence, neurons depolarize excessively, triggering the release of toxic concentrations of excitatory neurotransmitters, which fail to be taken up [5]. In fact, accumulation of glutamate in the extracellular space plays a central role in the mediation of ischemic cell death by initiating prolonged stimulation of glutamate receptors, which in turn leads to the excessive influx of Ca2+, Na+ and H2O (excitotoxicity). Whereas Na+ influx is mainly associated with the formation of cytotoxic oedema, intracellular Ca2+ overload triggers various cytotoxic signaling pathways, such as the dysregulation of Ca2+-dependent enzymes including CaMKII (Figure 2) [4,21]. Notably, various enzymes (proteases, lipases or nucleases) get overactivated as a consequence of Ca2+ overload, which are responsible for diminishing cellular integrity [4]. For instance, a major contributor to excitotoxic cell death is calpain, a Ca2+-dependent protease, which alters the stability but also function of several proteins critically involved in cellular survival by proteolytic processing [83].
The initial period after vascular occlusion is termed the acute phase (<3 days). Of note, in addition to the described neuronal changes, the acute phase is characterized by interlinked pathological processes involving all cell types of the neurovascular unit. As such, ischemia also triggers inflammation, oxidative stress, apoptosis and breakdown of the blood-brain barrier (BBB), which together with excitotoxicity ultimately cumulates into infarct formation (Figure 2) [4,82]. After the acute phase, damaged tissue transitions from injury to repair in the subacute (>3 days) and chronic phase (>1 month) [84,85]. The repair phase is characterized by a period of heightened plasticity, where neurons within the area surrounding the infarct (peri-infarct) and their connected areas facilitate structural plasticity and functional remodeling. This involves axonal growth, dendritic remodeling and the formation of new synapses leading to remapping of sensorimotor functions, which is believed to underlie a limited degree of spontaneous recovery of functional deficits after stroke [15,85,86,87]. On a molecular level, these changes are mediated by similar mechanisms that underlie synaptic plasticity during learning and memory, such as LTP and dendritic remodeling [88,89,90].

3.2. CaMKII Dysregulation after Ischemia

Several studies suggest the involvement and dysregulation of CaMKII in cell death in the acute phase after ischemia [21,91], which is also the focus of this review. Interestingly and despite its major role in synaptic plasticity [36], CaMKII signaling has, to our knowledge not been investigated during the repair phase after stroke.

3.2.1. Increased Autophosphorylation

Acute CaMKII inhibition has been shown to protect neurons from excitotoxic cell death, clearly highlighting a role of CaMKII signaling in ischemic cell death [21,24,26,92]. However, at this point it is unclear exactly how CaMKII mediates ischemic cell death. Several mechanisms for its dysregulation have been described, and the majority of studies are in agreement with the following overall sequence of events. Initially, intracellular Ca2+ overload introduced by excitotoxic glutamate signaling results in rapid activation of CaMKII via Ca2+/CaM binding. Subsequently, CaMKII undergoes increased and sustained autophosphorylation at Thr286 after ischemia in comparison to sham non-stroke controls, rendering CaMKII autonomously active. Moreover, CaMKII gets persistently translocated to the PSD, where it co-localizes with the NMDA receptor subunit GluN2B. Here, CaMKII is centrally located in close proximity of a number of substrates implicated in both cellular survival and excitotoxic cell death, and ischemia-induced hyperactivation of CaMKII is believed to lead to excessive substrate phosphorylation (Figure 3) [21,91].
Specifically, multiple in vivo models of ischemic injury could demonstrate persistent translocation of CaMKII to the PSD concomitant with elevated pThr286 levels in this fraction, while pThr286 levels and total CaMKII expression were decreased in the cytosolic fraction [26,93,94,95,96,97,98]. This pattern of CaMKII dysregulation has for instance been shown by Matsumoto and colleagues in the ischemic cortex following 1 and 2 h of ischemia as well as following 2 h of reperfusion in a mouse model of focal ischemic stroke (transient middle cerebral artery occlusion, tMCAO) [95]. In addition, rapid increase in autophosphorylation at Thr253 was observed after tMCAO [94], and similar autophosphorylation patterns for both Thr286 and Thr253 could also be demonstrated after global ischemia [26,97,98,99], which shares important pathophysiological mechanisms with ischemic stroke. However, in contrast to focal ischemic injury, global ischemia occurs after transient loss of oxygen supply to the whole brain, for example after cardiac arrest, and is characterized by neuronal death in brain regions most susceptible to ischemic injury, such as the hippocampus and striatum [100,101].

3.2.2. Self-Association

In contrast to the apparent increase in Thr286 autophosphorylation after ischemia, it has been shown that the catalytic activity of CaMKII is considerably decreased in the membrane and cytosolic fraction after both global and focal ischemia [93,102,103,104,105,106]. It has been suggested that the CaMKII inactivation after ischemia is mediated by vast self-association of multiple CaMKII holoenzymes (aggregation). As such, CaMKII aggregation is favored in vitro after Ca2+/CaM activation under conditions mimicking ischemia (low ATP, high adenosine diphosphate (ADP) and low pH) and involves the interaction of a kinase domain with the regulatory domain of a neighboring holoenzyme, which results in reduced catalytic activity [107,108,109,110]. Consequently, CaMKII aggregation has been proposed as a natural protective mechanism to minimize excessive kinase activity after ischemia, yet the potential neuroprotective effect of aggregation requires further studies [21]. However, the fact that pThr286 levels are increased after ischemia clearly indicates that at least a subpopulation of total CaMKII is hyperactive and is likely contributing to ischemic cell death, since inhibition of CaMKII activity has been shown to be neuroprotective [24,26].

3.2.3. GluN2B Co-Localization

Co-localization of CaMKIIα with GluN2B has been shown to be crucially involved in the mediation of ischemic cell death since mice carrying a mutation in the binding site for CaMKIIα on GluN2B (L1298A/R1300Q) are protected from global ischemia [111]. Interestingly, it appears that the generation of autonomous activity and/or the strategic positioning of the holoenzyme at the PSD as a consequence of CaMKIIα-GluN2B co-localization could be the decisive factors for the neurotoxic role of this interaction as opposed to CaMKII-mediated phosphorylation of GluN2B at Ser1303. This is based on the fact that the level of Ser1303 phosphorylation of GluN2B was not changed in the hippocampus after global or in cortex after focal ischemia, thus indicating that it is not involved in ischemic cell death [94,112].

3.2.4. Calpain-Mediated Proteolytic Processing

Recently, another pathological regulatory mechanism of CaMKII was described, which could contribute to the hyperactivity of a subpopulation of CaMKII after ischemia. Specifically, CaMKII was found to be proteolytically processed by the protease calpain after ischemia, which generates a stable kinase fragment with uncontrolled kinase activity (∆CaMKII, 31 kDa) [34,113]. As the cleavage site was mapped to the beginning of the regulatory domain (i.e., M281↓H282), cleavage of CaMKII at this site generates a N-terminal fragment consisting of the kinase domain only (Figure 4). In fact, ∆CaMKII is missing the regulatory motif responsible for kinase autoinhibition and activity regulation by Ca2+/CaM, making this fragment constitutively active [34,70,114,115]. Although proteolytic processing of CaMKII by calpain could already be described in vitro in 1989 [116,117,118], its relevance in vivo was only recently addressed. Intriguingly, ∆CaMKII has been detected exclusively after ischemic stroke (photothrombotic stroke) [113]. Furthermore, in vitro experiments showed that proteolytic processing of CaMKII at this site requires prior Ca2+-stimulation and Thr286 autophosphorylation [113], resembling ischemic conditions of CaMKII [26]. Proteolytic processing of CaMKII might thus represent a pathological mechanism downstream of ischemia-activated and Thr286 phosphorylated CaMKII, and the uncontrolled kinase activity of ∆CaMKII might contribute to the neurotoxic role of sustained CaMKII activity after ischemia by phosphorylating specific neuronal proteins to direct cell death. Although it is well-established that calpain overactivation contributes to excitotoxic cell death [119,120,121], the neurotoxic role of ∆CaMKII requires further experimental validation.

3.2.5. Potential Downstream Effectors

Collectively, ischemia might differently affect individual subpopulations of CaMKII. As such, one CaMKII pool might be inactivated by inter-holoenzyme aggregation, whereas another pool might undergo sustained activation involving increased autophosphorylation, translocation to the PSD as well as proteolytic activation. Currently it is unclear which one of these pathological mechanisms, leading to aberrant CaMKII activity, are most centrally involved in the mediation of excitotoxic cell death. Furthermore, it is unknown which downstream pathways of CaMKII are affected. Yet, a variety of substrates of CaMKII have been suggested as potential downstream effectors contributing to cell death (reviewed in [21]). For instance, CaMKII can phosphorylate the GluA1 subunit of AMPA receptors at Ser831, which mediates increased channel conductance for Ca2+ and may thus enhance excitotoxicity [122]. Notably, increased Ser831 phosphorylation after ischemia has been demonstrated in vivo [94,123]. CaMKII was also shown to phosphorylate the GABAB1 subunit of GABAB receptors at Ser867, which leads to enhanced GABAB receptor internalization [124]. Interestingly, phosphorylation of Ser867 was increased after sustained activation of glutamate receptors in cultured neurons [124], potentially contributing to downregulation of GABAB receptors and consequently reduced inhibition in the acute phase after ischemia [125]. In addition, CaMKII has been shown to modulate signaling of several other proteins with a previously described role in ischemic cell death [21], such as L-type voltage-dependent Ca2+ channels [126], connexin hemichannels [127] and acid-sensing ion channels [128].
In contrast, other downstream effects of CaMKII could mediate cell survival instead [21]. As such, phosphorylation of neuronal nitric oxide synthase (nNOS) by CaMKII results in reduced production of neurotoxic nitric oxide [129]. Furthermore, multiple studies have associated CaMKII with the activation of the transcription factor cAMP response element—binding protein (CREB), which is a well-described regulator of neuronal survival [130,131,132]. A potential role of CaMKII signaling in cell survival is supported by the fact that CaMKIIα knock-out (Camk2a−/−) mice display increased vulnerability to ischemic injury after tMCAO and photothrombotic stroke, as evidenced by increased cortical infarct sizes compared to wildtype (Camk2a+/+) littermates [25,133]. Correspondingly, prolonged CaMKII inhibition in cortical primary neurons was shown to enhance cell death in response to an excitotoxic insult [92]. Together, this led to the hypothesis that a sustained loss of CaMKII activity increases neuronal susceptibility to ischemic injury, whereas acute short-term CaMKII inhibition after injury is neuroprotective [21,92]. However, a recent study by Rumian and colleagues could not reproduce the sensitizing effect of long-term CaMKII inhibition in vitro, and they further showed that Camk2a−/− mice are protected from ischemic injury in the hippocampus after global ischemia [134]. Although discrepancies between these studies could be explained by technical differences such as divergent inhibitor concentrations or expressional changes in the employed Camk2a−/− mouse lines [134], this clearly highlights the complexity of CaMKII signaling after ischemia. Simultaneously this indicates that cell and/or injury type might determine which downstream pathway of CaMKII will dominate. In line, tissue specific responses of CaMKII to ischemia were shown by Skelding and colleagues, who found differential rates of Thr253 autophosphorylation between striatum and cortex after tMCAO [94].
Further studies are needed to elucidate the downstream signaling of CaMKII after ischemia, which would ideally require more specific genetic approaches, such as conditional gene knock-out, to overcome compensatory mechanisms, or developmental effects using germline knock-out models. Moreover, most studies evaluating downstream effects of CaMKII have been performed with inhibitors lacking subtype selectivity and/or showing off-target effects (CaMKII inhibitors will be addressed below). Consequently, better inhibitors would be desirable to study CaMKIIα signaling more specifically.

4. Targeting CaMKIIα after Ischemia

4.1. CaMKII Inhibitors

The most widely used CaMKII inhibitors are the small molecule KN93 and the peptide CN21 [135,136]. KN93 was developed in 1991 and was commonly believed to interact directly with CaMKII to prevent kinase activation by competing with Ca2+/CaM [135]. Yet, a recent study showed that KN93 inhibits CaMKII activation by directly binding to Ca2+/CaM instead [137]. In accordance with reported cellular effects, KN93 indeed only inhibits the stimulated and not the autonomous kinase activity, and its binding to Ca2+/CaM might also explain several off-target effects of the compound. For instance, Ca2+/calmodulin-dependent protein kinase IV (CaMKIV), another member of the CaM-kinase family, as well as voltage-gated potassium and calcium channels have been shown to be inhibited by KN93 [138,139,140]. In contrast, CN21 is a peptide derived from the natural and specific CaMKII inhibitor protein CaM-KIIN, which was fused to the cell penetrating peptide tat, thus enabling cellular and brain permeability [136]. Tat-CN21 inhibits CaMKII activity by binding to the kinase domain. Specifically, it interacts with the region responsible for the binding of the regulatory domain around Thr286 when CaMKII is autoinhibited. It means that prior enzyme activation by Ca2+/CaM is required for tat-CN21 binding, but also that it is able to inhibit both Ca2+/CaM-stimulated and autonomous activity [136]. Since GluN2B interacts with an overlapping site on the CaMKIIα kinase domain, tat-CN21 interferes with the interaction of CaMKIIα with GluN2B [24,136]. Furthermore, tat-CN21 displays higher selectivity for CaMKII compared to KN93, yet it does not discriminate between CaMKII subtypes [136]. Of note, further improvements of CN21 led to peptides with increased potency, such as CN19o (IC50 for CN21 0.1 μM [136] and IC50 for CN19o 0.4 nM determined for the substrate syntide2 at CaMKIIα) [141]. Moreover, as an alternative to tat-conjugated peptides, myristoylated CaM-KIIN (myr-CN27) has been used [142]. These peptides are collectively referred to as CN-peptides.

4.2. The Kinase Domain as a Target for Neuroprotection

CN-peptides are neuroprotective in vitro and in vivo (Table 1) [24,26,143]. Specifically, tat-CN21 could mediate substantial neuroprotection as evidenced in an infarct size reduction of 70% when treatment was initiated 1 h after artery occlusion at the same time as reperfusion induction in the tMCAO model [24]. Moreover, post-insult treatment with CN-peptides facilitates neuroprotection after global ischemia [26,143]. These studies were supported by in vitro findings, showing that tat-CN21 reduces glutamate-induced neuronal cell death in primary cortical and hippocampal cultures when applied 2 and 6 h after the insult, respectively [24,92]. In contrast, KN93 is only neuroprotective in vitro when present during, but not after the insult [24,92]. Since KN93 only inhibits Ca2+/CaM-stimulated activity, this led to the hypothesis that autonomous CaMKII activity is crucial for ischemic cell death and thus the relevant molecular species of CaMKIIα to target in neuroprotection. Although the inhibitor studies with KN93 are complicated by its off-target effects, this hypothesis was further supported by a recent study showing that CaMKIIα knock-in mice with impaired Thr286 autophosphorylation (T286A) are protected from hippocampal cell death induced by global ischemia [26]. However, the same mutation did not affect infarct sizes in mice after focal ischemic injury introduced by tMCAO [144], which again points towards tissue and/or injury specific responses of CaMKII. As such, Ca2+/CaM-stimulated CaMKII activity might play a bigger role than autonomous activity in mediating excitotoxic cell death after focal ischemia compared to global ischemia. Alternatively, autonomous activity could be generated by mechanisms other than Thr286 autophosphorylation or other autophosphorylation sites might be involved.

4.3. Ligands Targeting the CaMKIIα Hub Domain

Recently, another class of compounds could be linked to CaMKII modulation, namely small molecules related to γ-hydroxybutyrate (GHB). GHB itself is a metabolite of γ-aminobutyric acid (GABA) and was shown to be neuroprotective in pre-clinical models of ischemic stroke in rodents [146,147]. Although the mechanism behind GHB’s neuroprotective effect is not completely understood, it likely involves two distinct binding sites within the CNS. As such, GHB is a partial agonist at GABAB receptors with millimolar potency, but was also recently found to target CaMKII alpha with low micromolar affinity (Figure 5) [25,148]. Several series of structural analogues of GHB have been developed, which display improved selectivity and affinity for CaMKIIα in comparison to GHB. Importantly, these compounds, referred to as GHB analogues, are devoid of GABAB receptor binding [148,149,150,151,152,153]. Furthermore, the binding site could be mapped to a deep cavity within the hub domain of CaMKIIα, thus making them not only the first subtype selective CaMKIIα ligands, but also the first ligands to interact selectively with the hub domain [25]. This striking selectivity was confirmed for the conformationally restricted and brain permeable GHB analogue 3-hydroxycyclopent-1-enecarboxylic acid (HOCPCA) exhibiting a complete lack of binding to Camk2a−/− mouse brain tissue when tested in in vitro autoradiography using its tritiated radioligand (Figure 5) [25,150,154]. Moreover, the selectivity of HOCPCA was further verified by showing that it does not bind to 45 known CNS targets [155].

4.4. The CaMKIIα Hub Domain as a Target for Neuroprotection

The exact functional consequences of GHB analogue binding to the CaMKIIα hub domain is not completely understood. Importantly, it was found that HOCPCA mediates neuroprotection in vitro and in vivo (Table 1) [25,145]. In fact, HOCPCA improved the functional outcome of mice after ischemic stroke with an extended treatment window in two permanent models, namely 3 h after permanent middle cerebral artery occlusion (pMCAO) and 12 h after photothrombotic stroke [25,145]. Similar neuroprotective effects were seen after early HOCPCA treatment 30 min post-stroke in a model characterized by gradual reperfusion (thromboembolic stroke) and when tested in combination with tPA [145]. Moreover, HOCPCA increased cellular survival of cultured cortical neurons after an excitotoxic insult only when applied 1 h after the insult, but had no effect when applied during the insult [25]. This is in contrast to tat-CN21, which decreases cell death when present during the excitotoxic insult [24,92], thus pointing towards differential mechanism of actions of tat-CN21 and HOCPCA. Indeed, whereas CN-peptides directly interfere with kinase activity [136], an allosteric effect on holoenzyme function is envisaged for HOCPCA based on its interaction with the hub domain [25].

A Longer Treatment Window via Targeting of the CaMKIIα Hub?

The late-onset effect of HOCPCA in vitro might indicate that a functional effect of HOCPCA depends on pathological dysregulation of CaMKIIα. In support, hitherto no effects on classical CaMKIIα signaling could be found under naïve nonpathological conditions. HOCPCA did not affect basal Thr286 autophosphorylation in cortical primary neurons, LTP in mouse hippocampal slices nor the catalytic activity of CaMKIIα when tested in a kinase assay (ADP-Glo) [25]. Instead, a range of GHB analogues were found to substantially increase stabilization of the purified CaMKIIα hub domain in a thermal shift assay [25,152,153], suggesting that GHB analogues might alter holoenzyme functionality by introducing a conformational change of the hub domain and thereby affecting CaMKII oligomerization or protein-protein interactions. At this point it is unclear how hub domain stabilization would translate into neuroprotection, yet it was shown that HOCPCA affects CaMKIIα biochemistry after ischemia in vivo [145]. As such, HOCPCA was able to alleviate ischemia-induced dysregulation of different subpopulation of CaMKIIα after pMCAO. Particularly, HOCPCA treatment both normalized an ischemia-induced decrease of cytosolic pThr286 after stroke and downregulated the expression of the constitutively active cleavage product ∆CaMKII [145]. Intriguingly, this shows that HOCPCA alters CaMKII activity exclusively after ischemia, which is ultimately expected to affect downstream CaMKII signaling pathways. It is tempting to speculate that a conformational change introduced by HOCPCA binding underlies both its effect on cytosolic pThr286 and proteolytic processing, yet this requires further studies. Nonetheless, these results further point towards a pathospecific mechanism of HOCPCA and highlight the therapeutic potential in modulation of the hub for achieving neuroprotection.

5. Concluding Remarks on Targeting CaMKIIα after Ischemia

Due to its central role in regulating physiological and pathological glutamate signaling directly downstream of NMDA receptors, CaMKIIα emerged as a promising target for neuroprotective treatments for ischemic stroke and potentially other diseases characterized by glutamate and Ca2+ dysregulation [21,24,26]. Such a treatment would ideally be CaMKII subtype specific and would only target pathological functions of ischemia-activated CaMKIIα to avoid the interference with crucial physiological functions of CaMKIIα. Accordingly, the therapeutic potential of CN-peptides is challenged by possible effects on learning and memory as it has been shown that tat-CN21 inhibits LTP induction [156]. Although a certain degree of learning or memory impairments might be tolerated for the acute treatment of ischemic stroke, treatment with CN-peptides might also entail cardiological side effects by inhibiting CaMKIIδ in cardiomyocytes [157]. In contrast, pharmacological modulation of the CaMKIIα hub domain by GHB analogues might offer a therapeutic strategy with increased selectivity based on HOCPCA’s lack of binding to CaMKIIβ/δ/γ and apparent pathospecific mechanism [25,145]. To this end, further studies are warranted to elucidate the exact mechanism of action of pharmacological modulation of the CaMKIIα hub domain by GHB analogues. Nonetheless, the emerging functional role of the hub domain involving its influence on kinase activity, oligomerization and potentially activation triggered subunit exchange highlight the hub domain as an interesting pharmacological target [34,49,59]. In line, as HOCPCA does not directly interfere with kinase activity [25], it would be interesting to test its effect on functional recovery when treatment is initiated during the subacute phase after stroke. In contrast to the acute phase, treatments during the subacute phase aim to enhance endogenous repair mechanisms, which are for instance involving brain plasticity mechanisms that parallel those in learning and memory and might thus depend on CaMKII signaling [86,87]. Of note, CN-peptides would likely be counter-indicated for treatment during the recovery phase due to their negative effect on LTP in contrast to HOCPCA [156].
Moreover, identification of downstream effectors and interacting proteins contributing to CaMKII subsequent to ischemic cell death could guide the design of novel treatments either by directly targeting of downstream effector proteins or their protein-protein interactions with CaMKII. This would possibly allow the targeting of a specific pathological pathway over physiological and/or cell survival functions of CaMKII. For instance, this could recently be achieved by inhibiting the interaction of CaMKIIβ with GABAB receptors using a small interfering peptide derived from the CaMKII phosphorylation site on GABAB1 [158]. This peptide could successfully restore ischemia-mediated downregulation of GABAB receptors and increased neuronal survival after glutamate insult in vitro [158]. Other intriguing strategies could be to target the interaction of calpain with CaMKII with the goal to prevent ischemia-specific proteolytic processing of CaMKII [113].
Temporal and mechanistic delineation of the ischemia-induced dysregulation of CaMKII would be helpful to guide dosing regimens of pharmaceutical interventions targeting this important brain kinase. This would ideally include different types of ischemic injury (e.g., permanent versus gradual reperfusion and grey matter versus white matter injury models) and the combination with reperfusion therapy modelling thrombectomy or drug-induced thrombolysis [17]. Furthermore, more studies are needed to investigate the contribution of CaMKII signaling to ischemic cell death in non-neuronal cells such as endothelial cells or oligodendrocytes [159] or even infiltrating immune cells. Finally, with the eventual biological validation of targeting CaMKII in ischemic stroke, a broader range of acute brain injuries might be interesting to consider.

Author Contributions

Conceptualization, N.G.-K., A.N.C. and P.W.; writing—original draft preparation, N.G.-K.; writing—review and editing, N.G.-K., A.N.C. and P.W.; visualization, N.G.-K.; supervision, A.N.C. and P.W.; funding acquisition, P.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Lundbeck Foundation (R277-2018-260).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The University of Copenhagen and Otago Innovation Ltd. have licensed patent rights (EP 3 746 064) for the use of small-molecule CaMKIIα ligands in acute brain injury to Ceremedy Ltd., of which P.W. is a co-founder.

References

  1. Feigin, V.L.; Brainin, M.; Norrving, B.; Martins, S.; Sacco, R.L.; Hacke, W.; Fisher, M.; Pandian, J.; Lindsay, P. World Stroke Organization (WSO): Global Stroke Fact Sheet 2022. Int. J. Stroke 2022, 17, 18–29. [Google Scholar] [CrossRef] [PubMed]
  2. Donkor, E.S. Stroke in the 21st Century: A Snapshot of the Burden, Epidemiology, and Quality of Life. Stroke Res. Treat. 2018, 2018, 3238165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Tapuwa, D.; Musuka, M.; Wilton, S.; Traboulsi, M. Diagnosis and Management of Acute Ischemic Stroke: Speed Is Critical. Cmaj 2015, 187, 887–893. [Google Scholar]
  4. Doyle, K.P.; Simon, R.P.; Stenzel-Poore, M.P. Mechanisms of Ischemic Brain Damage. Neuropharmacology 2008, 55, 310–318. [Google Scholar] [CrossRef] [Green Version]
  5. Moskowitz, M.A.; Lo, E.H.; Iadecola, C. The Science of Stroke: Mechanisms in Search of Treatments. Neuron 2010, 67, 181–198. [Google Scholar] [CrossRef] [Green Version]
  6. Szydlowska, K.; Tymianski, M. Calcium, Ischemia and Excitotoxicity. Cell Calcium 2010, 47, 122–129. [Google Scholar] [CrossRef]
  7. Dirnagl, U.; Iadecola, C.; Moskowitz, M.A. Pathobiology of Ischaemic Stroke: An Integrated View. Trends Neurosci. 1999, 22, 391–397. [Google Scholar] [CrossRef] [PubMed]
  8. Heiss, W.-D.; Rosner, G. Functional Recovery of Cortical Neurons as Related to Degree and Duration of Ischemia. Ann. Neurol. 1983, 14, 294–301. [Google Scholar] [CrossRef]
  9. The National Institute of Neurological Disorders and Stroke Rt-PA Stroke Study Group. Tissue Plasminogen Activator for Acute Ischemic Stroke. N. Engl. J. Med. 1995, 333, 1581–1587. [Google Scholar] [CrossRef]
  10. Nogueira, R.G.; Jadhav, A.P.; Haussen, D.C.; Bonafe, A.; Budzik, R.F.; Bhuva, P.; Yavagal, D.R.; Ribo, M.; Cognard, C.; Hanel, R.A.; et al. Thrombectomy 6 to 24 Hours after Stroke with a Mismatch between Deficit and Infarct. N. Engl. J. Med. 2018, 378, 11–21. [Google Scholar] [CrossRef]
  11. De Los Ríos La Rosa, F.; Khoury, J.; Kissela, B.M.; Flaherty, M.L.; Alwell, K.; Moomaw, C.J.; Khatri, P.; Adeoye, O.; Woo, D.; Ferioli, S.; et al. Eligibility for Intravenous Recombinant Tissue-Type Plasminogen Activator within a Population: The Effect of the European Cooperative Acute Stroke Study (ECASS) III Trial. Stroke 2012, 43, 1591–1595. [Google Scholar] [CrossRef] [PubMed]
  12. Grefkes, C.; Fink, G.R. Recovery from Stroke: Current Concepts and Future Perspectives. Neurol. Res. Pract. 2020, 2, 17. [Google Scholar] [CrossRef] [PubMed]
  13. Vanacker, P.; Lambrou, D.; Eskandari, A.; Mosimann, P.J.; Maghraoui, A.; Michel, P. Eligibility and Predictors for Acute Revascularization Procedures in a Stroke Center. Stroke 2016, 47, 1844–1849. [Google Scholar] [CrossRef] [PubMed]
  14. Whiteley, W.N.; Emberson, J.; Lees, K.R.; Blackwell, L.; Albers, G.; Bluhmki, E.; Brott, T.; Cohen, G.; Davis, S.; Donnan, G.; et al. Risk of Intracerebral Haemorrhage with Alteplase after Acute Ischaemic Stroke: A Secondary Analysis of an Individual Patient Data Meta-Analysis. Lancet Neurol. 2016, 15, 925–933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Regenhardt, R.W.; Takase, H.; Lo, E.H.; Lin, D.J. Translating Concepts of Neural Repair after Stroke: Structural and Functional Targets for Recovery. Restor. Neurol. Neurosci. 2020, 38, 67–92. [Google Scholar] [CrossRef]
  16. Lyden, P.; Buchan, A.; Boltze, J.; Fisher, M. Top Priorities for Cerebroprotective Studies-A Paradigm Shift: Report from STAIR XI. Stroke 2021, 52, 3063–3071. [Google Scholar] [CrossRef]
  17. Fisher, M.; Savitz, S.I. Pharmacological Brain Cytoprotection in Acute Ischaemic Stroke—Renewed Hope in the Reperfusion Era. Nat. Rev. Neurol. 2022, 18, 193–202. [Google Scholar] [CrossRef]
  18. Hill, M.D.; Goyal, M.; Menon, B.K.; Nogueira, R.G.; McTaggart, R.A.; Demchuk, A.M.; Poppe, A.Y.; Buck, B.H.; Field, T.S.; Dowlatshahi, D.; et al. Efficacy and Safety of Nerinetide for the Treatment of Acute Ischaemic Stroke (ESCAPE-NA1): A Multicentre, Double-Blind, Randomised Controlled Trial. Lancet 2020, 395, 878–887. [Google Scholar] [CrossRef]
  19. Aarts, M.; Liu, Y.; Liu, L.; Besshoh, S.; Arundine, M.; Gurd, J.W.; Wang, Y.T.; Salter, M.W.; Tymianski, M. Treatment of Ischemic Brain Damage by Perturbing NMDA Receptor-PSD-95 Protein Interactions. Science 2002, 298, 846–850. [Google Scholar] [CrossRef]
  20. Silva, A.J.; Stevens, C.F.; Tonegawa, S.; Wang, Y. Deficient Hippocampal Long-Term Potentiation in α-Calcium-Calmodulin Kinase Mutant Mice. Science 1992, 257, 201–206. [Google Scholar] [CrossRef]
  21. Coultrap, S.J.; Vest, R.S.; Ashpole, N.M.; Hudmon, A.; Bayer, K.U. CaMKII in Cerebral Ischemia. Acta Pharmacol. Sin. 2011, 32, 861–872. [Google Scholar] [CrossRef] [PubMed]
  22. Kool, M.J.; Onori, M.P.; Borgesius, N.Z.; van de Bree, J.E.; Elgersma-Hooisma, M.; Nio, E.; Bezstarosti, K.; Buitendijk, G.H.S.; Aghadavoud Jolfaei, M.; Demmers, J.A.A.; et al. CAMK2-Dependent Signaling in Neurons Is Essential for Survival. J. Neurosci. 2019, 39, 5424–5439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Maier, L.S.; Bers, D.M. Role of Ca2+/Calmodulin-Dependent Protein Kinase (CaMK) in Excitation-Contraction Coupling in the Heart. Cardiovasc. Res. 2007, 73, 631–640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Vest, R.S.; O’Leary, H.; Coultrap, S.J.; Kindy, M.S.; Bayer, K.U. Effective Post-Insult Neuroprotection by a Novel Ca2+/ Calmodulin-Dependent Protein Kinase II (CaMKII) Inhibitor. J. Biol. Chem. 2010, 285, 20675–20682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Leurs, U.; Klein, A.B.; McSpadden, E.D.; Griem-Krey, N.; Solbak, S.M.Ø.; Houlton, J.; Villumsen, I.S.; Vogensen, S.B.; Hamborg, L.; Gauger, S.J.; et al. GHB Analogs Confer Neuroprotection through Specific Interaction with the CaMKIIα Hub Domain. Proc. Natl. Acad. Sci. USA 2021, 118, e2108079118. [Google Scholar] [CrossRef] [PubMed]
  26. Deng, G.; Orfila, J.E.; Dietz, R.M.; Moreno-Garcia, M.; Rodgers, K.M.; Coultrap, S.J.; Quillinan, N.; Traystman, R.J.; Bayer, K.U.; Herson, P.S. Autonomous CaMKII Activity as a Drug Target for Histological and Functional Neuroprotection after Resuscitation from Cardiac Arrest. Cell Rep. 2017, 18, 1109–1117. [Google Scholar] [CrossRef]
  27. Gaido, O.E.R.; Nkashama, L.J.; Schole, K.L.; Wang, Q.; Umapathi, P.; Mesubi, O.O.; Konstantinidis, K.; Luczak, E.D.; Anderson, M.E. CaMKII as a Therapeutic Target in Cardiovascular Disease. Annu. Rev. Pharmacol. Toxicol. 2023, 63, 1–62. [Google Scholar] [CrossRef]
  28. Miller, S.G.; Kennedy, M.B. Distinct Forebrain and Cerebellar Isozymes of Type II Ca2+/Calmodulin-Dependent Protein Kinase Associate Differently with the Postsynaptic Density Fraction. J. Biol. Chem. 1985, 260, 9039–9046. [Google Scholar] [CrossRef]
  29. Giese, K.P.; Fedorov, N.B.; Filipkowski, R.K.; Silva, A.J. Autophosphorylation at Thr286 of the α Calcium-Calmodulin Kinase II in LTP and Learning. Science 1998, 279, 870–873. [Google Scholar] [CrossRef]
  30. Hell, J.W. CaMKII: Claiming Center Stage in Postsynaptic Function and Organization. Neuron 2014, 81, 249–265. [Google Scholar] [CrossRef] [Green Version]
  31. Yasuda, R.; Hayashi, Y.; Hell, J.W. CaMKII: A Central Molecular Organizer of Synaptic Plasticity, Learning and Memory. Nat. Rev. Neurosci. 2022, 23, 666–682. [Google Scholar] [CrossRef] [PubMed]
  32. Bhattacharyya, M.; Karandur, D.; Kuriyan, J. Structural Insights into the Regulation of Ca2+/Calmodulin-Dependent Protein Kinase II (Camkii). Cold Spring Harb. Perspect. Biol. 2020, 12, a035147. [Google Scholar] [CrossRef] [PubMed]
  33. Rosenberg, O.S.; Deindl, S.; Comolli, L.R.; Downing, K.H.; Nairn, A.C.; Kuriyan, J. Oligomerization States of the Association Domain and the Holoenyzme of Ca2+/CaM Kinase II. FEBS J. 2006, 273, 682–694. [Google Scholar] [CrossRef] [PubMed]
  34. Chao, L.H.; Stratton, M.M.; Lee, I.H.; Rosenberg, O.S.; Levitz, J.; Mandell, D.J.; Kortemme, T.; Groves, J.T.; Schulman, H.; Kuriyan, J. A Mechanism for Tunable Autoinhibition in the Structure of a Human Ca2+/Calmodulin-Dependent Kinase II Holoenzyme. Cell 2011, 146, 732–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Colbran, R.J.; Smith, M.K.; Schworer, C.M.; Fong, Y.L.; Soderling, T.R. Regulatory Domain of Calcium/Calmodulin-Dependent Protein Kinase II. J. Biol. Chem. 1989, 264, 4800–4804. [Google Scholar] [CrossRef]
  36. Lisman, J.; Schulman, H.; Cline, H. The Molecular Basis of CaMKII Function in Synaptic and Behavioural Memory. Nat. Rev. Neurosci. 2002, 3, 175–190. [Google Scholar] [CrossRef]
  37. Yang, E.; Schulman, H. Structural Examination of Autoregulation of Multifunctional Calcium/Calmodulin-Dependent Protein Kinase II. J. Biol. Chem. 1999, 274, 26199–26208. [Google Scholar] [CrossRef] [Green Version]
  38. Miller, S.G.; Kennedy, M.B. Regulation of Brain Type II Ca2+/Calmodulin- Dependent Protein Kinase by Autophosphorylation: A Ca2+-Triggered Molecular Switch. Cell 1986, 44, 861–870. [Google Scholar] [CrossRef]
  39. Saitoh, T.; Schwartz, J.H. Phosphorylation-Dependent Subcellular Translocation of a Ca2+/Calmodulin-Dependent Protein Kinase Produces an Autonomous Enyzme in Aplysia Neurons. J. Cell Biol. 1985, 100, 835–842. [Google Scholar] [CrossRef]
  40. Coultrap, S.J.; Buard, I.; Kulbe, J.R.; Acqua, M.L.D.; Bayer, K.U. CaMKII Autonomy Is Substrate-Dependent and Further Stimulated by Ca2+/Calmodulin. J. Biol. Chem. 2010, 285, 17930–17937. [Google Scholar] [CrossRef] [Green Version]
  41. Coultrap, S.J.; Bayer, K.U. CaMKII Regulation in Information Processing and Storage. Trends Neurosci. 2012, 35, 607–618. [Google Scholar] [CrossRef] [PubMed]
  42. Meyer, T.; Hanson, P.I.; Stryer, L.; Schulman, H. Calmodulin Trapping by Calcium-Calmodulin-Dependent Protein Kinase. Science 1992, 256, 1199–1202. [Google Scholar] [CrossRef] [PubMed]
  43. Lou, L.L.; Schulman, H. Distinct Autophosphorylation Sites Sequentially Produce Autonomy and Inhibition of the Multifunctional Ca2+/Calmodulin-Dependent Protein Kinase. J. Neurosci. 1989, 9, 2020–2032. [Google Scholar] [CrossRef] [PubMed]
  44. Baucum, A.J. Proteomic Analysis of Postsynaptic Protein Complexes Underlying Neuronal Plasticity. ACS Chem. Neurosci. 2017, 8, 689–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Migues, P.V.; Lehmann, I.T.; Fluechter, L.; Cammarota, M.; Gurd, J.W.; Sim, A.T.R.; Dickson, P.W.; Rostas, J.A.P. Phosphorylation of CaMKII at Thr253 Occurs in Vivo and Enhances Binding to Isolated Postsynaptic Densities. J. Neurochem. 2006, 98, 289–299. [Google Scholar] [CrossRef]
  46. Bhattacharyya, M.; Lee, Y.K.; Muratcioglu, S.; Qiu, B.; Nyayapati, P.; Schulman, H.; Groves, J.T.; Kuriyan, J. Flexible Linkers in CaMKII Control the Balance between Activating and Inhibitory Autophosphorylation. Elife 2020, 9, e53670. [Google Scholar] [CrossRef] [Green Version]
  47. Strack, S.; Barban, M.A.; Wadzinski, B.E.; Coibran, R.J. Differential Inactivation of Postsynaptic Density-Associated and Soluble Ca2+/Calmodulin-Dependent Protein Kinase II by Protein Phosphatases 1 and 2A. J. Neurochem. 1997, 68, 2119–2128. [Google Scholar] [CrossRef] [Green Version]
  48. Tombes, R.M.; Faison, M.O.; Turbeville, J.M. Organization and Evolution of Multifunctional Ca2+/CaM- Dependent Protein Kinase Genes. Gene 2003, 322, 17–31. [Google Scholar] [CrossRef]
  49. Sloutsky, R.; Dziedzic, N.; Dunn, M.J.; Bates, R.M.; Torres-Ocampo, A.P.; Boopathy, S.; Page, B.; Weeks, J.G.; Chao, L.H.; Stratton, M.M. Heterogeneity in Human Hippocampal CaMKII Transcripts Reveals Allosteric Hub-Dependent Regulation. Sci. Signal. 2020, 13, eaaz0240. [Google Scholar] [CrossRef]
  50. Cook, S.G.; Bourke, A.M.; O’Leary, H.; Zaegel, V.; Lasda, E.; Mize-Berge, J.; Quillinan, N.; Tucker, C.L.; Coultrap, S.J.; Herson, P.S.; et al. Analysis of the CaMKIIα and β Splice-Variant Distribution among Brain Regions Reveals Isoform-Specific Differences in Holoenzyme Formation. Sci. Rep. 2018, 8, 5448. [Google Scholar] [CrossRef] [Green Version]
  51. O’Leary, H.; Lasda, E.; Bayer, K.U. CaMKIIb Association with the Actin Cytoskeleton Is Regulated by Alternative Splicing. Mol. Biol. Cell 2006, 17, 4656–4665. [Google Scholar] [CrossRef] [PubMed]
  52. Brocke, L.; Chiang, L.W.; Wagner, P.D.; Schulman, H. Functional Implications of the Subunit Composition of Neuronal CaM Kinase II. J. Biol. Chem. 1999, 274, 22713–22722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Lantsman, K.; Tombes, R.M. CaMK-II Oligomerization Potential Determined Using CFP/YFP FRET. Biochim. Biophys. Acta-Mol. Cell Res. 2005, 1746, 45–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Hoelz, A.; Nairn, A.C.; Kuriyan, J. Crystal Structure of a Tetradecameric Assembly of the Association Domain of Ca2+/Calmodulin-Dependent Kinase II. Mol. Cell 2003, 11, 1241–1251. [Google Scholar] [CrossRef] [PubMed]
  55. De Koninck, P.; Schulman, H. Sensitivity of CaM Kinase II to the Frequency of Ca2+ Oscillations. Science 1998, 279, 227–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Hudmon, A.; Schulman, H. Neuronal Ca2+/Calmodulin-Dependent Protein Kinase II: The Role of Structure and Autoregulation in Cellular Function. Annu. Rev. Biochem. 2002, 71, 473–510. [Google Scholar] [CrossRef] [PubMed]
  57. Chia, P.H.; Zhong, F.L.; Niwa, S.; Bonnard, C.; Utami, K.H.; Zeng, R.; Lee, H.; Eskin, A.; Nelson, S.F.; Xie, W.H.; et al. A Homozygous Loss-of-Function CAMK2A Mutation Causes Growth Delay, Frequent Seizures and Severe Intellectual Disability. Elife 2018, 7, e32451. [Google Scholar] [CrossRef]
  58. Bhattacharyya, M.; Stratton, M.M.; Going, C.C.; McSpadden, E.D.; Huang, Y.; Susa, A.C.; Elleman, A.; Cao, Y.M.; Pappireddi, N.; Burkhardt, P.; et al. Molecular Mechanism of Activation-Triggered Subunit Exchange in Ca2+/Calmodulin-Dependent Protein Kinase II. Elife 2016, 5, e13405. [Google Scholar] [CrossRef]
  59. Stratton, M.; Lee, I.H.; Bhattacharyya, M.; Christensen, S.M.; Chao, L.H.; Schulman, H.; Groves, J.T.; Kuriyan, J. Activation-Triggered Subunit Exchange between CaMKII Holoenzymes Facilitates the Spread of Kinase Activity. Elife 2014, 3, e01610. [Google Scholar] [CrossRef]
  60. Karandur, D.; Bhattacharyya, M.; Xia, Z.; Lee, Y.K.; Muratcioglu, S.; McAffee, D.; McSpadden, E.D.; Qiu, B.; Groves, J.T.; Williams, E.R.; et al. Breakage of the Oligomeric CaMKII Hub by the Regulatory Segment of the Kinase. Elife 2020, 9, e57784. [Google Scholar] [CrossRef]
  61. Lučić, I.; Héluin, L.; Jiang, P.-L.; Castro Scalise, A.G.; Wang, C.; Franz, A.; Wahl, M.; Liu, F.; Plested, A.J.R. CaMKII Activity Spreads by Inter-Holoenzyme Phosphorylation. bioRxiv 2022, 1–35. [Google Scholar] [CrossRef]
  62. Torres-Ocampo, A.P.; Özden, C.; Hommer, A.; Gardella, A.; Lapinskas, E.; Samkutty, A.; Esposito, E.; Garman, S.; Stratton, M. Characterization of CaMKIIα Holoenzyme Stability. Protein Sci. 2020, 29, 1524–1534. [Google Scholar] [CrossRef] [PubMed]
  63. Feng, B.; Raghavachari, S.; Lisman, J. Quantitative Estimates of the Cytoplasmic, PSD, and NMDAR-Bound Pools of CaMKII in Dendritic Spines. Brain Res. 2011, 1419, 46–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bayer, K.U.; De Koninck, P.; Leonard, A.S.; Hell, J.W.; Schulman, H. Interaction with the NMDA Receptor Locks CaMKII in an Active Conformation. Nature 2001, 411, 801–805. [Google Scholar] [CrossRef] [PubMed]
  65. Elgersma, Y.; Fedorov, N.B.; Ikonen, S.; Choi, E.S.; Elgersma, M.; Carvalho, O.M.; Giese, K.P.; Silva, A.J. Inhibitory Autophosphorylation of CaMKII Controls PSD Association, Plasticity, and Learning. Neuron 2002, 36, 493–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Strack, S.; Choi, S.; Lovinger, D.M.; Colbran, R.J. Translocation of Autophosphorylated Calcium/Calmodulin-Dependent Protein Kinase II to the Postsynaptic Density. J. Biol. Chem. 1997, 272, 13467–13470. [Google Scholar] [CrossRef] [Green Version]
  67. Shen, K.; Meyer, T. Dynamic Control of CaMKII Translocation and Localization in Hippocampal Neurons by NMDA Receptor Stimulation. Science 1999, 284, 162–166. [Google Scholar] [CrossRef]
  68. Bayer, K.U.; Schulman, H. Regulation of Signal Transduction by Protein Targeting: The Case for CaMKII. Biochem. Biophys. Res. Commun. 2001, 289, 917–923. [Google Scholar] [CrossRef]
  69. Özden, C.; Sloutsky, R.; Mitsugi, T.; Santos, N.; Agnello, E.; Gaubitz, C.; Foster, J.; Lapinskas, E.; Esposito, E.A.; Saneyoshi, T.; et al. CaMKII Binds Both Substrates and Activators at the Active Site. Cell Rep. 2022, 40, 111064. [Google Scholar] [CrossRef]
  70. Hayashi, Y.; Shi, S.; Esteban, J.A.; Piccini, A.; Poncer, J.-C.; Malinow, R. Driving AMPA Receptors into Synapses by LTP and CaMKII: Requirement for GluR1 and PDZ Domain Interaction. Science 2000, 287, 2262–2268. [Google Scholar] [CrossRef] [Green Version]
  71. Derkach, V.; Barria, A.; Soderling, T.R. Ca2+/Calmodulin-Kinase II Enhances Channel Conductance of α-Amino-3-Hydroxy-5-Methyl-4-Isoxazolepropionate Type Glutamate Receptors. Proc. Natl. Acad. Sci. USA 1999, 96, 3269–3274. [Google Scholar] [CrossRef] [PubMed]
  72. Borgesius, N.Z.; van Woerden, G.M.; Buitendijk, G.H.S.; Keijzer, N.; Jaarsma, D.; Hoogenraad, C.C.; Elgersma, Y. ΒCaMKII Plays a Nonenzymatic Role in Hippocampal Synaptic Plasticity and Learning by Targeting ACaMKII to Synapses. J. Neurosci. 2011, 31, 10141–10148. [Google Scholar] [CrossRef] [PubMed]
  73. Halt, A.R.; Dallapiazza, R.F.; Zhou, Y.; Stein, I.S.; Qian, H.; Juntti, S.; Wojcik, S.; Brose, N.; Silva, A.J.; Hell, J.W. CaMKII Binding to GluN2B Is Critical during Memory Consolidation. EMBO J. 2012, 31, 1203–1216. [Google Scholar] [CrossRef] [PubMed]
  74. Jourdain, P.; Fukunaga, K.; Muller, D. Calcium/Calmodulin-Dependent Protein Kinase II Contributes to Activity-Dependent Filopodia Growth and Spine Formation. J. Neurosci. 2003, 23, 10645–10649. [Google Scholar] [CrossRef] [Green Version]
  75. Pratt, K.G.; Watt, A.J.; Griffith, L.C.; Nelson, S.B.; Turrigiano, G.G. Activity-Dependent Remodeling of Presynaptic Inputs by Postsynaptic Expression of Activated CaMKII. Neuron 2003, 39, 269–281. [Google Scholar] [CrossRef] [Green Version]
  76. Coultrap, S.J.; Freund, R.K.; O’Leary, H.; Sanderson, J.L.; Roche, K.W.; Dell’Acqua, M.L.; Bayer, K.U. Autonomous CaMKII Mediates Both LTP and LTD Using a Mechanism for Differential Substrate Site Selection. Cell Rep. 2014, 6, 431–437. [Google Scholar] [CrossRef] [Green Version]
  77. Bayer, K.U.; Schulman, H. CaM Kinase: Still Inspiring at 40. Neuron 2019, 2, 380–394. [Google Scholar] [CrossRef]
  78. Cook, S.G.; Buonarati, O.R.; Coultrap, S.J.; Bayer, K.U. CaMKII Holoenzyme Mechanisms That Govern the LTP versus LTD Decision. Sci. Adv. 2021, 7, eabe2300. [Google Scholar] [CrossRef]
  79. Ginsberg, M.D. Current Status of Neuroprotection for Cerebral Ischemia Synoptic Overview. Stroke 2009, 40, S111–S114. [Google Scholar] [CrossRef] [Green Version]
  80. Kostandy, B.B. The Role of Glutamate in Neuronal Ischemic Injury: The Role of Spark in Fire. Neurol. Sci. 2012, 33, 223–237. [Google Scholar] [CrossRef]
  81. Astrup, J.; Symon, L.; Branston, N.M.; Lassen, N.A. Cortical Evoked Potential and Extracellular K+ and H+ at Critical Levels of Brain Ischemia. Stroke 1977, 8, 51–57. [Google Scholar] [CrossRef] [PubMed]
  82. Kunz, A.; Dirnagl, U.; Mergenthaler, P. Acute Pathophysiological Processes after Ischaemic and Traumatic Brain Injury. Best Pract. Res. Clin. Anaesthesiol. 2010, 24, 495–509. [Google Scholar] [CrossRef] [PubMed]
  83. Bevers, M.B.; Neumar, R.W. Mechanistic Role of Calpains in Postischemic Neurodegeneration. J. Cereb. Blood Flow Metab. 2008, 28, 655–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Bernhardt, J.; Hayward, K.S.; Kwakkel, G.; Ward, N.S.; Wolf, S.L.; Borschmann, K.; Krakauer, J.W.; Boyd, L.A.; Carmichael, S.T.; Corbett, D.; et al. Agreed Definitions and a Shared Vision for New Standards in Stroke Recovery Research: The Stroke Recovery and Rehabilitation Roundtable Taskforce. Neurorehabil. Neural Repair 2017, 31, 793–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Joy, M.T.; Carmichael, S.T. Encouraging an Excitable Brain State: Mechanisms of Brain Repair in Stroke. Nat. Rev. Neurosci. 2021, 22, 38–53. [Google Scholar] [CrossRef]
  86. Murphy, T.H.; Corbett, D. Plasticity during Stroke Recovery: From Synapse to Behaviour. Nat. Rev. Neurosci. 2009, 10, 861–872. [Google Scholar] [CrossRef] [PubMed]
  87. Clarkson, A.N.; López-Valdés, H.E.; Overman, J.J.; Charles, A.C.; Brennan, K.C.; Thomas Carmichael, S. Multimodal Examination of Structural and Functional Remapping in the Mouse Photothrombotic Stroke Model. J. Cereb. Blood Flow Metab. 2013, 33, 716–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Carmichael, S.T. Cellular and Molecular Mechanisms of Neural Repair after Stroke: Making Waves. Ann. Neurol. 2006, 59, 735–742. [Google Scholar] [CrossRef]
  89. Hagemann, G.; Redecker, C.; Neumann-Haefelin, T.; Freund, H.J.; Witte, O.W. Increased Long-Term Potentiation in the Surround of Experimentally Induced Focal Cortical Infarction. Ann. Neurol. 1998, 44, 255–258. [Google Scholar] [CrossRef]
  90. Brown, C.E.; Li, P.; Boyd, J.D.; Delaney, K.R.; Murphy, T.H. Extensive Turnover of Dendritic Spines and Vascular Remodeling in Cortical Tissues Recovering from Stroke. J. Neurosci. 2007, 27, 4101–4109. [Google Scholar] [CrossRef] [Green Version]
  91. Rostas, J.A.P.; Spratt, N.J.; Dickson, P.W.; Skelding, K.A. The Role of Ca2+-Calmodulin Stimulated Protein Kinase II in Ischaemic Stroke—A Potential Target for Neuroprotective Therapies. Neurochem. Int. 2017, 107, 33–42. [Google Scholar] [CrossRef] [PubMed]
  92. Ashpole, N.M.; Hudmon, A. Excitotoxic Neuroprotection and Vulnerability with CaMKII Inhibition. Mol. Cell. Neurosci. 2011, 46, 720–730. [Google Scholar] [CrossRef] [PubMed]
  93. Otmakhov, N.; Gorbacheva, E.V.; Regmi, S.; Yasuda, R.; Hudmon, A.; Lisman, J. Excitotoxic Insult Results in a Long-Lasting Activation of CaMKIIα and Mitochondrial Damage in Living Hippocampal Neurons. PLoS ONE 2015, 10, e0120881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Skelding, K.A.; Spratt, N.J.; Fluechter, L.; Dickson, P.W.; Rostas, J.A.P. ACaMKII Is Differentially Regulated in Brain Regions That Exhibit Differing Sensitivities to Ischemia and Excitotoxicity. J. Cereb. Blood Flow Metab. 2012, 32, 2181–2192. [Google Scholar] [CrossRef] [Green Version]
  95. Matsumoto, S.; Shamloo, M.; Isshiki, A.; Wieloch, T. Persistent Phosphorylation of Synaptic Proteins Following Middle Cerebral Artery Occlusion. J. Cereb. Blood Flow Metab. 2002, 22, 1107–1113. [Google Scholar] [CrossRef] [Green Version]
  96. Matsumoto, S.; Shamloo, M.; Matsumoto, E.; Isshiki, A.; Wieloch, T. Protein Kinase C-γ and Calcium/Calmodulin-Dependent Protein Kinase II-α Are Persistently Translocated to Cell Membranes of the Rat Brain during and after Middle Cerebral Artery Occlusion. J. Cereb. Blood Flow Metab. 2004, 24, 54–61. [Google Scholar] [CrossRef] [Green Version]
  97. Gurd, J.W.; Rawof, S.; Zhen Huo, J.; Dykstra, C.; Bissoon, N.; Teves, L.; Wallace, M.C.; Rostas, J.A.P. Ischemia and Status Epilepitcus Result in Enhanced Phosphorylation of Calcium and Calmodulin-Stimulated Protein Kinase II on Threonine 253. Brain Res. 2008, 1218, 158–165. [Google Scholar] [CrossRef]
  98. Shamloo, M.; Kamme, F.; Wieloch, T. Subcellular Distribution and Autophosphorylation of Calcium/Calmodulin- Dependent Protein Kinase II-α in Rat Hippocampus in a Model of Ischemic Tolerance. Neuroscience 2000, 96, 665–674. [Google Scholar] [CrossRef]
  99. Meng, F.; Guo, J.; Zhang, Q.; Song, B.; Zhang, G. Autophosphorylated Calcium/Calmodulin-Dependent Protein Kinase IIa (CaMKIIa) Reversibly Targets to and Phosphorylates N-Methyl-D-Aspartate Receptor Subunit 2B (NR2B) in Cerebral Ischemia and Reperfusion in Hippocampus of Rats. Brain Res. 2003, 967, 161–169. [Google Scholar] [CrossRef]
  100. Kirino, T.; Tamura, A.; Sano, K. Selective Vulnerability of the Hippocampus to Ischemia—Reversible and Irreversible Types of Ischemic Cell Damage. Prog. Brain Res. 1985, 63, 39–58. [Google Scholar] [CrossRef]
  101. Mangus, D.B.; Huang, L.; Applegate, P.M.; Gatling, J.W.; Zhang, J.; Applegate, R.L. A Systematic Review of Neuroprotective Strategies after Cardiac Arrest: From Bench to Bedside (Part I-Protection via Specific Pathways). Med. Gas Res. 2014, 4, 9. [Google Scholar] [CrossRef] [PubMed]
  102. Hanson, S.; Grotta, J.; Waxham, M.; Aronowski, J.; Ostrow, P. Calcium/Calmodulin-Dependent Protein Kinase II Activity in Focal Ischemia with Reperfusion in Rats. Stroke 1994, 25, 466–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Aronowski, J.; Grotta, J.C.; Waxham, M.N. Ischemia-Induced Translocation of Ca2+/Calmodulin-Dependent Protein Kinase II: Potential Role in Neuronal Damage. J. Neurochem. 1992, 58, 1743–1753. [Google Scholar] [CrossRef] [PubMed]
  104. Westgate, S.A.; Brown, J.; Aronowski, J.; Waxham, M.N. Activity of Ca2+/Calmodulin-Dependent Protein Kinase II Following Ischemia: A Comparison Between CA1 and Dentate Gyrus in a Hippocampal Slice Model. J. Neurochem. 1994, 63, 2217–2224. [Google Scholar] [CrossRef] [PubMed]
  105. Kolb, S.J.; Hudmon, A.; Waxham, M.N. Ca2+/Calmodulin Kinase II Translocates in a Hippocampal Slice Model of Ischemia. J. Neurochem. 1995, 64, 2147–2152. [Google Scholar] [CrossRef]
  106. Mengesdorf, T.; Althausen, S.; Mies, G.; Olah, L.; Paschen, W. Phosphorylation State, Solubility and Activity of Calcium/Calmodulin-Dependent Protein Kinase IIa in Transient Focal Ischemia in Mouse Brain. Neurochem. Res. 2002, 27, 477–484. [Google Scholar] [CrossRef]
  107. Hudmon, A.; Kim, S.A.; Kolb, S.J.; Stoops, J.K.; Neal Waxham, M. Light Scattering and Transmission Electron Microscopy Studies Reveal a Mechanism for Calcium/Calmodulin-Dependent Protein Kinase II Self-Association. J. Neurochem. 2001, 76, 1364–1375. [Google Scholar] [CrossRef]
  108. Dosemeci, A.; Reese, T.S.; Petersen, J.; Tao-Cheng, J.-H. A Novel Particulate Form of Ca2+/Calmodulin-Dependent Protein Kinase II in Neurons. J. Neurosci. 2000, 20, 3076–3084. [Google Scholar] [CrossRef] [Green Version]
  109. Buonarati, O.R.; Miller, A.P.; Coultrap, S.J.; Bayer, K.U.; Reichow, S.L. Conserved and Divergent Features of Neuronal CaMKII Holoenzyme Structure, Function, and High-Order Assembly. Cell Rep. 2021, 37, 110168. [Google Scholar] [CrossRef]
  110. Hudmon, A.; Aronowski, J.; Kolb, S.; Waxham, M. Inactivation and Self-Association of Ca2+/Calmodulin-Dependent Protein Kinase II during Autophosphorylation. J. Biol. Chem. 1996, 271, 8800–8808. [Google Scholar] [CrossRef] [Green Version]
  111. Buonarati, O.R.; Cook, S.G.; Goodell, D.J.; Chalmers, N.E.; Rumian, N.L.; Tullis, J.E.; Restrepo, S.; Coultrap, S.J.; Quillinan, N.; Herson, P.S.; et al. CaMKII versus DAPK1 Binding to GluN2B in Ischemic Neuronal Cell Death after Resuscitation from Cardiac Arrest. Cell Rep. 2020, 30, 1–8.e4. [Google Scholar] [CrossRef] [PubMed]
  112. Tullis, J.E.; Buonarati, O.R.; Coultrap, S.J.; Bourke, A.M.; Tiemeier, E.L.; Kennedy, M.J.; Herson, P.S.; Bayer, K.U. GluN2B S1303 Phosphorylation by CaMKII or DAPK1: No Indication for Involvement in Ischemia or LTP. iScience 2021, 24, 103214. [Google Scholar] [CrossRef] [PubMed]
  113. Ameen, S.S.; Griem-Krey, N.; Dufour, A.; Hossain, M.I.; Hoque, A.; Sturgeon, S.; Nandurkar, H.; Draxler, D.F.; Medcalf, R.L.; Kamaruddin, M.A.; et al. N-Terminomic Changes of Neurons During Excitotoxicity Reveals Proteolytic Events Associated with Synaptic Dysfunctions and Potential Targets for Neuroprotection. bioRxiv 2022. [Google Scholar] [CrossRef]
  114. Cruzalegui, F.H.; Kapilofft, M.S.; Morfint, J.; Kemp, B.E.; Rosenfeldt, M.G.; Means, A.R. Regulation of Intrasteric Inhibition of the Multifunctional Calcium/Calmodulin-Dependent Protein Kinase. Proc. Natl. Acad. Sci. USA 1992, 89, 12127–12131. [Google Scholar] [CrossRef] [Green Version]
  115. Yamagata, Y.; Czernik, A.J.; Greengard, P. Active Catalytic Fragment of Ca2+/Calmodulin-Dependent Protein Kinase II: Purification, Characterization, and Structural Analysis. J. Biol. Chem. 1991, 266, 15391–15397. [Google Scholar] [CrossRef] [PubMed]
  116. Kwiatkowski, A.P.; King, M.M. Autophosphorylation of the Type II Calmodulin-Dependent Protein Kinase Is Essential for Formation of a Proteolytic Fragment with Catalytic Activity. Implications for Long-Term Synaptic Potentiation. Biochemistry 1989, 28, 5380–5385. [Google Scholar] [CrossRef]
  117. Rich, D.P.; Schworer, C.M.; Colbran, R.J.; Soderling, T.R. Proteolytic Activation of Calcium/Calmodulin-Dependent Protein Kinase II: Putative Function in Synaptic Plasticity. Mol. Cell. Neurosci. 1990, 1, 107–116. [Google Scholar] [CrossRef] [PubMed]
  118. Yoshimura, Y.; Nomura, T.; Yamauchi, T. Purification and Characterization of Active Fragment of Ca2+/ Calmodulin-Dependent Protein Kinase II from the Post-Synaptic Density in the Rat Forebrain. J. Biochem. 1996, 119, 268–273. [Google Scholar] [CrossRef]
  119. Gascón, S.; Sobrado, M.; Roda, J.M.; Rodríguez-Pẽa, A.; Díaz-Guerra, M. Excitotoxicity and Focal Cerebral Ischemia Induce Truncation of the NR2A and NR2B Subunits of the NMDA Receptor and Cleavage of the Scaffolding Protein PSD-95. Mol. Psychiatry 2008, 13, 99–114. [Google Scholar] [CrossRef]
  120. Hossain, M.I.; Roulston, C.L.; Kamaruddin, M.A.; Chu, P.W.Y.; Ng, D.C.H.; Dusting, G.J.; Bjorge, J.D.; Williamson, N.A.; Fujita, D.J.; Cheung, S.N.; et al. A Truncated Fragment of Src Protein Kinase Generated by Calpain-Mediated Cleavage Is a Mediator of Neuronal Death in Excitotoxicity. J. Biol. Chem. 2013, 288, 9696–9709. [Google Scholar] [CrossRef] [Green Version]
  121. Xu, W.; Wong, T.P.; Chery, N.; Gaertner, T.; Wang, Y.T.; Baudry, M. Calpain-Mediated MGluR1α Truncation: A Key Step in Excitotoxicity. Neuron 2007, 53, 399–412. [Google Scholar] [CrossRef] [PubMed]
  122. Barria, A.; Muller, D.; Derkach, V.; Griffith, L.C.; Soderling, T.R. Regulatory Phosphorylation of AMPA-Type Glutamate Receptors by CaM-KII during Long-Term Potentiation. Science 1997, 276, 2042–2045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Fu, X.Z.; Zhang, Q.G.; Meng, F.J.; Zhang, G.Y. NMDA Receptor-Mediated Immediate Ser831 Phosphorylation of GluR1 through CaMKIIα in Rat Hippocampus during Early Global Ischemia. Neurosci. Res. 2004, 48, 85–91. [Google Scholar] [CrossRef] [PubMed]
  124. Guetg, N.; Aziz, S.A.; Holbro, N.; Turecek, R.; Rose, T.; Seddik, R.; Gassmann, M.; Moes, S.; Jenoe, P.; Oertner, T.G.; et al. NMDA Receptor-Dependent GABAB Receptor Internalization via CaMKII Phosphorylation of Serine 867 in GABAB1. Proc. Natl. Acad. Sci. USA 2010, 107, 13924–13929. [Google Scholar] [CrossRef] [Green Version]
  125. Hleihil, M.; Vaas, M.; Bhat, M.A.; Balakrishnan, K.; Benke, D. Sustained Baclofen-Induced Activation of GABAB Receptors After Cerebral Ischemia Restores Receptor Expression and Function and Limits Progressing Loss of Neurons. Front. Mol. Neurosci. 2021, 14, 188. [Google Scholar] [CrossRef]
  126. Hudmon, A.; Schulman, H.; Kim, J.; Maltez, J.M.; Tsien, R.W.; Pitt, G.S. CaMKII Tethers to L-Type Ca2+ Channels, Establishing a Local and Dedicated Integrator of Ca2+ Signals for Facilitation. J. Cell Biol. 2005, 171, 537–547. [Google Scholar] [CrossRef] [Green Version]
  127. Alev, C.; Urscheld, S.; Sonntag, S.; Zoidl, G.; Fort, A.G.; Höher, T.; Matsubara, M.; Willecke, K.; Spray, D.C.; Dermietzel, R. The Neuronal Connexin36 Interacts with and Is Phosphorylated by CaMKII in a Way Similar to CaMKII Interaction with Glutamate Receptors. Proc. Natl. Acad. Sci. USA 2008, 105, 20964–20969. [Google Scholar] [CrossRef] [Green Version]
  128. Gao, J.; Duan, B.; Wang, D.G.; Deng, X.H.; Zhang, G.Y.; Xu, L.; Xu, T. Le Coupling between NMDA Receptor and Acid-Sensing Ion Channel Contributes to Ischemic Neuronal Death. Neuron 2005, 48, 635–646. [Google Scholar] [CrossRef] [Green Version]
  129. Osuka, K.; Watanabe, Y.; Usuda, N.; Atsuzawa, K.; Takayasu, M. Phosphorylation of Neuronal Nitric Oxide Synthase at Ser1412 in the Dentate Gyrus of Rat Brain after Transient Forebrain Ischemia. Neurochem. Int. 2013, 63, 269–274. [Google Scholar] [CrossRef]
  130. Wheeler, D.G.; Barrett, C.F.; Groth, R.D.; Safa, P.; Tsien, R.W. CaMKII Locally Encodes L-Type Channel Activity to Signal to Nuclear CREB in Excitation-Transcription Coupling. J. Cell Biol. 2008, 183, 849–863. [Google Scholar] [CrossRef]
  131. Guo, X.; Zhou, J.; Starr, C.; Mohns, E.J.; Li, Y.; Chen, E.P.; Yoon, Y.; Kellner, C.P.; Tanaka, K.; Wang, H.; et al. Preservation of Vision after CaMKII-Mediated Protection of Retinal Ganglion Cells. Cell 2021, 184, 4299–4314.e12. [Google Scholar] [CrossRef]
  132. Mabuchi, T.; Kitagawa, K.; Kuwabara, K.; Takasawa, K.; Ohtsuki, T.; Xia, Z.; Storm, D.; Yanagihara, T.; Hori, M.; Matsumoto, M. Phosphorylation of CAMP Response Element-Binding Protein in Hippocampal Neurons as a Protective Response after Exposure to Glutamate in Vitro and Ischemia in Vivo. J. Neurosci. 2001, 21, 9204–9213. [Google Scholar] [CrossRef] [Green Version]
  133. Waxham, M.N.; Grotta, J.C.; Silva, A.J.; Strong, R.; Aronowski, J. Ischemia-Induced Neuronal Damage: A Role for Calcium/Calmodulin-Dependent Protein Kinase II. J. Cereb. Blood Flow Metab. 1996, 16, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Rumian, N.L.; Chalmers, N.E.; Tullis, J.E.; Herson, P.S.; Bayer, K.U. CaMKIIα Knockout Protects from Ischemic Neuronal Cell Death after Resuscitation from Cardiac Arrest. Brain Res. 2021, 1773, 147699. [Google Scholar] [CrossRef] [PubMed]
  135. Sumi, M.; Kiuchi, K.; Ishikawa, T.; Ishii, A.; Hagiwara, M.; Nagatsu, T.; Hidaka, H. The Newly Synthesized Selective Ca2+ calmodulin Dependent Protein Kinase II Inhibitor KN-93 Reduces Dopamine Contents in PC12h Cells. Biochem. Biophys. Res. Commun. 1991, 181, 968–975. [Google Scholar] [CrossRef] [PubMed]
  136. Vest, R.S.; Davies, K.D.; O’Leary, H.; Port, J.D.; Bayer, K.U. Dual Mechanism of a Natural CaMKII Inhibitor. Mol. Biol. Cell 2007, 18, 5024–5033. [Google Scholar] [CrossRef] [Green Version]
  137. Wong, M.H.; Samal, A.B.; Lee, M.; Vlach, J.; Novikov, N.; Niedziela-Majka, A.; Feng, J.Y.; Koltun, D.O.; Brendza, K.M.; Kwon, H.J.; et al. The KN-93 Molecule Inhibits Calcium/Calmodulin-Dependent Protein Kinase II (CaMKII) Activity by Binding to Ca2+/CaM. J. Mol. Biol. 2019, 431, 1440–1459. [Google Scholar] [CrossRef]
  138. Enslen, H.; Sun, P.; Brickey, D.; Soderling, S.H.; Klamo, E.; Soderling, T.R. Characterization of Ca2+/Calmodulin-Dependent Protein Kinase IV. Role in Transcriptional Regulation. J. Biol. Chem. 1994, 269, 15520–15527. [Google Scholar] [CrossRef]
  139. Ledoux, J.; Chartier, D.; Leblanc, N. Inhibitors of Calmodulin-Dependent Protein Kinase Are Nonspecific Blockers of Voltage-Dependent K+ Channels in Vascular Myocytes. J. Pharmacol. Exp. Ther. 1999, 290, 1165–1174. [Google Scholar]
  140. Li, G.; Hidaka, H.; Wollheim, C.B. Inhibition of Voltage-Gated Ca2+ Channels and Insulin Secretion in HIT Cells by the Ca2+/Calmodulin-Dependent Protein Kinase II Inhibitor KN-62: Comparison with Antagonists of Calmodulin and L-Type Ca2+ Channels. Mol. Pharmacol. 1992, 42, 488–489. [Google Scholar]
  141. Coultrap, S.J.; Bayer, K.U. Improving a Natural CaMKII Inhibitor by Random and Rational Design. PLoS ONE 2011, 6, e25245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Goold, C.P.; Nicoll, R.A. Single-Cell Optogenetic Excitation Drives Homeostatic Synaptic Depression. Neuron 2010, 68, 512–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Ahmed, M.E.; Dong, Y.; Lu, Y.; Tucker, D.; Wang, R.; Zhang, Q. Beneficial Effects of a CaMKIIα Inhibitor TatCN21 Peptide in Global Cerebral Ischemia. J. Mol. Neurosci. 2017, 61, 42–51. [Google Scholar] [CrossRef] [PubMed]
  144. Rostas, J.A.P.; Hoffman, A.; Murtha, L.A.; Pepperall, D.; Mcleod, D.D.; Dickson, P.W.; Spratt, N.J.; Skelding, K.A. Ischaemia- and Excitotoxicity-Induced CaMKII-Mediated Neuronal Cell Death: The Relative Roles of CaMKII Autophosphorylation at T286 and T253. Neurochem. Int. 2017, 104, 6–10. [Google Scholar] [CrossRef]
  145. Griem-krey, N.; Klein, A.B.; Clausen, B.H.; Namini, M.R.J.; Pernille, V. The GHB Analogue HOCPCA Improves Sensorimotor Function after MCAO via CaMKII A. bioRxiv 2022, 1–26. [Google Scholar] [CrossRef]
  146. Ottani, A.; Saltini, S.; Bartiromo, M.; Zaffe, D.; Renzo Botticelli, A.; Ferrari, A.; Bertolini, A.; Genedani, S. Effect of γ-Hydroxybutyrate in Two Rat Models of Focal Cerebral Damage. Brain Res. 2003, 986, 181–190. [Google Scholar] [CrossRef]
  147. Gao, B.; Kilic, E.; Baumann, C.R.; Hermann, D.M.; Bassetti, C.L. γ-Hydroxybutyrate Accelerates Functional Recovery after Focal Cerebral Ischemia. Cerebrovasc. Dis. 2008, 26, 413–419. [Google Scholar] [CrossRef] [Green Version]
  148. Bay, T.; Eghorn, L.F.; Klein, A.B.; Wellendorph, P. GHB Receptor Targets in the CNS: Focus on High-Affinity Binding Sites. Biochem. Pharmacol. 2014, 87, 220–228. [Google Scholar] [CrossRef]
  149. Maitre, M.; Hechler, V.; Vayer, P.; Gobaille, S.; Cash, C.D.; Schmitt, M.; Bourguignon, J.J. A Specific γ-Hydroxybutyrate Receptor Ligand Possesses Both Antagonistic and Anticonvulsant Properties. J. Pharmacol. Exp. Ther. 1990, 255, 657–663. [Google Scholar]
  150. Wellendorph, P.; Høg, S.; Greenwood, J.R.; de Lichtenberg, A.; Nielsen, B.; Frølund, B.; Brehm, L.; Clausen, R.P.; Bräuner-Osborne, H. Novel Cyclic γ-Hydroxybutyrate (GHB) Analogs with High Affinity and Stereoselectivity of Binding to GHB Sites in Rat Brain. J. Pharmacol. Exp. Ther. 2005, 315, 346–351. [Google Scholar] [CrossRef] [Green Version]
  151. Krall, J.; Jensen, C.H.; Bavo, F.; Falk-Petersen, C.B.; Haugaard, A.S.; Vogensen, S.B.; Tian, Y.; Nittegaard-Nielsen, M.; Sigurdardóttir, S.B.; Kehler, J.; et al. Molecular Hybridization of Potent and Selective γ-Hydroxybutyric Acid (GHB) Ligands: Design, Synthesis, Binding Studies, and Molecular Modeling of Novel 3-Hydroxycyclopent-1-Enecarboxylic Acid (HOCPCA) and Trans-γ-Hydroxycrotonic Acid (T-HCA) Analogs. J. Med. Chem. 2017, 60, 9022–9039. [Google Scholar] [CrossRef] [PubMed]
  152. Tian, Y.; Shehata, M.A.; Gauger, S.J.; Hamborg, L.; Thiesen, L.; Bruus-Jensen, J.; Leurs, U.; Larsen, A.S.G.; Veronesi, C.; Krall, J.; et al. Exploring the NCS-382 Scaffold for CaMKIIα Modulation: Synthesis, Pharmacology and Biophysical Characterization of Ph-HTBA as a Novel High-Affinity Brain-Penetrant Stabilizer for the CaMKIIα Hub Domain. J. Med. Chem. 2022, 65, 15066–15084. [Google Scholar] [CrossRef] [PubMed]
  153. Tian, Y.; Shehata, M.A.; Gauger, S.J.; Ng, C.K.L.; Solbak, S.; Thiesen, L.; Bruus-Jensen, J.; Krall, J.; Gibson, K.M.; Wellendorph, P.; et al. Discovery and Optimization of 5-Hydroxy-Diclofenac toward a New Class of Ligands with Nanomolar Affinity for the CaMKIIα Hub Domain. J. Med. Chem. 2022, 65, 6656–6676. [Google Scholar] [CrossRef]
  154. Thiesen, L.; Kehler, J.; Clausen, R.P.; Frølund, B.; Bundgaard, C.; Wellendorph, P. In Vitro and in Vivo Evidence for Active Brain Uptake of the GHB Analog HOCPCA by the Monocarboxylate Transporter Subtype 1. J. Pharmacol. Exp. Ther. 2015, 354, 166–174. [Google Scholar] [CrossRef]
  155. Vogensen, S.B.; Marek, A.; Bay, T.; Wellendorph, P.; Kehler, J.; Frølund, B.; Pedersen, M.H.F.; Clausen, R.P. New Synthesis and Tritium Labeling of a Selective Ligand for Studying High-Affinity γ-Hydroxybutyrate (GHB) Binding Sites. J. Med. Chem. 2013, 56, 8201–8205. [Google Scholar] [CrossRef] [Green Version]
  156. Buard, I.; Coultrap, S.J.; Freund, R.K.; Lee, Y.S.; Dell’Acqua, M.L.; Silva, A.J.; Bayer, K.U. CaMKII “Autonomy” Is Required for Initiating but Not for Maintaining Neuronal Long-Term Information Storage. J. Neurosci. 2010, 30, 8214–8220. [Google Scholar] [CrossRef] [Green Version]
  157. Nassal, D.; Gratz, D.; Hund, T.J. Challenges and Opportunities for Therapeutic Targeting of Calmodulin Kinase II in Heart. Front. Pharmacol. 2020, 11. [Google Scholar] [CrossRef]
  158. Balakrishnan, K.; Hleihil, M.; Bhat, M.A.; Ganley, R.P.; Vaas, M.; Klohs, J.; Zeilhofer, H.U.; Benke, D. Targeting the Interaction of GABAB Receptors with CaMKII with an Interfering Peptide Restores Receptor Expression after Cerebral Ischemia and Inhibits Progressive Neuronal Death in Mouse Brain Cells and Slices. Brain Pathol. 2022, e13099. [Google Scholar] [CrossRef]
  159. Zhang, X.; Connelly, J.; Levitan, E.S.; Sun, D.; Wang, J.Q. Calcium/Calmodulin–Dependent Protein Kinase II in Cerebrovascular Diseases. Transl. Stroke Res. 2021, 12, 513–529. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structure and activation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) [32,41]. (A) (Left) Domains of individual CaMKII subunit: kinase (grey), regulatory (teal), linker (yellow) and hub (purple) domain. The regulatory domain contains important autophosphorylation sites Thr286 (red) and Thr305/306 (blue). Thr305/306 are located within the calmodulin binding element (light blue). (Right) Holoenzyme assembly made up of 12 subunits. (B) Regulation of CaMKII activity. Of note, the hub domain is not shown. During basal states, the regulatory domain binds to the kinase domain and blocks access to the substrate binding site (autoinhibition). CaMKII gets activated by Ca2+/CaM binding to the regulatory segment, which releases autoinhibition of the kinase domain and subsequently exposes Thr286. Trans autophosphorylation of Thr286 by a neighboring subunit infers autonomous enzyme activity. CaMKII can get autophosphorylated in an intra-subunit reaction at Thr305/306 after Ca2+/CaM dissociation [46]. Thr305/306 autophosphorylation inhibits re-binding of Ca2+/CaM. Architecture is shown for CaMKIIα but overall domain structure, assembly and activation mechanism is similar for other CaMKII subtypes.
Figure 1. Structure and activation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) [32,41]. (A) (Left) Domains of individual CaMKII subunit: kinase (grey), regulatory (teal), linker (yellow) and hub (purple) domain. The regulatory domain contains important autophosphorylation sites Thr286 (red) and Thr305/306 (blue). Thr305/306 are located within the calmodulin binding element (light blue). (Right) Holoenzyme assembly made up of 12 subunits. (B) Regulation of CaMKII activity. Of note, the hub domain is not shown. During basal states, the regulatory domain binds to the kinase domain and blocks access to the substrate binding site (autoinhibition). CaMKII gets activated by Ca2+/CaM binding to the regulatory segment, which releases autoinhibition of the kinase domain and subsequently exposes Thr286. Trans autophosphorylation of Thr286 by a neighboring subunit infers autonomous enzyme activity. CaMKII can get autophosphorylated in an intra-subunit reaction at Thr305/306 after Ca2+/CaM dissociation [46]. Thr305/306 autophosphorylation inhibits re-binding of Ca2+/CaM. Architecture is shown for CaMKIIα but overall domain structure, assembly and activation mechanism is similar for other CaMKII subtypes.
Brainsci 12 01639 g001
Figure 2. Simplified overview of excitotoxicity after ischemic stroke [4]. Obstruction of blood flow leads to energy failure and excessive depolarization of neurons. Glutamate accumulates in the synaptic cleft and overstimulates glutamate receptors, such as N-methyl-D-aspartate (NMDA)– and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors. Overstimulation of glutamate receptors results in increased cell permeability and dramatic increases in Ca2+ and Na+ influx, creating an osmotic gradient resulting in cell swelling and oedema. Intracellular Ca2+ overload dysregulates various Ca2+-dependent pathways and overstimulates enzymes, including CaMKII. Overactivity of phospholipases and proteases diminishes cellular integrity, as well as overstimulation of enzymes responsible for producing excessive amounts of reactive oxygen species (ROS), which in turn damage any cellular component. Ca2+ overload and ROS also mediate mitochondrial damage, ultimately promoting apoptotic cell death. Excitotoxicity, oxidative stress and apoptosis lead to ischemic cell death and infarct formation in conjunction with inflammation.
Figure 2. Simplified overview of excitotoxicity after ischemic stroke [4]. Obstruction of blood flow leads to energy failure and excessive depolarization of neurons. Glutamate accumulates in the synaptic cleft and overstimulates glutamate receptors, such as N-methyl-D-aspartate (NMDA)– and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors. Overstimulation of glutamate receptors results in increased cell permeability and dramatic increases in Ca2+ and Na+ influx, creating an osmotic gradient resulting in cell swelling and oedema. Intracellular Ca2+ overload dysregulates various Ca2+-dependent pathways and overstimulates enzymes, including CaMKII. Overactivity of phospholipases and proteases diminishes cellular integrity, as well as overstimulation of enzymes responsible for producing excessive amounts of reactive oxygen species (ROS), which in turn damage any cellular component. Ca2+ overload and ROS also mediate mitochondrial damage, ultimately promoting apoptotic cell death. Excitotoxicity, oxidative stress and apoptosis lead to ischemic cell death and infarct formation in conjunction with inflammation.
Brainsci 12 01639 g002
Figure 3. CaMKII translocation under physiological and ischemic conditions [21]. (A) Physiological synaptic activity at excitatory glutamatergic neurons leads to Ca2+ influx through NMDA receptors. Subsequent Ca2+/CaM binding leads to CaMKII autophosphorylation at Thr286 and induces its translocation from the center of the dendritic spine to the post-synaptic density (PSD). Here, CaMKII co-localizes with NMDA receptors, positioning the holoenzyme in proximity to multiple substrates, such as AMPA receptors. (B) Ischemia leads to excitotoxicity concomitant with the excessive release of glutamate, overstimulation of glutamate receptors and intracellular Ca2+ overload. Increased Ca2+ influx rapidly activates CaMKII and leads to excessive autophosphorylation and translocation to the PSD.
Figure 3. CaMKII translocation under physiological and ischemic conditions [21]. (A) Physiological synaptic activity at excitatory glutamatergic neurons leads to Ca2+ influx through NMDA receptors. Subsequent Ca2+/CaM binding leads to CaMKII autophosphorylation at Thr286 and induces its translocation from the center of the dendritic spine to the post-synaptic density (PSD). Here, CaMKII co-localizes with NMDA receptors, positioning the holoenzyme in proximity to multiple substrates, such as AMPA receptors. (B) Ischemia leads to excitotoxicity concomitant with the excessive release of glutamate, overstimulation of glutamate receptors and intracellular Ca2+ overload. Increased Ca2+ influx rapidly activates CaMKII and leads to excessive autophosphorylation and translocation to the PSD.
Brainsci 12 01639 g003
Figure 4. CaMKII cleavage by calpain after ischemia [113]. Ischemic conditions lead to overactivation of calpains (red scissors) as well as increased Thr286/287 CaMKII autophosphorylation, facilitating cleavage of CaMKII by overactivated calpains at M281↓H282. This leads to the ischemia-specific expression of ΔCaMKII consisting of the kinase domain and the N-terminal end of the regulatory domain, which is devoid of the Thr286/287 and CaM-binding region responsible for autoinhibition of the kinase.
Figure 4. CaMKII cleavage by calpain after ischemia [113]. Ischemic conditions lead to overactivation of calpains (red scissors) as well as increased Thr286/287 CaMKII autophosphorylation, facilitating cleavage of CaMKII by overactivated calpains at M281↓H282. This leads to the ischemia-specific expression of ΔCaMKII consisting of the kinase domain and the N-terminal end of the regulatory domain, which is devoid of the Thr286/287 and CaM-binding region responsible for autoinhibition of the kinase.
Brainsci 12 01639 g004
Figure 5. CaMKIIα as high-affinity binding site for γ-hydroxybutyrate (GHB) [25,148]. GHB targets both high- and low-affinity binding sites within the CNS. One well-established low-affinity binding site is the GABAB receptor, and the high-affinity binding site was recently identified as CaMKIIα. GHB analogues display improved affinity and selectivity for CaMKIIα compared to GHB. E.g., the GHB analogue 3-hydroxycyclopent-1-enecarboxylic acid (HOCPCA) does not target GABAB receptors and its selectivity was studied by showing the complete absence of [3H]HOCPCA binding to tissue from Camk2a−/− mice. In contrast, binding to Camk2a+/+, Camk2b−/− and Camk2b+/+ mouse brain tissue was preserved.
Figure 5. CaMKIIα as high-affinity binding site for γ-hydroxybutyrate (GHB) [25,148]. GHB targets both high- and low-affinity binding sites within the CNS. One well-established low-affinity binding site is the GABAB receptor, and the high-affinity binding site was recently identified as CaMKIIα. GHB analogues display improved affinity and selectivity for CaMKIIα compared to GHB. E.g., the GHB analogue 3-hydroxycyclopent-1-enecarboxylic acid (HOCPCA) does not target GABAB receptors and its selectivity was studied by showing the complete absence of [3H]HOCPCA binding to tissue from Camk2a−/− mice. In contrast, binding to Camk2a+/+, Camk2b−/− and Camk2b+/+ mouse brain tissue was preserved.
Brainsci 12 01639 g005
Table 1. Summary of in vivo neuroprotective effects of the Tat peptide CN21 and CN19o as well as the GHB analogue 3-hydroxycyclopent-1-enecarboxylic acid (HOCPCA). Of note, CN-peptides are administered intravenously and HOCPCA intraperitoneally [24,25,26,145].
Table 1. Summary of in vivo neuroprotective effects of the Tat peptide CN21 and CN19o as well as the GHB analogue 3-hydroxycyclopent-1-enecarboxylic acid (HOCPCA). Of note, CN-peptides are administered intravenously and HOCPCA intraperitoneally [24,25,26,145].
CN-PeptidesHOCPCA
In vivo
Ischemic stroke
1 mg/kg tat-CN21 at 1 h post tMCAO 1 improves infarct volume in young male mice [24]175 mg/kg HOCPCA at 30 min, 3 h, 6, h and 12 h and 90 mg/kg HOCPCA at 3 h post photothrombotic stroke improves infarct size and motor function (cylinder and grid walking task) in young male mice [25]
175 mg/kg HOCPCA at 3 h post photothrombotic stroke improves infarct size and motor function (cylinder and grid walking task) in aged female mice [25]
175 mg/kg HOCPCA at 30 min post pMCAO 1 improves infarct size and motor function (grip strength) in young male mice [145]
175 mg/kg HOCPCA at 3 h post pMCAO improves motor function (grip strength) in young male mice [145]
175 mg/kg HOCPCA at 30 min post thromboembolic stroke improves motor function (grip strength) in young male mice [145]
In vivo
Global ischemia
Intracerebroventricular injection of 50 mg tat-CN21 at 3 h after global ischemia improves neuronal survival in the hippocampus and improved memory function (Barnes maze) [146]
0.01 mg/kg, 0.1 mg/kg and 1 mg/kg of tat-CN19o at 30 min post cardiac arrest and cardiopulmonary resuscitation (CA/CPR)
improves neuronal survival in the hippocampus and improved memory function (contextual fear conditioning) [26]
1 tMCAO: transient middle cerebral artery occlusion; pMCAO: permanent middle cerebral artery occlusion.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Griem-Krey, N.; Clarkson, A.N.; Wellendorph, P. CaMKIIα as a Promising Drug Target for Ischemic Grey Matter. Brain Sci. 2022, 12, 1639. https://doi.org/10.3390/brainsci12121639

AMA Style

Griem-Krey N, Clarkson AN, Wellendorph P. CaMKIIα as a Promising Drug Target for Ischemic Grey Matter. Brain Sciences. 2022; 12(12):1639. https://doi.org/10.3390/brainsci12121639

Chicago/Turabian Style

Griem-Krey, Nane, Andrew N. Clarkson, and Petrine Wellendorph. 2022. "CaMKIIα as a Promising Drug Target for Ischemic Grey Matter" Brain Sciences 12, no. 12: 1639. https://doi.org/10.3390/brainsci12121639

APA Style

Griem-Krey, N., Clarkson, A. N., & Wellendorph, P. (2022). CaMKIIα as a Promising Drug Target for Ischemic Grey Matter. Brain Sciences, 12(12), 1639. https://doi.org/10.3390/brainsci12121639

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop