Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4646 2022-10-03 09:21:47 |
2 format corrected. -3 word(s) 4643 2022-10-13 10:19:00 | |
3 format corrected. Meta information modification 4085 2022-10-13 10:19:53 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Maudsley, S. Multidimensional Roles of GRK5 in Molecular Aging. Encyclopedia. Available online: https://encyclopedia.pub/entry/28231 (accessed on 03 May 2024).
Maudsley S. Multidimensional Roles of GRK5 in Molecular Aging. Encyclopedia. Available at: https://encyclopedia.pub/entry/28231. Accessed May 03, 2024.
Maudsley, Stuart. "Multidimensional Roles of GRK5 in Molecular Aging" Encyclopedia, https://encyclopedia.pub/entry/28231 (accessed May 03, 2024).
Maudsley, S. (2022, October 03). Multidimensional Roles of GRK5 in Molecular Aging. In Encyclopedia. https://encyclopedia.pub/entry/28231
Maudsley, Stuart. "Multidimensional Roles of GRK5 in Molecular Aging." Encyclopedia. Web. 03 October, 2022.
Multidimensional Roles of GRK5 in Molecular Aging
Edit

Considerable evidence now suggests that GRK5 can act as a molecular 'bridging' factor, allowing signaling regulation in pathophysiological settings that can control the connectivity between both the cardiovascular and neurophysiological complications of aging. 

G-protein coupled receptor kinase 5 aging cardiovascular disease neurodegeneration GRK5 interactors

1. Role(s) of GRK5 in Molecular Aging

GRK5 (G protein-coupled receptor kinase 5) activity has been linked with multiple age-associated neoplastic, metabolic, neurodegenerative and cardiovascular ailments [1][2]. At the specific disease level, the expressional regulation and activity of GRK5 has been linked with multiple age-related diseases such as type 2 diabetes mellitus (T2DM) [3], cardiac hypertrophy [4], hypertension [5], Parkinson’s disease [6][7], and Alzheimer’s pathology in mice and humans [8]. As we age, a progressive dysfunction of multiple receptor signaling systems, across a broad range of tissues, takes place. In this context of age-related receptor system dysfunction, the loss of signaling system sensitivity has been most intensively studies for the insulinotropic signaling cascade. Disruption of the ability to effectively sense, uptake and eventually metabolize glucose has been identified as a pivotal regulator of the rate of aging in nearly every animal model tested [9]. Many of the first genes identified in lower species that control animal longevity were almost exclusively associated with the insulinotropic/insulin-like growth factor system [10]. The glycometabolic system, as well as the somatic sensitivity to insulin receptor functionality, is also strongly controlled through the functional status of adipose tissue in the body, e.g., adiponectin release from white adipose tissue is a potent insulin sensitizing factor [11]. Commensurate with a potentially important role of GRK5 in aging, it has been shown to be strongly expressed in adipose tissues, suggesting that its functionality may impact the glycoregulatory system. [12] demonstrated that GRK5 genomic deletion in murine models resulted in the generation of significant insulin resistance. In addition to this, genetic polymorphisms of GRK5 have been strongly associated with the generation of T2DM [13] and the efficacy profile of anti-diabetic therapeutic agents [14]. Furthermore, previous studies performed in GRK5 knock-out mice (GRK5-KO) reinforced the importance of GRK5 in metabolism as these animals displayed a decreased white adipose tissue mass, a lower weight gain, a decreased expression of adipogenic genes and a reduced adipocyte differentiation when fed a high-fat diet [12][15]. Although human data linking GRK5 to metabolism are sparse, a recent genome-wide association study found a robust association of two single nucleotide polymorphisms (SNPs) in the GRK5 gene with apoB levels and total LDL-cholesterol, highlighting the role of GRK5 in cholesterol metabolism.

As well as long-term dysfunction of metabolic signaling systems in the aging process, significant disruptions of inflammatory mediator receptor systems are evident. This inflammatory signaling perturbation typically results in the creation of chronic low-grade inflammatory syndrome, recently codified as “inflammaging” [16][17][18]. Inflammaging, as a process, has been proposed to be functionally independent of exogenous systemic infection [19][20]. This chronic inflammatory condition has been linked to potentiated circulatory C-reactive peptide and IL-6 (interleukin 6) concentrations. Protracted exposure to these pro-inflammatory agents predisposes patients to an increased incidence of obesity, premature immunological aging, vascular sclerosis and neurodegenerative phenotypes [21][22]. The inflammaging process itself appears to be closely tied to the mechanisms of whole-somatic aging trajectory control. Hence, inflammaging has been strongly linked to the potentiation of nuclear factor-κB (NF-κB) activity – a process that at the hypothalamic level seems to act as an arbiter of the aging process [1][23].

At a fundamental level NF-κB has been shown possess the ability to regulate the expression of GRK5 [24] – therefore these two systems in fact potentiate each other’s activity in a feed-forward loop, a mode of signaling highly characteristic of the aging process itself. Demonstrating the intersection of GRK5 with the inflammatory aging process, GRK5 has been shown to antagonize TLR4 (Toll-like receptor 4) mediated phosphorylation of the NF-κB p105 protein. This action inhibits inflammatory mediator (lipopolysaccharide) sensitivity in macrophages [25]. Subsequent to the discovery of GRK5 regulation of p105, [26] reported that GRK5 binding to IκBα stabilizes this protein and facilitates the nuclear accumulation of IκBα by masking and thus inhibiting its nuclear export signal sequence. This nuclear accumulation of IκBα can then lead to decreased NF-κB activation in vascular endothelial cells. Research from [27] employing a GRK5 knockout (KO) murine model confirmed that endothelial GRK5 likely stabilizes IκBα in a manner reminiscent to previous studies [26][28]. Using this model, [24] further demonstrated an NF-κB inhibitory action of GRK5 in cardiac muscle cells.

Taken together, GRK5 is clearly a vital component in both energy metabolism and chronic inflammation paradigms. It is interesting to note that both of these systems are known to strongly control molecular aging pathologies implicated in many different human disorders and consequently in inflammatory pathways [29]. These findings therefore make GRK5 a potentially important therapeutic target in the treatment of age-related diseases such as cardiovascular disease, neurological and metabolic disorders. In this review, we discuss the role of GRK5 in the context of cardiovascular and neurodegenerative disease to emphasize its function in inflammaging.

2. The Role of GRK5 in Cardiovascular Disease Pathology

Cardiovascular pathophysiologies, such as myocardial ischemia, myocardial infarction or hypertension involve the dysregulation of cardiac GPCR responsiveness, which in turn is partly induced by deleterious GRK signaling activity profiles [30][31]. The first cardiac GRK form identified was GRK2 [32], while the discovery of cardiac GRK5 came later [33]. GRK5 was found to be highly expressed in the myocardium through several studies employing genetically engineered mice with altered GRK5 levels [2][33][34]. Homozygous GRK5-KO mice are born with a normal basal phenotype, although a loss of both GRK5 and GRK6 in mice results in lethality [1] [35]. Further studies in zebrafish lacking the GRK5 homolog Grk5l, suggested the importance of GRK5 fine tuning capacity in cardiac development through the mTOR pathway. Hence, these Grk5l deficient fish demonstrated altered cardiac tissue generation associated with premature loss of muscle cell progenitors leading to an imbalance of gross structure [1][36]. Of note, GRK5 is shown to be up-regulated in heart failure [37]. It has been demonstrated that elevation of GRK5 expression in vascular smooth muscle cells (VSMCs) can also induce the development of high blood pressure [7] via altered β1-adrenergic receptor (β1-AR) and angiotensin II (Ang II) receptor signaling dynamics [38][39]. GRK5 functionality also appears to be linked to the generation of atherosclerotic vascular pathophysiologies. Hence, the genomic deletion of GRK5 in an ApoE4-deficient murine background significantly accelerated the creation of aortic atherosclerosis compared to control mice [28].

2.1. Cardiac failure

GRK5 appears to exert a pivotal role in cardiac failure and several cardiomyopathies including cardiac hypertrophy [6][40]. Cardiac hypertrophy refers to the abnormal enlargement, or thickening, of cardiac muscle. This thickening can be caused by increases in cardiomyocyte size themselves or via changes in other cardiac muscular components, such as extracellular matrix. Cardiac hypertrophy can be induced via physiological effects (e.g., elevated cardiovascular exercise) or as the result of pathophysiology (e.g., hypertension or valvular disease) [41].

In humans, four non-synonymous SNPs of GRK5 with translational significance have been demonstrated. Of these known SNPs, the RH-domain resident Q41L polymorphism [leucine (L) converted to a glutamine (Q)] is highly enriched amongst African-American (A-A) individuals [42]. This divergent form of GRK5 possesses an augmented capacity to desensitize β2-adrenergic receptors (β2ARs) [43], thus engendering a population specific cardiovascular effect. The Q41L GRK5 variant appears to afford protection against congestive cardiac failure amongst A-A heart failure patients [1][44]. Reinforcing the potential protective capacity of GRK5 in the cardiac setting, increased GRK5 expression has been shown to attenuate cardiac burden in response to intense adrenergic stimulation [38][45]. As expected, a GRK5 activity blockade mediates the opposite effect, i.e., increased cardiac performance as well as improved resilience in the context of heart failure [46][47]. It has also been shown that functional GRK5 inhibition, performed by ectopic expression of an N-terminal GRK5 peptide fragment of GRK5, reduces the extent of cardiac muscle damage and attenuates the risk of heart failure [48].

During cardiac failure, the expression and activity of GRK5 are reflexively increased to enhance β-adrenergic receptor desensitization and thus attenuate contractility [38]. Activation of GPCRs by hypertrophic agonists, such as phenylephrine and/or Ang II, engages a number of intracellular signaling pathways, including calcineurin-nuclear factor of activated T cells (NFAT) [49], Ca2+/CaM – dependent kinase II (CamK II) [50][51], MAPKs [52][53] and the Akt-mechanistic target of rapamycin (mTOR) pathway [54][55] among many others, that are important transducers of the hypertrophic response.

GRK5 can undergo nuclear translocation in a calmodulin-dependent manner following Gαq-based signals emanating from α-adrenergic and Ang II receptors. This nuclear translocation of GRK5 has been shown to be mutually exclusive with its interaction with plasma membrane GPCRs – thus distinguishing canonical and non-canonical GRK5 functions [7]. This cellular redistribution is proposed to help mitigate the deleterious functions of cardiac hypertrophy [56][57][58][59][60]. Nuclear GRK5 migration is assisted through a productive interaction with calcium sensing proteins (CSP) [61] – thus GRK5 is specifically sensitive to the presence of Ca2+/CaM [62][63]. Indeed, GRK5, possessing a high affinity for CaM, is rapidly inactivated in cells upon elevations in cytosolic calcium. This aspect of GRK5 biology reinforces its pivotal role in the modulation of calcium-associated muscular contractility [64][65].

It has been demonstrated that nuclear GRK5 acts as a class II histone deacetylase kinase (HDAC). In this scenario it has been reported that GRK5 is able to phosphorylate HDAC5 (histone deactylase 5) [66]. This GRK5-mediated phosphorylation causes redistribution of HDCA5 out of the nucleus resulting in a function alleviation of its MEF2 (myocyte enhancer factor 2) transcription factor repression – leading to “de-repression” of MEF2. Demonstrating the important role of GRK5 in cardiovascular aging this GRK5-mediated MEF2 activation transcribes multiple genes associated with cardiac hypertrophy [59][66]. GRK5 activity has further been shown to control hypertrophic responses via its interaction in the nucleus with components of the NFAT pathway [67]. GRK5 interacts with the NFAT-pathway in the nucleus during pathological hypertrophy. In addition, it is clear that GRK5 is strongly connected with the NF-κB signaling cascade [26][27][28][29][26][68][69][70] as an NF-κB binding element has been identified within the GRK5 DNA promoter region. This functional signaling region has subsequently been demonstrated to orchestrate the expression pattern of GRK5 in cardiac muscle cells [70].

Physiological hypertrophy does not only occur naturally in the heart due to augmented exercise regimens but also during pregnancy [71] Non-pathological cardiac hypertrophy is a process typified by relatively normal and proportionate myocyte growth – this reflexive response increases the capacity for cytoprotective cardiac activity [72][73]. In contrast, pathological hypertrophy involves a disruption of the proportions of the new myocytes that causes an eventual diminution of heart chamber volume with a concomitant augmentation of septal wall thickness [73]. Recent research has suggested that GRK5 is only a controller of non-pathological hypertrophy [74]. In this research, using TgGRK5 mice, it was shown that physical exercise induced a classical physiological cardiac hypertrophy response. With specific respect to the nuclear functionality of GRK5 in cardiac hypertrophy it was shown in this exercise context that minimal nuclear GRK5 activity was found [74]. This corresponds with a study demonstrating that NFAT was not shown to regulate physiological hypertrophy [75]. While elevated levels of intracellular Ca2+ levels are common to both physiological and pathological cardiac hypertrophy, it has been proposed that pathological hypertrophic effects are differentially controlled through distinct intracellular calcium stores. Thus differential sources of “activating” calcium may allow the specific stimulation of the GRK5-related hallmarks of pathological hypertrophy, i.e., nuclear GRK5 accumulation, HDAC kinase activity and increased NFAT activity. Reinforcing the concept of differential hypertrophic mechanisms, none of these selective events are routinely found in standard exercise-induced hypertrophic paradigms [74].

While GRK5 is evidently associated with deleterious cardiac signaling and cell growth, GRK5 does appear to possess additional non-pathological roles in heart functionality. For example, GRK5 has been shown to be an important intermediate in the mitogenic and pro-survival signaling cascades emanating from the β1-adrenergic receptor-mediated transactivation [76] of the epidermal growth factor receptor (EGFR) [77]. Therefore, it appears that GRK5 possesses a dual functionality with respect to cardiac activity, i.e., GRK5 is involved in both protective and detrimental signaling events that are delineated via differential subcellular compartmentalization between nuclear and non-nuclear sites.

2.2. Hypertension

The maintenance of well-controlled vascular blood pressure is imperative for effective and reliable delivery of oxygenated blood to all major life-preserving organs. Significantly and chronically elevated blood pressure, i.e., hypertension, is a prominent risk-determining player in the etiological profile of multiple chronic conditions including ischemic heart disease with associated subsequent cardiac and renal failure [78][79]. The major organs and processes that endogenously regulate vascular pressure include the kidney, heart and the contractile state of VSMCs which regulates radial changes of blood vessels, thus modulating peripheral vascular resistance.

High blood pressure, with its associated stressful effects on vascular wall integrity, can result in the potentiation of GRK5 expression within VSMCs. Associated with these observations it has been shown that Ang II stimulation of VSMCs can also increase GRK5 expression levels in a calcium dependent manner [80]. This association between VSMC-based GRK5 expression and hypertension was again studied by [40] in which an ectopic increase of GRK5 expression in vessels was genetically engineered. GRK5 overexpression was subsequently found to induce a gender-specific hypertensive response, i.e., blood pressure increases were much more profound in males compared to females [40]. Both male and female hypertension in these GRK5-overexpressing mice was ablated upon treatment with the inhibitor of Gαi signaling, pertussis toxin. Further gender-specific effects on the cardiovascular parameters of these GRK5-overexpressing mice were also apparent, e.g., β1-adrenergi receptor signaling in males was altered while Ang II-mediated increased vascular tone was only found in females [40]. Interestingly, and in contrast to the reported overexpression of GRK2 in VSMCs, the elevation of GRK5 expression failed to induce any significant cardiac hypertrophy [39].

2.3. Atherosclerosis

Atherosclerosis presents as a long-term inflammatory disease found primarily in the major arteries. This condition is typified by the accumulation of oxidized low-density lipoproteins (LDL) within the arterial wall and a progressive inflammatory cell infiltration into the vessel [81][82]. The recruitment of inflammatory cells to these lesions is triggered by the production of chemokines within the plaque microenvironment [82]. Chemokine-stimulated GPCRs initiate several downstream effectors, promoting actin polarization, shape changes and directed cell movement which ultimately leads to atherosclerotic plaque formation [83].

GRK5 possesses the capacity to regulate signaling through multiple heptahelical receptors [4][84] including multiple types that have been strongly linked to etiological activities in the atherosclerotic process [85][86][87][88][89][90]. Interestingly GRK5 has also been shown to phosphorylate other signal transduction proteins that can influence the atherosclerotic process too, including p53 [91], IκBα [68][92], platelet derived growth factor receptor-β (PDGFRβ) [93] [94] and HDCA5, via MEF2 activation [59]. GRK5 can also stimulate anti-atherogenic signaling activity in model systems. For example, GRK5-KO mice have an increase in lesion area when compared to wildtype mice through two different cell-type regulatory mechanisms in monocyte/macrophages and VSMCs [29]. In VSMCs, GRK5 is able to promote the degradation of the pro-atherogenic platelet-derived growth factor receptor-β in lysosomes which is thought to reduce platelet-derived growth factor-mediated VSMC proliferation and migration [29]. GRK5 also regulates monocyte chemotaxis; i.e., in vitro GRK5-KO monocytes possess increased migration capacity in response to C-C chemokine ligand 2 (CCL2) (a ligand for the C-C chemokine receptor type 2 (CCR2) receptor) and colony stimulating factor-1 (CSF1) (a ligand for the colony stimulating factor 1 receptor (CSF1R) tyrosine kinase) [29]. CCL2-mediated leukocyte migration is instrumental in atherosclerotic lesion progression and responsible for the increased macrophage content in lesions from GRK5-KO mice. These findings highlight the potential mechanisms in both monocyte retention and emigration after their migration across the endothelium and present new strategies to limit atherosclerotic lesion progression.

3. GRK5 in Neurodegeneration

In contrast to its expression profile in cardiovascular organs, central nervous system (CNS) expression of GRK5 is comparatively sparse [36][35] because of a low GRK5 expression in the majority of cortical areas, except for the limbic system [95]. As we have outlined previously there is emerging evidence that demonstrates the multiple non-canonical roles of GRK5 outside of GPCR activity regulation. These novel effects of GRK5 are also associated with multiple important neurophysiological functions. For example, GRK5 deficient mice display a specific and nuanced subtype-specific muscarinic receptor dysfunction while closely-associated adrenergic and opioid receptor activity was not altered [96][97][98]. CNS muscarinic receptor activity has long been associated with the maintenance of learning and memory behavior [99]. Thus, it is unsurprising that GRK5-KO mice present with cognitive dysfunction shown to correlate with hippocampal neurosynaptic failure [98]. Again, as with the cardiovascular effects, gender differences in GRK5 activity were seen with respect to neurodegenerative phenotypes, i.e., augmented axonal defects and synaptic degenerative changes, were shown to be greater in female experimental animals as opposed to males. In addition, at the molecular signaling level, hippocampal levels of the synaptosomal-associated protein 25 (SNAP25) and synaptophysin were significantly lower in females compared to males [98].

It has also been proposed that the involvement of GRK5 in dementia-related conditions is likely associated with its potent role in regulating neurite outgrowth that is required c [100].

Obstructive sleep apnea (OSA) occurs in approximately 2 to 4% of middle-aged women and men, respectively. Among these, OSA is also observed to be more common in obese patients, potentially due to increased tracheal occlusion caused by excessive cervical adipose deposits. While OSA can induce health concerns with respect to lack of effective sleep patterns, it is evident that OSA is also closely associated with intermittent cerebral hypoxia. Considering this deleterious hemodynamic effect it is unsurprising that OSA has been shown to be a potent risk factor for associated cognitive impairment in nearly a quarter of diagnosed OSA patients [101]. At the molecular level CNS hypoxic episodes can often result in the increased production rate of ROS – these oxygen species can rapidly interact and modify a broad range of CNS lipids, nucleic acids and proteins. Enhanced CNS ROS production has therefore been associated vascular endothelial dysfunction, perturbations of blood-brain barrier integrity and eventual neurosynaptic signal transduction dysfunction. Rodent models of intermittent hypoxia have been developed to effectively replicate the OSA found in human patients [1][102]. Using these, it has been demonstrated that intermittent hypoxia effects upon behavioral rodent activity (anxiety, balance, short-term memory) are acutely sensitive to, and potently augmented by, the genetic deletion of GRK5 [1][102]. Such research suggests that part of the CNS functionality of GRK5 may be associated with oxygen sensation neurochemistry, potentially via controlling astrocytic functions.

3.1. GRK5 and Alzheimer’s disease (AD) pathology

For a significant period of time, undue focus on amyloid pathologies and their subsequent association with Alzheimer’s disease (AD) has been in effect [103][104][105][106]. However, and from a more therapeutically important aspect, there has long been known to be an extant cholinergic receptor (post-synaptic nicotinic and M1 muscarinic acetylcholine) hypofunction evident in AD [107]). In AD it has been demonstrated that augmented presynaptic cholinergic activity results in the reflexive attenuation of synaptic acetylcholine release. This reduced release therefore results in diminished level of activity at the post-synaptic muscarinic M1 GPCRs. Indicating the importance of muscarinic signaling in AD pathophysiology, muscarinic M1 receptor signaling cascades can inhibit β-amyloidogenic (Aβ) amyloid precursor protein (APP) processing, resulting in a decreased level of cytotoxic β-amyloid accumulation [108]. From genetic deletion mouse models (i.e., GRK5-KO) it has been shown that GRK5 functionality is associated with severe hippocampal dysfunction (loss of neurosynaptic proteins and axonal swelling) as well as increased amyloidosis [13][109].

When combined with murine AD models (Tg2576) GRK5 deficiency was found to cause increased inflammatory astrogliosis in both hippocampal and cortical brain areas [110]. In addition to this effect, the GRK5 deficiency was also linked with both increased soluble Aβ levels as well as increased insoluble Aβ plaque load [111]. These findings were proposed to be due to a GRK5-induced potentiation of presynaptic muscarinic M2 receptor activity that resulted in a significant reduction of synaptic acetylcholine transmission levels [100][111]. This GRK5-associated alteration of synaptic receptor activity in murine models of AD has been shown to be linked to disruptions in sub-cellular compartmentalization of GRK5. Hence, [112] were able to demonstrate that aged AD model mice possess a highly specific plasma membrane deficiency of GRK5 [112]. A paucity of pre-synaptic GRK5, with its concomitant detrimental effect on M2-acetylcholine receptor-controlled acetylcholine release, has been subsequently linked to an exacerbation of tau hyperphosphorylation and further neuronal dysfunction. Using chemical blockade of these hyperactivated M2 receptors [112] were able to attenuate this tau hyperphosphorylation in a GSK3β-dependent manner.

It is thus apparent that GRK5 may indeed hold the key to the connection between the current major theories of AD, i.e., the amyloid and the cholinergic hypotheses. The cholinergic hypothesis suggests that cholinergic CNS dysfunction is responsible for the cognitive decline [113] while the amyloid hypothesis proposes that Aβ is the AD-causative factor [114][115][116][117][118]. Interestingly, as we have previously outlined, Aβ is thought to be one of the driving forces for alterations of membrane associated GRK5 in AD [119]. GRK5 plasma membrane deficiencies can mediate presynaptic M2 acetylcholine autoreceptor hyperactivation that, in turn, causes post-synaptic cholinergic hypoactivity through the functional attenuation of cholinergic neurotransmission. This disrupted cholinergic transmission then serves to augment Aβ amyloid production leading to a “feed-forward” process of progressive neurosynaptic dysfunction and amyloid toxicity. In this recursive process both amyloid deposition and cholinergic dysfunction each can serve as a cause and/or consequence of each other, with the extant GRK5 dysfunction as the pivotal mediator. Given the present interest in these hypotheses in AD pharmacotherapy, the importance of GRK5 as a drug target in this system may increase significantly in the future.

As a prelude to our next section, it is intriguing to note that GRK5 can be further connected with AD through its ability to phosphorylate α-synuclein (SNCA) [12][120][121], tubulin as well as the AD-associated tau protein [112]. This pathological effect has been proposed to occur through GRK5-mediated phosphorylation causing increased SNCA polymerization and eventual aggregation – in a similar manner to that seen with Aβ in the context of AD [122].

3.2. Parkinson’s disease (PD)

Parkinson’s disease (PD) is one of the most commonly encountered neurodegenerative diseases at the present time, just behind AD with respect to world prevalence. The pathological effects of PD impact the primary fine motor systems of the body. PD is clinically typified by progressive deterioration of tremor, rigidity, bradykinesia/akinesia, gait disturbance, and postural instability. The major defining neuropathological feature of PD has long been considered to be the loss of neurons in the substantia nigra that provide dopaminergic innervation to the striatum, the CNS region most heavily implicated in fine motor control. Since the molecular mechanism causing dopaminergic neuron dysfunction are yet to be comprehensively defined, there are unfortunately no effective current pharmacotherapeutic interventions capable of retarding, or reversing, the disease [123]. One of the lesser known aspects of PD is the fact that advancing age is arguably the strongest risk factor for its generation [124]. In this light it is unsurprising that PD is typically is presented after the age of 60.

With respect to the functional intersection between GRK5 and PD pathology, it has been demonstrated by multiple research groups that GRK5 represents one of the major kinases that can phosphorylate SNCA. This classical function of GRK5 results in the promotion of the oligomerization of PD (with actual co-localization of GRK5 and SNCA), facilitating the creation of pathological Lewy bodies in the substantia nigra and locus coeruleus of PD patients [12][120]. The nuclear functionality of GRK5 is one of its defining functional features among GRK proteins – GRK5 activity itself has also been shown to promote the nuclear translocation of SNCA and its associated factors PLK2 and 3 (Polo-like kinase 2 and 3) [125][126]. While the full ramifications of nuclear SNCA remain currently cryptic, it has been proposed that this aspect of SNCA biology may be independent of the classically-pathological SNCA aggregation modality. It is important to note, especially with respect to aging pathomechanisms, that oxidative stress environments promote the enhanced nuclear localization of SNCA [127][128][129]. Within the nuclear domain SNCA has been shown to functionally antagonize histone acetylation, resulting in increased neurotoxicity [130][131]). Nuclear SNCA has also been found to be a transcriptional regulator capable of binding to PGC1-α (Peroxisome proliferator activated receptor gamma coactivator 1-alpha) promoter regions, and in doing so, potentially regulate mitochondrial gene transcription and thus neurometabolic ROS-associated activity [129]. In addition to these cell signaling-based analyses, genetic association studies have proposed a haplotypic association of GRK5 gene with the clinical presentation of sporadic PD. These pathological haplotypes associated with functional GRK5 SNPs that can control multiple transcription factors (Yin Yang-1 (YY1) and cAMP response element-binding protein (CREB-1)) that together are capable of potentiating SNCA transcription [121]. Unfortunately, and as is quite common with genetic association studies, subsequent studies have failed to reproduce some of these propositions. Hence, studies employing GRK5 deletion in cells have failed to find a resultant attenuation of SNCA phosphorylation[132][133]. In addition, further studies have not observed a strong localization of GRK5 in Lewy bodies [134] or a firm association of GRK5 SNPs with PD [135].

References

  1. Selkoe, D. J., and Hardy, J. (2016). The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 8, 595–608. doi: 10.15252/emmm.201606210
  2. Singh, P., Peng, W., Zhang, Q., Ding, X., and Suo, W. Z. (2016). GRK5 deficiency leads to susceptibility to intermittent hypoxia-induced cognitive impairment. Behav. Brain Res. 302, 29–34. doi: 10.1016/j.bbr.2016.01.019
  3. Liggett, S. B., Cresci, S., Kelly, R. J., Syed, F. M., Matkovich, S. J., Hahn, H. S., et al. (2008). A GRK5 polymorphism that inhibits β-adrenergic receptor signaling is protective in heart failure. Nat. Med. 14, 510–517. doi: 10.1038/nm1750
  4. Burkhalter, M. D., Fralish, G. B., Premont, R. T., Caron, M. G., and Philipp, M. (2013). Grk5l controls heart development by limiting mTOR signaling during symmetry breaking. Cell Rep. 4, 625–632. doi: 10.1016/j.celrep.2013.07.036
  5. Zhang, G., Li, J., Purkayastha, S., Tang, Y., Zhang, H., Yin, Y. I., et al. (2013). Hypothalamic programming of systemic aging involving IKKβ/NF-κB and GnRH. Nature 497, 211–216. doi: 10.1038/nature12143
  6. Premont, R. T., and Gainetdinov, R. R. (2007). Physiological roles of G protein–coupled receptor kinases and arrestins. Annu. Rev. Physiol. 69, 511–534. doi: 10.1146/annurev.physiol.69.022405.154731
  7. Gurevich, E. V., Tesmer, J. J., Mushegian, A., and Gurevich, V. V. (2012). G protein-coupled receptor kinases: more than just kinases and not only for GPCRs. Pharmacol. Ther. 133, 40–69. doi: 10.1016/j.pharmthera.2011.08.001
  8. Li, H., Gan, W., Lu, L., Dong, X., Han, X., Hu, C., et al. (2013). A genome-wide association study identifies GRK5 and RASGRP1 as type 2 diabetes loci in Chinese hans. Diabetes Metab. Res. Rev. 62, 291–298. doi: 10.2337/db12-0454
  9. Gold, J. I., Gao, E., Shang, X., Premont, R. T., and Koch, W. J. (2012). Determining the absolute requirement of G protein–coupled receptor kinase 5 for pathological cardiac hypertrophy novelty and significance. Circ. Res. 111, 1048–1053. doi: 10.1161/CIRCRESAHA.112.273367
  10. Harris, D. M., Cohn, H. I., Pesant, S., and Eckhart, A. D. (2008). GPCR signalling in hypertension: role of GRKs. Clin. Sci. 115, 79–89. doi: 10.1042/CS20070442
  11. Arawaka, S., Wada, M., Goto, S., Karube, H., Sakamoto, M., Ren, C.-H., et al. (2006). The role of g-protein-coupled receptor kinase 5 in pathogenesis of sporadic Parkinson’s disease. J. Neurosci. 26, 9227–9238. doi: 10.1523/JNEUROSCI.0341-06.2006.
  12. Bychkov, E., Gurevich, V., Joyce, J., Benovic, J., and Gurevich, E. (2008). Arrestins and two receptor kinases are upregulated in Parkinson’s disease with dementia. Neurobiol. Aging 29, 379–396. doi: 10.1016/j.neurobiolaging.2006.10.012
  13. Suo, Z., Cox, A. A., Bartelli, N., Rasul, I., Festoff, B. W., Premont, R. T., et al. (2007). GRK5 deficiency leads to early alzheimer-like pathology and working memory impairment. Neurobiol. Aging 28, 1873–1888. doi: 10.1016/j.neurobiolaging.2006.08.013
  14. Ma, S., and Gladyshev, V. N. (2017). Molecular signatures of longevity: insights from cross-species comparative studies. Semin. Cell Dev. Biol. 70, 190–203. doi: 10.1016/j.semcdb.2017.08.007
  15. Mathew, R., Bhadra, M. P., and Bhadra, U. (2016). Insulin/insulin-like growth factor-1 signalling (IIS) based regulation of lifespan across species. Biogerontology 18, 35–53. doi: 10.1007/s10522-016-9670-8
  16. Diez, J. J. R., and Iglesias, P. (2003). The role of the novel adipocyte-derived hormone adiponectin in human disease. Eur. J. Endocrinol. 148, 293–300. doi: 10.1530/eje.0.1480293
  17. Wang, F., Wang, L., Shen, M., and Ma, L. (2012). GRK5 deficiency decreases diet-induced obesity and adipogenesis. Biochem. Biophys. Res. Commun. 421, 312–317. doi: 10.1016/j.bbrc.2012.04.006
  18. Xia, Z., Yang, T., Wang, Z., Dong, J., and Liang, C. (2014). GRK5 intronic (CA)n polymorphisms associated with type 2 diabetes in Chinese Hainan island. PLoS One 9:e90597. doi: 10.1371/journal.pone.0090597
  19. Shang, Z., Han, F., Zhou, X., Bao, Z., Zhu, J., Wang, T., et al. (2018). A variant of GRK5 is associated with the therapeutic efficacy of repaglinide in Chinese Han patients with type 2 diabetes mellitus. Drug Dev. Res. 79, 129–135. doi: 10.1002/ddr.21426
  20. Wang, L., Shen, M., Wang, F., and Ma, L. (2012). GRK5 ablation contributes to insulin resistance. Biochem. Biophys. Res. Commun. 429, 99–104. doi: 10.1016/j.bbrc.2012.10.077
  21. Baylis, D., Bartlett, D. B., Patel, H. P., and Roberts, H. C. (2013). Understanding how we age: insights into inflammaging. Longev. Healthspan 2:8. doi: 10.1186/s12979-016-0070-3
  22. Franceschi, C., Garagnani, P., Parini, P., Giuliani, C., and Santoro, A. (2018). Inflammaging: a new immune–metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 14, 576–590. doi: 10.1038/s41574-018-0059-4
  23. Olivieri, F., Prattichizzo, F., Grillari, J., and Balistreri, C. R. (2018). Cellular senescence and inflammaging in age-related diseases. Mediators Inflamm. 2018:9076485. doi: 10.1155/2018/9076485
  24. Franceschi, C., Bonafe, M., Valensin, S., Olivieri, F., De Luca, M., Ottaviani, E., et al. (2000). Inflamm-aging: an evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 908, 244–254. doi: 10.1111/j.1749-6632.2000.tb06651.x
  25. Frasca, D., and Blomberg, B. B. (2016). Inflammaging decreases adaptive and innate immune responses in mice and humans. Biogerontology 17, 7–19. doi: 10.1007/s10522-015-9578-8
  26. Tabas, I. (2010). Macrophage death and defective inflammation resolution in atherosclerosis. Nat. Rev. Immunol. 10, 36–46. doi: 10.1038/nri2675
  27. Barzilai, N., Huffman, D. M., Muzumdar, R. H., and Bartke, A. (2012). The critical role of metabolic pathways in aging. Diabetes Metab. Res. Rev. 61, 1315–1322. doi: 10.2337/db11-1300
  28. Islam, K. N., Bae, J.-W., Gao, E., and Koch, W. J. (2013). Regulation of nuclear factor κb (NF-κB) in the nucleus of cardiomyocytes by G protein-coupled receptor kinase 5 (GRK5). J. Biol. Chem. 288, 35683–35689. doi: 10.1074/jbc.M113.529347
  29. Parameswaran, N., Pao, C. S., Leonhard, K. S., Kang, D. S., Kratz, M., Ley, S. C., et al. (2006). Arrestin-2 and G protein-coupled receptor kinase 5 interact with NFκB1 p105 and negatively regulate lipopolysaccharide-stimulated ERK1/2 activation in macrophages. J. Biol. Chem. 281, 34159–34170. doi: 10.1074/jbc.M605376200
  30. Sorriento, D., Ciccarelli, M., Santulli, G., Campanile, A., Altobelli, G. G., Cimini, V., et al. (2008). The G-protein-coupled receptor kinase 5 inhibits NFκB transcriptional activity by inducing nuclear accumulation of IκBα. Proc. Natl. Acad. Sci. U.S.A. 105, 17818–17823. doi: 10.1073/pnas.0804446105
  31. Wu, J.-H., Zhang, L., Fanaroff, A. C., Cai, X., Sharma, K. C., Brian, L., et al. (2012). G protein–coupled receptor kinase-5 attenuates atherosclerosis by regulating receptor tyrosine kinases and 7-transmembrane receptors. Arterioscler. Thromb. Vasc. Biol. 32, 308–316. doi: 10.1161/ATVBAHA.111.239608
  32. Rockman, H. A., Choi, D.-J., Rahman, N. U., Akhter, S. A., Lefkowitz, R. J., and Koch, W. J. (1996). Receptor-specific in vivo desensitization by the G protein-coupled receptor kinase-5 in transgenic mice. Proc. Natl. Acad. Sci. U.S.A. 93, 9954–9959. doi: 10.1073/pnas.93.18.9954
  33. Packiriswamy, N., Lee, T., Raghavendra, P. B., Durairaj, H., Wang, H., and Parameswaran, N. (2013). G-protein-coupled receptor kinase-5 mediates inflammation but does not regulate cellular infiltration or bacterial load in a polymicrobial sepsis model in mice. J. Innate Immun. 5, 401–413. doi: 10.1159/000347002
  34. Dorn, G. W. (2009). GRK mythology: G-protein receptor kinases in cardiovascular disease. J. Mol. Med. 87, 455–463. doi: 10.1007/s00109-009-0450-7
  35. Cannavo, A., Liccardo, D., and Koch, W. J. (2013). Targeting cardiac β-adrenergic signaling via GRK2 inhibition for heart failure therapy. Front. Physiol. 4:264. doi: 10.3389/fphys.2013.00264
  36. Kwatra, M. M., Benovic, J. L., Caron, M. G., Lefkowitz, R. J., and Hosey, M. M. (1989). Phosphorylation of chick heart muscarinic cholinergic receptors by the β-adrenergic receptor kinase. Biochemistry 28, 4543–4547. doi: 10.1021/bi00437a005
  37. Premont, R., Koch, W., Inglese, J., and Lefkowitz, R. (1994). Identification, purification, and characterization of GRK5, a member of the family of G protein-coupled receptor kinases. J. Biol. Chem. 269, 6832–6841.
  38. Kunapuli, P., and Benovic, J. L. (1993). Cloning and expression of GRK5: a member of the G protein-coupled receptor kinase family. Proc. Natl. Acad. Sci. U.S.A. 90, 5588–5592. doi: 10.1073/pnas.90.12.5588
  39. Philipp, M., Berger, I. M., Just, S., and Caron, M. G. (2014). Overlapping and opposing functions of G protein-coupled receptor kinase 2 (GRK2) and GRK5 during heart development. J. Biol. Chem. 289, 26119–26130. doi: 10.1074/jbc.M114.551952
  40. Chen, E. P., Bittner, H. B., Akhter, S. A., Koch, W. J., and Davis, R. D. (2001). Myocardial function in hearts with transgenic overexpression of the G protein-coupled receptor kinase 5. Ann. Thorac. Surg. 71, 1320–1324. doi: 10.1016/S0003-4975(00)01754-9
  41. Eckhart, A. D., Ozaki, T., Tevaearai, H., Rockman, H. A., and Koch, W. J. (2002). Vascular-targeted overexpression of G protein-coupled receptor kinase-2 in transgenic mice attenuates β-adrenergic receptor signaling and increases resting blood pressure. Mol. Pharmacol. 61, 749–758. doi: 10.1124/mol.61.4.749
  42. Keys, J. R., Zhou, R.-H., Harris, D. M., Druckman, C. A., and Eckhart, A. D. (2005). Vascular smooth muscle overexpression of G protein–coupled receptor kinase 5 elevates blood pressure, which segregates with sex and is dependent on GI-mediated signaling. Circulation 112, 1145–1153. doi: 10.1161/CIRCULATIONAHA.104.531657
  43. Dzimiri, N., Muiya, P., Andres, E., and Al-Halees, Z. (2004). Differential functional expression of human myocardial G protein receptor kinases in left ventricular cardiac diseases. Eur. J. Pharmacol. 489, 167–177. doi: 10.1016/j.ejphar.2004.03.015
  44. Tardiff, J. C. (2006). Cardiac hypertrophy: stressing out the heart. J. Clin. Invest. 116, 1467–1470. doi: 10.1172/JCI28884
  45. Wang, W. C., Mihlbachler, K. A., Bleecker, E. R., Weiss, S. T., and Liggett, S. B. (2008). A polymorphism of GRK5 alters agonist-promoted desensitization of β2-adrenergic receptors. Pharmacogenet. Genomics 18, 729–732. doi: 10.1097/FPC.0b013e32830967e9
  46. Eijgelsheim, M., Visser, L. E., Uitterlinden, A. G., and Stricker, B. H. C. (2008). Protective effect of a GRK5 polymorphism on heart failure and its interaction with β-adrenergic receptor antagonists. Pharmacogenomics 9, 1551–1555. doi: 10.2217/14622416.9.10.1551
  47. Brinks, H., Boucher, M., Gao, E., Chuprun, J. K., Pesant, S., Raake, P. W., et al. (2010). Level of G protein–coupled receptor kinase-2 determines myocardial ischemia/reperfusion injury via pro-and anti-apoptotic mechanisms novelty and significance. Circ. Res. 107, 1140–1149. doi: 10.1161/CIRCRESAHA.110.221010
  48. Raake, P. W., Vinge, L. E., Gao, E., Boucher, M., Rengo, G., Chen, X., et al. (2008). G protein–coupled receptor kinase 2 ablation in cardiac myocytes before or after myocardial infarction prevents heart failure. Circ. Res. 103, 413–422. doi: 10.1161/CIRCRESAHA.107.168336
  49. Vinge, L. E., Von Lueder, T. G., Aasum, E., Qvigstad, E., Gravning, J. A., How, O.-J., et al. (2008). Cardiac-restricted expression of the carboxyl-terminal fragment of GRK3 uncovers distinct functions of GRK3 in regulation of cardiac contractility and growth GRK3 controls cardiac α1-adrenergic receptor responsiveness. J. Biol. Chem. 283, 10601–10610. doi: 10.1074/jbc.M708912200
  50. Sorriento, D., Santulli, G., Fusco, A., Anastasio, A., Trimarco, B., and Iaccarino, G. (2010). Intracardiac injection of AdGRK5-NT reduces left ventricular hypertrophy by inhibiting NF-κB–dependent hypertrophic gene expression. Hypertension 56, 696–704. doi: 10.1161/HYPERTENSIONAHA.110.155960
  51. Molkentin, J. D., Lu, J.-R., Antos, C. L., Markham, B., Richardson, J., Robbins, J., et al. (1998). A calcineurin-dependent transcriptional pathway for cardiac hypertrophy. Cell 93, 215–228. doi: 10.1016/S0092-8674(00)81573-1
  52. Bossuyt, J., Helmstadter, K., Wu, X., Clements-Jewery, H., Haworth, R. S., Avkiran, M., et al. (2008). Ca2+/calmodulin-dependent protein kinase IIδ and protein kinase d overexpression reinforce the histone deacetylase 5 redistribution in heart failure. Circ. Res. 102, 695–702. doi: 10.1161/CIRCRESAHA.107.169755
  53. Zhang, T., Kohlhaas, M., Backs, J., Mishra, S., Phillips, W., Dybkova, N., et al. (2007). CaMKIIδ isoforms differentially affect calcium handling but similarly regulate HDAC/MEF2 transcriptional responses. J. Biol. Chem. 282, 35078–35087. doi: 10.1074/jbc.M707083200
  54. Purcell, N. H., Wilkins, B. J., York, A., Saba-El-Leil, M. K., Meloche, S., Robbins, J., et al. (2007). Genetic inhibition of cardiac ERK1/2 promotes stress-induced apoptosis and heart failure but has no effect on hypertrophy in vivo. Proc. Natl. Acad. Sci. U.S.A. 104, 14074–14079. doi: 10.1073/pnas.0610906104
  55. Kehat, I., Davis, J., Tiburcy, M., Accornero, F., Saba-El-Leil, M. K., Maillet, M., et al. (2011). Extracellular signal-regulated kinases 1 and 2 regulate the balance between eccentric and concentric cardiac growth novelty and significance. Circ. Res. 108, 176–183. doi: 10.1161/CIRCRESAHA.110.231514
  56. Shioi, T., McMullen, J. R., Kang, P. M., Douglas, P. S., Obata, T., Franke, T. F., et al. (2002). Akt/protein kinase b promotes organ growth in transgenic mice. Mol. Cell. Biol. 22, 2799–2809. doi: 10.1128/MCB.22.8.2799-2809.2002
  57. Sussman, M. A., Volkers, M., Fischer, K., Bailey, B., Cottage, C. T., Din, S., et al. (2011). Myocardial AKT: the omnipresent nexus. Physiol. Rev. 91, 1023–1070. doi: 10.1152/physrev.00024.2010
  58. Johnson, L. R., Scott, M. G., and Pitcher, J. A. (2004). G protein-coupled receptor kinase 5 contains a DNA-binding nuclear localization sequence. Mol. Cell. Biol. 24, 10169–10179. doi: 10.1128/MCB.24.23.10169-10179.2004
  59. Gold, J. I., Martini, J. S., Hullmann, J., Gao, E., Chuprun, J. K., Lee, L., et al. (2013). Nuclear translocation of cardiac G protein-coupled receptor kinase 5 downstream of select gq-activating hypertrophic ligands is a calmodulin-dependent process. PLoS One 8:e57324. doi: 10.1371/journal.pone.0057324
  60. Yi, X. P., Gerdes, A. M., and Li, F. (2002). Myocyte redistribution of GRK2 and GRK5 in hypertensive, heart-failure–prone rats. Hypertension 39, 1058–1063. doi: 10.1161/01.HYP.0000019130.09167.3B
  61. Martini, J. S., Raake, P., Vinge, L. E., DeGeorge, B. R., Chuprun, J. K., Harris, D. M., et al. (2008). Uncovering G protein-coupled receptor kinase-5 as a histone deacetylase kinase in the nucleus of cardiomyocytes. Proc. Natl. Acad. Sci. U.S.A. 105, 12457–12462. doi: 10.1073/pnas.0803153105
  62. Zhang, Y., Matkovich, S. J., Duan, X., Gold, J. I., Koch, W. J., and Dorn, G. W. II (2011). Nuclear effects of G-protein receptor kinase 5 on histone deacetylase 5-regulated gene transcription in heart failure. Circ. Heart Fail. 4, 659–668. doi: 10.1161/CIRCHEARTFAILURE.111.962563
  63. Akhter, S. A., Luttrell, L. M., Rockman, H. A., Iaccarino, G., Lefkowitz, R. J., and Koch, W. J. (1998). Targeting the receptor-gq interface to inhibit in vivo pressure overload myocardial hypertrophy. Science 280, 574–577. doi: 10.1126/science.280.5363.574
  64. Freeman, J. L., Enrique, M., Pollard, T. D., Lefkowitz, R. J., and Pitcher, J. A. (1998). Regulation of G protein-coupled receptor kinase 5 (GRK5) by actin. J. Biol. Chem. 273, 20653–20657. doi: 10.1074/jbc.273.32.20653
  65. Haeseleer, F., Sokal, I., Verlinde, C. L., Erdjument-Bromage, H., Tempst, P., Pronin, A. N., et al. (2000). Five members of a novel Ca2+-binding protein (CABP) subfamily with similarity to calmodulin. J. Biol. Chem. 275, 1247–1260. doi: 10.1074/jbc.275.2.1247
  66. Schafer, B. W., and Heizmann, C. W. (1996). The s100 family of EF-hand calcium-binding proteins: functions and pathology. Trends Biochem. Sci. 21, 134–140. doi: 10.1016/S0968-0004(96)80167-8
  67. Ikura, M. (1996). Calcium binding and conformational response in EF-hand proteins. Trends Biochem. Sci. 21, 14–17. doi: 10.1016/S0968-0004(06)80021-6
  68. Johnson, L. R., Robinson, J. D., Lester, K. N., and Pitcher, J. A. (2013). Distinct structural features of G protein-coupled receptor kinase 5 (GRK5) regulate its nuclear localization and DNA-binding ability. PLoS One 8:e62508. doi: 10.1371/journal.pone.0062508
  69. Hullmann, J. E., Grisanti, L. A., Makarewich, C. A., Gao, E., Gold, J. I., Chuprun, J. K., et al. (2014). GRK5-mediated exacerbation of pathological cardiac hypertrophy involves facilitation of nuclear NFAT activity novelty and significance. Circ. Res. 115, 976–985. doi: 10.1161/CIRCRESAHA.116.304475
  70. Patial, S., Luo, J., Porter, K. J., Benovic, J. L., and Parameswaran, N. (2009). G-protein coupled receptor kinases mediate TNF alpha-induced NF kappab signalling via direct interaction with and phosphorylation of I kappa B alpha. Biochem. J. 425, 169–178. doi: 10.1042/BJ20090908
  71. Valanne, S., Myllymaki, H., Kallio, J., Schmid, M. R., Kleino, A., Murumägi, A., et al. (2010). Genome-wide RNA interference in drosophila cells identifies G protein-coupled receptor kinase 2 as a conserved regulator of NF-κB signaling. J. Immunol. 184, 6188–6198. doi: 10.4049/jimmunol.1000261
  72. Islam, K. N., and Koch, W. J. (2012). Involvement of nuclear factor κB (NF-κB) signaling pathway in regulation of cardiac G protein-coupled receptor kinase 5 (GRK5) expression. J. Biol. Chem. 287, 12771–12778. doi: 10.1074/jbc.M111.324566
  73. Dorn, G. W. (2007). The fuzzy logic of physiological cardiac hypertrophy. Hypertension 49, 962–970. doi: 10.1161/HYPERTENSIONAHA.106.079426
  74. Huang, C.-Y., Yang, A.-L., Lin, Y.-M., Wu, F.-N., Lin, J. A., Chan, Y.-S., et al. (2011). Anti-apoptotic and pro-survival effects of exercise training on hypertensive hearts. J. Appl. Physiol. 112, 883–891. doi: 10.1152/japplphysiol.00605.2011
  75. van Berlo, J. H., Maillet, M., and Molkentin, J. D. (2013). Signaling effectors underlying pathologic growth and remodeling of the heart. J. Clin. Invest. 123, 37–45. doi: 10.1172/JCI62839
  76. Traynham, C. J., Cannavo, A., Zhou, Y., Vouga, A., Woodall, B. P., Hullmann, J. E., et al. (2015). Differential role of G protein-coupled receptor kinase 5 in physiological versus pathological cardiac hypertrophy. Circ. Res. 117, 1001–1012. doi: 10.1161/CIRCRESAHA.115.306961
  77. Wilkins, B. J., Dai, Y.-S., Bueno, O. F., Parsons, S. A., Xu, J., Plank, D. M., et al. (2004). Calcineurin/NFAT coupling participates in pathological, but not physiological, cardiac hypertrophy. Circ. Res. 94, 110–118. doi: 10.1161/01.RES.0000109415.17511.18
  78. Maudsley, S., Pierce, K. L., Zamah, A. M., Miller, W. E., Ahn, S., Daaka, Y., et al. (2000). The β2-adrenergic receptor mediates extracellular signal-regulated kinase activation via assembly of a multireceptor complex with the epidermal growth factor receptor. J. Biol. Chem. 275, 9572–9580. doi: 10.1074/jbc.275.13.9572
  79. Noma, T., Lemaire, A., Prasad, S. V. N., Barki-Harrington, L., Tilley, D. G., Chen, J., et al. (2007). β-arrestin–mediated β 1-adrenergic receptor transactivation of the EGFR confers cardioprotection. J. Clin. Invest. 117, 2445–2458. doi: 10.1172/JCI31901
  80. Bath, P., Chalmers, J., Powers, W., Beilin, L., Davis, S., Lenfant, C., et al. (2003). International society of hypertension (ISH): statement on the management of blood pressure in acute stroke. J. Hypertens. 21, 665–672. doi: 10.1097/00004872-200304000-00003
  81. Chobanian, A. V., Bakris, G. L., Black, H. R., Cushman, W. C., Green, L. A., Izzo, J. L., et al. (2003). Seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure. Hypertension 42, 1206–1252. doi: 10.1161/01.HYP.0000107251.49515.c2
  82. Ishizaka, N., Alexander, R. W., Laursen, J. B., Kai, H., Fukui, T., Oppermann, M., et al. (1997). G protein-coupled receptor kinase 5 in cultured vascular smooth muscle cells and rat aorta regulation by angiotensin II and hypertension. J. Biol. Chem. 272, 32482–32488. doi: 10.1074/jbc.272.51.32482
  83. Ross, R. (1999). Atherosclerosis—an inflammatory disease. N. Engl. J. Med. 340, 115–126. doi: 10.1056/NEJM199901143400207
  84. Braunersreuther, V., Mach, F., and Steffens, S. (2007). The specific role of chemokines in atherosclerosis. Thromb. Haemost. 97, 714–721. doi: 10.1160/TH07-01-0036
  85. Kehrl, J. H. (1998). Heterotrimeric G protein signaling: roles in immune function and fine-tuning by RGS proteins. Immunity 8, 1–10. doi: 10.1016/S1074-7613(00)80453-7
  86. Pitcher, J. A., Freedman, N. J., and Lefkowitz, R. J. (1998). G protein–coupled receptor kinases. Annu. Rev. Biochem. 67, 653–692. doi: 10.1146/annurev.biochem.67.1.653
  87. Hayek, T., Attias, J., Coleman, R., Brodsky, S., Smith, J., Breslow, J. L., et al. (1999). The angiotensin converting enzyme inhibitor, fosinopril, and the angiotensin II receptor antagonist, losartan, inhibit LDL oxidation and attenuate atherosclerosis independent of lowering blood pressure in apolipoprotein e deficient mice. Cardiovasc. Res. 44, 579–587. doi: 10.1016/S0008-6363(99)00239-4
  88. Tiruppathi, C., Yan, W., Sandoval, R., Naqvi, T., Pronin, A. N., Benovic, J. L., et al. (2000). G protein coupled receptor kinase-5 regulates thrombin-activated signaling in endothelial cells. Proc. Natl. Acad. Sci. U.S.A. 97, 7440–7445. doi: 10.1073/pnas.97.13.7440
  89. Fan, J., and Malik, A. B. (2003). Toll-like receptor-4 (TLR4) signaling augments chemokine-induced neutrophil migration by modulating cell surface expression of chemokine receptors. Nat. Med. 9, 315–321. doi: 10.1038/nm832
  90. Kim, J., Ahn, S., Ren, X.-R., Whalen, E. J., Reiter, E., Wei, H., et al. (2005). Functional antagonism of different G protein-coupled receptor kinases for β-arrestin-mediated angiotensin II receptor signaling. Proc. Natl. Acad. Sci. U.S.A. 102, 1442–1447. doi: 10.1073/pnas.0409532102
  91. Bea, F., Kreuzer, J., Preusch, M., Schaab, S., Isermann, B., Rosenfeld, M. E., et al. (2006). Melagatran reduces advanced atherosclerotic lesion size and may promote plaque stability in apolipoprotein e– deficient mice. Arterioscler. Thromb. Vasc. Biol. 26, 2787–2792. doi: 10.1161/01.ATV.0000246797.05781.ad
  92. Zernecke, A., Shagdarsuren, E., and Weber, C. (2008). Chemokines in atherosclerosis: an update. Arterioscler. Thromb. Vasc. Biol. 28, 1897–1908. doi: 10.1161/ATVBAHA.107.161174
  93. Mercer, J., Figg, N., Stoneman, V., Braganza, D., and Bennett, M. R. (2005). Endogenous p53 protects vascular smooth muscle cells from apoptosis and reduces atherosclerosis in APoE knockout mice. Circ. Res. 96, 667–674. doi: 10.1161/01.RES.0000161069.15577.ca
  94. Patial, S., Shahi, S., Saini, Y., Lee, T., Packiriswamy, N., Appledorn, D. M., et al. (2011). G-protein coupled receptor kinase 5 mediates lipopolysaccharide-induced NFκB activation in primary macrophages and modulates inflammation in vivo in mice. J. Cell. Physiol. 226, 1323–1333. doi: 10.1002/jcp.22460
  95. Wu, J.-H., Goswami, R., Cai, X., Exum, S. T., Huang, X., Zhang, L., et al. (2006). Regulation of the platelet-derived growth factor receptor-β by G protein-coupled receptor kinase-5 in vascular smooth muscle cells involves the phosphatase SHP2. J. Biol. Chem. 281, 37758–37772. doi: 10.1074/jbc.M605756200
  96. Cai, X., Wu, J.-H., Exum, S. T., Oppermann, M., Premont, R. T., Shenoy, S. K., et al. (2008). Reciprocal regulation of the platelet-derived growth factor receptor-β and G protein-coupled receptor kinase 5 by cross-phosphorylation: effects on catalysis. Mol. Pharmacol. 75, 626–636. doi: 10.1124/mol.108.050278
  97. Erdtmann-Vourliotis, M., Mayer, P., Ammon, S., Riechert, U., and Hollt, V. (2001). Distribution of g-protein-coupled receptor kinase (GRK) isoforms 2, 3, 5 and 6 mRNA in the rat brain. Mol. Brain Res. 95, 129–137. doi: 10.1016/S0006-8993(01)03046-3
  98. Gainetdinov, R. R., Bohn, L. M., Walker, J. K., Laporte, S. A., Macrae, A. D., Caron, M. G., et al. (1999). Muscarinic supersensitivity and impaired receptor desensitization in G protein–coupled receptor kinase 5–deficient mice. Neuron 24, 1029–1036. doi: 10.1016/S0896-6273(00)81048-X
  99. Matsui, M., Yamada, S., Oki, T., Manabe, T., Taketo, M. M., and Ehlert, F. J. (2004). Functional analysis of muscarinic acetylcholine receptors using knockout mice. Life Sci. 75, 2971–2981. doi: 10.1016/j.lfs.2004.05.034
  100. Liu, J., Rasul, I., Sun, Y., Wu, G., Li, L., Premont, R. T., et al. (2009). GRK5 deficiency leads to reduced hippocampal acetylcholine level via impaired presynaptic M2/M4 autoreceptor desensitization. J. Biol. Chem. 284, 19564–19571. doi: 10.1074/jbc.M109.005959
  101. Blokland, A. (1995). Acetylcholine: a neurotransmitter for learning and memory? Brain Res. Rev. 21, 285–300.
  102. Chen, Y., Wang, F., Long, H., Chen, Y., Wu, Z., and Ma, L. (2011). Grk5 promotes f-actin bundling and targets bundles to membrane structures to control neuronal morphogenesis. J. Cell Biol. 194, 905–920. doi: 10.1083/jcb.201104114
  103. Selkoe, D. J., and Hardy, J. (2016). The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 8, 595–608. doi: 10.15252/emmm.201606210
  104. Badran, M., Ayas, N. T., and Laher, I. (2014). Cardiovascular complications of sleep apnea: role of oxidative stress. Oxid. Med. Cell. Longev. 2014:985258. doi: 10.1155/2014/985258
  105. Tanzi, R. E., and Bertram, L. (2005). Twenty years of the Alzheimer’s disease amyloid hypothesis: a genetic perspective. Cell 120, 545–555. doi: 10.1016/j.cell.2005.02.
  106. Mawuenyega, K. G., Sigurdson, W. C., Ovod, V., Munsell, L. Y., Kasten, T. P., Morris, J. C., et al. (2010). Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science 330:1774. doi: 10.1126/science.1197623
  107. Terry, A. V., and Buccafusco, J. (2003). The cholinergic hypothesis of age and Alzheimer’s disease-related cognitive deficits: recent challenges and their implications for novel drug development. J. Pharmacol. Exp. Ther. 306, 821–827. doi: 10.1124/jpet.102.041616
  108. Sadot, E., Gurwitz, D., Barg, J., Behar, L., Ginzburg, I., and Fisher, A. (1996). Activation of m1 muscarinic acetylcholine receptor regulates τ phosphorylation in transfected PC12 cells. J. Neurochem. 66, 877–880. doi: 10.1046/j.1471-4159.1996.66020877.x
  109. Li, L., Rasul, I., Liu, J., Zhao, B., Tang, R., Premont, R. T., et al. (2009). Augmented axonal defects and synaptic degenerative changes in female GRK5 deficient mice. Brain Res. Bull. 78, 145–151. doi: 10.1016/j.brainresbull.2008.09.019
  110. Li, L., Liu, J., and Suo, W. Z. (2008). Grk5 deficiency exaggerates inflammatory changes in TgAPPsw mice. J. Neuroinflammation 5:24. doi: 10.1186/1742-2094-5-24
  111. Cheng, S., Li, L., He, S., Liu, J., Sun, Y., He, M., et al. (2010). Grk5 deficiency accelerates β-amyloid accumulation in TG2576 mice via impaired cholinergic activity. J. Biol. Chem. 285, 41541–41548. doi: 10.1074/jbc.M110.170894
  112. Zhang, Y., Chen, L., Shen, G., Zhao, Q., Shangguan, L., and He, M. (2014). GRK5 dysfunction accelerates tau hyperphosphorylation in APP (swe) mice through impaired cholinergic activity. Neuroreport 25, 542–547. doi: 10.1097/WNR.0000000000000142
  113. Bartus, R. T., Dean, R. L., Beer, B., and Lippa, A. S. (1982). The cholinergic hypothesis of geriatric memory dysfunction. Science 217, 408–414. doi: 10.1126/science.7046051
  114. Bartus, R. T., Dean, R. L., Pontecorvo, M. J., and Flicker, C. (1985). The cholinergic hypothesis: a historical overview, current perspective, and future directions. Ann. N. Y. Acad. Sci. 444, 332–358. doi: 10.1111/j.1749-6632.1985.tb37600.x
  115. Woolf, N. J. (1996). The critical role of cholinergic basal forebrain neurons in morphological change and memory encoding: a hypothesis. Neurobiol. Learn. Mem. 66, 258–266. doi: 10.1006/nlme.1996.0068
  116. Hardy, J., and Selkoe, D. J. (2002). The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 297, 353–356. doi: 10.1126/science.1072994
  117. Small, D. H., and Cappai, R. (2006). Alois Alzheimer and Alzheimer’s disease: a centennial perspective. J. Neurochem. 99, 708–710. doi: 10.1111/j.1471-4159.2006.04212.x
  118. Fisher, A. (2008). Cholinergic treatments with emphasis on m1 muscarinic agonists as potential disease modifying agents for Alzheimer’s disease. Neurotherapeutics 5, 433–442. doi: 10.1016/j.nurt.2008.05.002
  119. Suo, Z., Wu, M., Citron, B. A., Wong, G. T., and Festoff, B. W. (2004). Abnormality of G-protein coupled receptor kinases at prodromal and early stages of Alzheimer’s disease: an association with early β-amyloid accumulation. J. Neurosci. 24, 3444–3452. doi: 10.1523/JNEUROSCI.4856-03.2004
  120. Pronin, A. N., Morris, A. J., Surguchov, A., and Benovic, J. L. (2000). Synucleins are a novel class of substrates for G protein-coupled receptor kinases. J. Biol. Chem. 275, 26515–26522. doi: 10.1074/jbc.M003542200
  121. Arawaka, S., Wada, M., Goto, S., Karube, H., Sakamoto, M., Ren, C.-H., et al. (2006). The role of g-protein-coupled receptor kinase 5 in pathogenesis of sporadic Parkinson’s disease. J. Neurosci. 26, 9227–9238. doi: 10.1523/JNEUROSCI.0341-06.2006
  122. Carman, C. V., Som, T., Kim, C. M., and Benovic, J. L. (1998). Binding and phosphorylation of tubulin by G protein-coupled receptor kinases. J. Biol. Chem. 273, 20308–20316. doi: 10.1074/jbc.273.32.20308
  123. Dawson, T. M., and Dawson, V. L. (2002). Neuroprotective and neurorestorative strategies for Parkinson’s disease. Nat. Neurosci. 5, 1058–1061. doi: 10.1038/nn941
  124. Rango, M., and Bresolin, N. (2018). Brain mitochondria, aging, and Parkinson’s disease. Genes 9:E250. doi: 10.3390/genes9050250
  125. Goncalves, S. A., and Outeiro, T. F. (2013). Assessing the subcellular dynamics of alpha-synuclein using photoactivation microscopy. Mol. Neurobiol. 47, 1081–1092. doi: 10.1007/s12035-013-8406-x
  126. Fares, M.-B., Ait-Bouziad, N., Dikiy, I., Mbefo, M. K., Jovičić, A., Kiely, A., et al. (2014). The novel Parkinson’s disease linked mutation G51D attenuates in vitro aggregation and membrane binding of α-synuclein, and enhances its secretion and nuclear localization in cells. Hum. Mol. Genet. 23, 4491–4509. doi: 10.1093/hmg/ddu165
  127. Xu, S., Zhou, M., Yu, S., Cai, Y., Zhang, A., Ueda, K., et al. (2006). Oxidative stress induces nuclear translocation of C-terminus of α-synuclein in dopaminergic cells. Biochem. Biophys. Res. Commun. 342, 330–335. doi: 10.1016/j.bbrc.2006.01.148
  128. Monti, B., Gatta, V., Piretti, F., Raffaelli, S. S., Virgili, M., and Contestabile, A. (2010). Valproic acid is neuroprotective in the rotenone rat model of Parkinson’s disease: involvement of α-synuclein. Neurotox. Res. 17, 130–141. doi: 10.1007/s12640-009-9090-5
  129. Siddiqui, A., Chinta, S. J., Mallajosyula, J. K., Rajagopolan, S., Hanson, I., Rane, A., et al. (2012). Selective binding of nuclear α-synuclein to the PGC1α promoter under conditions of oxidative stress may contribute to losses in mitochondrial function: implications for Parkinson’s disease. Free Radic. Biol. Med. 53, 993–1003. doi: 10.1016/j.freeradbiomed.2012.05.024
  130. Goers, J., Manning-Bog, A. B., McCormack, A. L., Millett, I. S., Doniach, S., Di Monte, D. A., et al. (2003). Nuclear localization of α-synuclein and its interaction with histones. Biochemistry 42, 8465–8471. doi: 10.1021/bi0341152
  131. Kontopoulos, E., Parvin, J. D., and Feany, M. B. (2006). α-synuclein acts in the nucleus to inhibit histone acetylation and promote neurotoxicity. Hum. Mol. Genet. 15, 3012–3023. doi: 10.1093/hmg/ddl243
  132. Sakamoto, M., Arawaka, S., Hara, S., Sato, H., Cui, C., Machiya, Y., et al. (2009). Contribution of endogenous G-protein-coupled receptor kinases to Ser129 phosphorylation of α-synuclein in HEK293 cells. Biochem. Biophys. Res. Commun. 384, 378–382. doi: 10.1016/j.bbrc.2009.04.130
  133. Liu, P., Wang, X., Gao, N., Zhu, H., Dai, X., Xu, Y., et al. (2010). G protein-coupled receptor kinase 5, overexpressed in the α-synuclein up-regulation model of Parkinson’s disease, regulates bcl-2 expression. Brain Res. 1307, 134–141. doi: 10.1016/j.brainres.2009.10.036
  134. Takahashi, M., Uchikado, H., Caprotti, D., Weidenheim, K. M., Dickson, D. W., Ksiezak-Reding, H., et al. (2006). Identification of G-protein coupled receptor kinase 2 in paired helical filaments and neurofibrillary tangles. J. Neuropathol. Exp. Neurol. 65, 1157–1169. doi: 10.1097/01.jnen.0000248542.82681.12
  135. Tarantino, P., De Marco, E. V., Annesi, G., Rocca, F. E., Annesi, F., Civitelli, D., et al. (2011). Lack of association between G-protein coupled receptor kinase 5 gene and Parkinson’s disease. Am. J. Med. Genet. B Neuropsychiatr. Genet. 156, 104–107. doi: 10.1002/ajmg.b.31129
  136. Liu, P., Wang, X., Gao, N., Zhu, H., Dai, X., Xu, Y., et al. (2010). G protein-coupled receptor kinase 5, overexpressed in the α-synuclein up-regulation model of Parkinson’s disease, regulates bcl-2 expression. Brain Res. 1307, 134–141. doi: 10.1016/j.brainres.2009.10.036
  137. Takahashi, M., Uchikado, H., Caprotti, D., Weidenheim, K. M., Dickson, D. W., Ksiezak-Reding, H., et al. (2006). Identification of G-protein coupled receptor kinase 2 in paired helical filaments and neurofibrillary tangles. J. Neuropathol. Exp. Neurol. 65, 1157–1169. doi: 10.1097/01.jnen.0000248542.82681.12
  138. Tarantino, P., De Marco, E. V., Annesi, G., Rocca, F. E., Annesi, F., Civitelli, D., et al. (2011). Lack of association between G-protein coupled receptor kinase 5 gene and Parkinson’s disease. Am. J. Med. Genet. B Neuropsychiatr. Genet. 156, 104–107. doi: 10.1002/ajmg.b.31129
More
Information
Subjects: Gerontology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 668
Entry Collection: Neurodegeneration
Revisions: 3 times (View History)
Update Date: 23 Oct 2022
1000/1000