Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jun 1.
Published in final edited form as: Pflugers Arch. 2014 Feb 12;466(6):1101–1111. doi: 10.1007/s00424-014-1456-8

Focal Adhesion Signaling in Heart Failure

Allen M Samarel 1
PMCID: PMC4037338  NIHMSID: NIHMS565010  PMID: 24515292

Summary

In this brief review, recent evidence is presented to indicate a role for specific components of the cardiomyocyte costamere (and its related structure the focal adhesion complex of cultured cardiomyocytes) in initiating and sustaining the aberrant signal transduction that contributes to myocardial remodeling and the progression to heart failure (HF). Special attention is devoted to the focal adhesion kinase family of nonreceptor protein tyrosine kinases in bi-directional signal transduction during cardiac remodeling and HF progression. Finally, some speculations and directions for future study are provided for this rapidly developing field of research.

Keywords: costameres, focal adhesion kinase, PYK2, talin, vinculin

Introduction

Heart failure (HF) results from the inability of the heart to pump blood forward at a sufficient rate to meet the metabolic demands of the body, or the ability to do so only at the expense of abnormally high filling pressures. Even before the onset of overt HF, reduced cardiac performance leads to profound mechanical and neurohormonal stresses on the remaining functional myocardium. These stresses trigger signal transduction pathways that ultimately result in additional structural changes in the cardiomyocytes and nonmuscle cells of the heart (so called “myocardial remodeling”) which contribute to further functional deterioration of the already diseased myocardium [7]. The downward spiral in cardiac performance during cardiac remodeling may be initiated by specific signaling molecules that function to integrate both neurohormonal and mechanical signals in order to elicit compensatory responses in an attempt to counteract the functional deterioration. However, some of these compensatory responses ultimately prove detrimental. In this brief review, I will focus on recent evidence to indicate a role for specific components of the costamere (and its related structure the focal adhesion complex) in initiating and sustaining the aberrant signal transduction that contributes to myocardial remodeling and the progression to HF.

Cardiomyocyte costameres are sites of attachment to the extracellular matrix (ECM)

Cardiomyocyte costameres (and their focal adhesion counterparts in cultured cells), are critical cytoskeletal elements involved in bi-directional mechanotransduction [66]. The term “costamere” was first used by Craig and colleagues [58,57] to describe vinculin-containing, rib-like bands that encircle the cardiomyocyte perpendicular to its long axis. In addition to vinculin, costameres contain many other structural elements that coalesce beneath the sarcolemmal membrane, and resemble the metal ribs of a wooden barrel. They flank the Z-disc and overlying I-bands of sarcomeres that run just beneath the cardiomyocyte plasma membrane (FIGURE 1). Immunoelectron and fluorescent microscopic analyses have revealed that cardiomyocyte costameres also form discrete physical attachments between the underlying, outer Z-discs of the muscle cell and their surrounding, 3-dimensional, stress-tolerant ECM. Attachment is mediated in part by integrins, which are cell-surface receptors for a variety of ECM proteins, and the dystrophin-glycoprotein complex, which mediates attachment to the ECM protein laminin. It is through these attachments that costameres directly transmit contractile forces generated within the cardiomyocyte to the surrounding ECM, and where longitudinal displacement of the ECM by adjacent muscle cells is transmitted directly to the internal contractile machinery. Thus, both externally applied and intrinsically generated mechanical loads are transmitted bi-directionally through costameres.

Figure 1. Cellular localization of costameres in striated muscle.

Figure 1

(A) A schematic diagram is depicted, illustrating costameres as circumferential elements that physically couple peripheral myofibrils to the sarcolemma in periodic register with the Z-disk. The protein composition of costameres includes integrins, a variety of cytoskeletal and adaptor proteins, and signaling kinases. (B) An inside-out sarcolemma that was mechanically peeled from a single myofiber and stained with antibodies to γ-actin to reveal the costameric cytoskeleton. Bar = 10 μm. The figure was reproduced from [17] by copyright permission of The American Society for Biochemistry and Molecular Biology.

Focal adhesion complexes in cultured cardiomyocytes

Costameres share many of the structural features of cell-to-matrix adherens junctions, and thus may be considered striated muscle-specific elaborations of focal adhesions found in cultured nonmuscle cells [17]. Immunolocalization studies have confirmed that many, if not all of the same proteins that comprise the costamere in vivo eventually reassemble within focal adhesions as isolated cardiomyocytes attach and spread in culture. Furthermore, cultured cardiomyocytes that retain contractile activity, or are stimulated to contract in culture, re-assemble their costameric proteins along the cell-substratum interface in register with the overlying Z-discs of their remodeled sarcomeres. These basal, costameric attachment sites, as well as the remodeled cell-to-cell adherens junctions derived from the remaining components of the intercalated disc, provide the major cell adhesion sites of both neonatal and adult cardiomyocytes in culture.

A detailed structural analysis of the various focal adhesion components of cardiomyocyte costameres and focal adhesions has not yet been obtained. However, using photoactivatable fusion proteins of several focal adhesion proteins and interferometric photoactivated localization microscopy, Kanchanawong et al. [34] have described the 3-dimensional organization of focal adhesion complexes in human osteosarcoma and mouse embryonic fibroblast cells (FIGURE 2). Their images reveal a highly organized, vertically layered structure consisting of at least 3 strata: a membrane-apposed integrin signaling layer containing integrin cytoplasmic tails, focal adhesion kinase (FAK) and the adaptor protein paxillin; an intermediate force-transduction layer containing talin and vinculin; and an uppermost actin-regulatory layer containing zyxin, vasodilator-stimulated phosphoprotein (VASP) and α-actinin overlying and attached to the actin cytoskeleton. The integrin cytoplasmic domains and the subcortical actin layer were separated by a distance of approximately 40nm, indicating an important role for talin, vinculin and other intermediary proteins above the plasma membrane in bi-directional force transmission.

Figure 2. Nanoscale architecture of focal adhesions.

Figure 2

Schematic model of focal adhesion molecular architecture, depicting experimentally determined protein positions. Note that the model does not depict protein stoichiometry. The figure was reproduced from [33] by copyright permission of the Nature Publishing Group.

These images provide enormous detail in describing the molecular architecture of focal adhesions, but do not reveal the dynamic nature of focal adhesion formation and dissolution. However, the rapid turnover of focal adhesion components plays a crucial role in cellular differentiation and migration during cardiac development [18,89,30,23,24], and may also be an important regulatory factor during new sarcomere addition in response to hypertrophic stimuli [47,10]. Using fluorescence recovery after photobleaching (FRAP) and mathematical modeling, Ingber and colleagues [42] showed that various components of the focal adhesion complex display residence times that vary from as little as 1 sec for vinculin, and up to 111 sec for talin. Sanger and co-workers [86] had previously observed a similar dynamic range of exchange between costamere/Z-disc proteins and the cytoplasm of spreading skeletal muscle myotubes. Using FRAP, we subsequently demonstrated that the phosphorylation of FAK, a critical component of the signaling layer of cardiomyocyte focal adhesions, regulates the stability of paxillin within cardiomyocyte focal adhesions, and ultimately controls the rate of cell spreading and myofibrillar organization of cultured cardiomyocytes in response to both static stretch, and the hypertrophic agonist endothelin-1 [10]. Thus, the dynamic nature of cytoskeletal assembly and disassembly within focal adhesion complexes appears critical during the response of cultured cardiomyocytes to neurohormonal and mechanical stimuli.

Focal adhesion complexes assemble in response to mechanical overload in vivo

In pressure-overloaded feline myocardium, Kuppaswamy and co-workers [39,41] first demonstrated the cytoskeletal assembly of c-Src and other signaling proteins, which was partially mimicked in vitro using adult feline cardiomyocytes embedded within a three-dimensional collagen matrix and stimulated with an integrin-binding Arg-Gly-Asp (RGD) peptide [41]. In subsequent elegant studies, Franchini and co-workers [20,15,85], analyzed the rapid assembly of focal adhesion complexes in response to pressure-loading of the isolated perfused rat heart. The assembly of focal adhesion complexes was also an early response of cultured cardiomyocytes to a variety of neurohormonal and mechanical stimuli that ultimately lead to cardiomyocyte hypertrophy [76,16,63,82,84].

Clustering of β1-integrins at the sarcolemmal membrane and their attachment to ECM proteins were critical factors in the regulation of focal adhesion assembly in these settings. Ross and colleagues [75] further demonstrated the importance of β-integrins in costamere assembly. They used Cre recombinase driven by the myosin light chain-2 ventricular (MLC-2v) promoter to induce β1-integrin gene excision exclusively in ventricular cardiomyocytes. They found that β1-integrin knockout mice had significantly depressed contractile performance with extensive cardiac fibrosis, and were intolerant to pressure-overload produced by transverse aortic coarctation (TAC). Surviving mice developed spontaneous heart failure by 6 months of age, with abnormal myocyte membrane integrity as determined by Evan’s blue dye staining. This group subsequently described the structural and functional consequences of cardiomyocyte-specific excision of the β1-integrin gene in adult cardiomyocytes [43]. Using a tamoxofen-inducible promoter to drive expression of Cre recombinase, even partial excision of the β1-integrin gene resulted in multiple defects in mechanotransduction signaling when hearts were stressed by TAC. Adaptive hypertrophy was blunted, and the response to adrenergic stimuli was also markedly impaired. Thus, these results demonstrated a critical function of β1-integrins in the postnatal myocardium, and first linked defects in β1-integrin-dependent costamere assembly to the development of cardiomyopathy.

In the majority of aforementioned studies, a hemodynamic stress in the form of an increase in ventricular afterload of the intact heart [39,41,20,85,75,43] was used to stimulate focal adhesion assembly. However, Domingos et al. [15] showed that increasing ventricular preload (by raising diastolic pressure from 0 to 15 mmHg in the beating, isolated perfused rat heart) also rapidly increased FAK tyrosine phosphorylation and binding of c-Src and Grb2 to FAK. This was paralleled by activation and binding of ERK1/2 to the cardiomyocyte cytoskeleton, indicating enhanced focal adhesion signaling in response to increased diastolic stress. Balloon inflation to raise intraventricular pressure in hearts perfused with cardioplegic solution also activated FAK and ERK1/2. In contrast, early induction of volume overload 4 weeks after surgically induced mitral regurgitation in the dog decreased focal adhesion signaling [65]. Similarly, rats with surgically induced aortocaval fistulae demonstrated increased ECM degradation, decreased focal adhesion signaling, and increased apoptosis [74]. Downregulation of focal adhesion signaling was mediated by the activation of PTEN, a dual lipid and protein phosphatase. Beta adrenergic blockade prevented the PTEN activation, the downregulation of focal adhesion signaling, and the increased rate of apoptosis, but had little effect on ECM degradation. These data suggest that volume overload per se is a relatively weak stimulator of focal adhesion signaling as compared to pressure overload of the intact heart. Attempts to model the differential effects of pressure vs. volume loading by subjecting cardiomyocytes to transverse vs. linear stretch support these observations [78,21,73].

β1-integrin cytoplasmic domains form cytoskeletal attachments through talin dimers

Unlike growth factor receptors, the cytoplasmic tails of integrins do not possess intrinsic catalytic activity. However, they interact with other cytoskeletal proteins to transmit extrinsically applied and internally generated mechanical force. A major binding partner of the β1-integrin cytoplasmic domain is the cytoskeletal protein talin, which is a rod-shaped, multi-domain protein involved in bi-directional activation of integrins [70]. Cell-surface integrins can exist in either low- or high-affinity states, and cellular modulation of integrin affinity is in part accomplished by reversible binding of the talin N-terminal, globular head domain to the C-terminal, β1-integrin cytoplasmic tail [8]. Cardiomyocytes predominantly express the muscle-specific β1D-integrin isoform, which has the highest binding affinity for talin of the various β-integrin cytoplasmic domains. This observation suggests that integrin engagement to the cardiac ECM favors a mechanically strong, activated integrin binding conformation [2].

The mammalian genome contains two genes for talin encoding two structurally similar proteins (talin-1 and talin-2) that share 74% sequence identity [92]. The specific function of each talin isoform is not known, but it appears that the talin-2 isoform plays a unique role in muscle development and disease. Localization of talin-2 is restricted to costameres and intercalated discs in striated muscle [72], but talin-2 appears dispensable in the presence of talin-1 for normal costamere assembly. However, deletion of both genes produced a severe defect in sarcomere assembly in striated muscle, with profound defects in the assembly of focal adhesion complexes and sarcomeres by cultured myoblasts isolated from double knockout embryos [11]. The failure of normal sarcomere development in these immature muscle cells also highlights the potential importance of talin (and perhaps other cytoskeletal linker proteins) in adaptation to hypertrophic stimuli.

Ross and colleagues [46] recently evaluated talin-1 and talin-2 expression in the normal embryonic and adult mouse heart, as well as in control and failing human adult myocardium. Using isoform-specific antibodies and nonfailing human cardiac tissue, they showed that talin-1 was only weakly detected in the cardiomyocyte plasma membrane, and rarely co-localized with dystrophin, a muscle-specific costameric protein (FIGURE 3). Talin-1 was also more readily detected in nonmuscle cells of the cardiac interstitium. In contrast, talin-2 was readily detected in a costameric pattern in cardiomyoyctes as demonstrated by strong co-localization with dystrophin. Talin-1 function was then tested in the basal and mechanically stressed myocardium after cardiomyocyte-specific excision of the talin-1 gene. They found that during embryogenesis, both talin isoforms are highly expressed, but talin-2 is the main isoform expressed in adult mammalian cardiomyocytes, where it was predominantly localized to costameres. However, talin-1 expression was up-regulated during cardiac hypertrophy, suggesting that it plays an important role in the compensatory response of the heart to stress. In human failing heart, cardiomyocyte talin-1 was also increased as compared to control samples from normal functioning myocardium. Talin-1 knockout mice showed normal basal cardiac structure and function, but when subjected to pressure overload, these animals showed blunted hypertrophy, less fibrosis, but improved cardiac function versus controls. Overall, these data suggested that reduction of cardiomyoycte talin-1 expression might lead to improved cardiac remodeling following pressure overload.

Figure 3. Expression of talin-1 (Tln1) and talin-2 (Tln2) in adult human cardiac tissue.

Figure 3

Adult human cardiac tissue was evaluated for expression of Tln1 and Tln2. Tln1 was weakly detected in the cardiomyocyte membrane, as shown by co-localization with Dystrophin (Dys), a muscle-specific membrane marker. It was also detected in non-myocyte cells (*). Tln2 was detected in a costameric pattern in cardiomyocytes as demonstrated by strong co-localization with Dys (arrows). DAPI staining (blue) shows the position of cell nuclei. Scale bar = 20 μm. The figure was reproduced from [45] by copyright permission of The American Society for Biochemistry and Molecular Biology.

Exactly how talin isoforms mediate integrin-dependent signal transduction in the heart remains unknown. Adhesion to ECM proteins via β1-integrins causes the recruitment of talin dimers to the cytoplasmic face of the sarcolemmal membrane, leading to integrin transition to its high affinity state. This process is an important, early step in “outside-in” signaling during cell attachment of cultured cardiomyocytes, and may play an important functional role in costamere assembly during the addition of new sarcomeres in vivo. Conversely, talin activation by a number of intracellular signaling pathways causes the physical displacement of α-integrin subunits, thereby allowing for high-affinity ECM engagement during “inside-out” signaling [80,2]. Intracellular talin binding alone is sufficient to alter the conformation of integrin extracellular domains and promote their attachment to ECM proteins [90]. The recruitment of talin involves the Src-dependent tyrosine phosphorylation of the β-integrin tail, and its recognition by the talin head region [1]. Thus, intracellular stimuli that cause Src-dependent integrin phosphorylation promote the activation of integrins via talin recruitment to costameres, and stimulate costamere formation during inside-out signaling. Subsequent recruitment of additional cytoplasmic linker proteins (such as paxillin and vinculin) to the costamere may also be required for Z-disc assembly and premyofibril formation during myofibrillogenesis [13,79].

FAK and cardiomyocyte mechanotransduction

Talin localization during integrin engagement and clustering may initiate outside-in signaling, but talin, like β1-integrin cytoplasmic domains, has no intrinsic catalytic activity capable of relaying biochemical signals into the cell interior. However, secondary recruitment and activation of protein tyrosine and serine/threonine kinases to the cytoplasmic adhesion plaque may accomplish this function. FAK is clearly one candidate enzyme that is responsible for integrin-mediated mechanotransduction within cardiomyocyte focal adhesions and costameres. As indicated above, FAK is a nonreceptor protein tyrosine kinase that functions as an “activatable scaffold” [69] in integrin-dependent signal transduction. FAK contains a central kinase domain flanked by long N- and C-terminal extensions (FIGURE 4). An autoinhibitory FERM domain, located within the N-terminal region of FAK, associates with the plasma membrane via its interaction with several different growth factor receptors. The C-terminal region of FAK comprises the focal adhesion targeting (FAT) domain. The FAK FAT domain binds directly to paxillin and talin, which in turn bind to the cytoplasmic tail of β1-integrins at sites of integrin clustering. Once localized, FAK phosphorylates itself at a single tyrosine residue (Y397). This autophosphorylation site serves as a high-affinity binding site for the SH2 domain of Src-family protein tyrosine kinases [68]. Once bound to FAK, active Src then phosphorylates FAK at residues Y576 and Y577 within its catalytic domain (which augments FAK kinase activity toward exogenous substrates), and at Y861 and Y925 near its C-terminus [19]. Phosphorylation of the Y861 site promotes the binding of p130Cas to FAK [45]. The Y925 phosphorylation site promotes the binding of Grb2 to FAK, and other adapter proteins and kinases containing SH2 domains. The FAK-Src complex can also phosphorylate paxillin, p130Cas, and other cytoskeletal proteins involved in costamere and myofibrillar assembly.

Figure 4. Structural domains of focal adhesion kinase (FAK) and proline-rich tyrosine kinase-2 (PYK2).

Figure 4

Proline-rich tyrosine kinase-2 (PYK2) shares a similar domain arrangement with focal adhesion kinase (FAK), with 60% sequence identity in the central kinase domain, conservation of proline-rich regions (PRRs), and identical positions of four tyrosine phosphorylation sites. PYK2 tyrosines 402, 579, 580 and 881 correspond to FAK tyrosines 397, 576, 577 and 925, respectively. FAK and PYK2 both contain a C-terminal focal adhesion targeting (FAT) domain that binds to paxillin. However, PYK2 shows a perinuclear distribution and is not strongly localized to focal contacts in many cells. The figure was reproduced from [49] by copyright permission of the Nature Publishing Group.

FAK serine phosphorylation and sarcomere assembly

In addition to multiple tyrosine phosphorylation sites, FAK contains several serine residues (S722, S843, S846, and S910) that undergo reversible phosphorylation in response to hypertrophic stimuli. These serine residues are in close proximity to critical protein-protein interaction sites within the C-terminal region of FAK, such as the binding site for p130Cas, Grb2, and the adjacent FAT domain. The functional role of FAK serine phosphorylation in cardiomyocytes is largely unknown, but one report indicates that serine (and tyrosine) phosphorylation of FAK increases dramatically in hypertensive rats, with different sites of phosphorylation appearing to regulate FAK subcellular localization [91]. Our group [10] recently demonstrated that endothelin-1 and other hypertrophic factors induced a time- and dose-dependent increase in FAK-S910 phosphorylation. Endothelin-induced FAK-S910 phosphorylation required endothelin Type A receptor-dependent activation of Protein Kinase Cδ (PKCδ) and Src via parallel Raf-1→ MEK1/2 →ERK1/2 and MEK5 →ERK5 signaling pathways. Replication-deficient adenoviruses expressing wild-type FAK and a non-phosphorylatable, S910A-FAK mutant were then used to examine the functional significance of FAK-S910 phosphorylation. Unlike wildtype FAK, S910A-FAK increased the half-life of GFP-tagged paxillin within costameres (as determined by total internal reflection fluorescence microscopy and FRAP) and increased the steady-state FAK-paxillin interaction (as determined by co-immunoprecipitation and Western blotting). These alterations resulted in reduced neonatal rat ventricular myocyte (NRVM) sarcomere reorganization and cell spreading. Finally, we found that FAK was serine-phosphorylated at multiple sites in non-failing, human left ventricular tissue, and both FAK-S910 phosphorylation and ERK5 expression were dramatically reduced in patients undergoing heart transplantation for end-stage dilated cardiomyopathy. These results suggest that reduced FAK-S910 phosphorylation may contribute to sarcomere disorganization that is frequently observed in end-stage heart failure patients [10].

The ERK-dependent, regulated exit of FAK from cardiomyocyte costameres [10] is reminiscent of the cyclic FAK tyrosine and serine phosphorylation that occurs during migration of nonmuscle cells [52]. In this scenario, FAK/Src tyrosine phosphorylation at Y397 and Y925 promotes the assembly of new costameres in response to hypertrophic agonists or mechanical strain. Subsequent ERK1/2/5 activation then leads to the serine phosphorylation of FAK at S910, which promotes FAK exit from newly formed costameres and its replacement by vinculin during premyofibril formation [67]. The dynamic nature of FAK entry and exit from focal adhesions and costameres is consistent with the time required for new sarcomere addition in response to longitudinal strain, and its dependence on FAK localization and activation [47].

FAK knockout mice and the development of HF

Studies of global and cardiomyocyte-specific FAK knockout mice lend further support for an important role for FAK in cardiomyocyte mechanotransduction and myofibrillar assembly. Global FAK deletion led to lethality at embryonic day 8.5, and the mutant embryos displayed a profound defect in development of all mesodermal structures, including the heart and vasculature [31,32]. Interestingly, the developmental defects found in FAK−/− embryos were phenotypically very similar in timing and phenotype to the morphological defects observed in fibronectin-null mice, suggesting an important relationship between fibronectin- and FAK-dependent signaling, especially with respect to development of the cardiovascular system [69]. In both cases, the developmental defects were attributed to the inability of mesodermal cells to migrate normally. Indeed, fibroblasts isolated from FAK−/− embryos displayed markedly reduced mobility and abnormally large focal adhesions, indicating a defect in focal adhesion turnover. However, cardiomyocyte-restricted FAK knockout mice have a variable cardiac phenotype depending on precisely when during development FAK is deleted. Embryonic deletion of FAK in cardiomyocytes caused perinatal lethality due to the presence of large ventricular septal defects and abnormalities in outflow tract alignment, indicating again that FAK predominantly regulates mammalian cardiomyocyte proliferation and migration during early cardiac development [24,61]. However, cardiomyocyte FAK deletion during later prenatal development was not associated with any congenital heart defects, but led to spontaneous dilated cardiomyopathy in aged animals [60], and a blunted hypertrophic response to angiotensin II infusion [60] or TAC [14] in the adult heart. The inability to respond normally to hypertrophic stimuli supported earlier cell culture studies that demonstrated impaired hypertrophic responses of cultured cardiomyocytes overexpressing FAK-related nonkinase (FRNK, a naturally occurring inhibitor of FAK), Y397F-FAK (a FAK autophosphorylation mutant), FAK antisense RNA, or just the FAK-FAT domain [63,16,27,38,84,47,53]. FAK also plays an important role in limiting stress-induced cardiomyocyte apoptosis, and improving cardiomyocyte survival after ischemia-reperfusion injury [25,87]. Furthermore, Vander Heide and co-workers recently showed that the tissue-specific, inducible knockout of FAK in adult cardiomyocytes substantially reduced the preconditioning response during ischemia-reperfusion injury [62]. Thus, there are substantial data indicating an important role for FAK and FAK-dependent signal transduction in preserving cardiac function, and preventing disease progression and HF during adaptation to hemodynamic, neurohormonal, and ischemic stress.

PYK2, the other member of the FAK family of protein tyrosine kinases

The FAK family of protein tyrosine kinases actually consists of 2 members [51], both of which are expressed in cardiomyocytes. Proline-rich Tyrosine Kinase 2 (PYK2; also known as FAK2, cell adhesion kinase-β (CAK-β); or Cell Adhesion Tyrosine Kinase (CADTK)), is the other member of the family, and shares ~45% protein sequence homology with FAK. PYK2 has a similar domain structure as FAK (FIGURE 4), and both kinases are believed to be derived from a common, ancient ancestral gene that diverged during early vertebrate evolution [12]. Whereas FAK is expressed at high levels in virtually all cells and tissues, PYK2 expression is much more restricted, and is preferentially expressed in cells of the endothelium, central nervous system and haematopoietic lineages.

Activation of PYK2 occurs in response to integrin engagement and clustering in some cell types, but its cellular distribution is predominantly cytoplasmic. This is particularly true in cardiomyocytes, where PYK2 is found throughout the cytoplasm of both neonatal and adult cells [3] with only a small portion localized to focal adhesions and costameres [28]. There are other important structural differences between PYK2 and FAK that reflect their distinct roles in cardiomyocyte signal transduction. Perhaps most important is the fact that unlike FAK, PYK2 activation is sensitive to intracellular Ca2+ ([Ca2+]i) signals, and undergoes dimerization in response to Ca2+-calmodulin binding to the FERM F2 subdomain within the N-terminus of the molecule [36]. The calcium dependence of PYK2 activation may be related to the dissociation of the PYK2 FERM domain from the central catalytic domain as occurs during FAK activation [44], resulting in dimerization and intermolecular trans-autophosphorylation. Indeed, overexpression of the PYK2 FERM domain (in the absence of calmodulin overexpression) inhibited the activation of full-length PYK2 by forming a complex with the inactive enzyme [Riggs, 2011 #7050]. These data indicate that the PYK2 FERM domain is also involved in the regulation of PYK2 activity by regulating the formation of PYK2 oligomers that are critical for its activity. Regardless of its role in oligomerization, Ca2+-calmodulin binding to the PYK2 FERM domain provided an answer to the question of how PYK2 activity was regulated by [Ca2+]i, and this may be an important issue with regard to its regulation during cardiac hypertrophy and HF.

As a Ca2+-calmodulin-dependent protein tyrosine kinase, PYK2 undergoes bimolecular transphosphorylation [59] in response to integrin engagement, increased [Ca2+]I, and activation of PKCs in many cell types, including cardiomyocytes [3,50,35,4,49,29,28,22,26]. Like FAK, PYK2 serves as an activatable scaffolding protein, and transduces signals from G-protein coupled receptors to the mitogen-activated protein kinases (MAPK) and the phosphoinositol-3-kinase (PI-(3)K)-PDK1-Akt signaling pathway depending upon which adaptor proteins bind to the phosphorylated enzyme [6,56,81,22]. Indeed, many of the same binding partners for FAK also bind PYK2, making it somewhat difficult to assign a specific role for PYK2-dependent signaling in cells expressing both PYK2 and FAK. Furthermore, PYK2 can functionally compensate for the loss of FAK in some cell types [88]. The adaptive capacity of cells to switch to PYK2-dependent signaling after deletion or kinase inhibition of FAK is evidence for some degree of signaling redundancy between the two protein tyrosine kinases. In fact, global deletion of the PTK2b gene that encodes PYK2 produces viable offspring without obvious defects in cardiovascular development [54], indicating that FAK may also compensate for the loss of PYK2 in most cell types.

Despite these caveats, PYK2 appears to serve a limited number of specific functions in cardiomyocytes, especially with respect to the recognition of, and response to a variety of stressful stimuli. Unlike FAK, PYK2 undergoes tyrosine phosphorylation during Ca2+ overload, UV irradiation, and H2O2 and TNF- treatment [83]. Hirotani et al. [29] demonstrated that PYK2 is an essential signaling component in endothelin- and phenylephrine-induced cardiomyocyte hypertrophy, perhaps acting via the Ca2+- and/or PKC-dependent activation of Rac1. Furthermore, recent studies have confirmed that PYK2 is an important upstream regulator of the stress activated protein kinases (p38MAPK and JNK1/2) in cardiomyocytes [49,28,22]. Thus, PYK2 activation has been implicated in hypertrophic gene expression changes during pathological cardiomyocyte hypertrophy [28,26] and in the induction of apoptosis [49].

Regulated PYK2 expression in cardiomyocytes

Unlike FAK, which appears to be constitutively expressed in most cell types, PYK2 expression also appears to vary in response to intracellular and extracellular stimuli. Although PYK2 is highly expressed in the neonatal cardiomyocyte, PYK2 expression in the adult heart is substantially reduced, but undergoes up-regulation in response to cardiomyocyte stress. The mechanisms underlying the regulated expression of PYK2 in cardiomyocytes and other cells are not known. As indicated above, embryonic fibroblasts isolated from FAK-knockout mice demonstrate a 2–3 fold increase in PYK2 expression levels, suggesting a compensatory role for PYK2 in maintaining integrin-dependent signaling events in FAK-deficient cells [77]. A similar increase in PYK2 expression and phosphorylation was observed in mice with conditional deletion of FAK in endothelial cells [88]. However, PYK2 levels did not increase with cardiomyocyte-restricted FAK deletion [14,61,24]. Nevertheless, our group was the first to demonstrate that PYK2 expression and phosphorylation were both significantly increased in cultured NRVM and adult rat ventricular myocytes (ARVM) by increases in [Ca2+]I and contractile activity [3], and in ARVM in vivo in response to acute LV pressure overload [5]. The increase in PYK2 levels was at least partly mediated by an increase in PYK2 mRNA [64]. Similarly, Melendez et al. [50] showed that PYK2 expression and PYK2 activity were substantially increased in a mouse model of dilated cardiomyopathy due to tropomodulin overexpression, and we have recently demonstrated that PYK2 is substantially up-regulated in the PKCε-overexpressing mouse during LV remodeling and HF [37]. Nevertheless, very little is known about the factors that regulate PYK2 expression in cardiomyocytes and other cell types.

PYK2-dependent signal transduction in pathological LV remodeling

The up-regulation and activation of PYK2 during cardiac hypertrophy and its transition to HF suggests that this focal adhesion kinase plays an important role in the pathogenesis of contractile dysfunction during cardiomyocyte stress. In general, PYK2 is considered a pro-apoptotic signaling molecule, in contradistinction to FAK, which has been shown to be pro-survival in many cell types. Indeed, our group has recently shown that adenovirally-mediated overexpression of CRNK, the C-terminal domain of PYK2, inhibits PYK2 activation in cardiomyocytes, improves LV contractile function in normal hearts, and improves LV fractional shortening, pathological changes in gene expression, and survival during post-MI ventricular remodeling [26]. These results are in stark contrast to those obtained by conditional deletion of FAK in adult cardiomyocytes, in which FAK depletion alone led to progressive LV dilatation and fibrosis [60] or the impairment of compensatory LVH in response to pressure overload [14]. Thus, FAK and PYK2 appear to have very different, opposing functions in cardiomyocytes.

Termination of FAK- and PYK2-dependent signaling in the heart

Activation of both FAK and PYK2 during mechanotransduction signaling is dependent on phosphorylation of specific tyrosine and serine residues, which either augment kinase activity, or induce conformational alterations that promote adaptor protein binding and release. Kinase activation, however, is transient, which suggests a role for cellular phosphatases in attenuating or terminating focal adhesion signaling. Two phosphatases, Shp2 and PTEN, have been implicated in this process.

PTEN is a dual specificity, lipid and protein phosphatase that is highly expressed in cardiomyocytes, and whose expression increases in response to hypertrophic stimuli [71]. PTEN is known to inhibit cardiomyocyte growth, in part via the dephosphorylation of FAK (and PYK2), and inhibition of downstream signaling to the PI(3)K-AKT signaling pathway. PTEN was shown by Seqqat et al. [74] to be activated in response to β1-adrenergic stimulation following acute volume overload in vivo, and by prolonged isoproterenol treatment in cultured cardiomyocytes. PTEN activation was accompanied by FAK and PYK2 dephosphorylation and the induction of apoptosis. In mice, muscle-specific knockout of PTEN resulted in basal hypertrophy and mild reduction in LV systolic function [55]. But in contrast to control mice, TAC in PTEN-knockout mice resulted in reduced pathological hypertrophy, less interstitial fibrosis, and reduced apoptosis with a marked preservation of LV function, indicating that loss of PTEN prevents the development of maladaptive ventricular remodeling in response to pathological biomechanical stress.

Shp2 is another FAK/PYK2 phosphatase, whose activity is reduced in response to biomechanical stress. Depletion of Shp2 by small interfering RNAs increased FAK phosphorylation and downstream focal adhesion signaling in cultured cardiomyocytes [48]. These findings demonstrate that basal Shp2 tyrosine phosphatase activity controls the size of cardiomyocytes by downregulating a pathway that involves FAK/Src and the PI(3)K-AKT-mTOR signaling pathways.

Speculation and future directions

The application of inducible gene excision technology to the study of focal adhesion signaling has already provided insight into the role of FAK and other components of the costamere in cardiomyocyte signal transduction. This approach, along with high-resolution fluorescence microscopy, should continue to provide important structure-function relationships between individual components of the cardiomyocyte costamere in health and disease. In contrast, the roles of PYK2 and PYK2-dependent signaling in HF progression remain largely unexplored, despite the fact that this kinase is up-regulated during pressure- and volume overload prior to the development of overt HF. Indeed, there is some controversy as to whether the kinase is cardioprotective in this setting, where it may protect the ventricular myocardium against tachyarrrhythmias during vagal stimulation [40]. However, this cardioprotective, electrophysiological effect was examined in global PYK2 knockout mice, where at least some of the deleterious effects of PYK2 deletion may have resulted from PYK2 ablation in noncardiac cell types, including neuronal, inflammatory and vascular cells which all express abundant amounts of PYK2. Inducible gene targeting in cardiomyoyctes should clarify this issue. FAK has already been considered a novel drug target for the development of small molecule inhibitors in the treatment of a variety of malignancies in which FAK overexpression is a common finding [33]. However, it remains to be determined whether FAK inhibition will result in significant cardiotoxicity in patients treated with these agents, as has been observed in patients treated with other tyrosine kinase inhibitors [9]. FAK inhibitors currently under evaluation also show significant inhibitory activity against PYK2, which will undoubtedly complicate the interpretation of toxicity studies. Conversely, specific small molecule inhibitors of PYK2 are not currently available, but might have utility in the treatment of HF if indeed PYK2 has deleterious effects during HF progression. Nevertheless, therapeutics aimed at focal adhesion signaling to prevent cardiac hypertrophy and HF may ultimately prove useful in reducing morbidity and mortality during LV remodeling.

Acknowledgments

The author is supported by NIH 2PO1 HL062426. The author also gratefully acknowledges the support of the Dr. Ralph and Marian Falk Medical Research Trust.

References

  • 1.Anthis NJ, Haling JR, Oxley CL, Memo M, Wegener KL, Lim CJ, Ginsberg MH, Campbell ID. Beta integrin tyrosine phosphorylation is a conserved mechanism for regulating talin-induced integrin activation. J Biol Chem. 2009;284 (52):36700–36710. doi: 10.1074/jbc.M109.061275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Anthis NJ, Wegener KL, Ye F, Kim C, Goult BT, Lowe ED, Vakonakis I, Bate N, Critchley DR, Ginsberg MH, Campbell ID. The structure of an integrin/talin complex reveals the basis of inside-out signal transduction. EMBO J. 2009;28 (22):3623–3632. doi: 10.1038/emboj.2009.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Bayer AL, Ferguson AG, Lucchesi PA, Samarel AM. Pyk2 expression and phosphorylation in neonatal and adult cardiomyocytes. J Mol Cell Cardiol. 2001;33 (5):1017–1030. doi: 10.1006/jmcc.2001.1369. [DOI] [PubMed] [Google Scholar]
  • 4.Bayer AL, Heidkamp MC, Howes AL, Heller Brown J, Byron KL, Samarel AM. Protein kinase Cε-dependent activation of proline-rich tyrosine kinase 2 in neonatal rat ventricular myocytes. J Mol Cell Cardiol. 2003;35 (9):1121–1133. doi: 10.1016/s0022-2828(03)00228-1. [DOI] [PubMed] [Google Scholar]
  • 5.Bayer AL, Heidkamp MC, Patel N, Porter MJ, Engman SJ, Samarel AM. PYK2 expression and phosphorylation increases in pressure overload-induced left ventricular hypertrophy. Am J Physiol Heart Circ Physiol. 2002;283 (2):H695–706. doi: 10.1152/ajpheart.00021.2002. [DOI] [PubMed] [Google Scholar]
  • 6.Blaukat A, Ivankovic-Dikic I, Gronroos E, Dolfi F, Tokiwa G, Vuori K, Dikic I. Adaptor proteins Grb2 and Crk couple Pyk2 with activation of specific mitogen-activated protein kinase cascades. J Biol Chem. 1999;274 (21):14893–14901. doi: 10.1074/jbc.274.21.14893. [DOI] [PubMed] [Google Scholar]
  • 7.Burchfield JS, Xie M, Hill JA. Pathological ventricular remodeling: mechanisms: part 1 of 2. Circulation. 2013;128 (4):388–400. doi: 10.1161/CIRCULATIONAHA.113.001878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Campbell ID, Ginsberg MH. The talin-tail interaction places integrin activation on FERM ground. Trends Biochem Sci. 2004;29 (8):429–435. doi: 10.1016/j.tibs.2004.06.005. [DOI] [PubMed] [Google Scholar]
  • 9.Chen MH, Kerkela R, Force T. Mechanisms of cardiac dysfunction associated with tyrosine kinase inhibitor cancer therapeutics. Circulation. 2008;118 (1):84–95. doi: 10.1161/CIRCULATIONAHA.108.776831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Chu M, Iyengar R, Koshman YE, Kim T, Russell B, Martin JL, Heroux AL, Robia SL, Samarel AM. Serine-910 phosphorylation of focal adhesion kinase is critical for sarcomere reorganization in cardiomyocyte hypertrophy. Cardiovasc Res. 2011;92 (3):409–419. doi: 10.1093/cvr/cvr247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Conti FJ, Monkley SJ, Wood MR, Critchley DR, Muller U. Talin 1 and 2 are required for myoblast fusion, sarcomere assembly and the maintenance of myotendinous junctions. Development. 2009;136 (21):3597–3606. doi: 10.1242/dev.035857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Corsi JM, Rouer E, Girault JA, Enslen H. Organization and post-transcriptional processing of focal adhesion kinase gene. BMC Genomics. 2006;7:198. doi: 10.1186/1471-2164-7-198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Dabiri GA, Turnacioglu KK, Sanger JM, Sanger JW. Myofibrillogenesis visualized in living embryonic cardiomyocytes. Proc Natl Acad Sci U S A. 1997;94 (17):9493–9498. doi: 10.1073/pnas.94.17.9493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.DiMichele LA, Doherty JT, Rojas M, Beggs HE, Reichardt LF, Mack CP, Taylor JM. Myocyte-restricted focal adhesion kinase deletion attenuates pressure overload-induced hypertrophy. Circ Res. 2006;99 (6):636–645. doi: 10.1161/01.RES.0000240498.44752.d6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Domingos PP, Fonseca PM, Nadruz W, Jr, Franchini KG. Load-induced focal adhesion kinase activation in the myocardium: role of stretch and contractile activity. Am J Physiol Heart Circ Physiol. 2002;282 (2):H556–564. doi: 10.1152/ajpheart.00534.2001. [DOI] [PubMed] [Google Scholar]
  • 16.Eble DM, Strait JB, Govindarajan G, Lou J, Byron KL, Samarel AM. Endothelin-induced cardiac myocyte hypertrophy: role for focal adhesion kinase. Am J Physiol Heart Circ Physiol. 2000;278 (5):H1695–1707. doi: 10.1152/ajpheart.2000.278.5.H1695. [DOI] [PubMed] [Google Scholar]
  • 17.Ervasti JM. Costameres: the Achilles’ heel of Herculean muscle. J Biol Chem. 2003;278 (16):13591–13594. doi: 10.1074/jbc.R200021200. [DOI] [PubMed] [Google Scholar]
  • 18.Fassler R, Rohwedel J, Maltsev V, Bloch W, Lentini S, Guan K, Gullberg D, Hescheler J, Addicks K, Wobus AM. Differentiation and integrity of cardiac muscle cells are impaired in the absence of β1 integrin. J Cell Sci. 1996;109 (Pt 13):2989–2999. doi: 10.1242/jcs.109.13.2989. [DOI] [PubMed] [Google Scholar]
  • 19.Frame MC, Patel H, Serrels B, Lietha D, Eck MJ. The FERM domain: organizing the structure and function of FAK. Nat Rev Mol Cell Biol. 2010;11 (11):802–814. doi: 10.1038/nrm2996. [DOI] [PubMed] [Google Scholar]
  • 20.Franchini KG, Torsoni AS, Soares PH, Saad MJ. Early activation of the multicomponent signaling complex associated with focal adhesion kinase induced by pressure overload in the rat heart. Circ Res. 2000;87 (7):558–565. doi: 10.1161/01.res.87.7.558. [DOI] [PubMed] [Google Scholar]
  • 21.Gopalan SM, Flaim C, Bhatia SN, Hoshijima M, Knoell R, Chien KR, Omens JH, McCulloch AD. Anisotropic stretch-induced hypertrophy in neonatal ventricular myocytes micropatterned on deformable elastomers. Biotechnol Bioeng. 2003;81 (5):578–587. doi: 10.1002/bit.10506. [DOI] [PubMed] [Google Scholar]
  • 22.Guo J, Sabri A, Elouardighi H, Rybin V, Steinberg SF. Alpha1-adrenergic receptors activate AKT via a Pyk2/PDK-1 pathway that is tonically inhibited by novel protein kinase C isoforms in cardiomyocytes. Circ Res. 2006;99 (12):1367–1375. doi: 10.1161/01.RES.0000252830.01581.fd. [DOI] [PubMed] [Google Scholar]
  • 23.Hagel M, George EL, Kim A, Tamimi R, Opitz SL, Turner CE, Imamoto A, Thomas SM. The adaptor protein paxillin is essential for normal development in the mouse and is a critical transducer of fibronectin signaling. Mol Cell Biol. 2002;22 (3):901–915. doi: 10.1128/MCB.22.3.901-915.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hakim ZS, DiMichele LA, Doherty JT, Homeister JW, Beggs HE, Reichardt LF, Schwartz RJ, Brackhan J, Smithies O, Mack CP, Taylor JM. Conditional deletion of focal adhesion kinase leads to defects in ventricular septation and outflow tract alignment. Mol Cell Biol. 2007;27 (15):5352–5364. doi: 10.1128/MCB.00068-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hakim ZS, DiMichele LA, Rojas M, Meredith D, Mack CP, Taylor JM. FAK regulates cardiomyocyte survival following ischemia/reperfusion. J Mol Cell Cardiol. 2009;46 (2):241–248. doi: 10.1016/j.yjmcc.2008.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hart DL, Heidkamp MC, Iyengar R, Vijayan K, Szotek EL, Barakat JA, Leya M, Henze M, Scrogin K, Henderson KK, Samarel AM. CRNK gene transfer improves function and reverses the myosin heavy chain isoenzyme switch during post-myocardial infarction left ventricular remodeling. J Mol Cell Cardiol. 2008;45 (1):93–105. doi: 10.1016/j.yjmcc.2008.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Heidkamp MC, Bayer AL, Kalina JA, Eble DM, Samarel AM. GFP-FRNK disrupts focal adhesions and induces anoikis in neonatal rat ventricular myocytes. Circ Res. 2002;90 (12):1282–1289. doi: 10.1161/01.res.0000023201.41774.ea. [DOI] [PubMed] [Google Scholar]
  • 28.Heidkamp MC, Scully BT, Vijayan K, Engman SJ, Szotek EL, Samarel AM. PYK2 regulates SERCA2 gene expression in neonatal rat ventricular myocytes. Am J Physiol Cell Physiol. 2005;289 (2):C471–482. doi: 10.1152/ajpcell.00130.2005. [DOI] [PubMed] [Google Scholar]
  • 29.Hirotani S, Higuchi Y, Nishida K, Nakayama H, Yamaguchi O, Hikoso S, Takeda T, Kashiwase K, Watanabe T, Asahi M, Taniike M, Tsujimoto I, Matsumura Y, Sasaki T, Hori M, Otsu K. Ca2+-sensitive tyrosine kinase Pyk2/CAKβ-dependent signaling is essential for G-protein-coupled receptor agonist-induced hypertrophy. J Mol Cell Cardiol. 2004;36 (6):799–807. doi: 10.1016/j.yjmcc.2004.03.002. [DOI] [PubMed] [Google Scholar]
  • 30.Honda H, Oda H, Nakamoto T, Honda Z, Sakai R, Suzuki T, Saito T, Nakamura K, Nakao K, Ishikawa T, Katsuki M, Yazaki Y, Hirai H. Cardiovascular anomaly, impaired actin bundling and resistance to Src-induced transformation in mice lacking p130Cas. Nat Genet. 1998;19 (4):361–365. doi: 10.1038/1246. [DOI] [PubMed] [Google Scholar]
  • 31.Ilic D, Furuta Y, Kanazawa S, Takeda N, Sobue K, Nakatsuji N, Nomura S, Fujimoto J, Okada M, Yamamoto T. Reduced cell motility and enhanced focal adhesion contact formation in cells from FAK-deficient mice. Nature. 1995;377 (6549):539–544. doi: 10.1038/377539a0. [DOI] [PubMed] [Google Scholar]
  • 32.Ilic D, Kovacic B, McDonagh S, Jin F, Baumbusch C, Gardner DG, Damsky CH. Focal adhesion kinase is required for blood vessel morphogenesis. Circ Res. 2003;92 (3):300–307. doi: 10.1161/01.res.0000055016.36679.23. [DOI] [PubMed] [Google Scholar]
  • 33.Infante JR, Camidge DR, Mileshkin LR, Chen EX, Hicks RJ, Rischin D, Fingert H, Pierce KJ, Xu H, Roberts WG, Shreeve SM, Burris HA, Siu LL. Safety, pharmacokinetic, and pharmacodynamic phase I dose-escalation trial of PF-00562271, an inhibitor of focal adhesion kinase, in advanced solid tumors. J Clin Oncol. 2012;30 (13):1527–1533. doi: 10.1200/JCO.2011.38.9346. [DOI] [PubMed] [Google Scholar]
  • 34.Kanchanawong P, Shtengel G, Pasapera AM, Ramko EB, Davidson MW, Hess HF, Waterman CM. Nanoscale architecture of integrin-based cell adhesions. Nature. 2010;468 (7323):580–584. doi: 10.1038/nature09621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kodama H, Fukuda K, Takahashi T, Sano M, Kato T, Tahara S, Hakuno D, Sato T, Manabe T, Konishi F, Ogawa S. Role of EGF Receptor and Pyk2 in endothelin-1-induced ERK activation in rat cardiomyocytes. J Mol Cell Cardiol. 2002;34 (2):139–150. doi: 10.1006/jmcc.2001.1496. [DOI] [PubMed] [Google Scholar]
  • 36.Kohno T, Matsuda E, Sasaki H, Sasaki T. Protein-tyrosine kinase CAKβ/PYK2 is activated by binding Ca2+/calmodulin to FERM F2 α2 helix and thus forming its dimer. Biochem J. 2008;410 (3):513–523. doi: 10.1042/BJ20070665. [DOI] [PubMed] [Google Scholar]
  • 37.Koshman YE, Patel N, Chu M, Iyengar R, Kim T, Lewis W, Goldspink PH, Samarel AM. Mechanical load up-regulates CTGF gene expression in isolated cardiomyocytes and in animal models of heart failure. J Mol Cell Cardiol. 2012;53:S18. (abstract) [Google Scholar]
  • 38.Kovacic-Milivojevic B, Roediger F, Almeida EA, Damsky CH, Gardner DG, Ilic D. Focal adhesion kinase and p130Cas mediate both sarcomeric organization and activation of genes associated with cardiac myocyte hypertrophy. Mol Biol Cell. 2001;12 (8):2290–2307. doi: 10.1091/mbc.12.8.2290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kuppuswamy D, Kerr C, Narishige T, Kasi VS, Menick DR, Cooper Gt. Association of tyrosine-phosphorylated c-Src with the cytoskeleton of hypertrophying myocardium. J Biol Chem. 1997;272 (7):4500–4508. doi: 10.1074/jbc.272.7.4500. [DOI] [PubMed] [Google Scholar]
  • 40.Lang D, Glukhov AV, Efimova T, Efimov IR. Role of Pyk2 in cardiac arrhythmogenesis. Am J Physiol Heart Circ Physiol. 2011;301 (3):H975–983. doi: 10.1152/ajpheart.00241.2011. [DOI] [PubMed] [Google Scholar]
  • 41.Laser M, Willey CD, Jiang W, Cooper Gt, Menick DR, Zile MR, Kuppuswamy D. Integrin activation and focal complex formation in cardiac hypertrophy. J Biol Chem. 2000;275 (45):35624–35630. doi: 10.1074/jbc.M006124200. [DOI] [PubMed] [Google Scholar]
  • 42.Lele TP, Thodeti CK, Pendse J, Ingber DE. Investigating complexity of protein-protein interactions in focal adhesions. Biochem Biophys Res Commun. 2008;369 (3):929–934. doi: 10.1016/j.bbrc.2008.02.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Li R, Wu Y, Manso AM, Gu Y, Liao P, Israeli S, Yajima T, Nguyen U, Huang MS, Dalton ND, Peterson KL, Ross RS. β1 integrin gene excision in the adult murine cardiac myocyte causes defective mechanical and signaling responses. Am J Pathol. 2012;180 (3):952–962. doi: 10.1016/j.ajpath.2011.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lietha D, Cai X, Ceccarelli DF, Li Y, Schaller MD, Eck MJ. Structural basis for the autoinhibition of focal adhesion kinase. Cell. 2007;129 (6):1177–1187. doi: 10.1016/j.cell.2007.05.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lim Y, Han I, Jeon J, Park H, Bahk YY, Oh ES. Phosphorylation of focal adhesion kinase at tyrosine 861 is crucial for Ras transformation of fibroblasts. J Biol Chem. 2004;279 (28):29060–29065. doi: 10.1074/jbc.M401183200. [DOI] [PubMed] [Google Scholar]
  • 46.Manso AM, Li R, Monkley SJ, Cruz NM, Ong S, Lao DH, Koshman YE, Gu Y, Peterson KL, Chen J, Abel ED, Samarel AM, Critchley DR, Ross RS. Talin1 has unique expression versus talin 2 in the heart and modifies the hypertrophic response to pressure overload. J Biol Chem. 2013;288 (6):4252–4264. doi: 10.1074/jbc.M112.427484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mansour H, de Tombe PP, Samarel AM, Russell B. Restoration of resting sarcomere length after uniaxial static strain is regulated by protein kinase Cε and focal adhesion kinase. Circ Res. 2004;94 (5):642–649. doi: 10.1161/01.RES.0000121101.32286.C8. [DOI] [PubMed] [Google Scholar]
  • 48.Marin TM, Clemente CF, Santos AM, Picardi PK, Pascoal VD, Lopes-Cendes I, Saad MJ, Franchini KG. Shp2 negatively regulates growth in cardiomyocytes by controlling focal adhesion kinase/Src and mTOR pathways. Circ Res. 2008;103 (8):813–824. doi: 10.1161/CIRCRESAHA.108.179754. [DOI] [PubMed] [Google Scholar]
  • 49.Melendez J, Turner C, Avraham H, Steinberg SF, Schaefer E, Sussman MA. Cardiomyocyte apoptosis triggered by RAFTK/Pyk2 via Src kinase is antagonized by paxillin. J Biol Chem. 2004;279 (51):53516–53523. doi: 10.1074/jbc.M408475200. [DOI] [PubMed] [Google Scholar]
  • 50.Melendez J, Welch S, Schaefer E, Moravec CS, Avraham S, Avraham H, Sussman MA. Activation of Pyk2/Related focal adhesion tyrosine kinase and focal adhesion kinase in cardiac remodeling. J Biol Chem. 2002;277 (47):45203–45210. doi: 10.1074/jbc.M204886200. [DOI] [PubMed] [Google Scholar]
  • 51.Mitra SK, Hanson DA, Schlaepfer DD. Focal adhesion kinase: in command and control of cell motility. Nat Rev Mol Cell Biol. 2005;6 (1):56–68. doi: 10.1038/nrm1549. [DOI] [PubMed] [Google Scholar]
  • 52.Mitra SK, Schlaepfer DD. Integrin-regulated FAK-Src signaling in normal and cancer cells. Curr Opin Cell Biol. 2006;18 (5):516–523. doi: 10.1016/j.ceb.2006.08.011. [DOI] [PubMed] [Google Scholar]
  • 53.Nadruz W, Jr, Corat MA, Marin TM, Guimaraes Pereira GA, Franchini KG. Focal adhesion kinase mediates MEF2 and c-Jun activation by stretch: role in the activation of the cardiac hypertrophic genetic program. Cardiovasc Res. 2005;68:87–97. doi: 10.1016/j.cardiores.2005.05.011. [DOI] [PubMed] [Google Scholar]
  • 54.Okigaki M, Davis C, Falasca M, Harroch S, Felsenfeld DP, Sheetz MP, Schlessinger J. Pyk2 regulates multiple signaling events crucial for macrophage morphology and migration. Proc Natl Acad Sci USA. 2003;100 (19):10740–10745. doi: 10.1073/pnas.1834348100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Oudit GY, Kassiri Z, Zhou J, Liu QC, Liu PP, Backx PH, Dawood F, Crackower MA, Scholey JW, Penninger JM. Loss of PTEN attenuates the development of pathological hypertrophy and heart failure in response to biomechanical stress. Cardiovasc Res. 2008;78 (3):505–514. doi: 10.1093/cvr/cvn041. [DOI] [PubMed] [Google Scholar]
  • 56.Pandey P, Avraham S, Kumar S, Nakazawa A, Place A, Ghanem L, Rana A, Kumar V, Majumder PK, Avraham H, Davis RJ, Kharbanda S. Activation of p38 mitogen-activated protein kinase by PYK2/related adhesion focal tyrosine kinase-dependent mechanism. J Biol Chem. 1999;274 (15):10140–10144. doi: 10.1074/jbc.274.15.10140. [DOI] [PubMed] [Google Scholar]
  • 57.Pardo JV, Siliciano JD, Craig SW. A vinculin-containing cortical lattice in skeletal muscle: transverse lattice elements (“costameres”) mark sites of attachment between myofibrils and sarcolemma. Proc Natl Acad Sci USA. 1983;80 (4):1008–1012. doi: 10.1073/pnas.80.4.1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Pardo JV, Siliciano JD, Craig SW. Vinculin is a component of an extensive network of myofibril-sarcolemma attachment regions in cardiac muscle fibers. J Cell Biol. 1983;97 (4):1081–1088. doi: 10.1083/jcb.97.4.1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Park SY, Avraham HK, Avraham S. RAFTK/Pyk2 activation is mediated by transacting autophosphorylation in a Src-independent manner. J Biol Chem. 2004;279 (32):33315–33322. doi: 10.1074/jbc.M313527200. [DOI] [PubMed] [Google Scholar]
  • 60.Peng X, Kraus MS, Wei H, Shen TL, Pariaut R, Alcaraz A, Ji G, Cheng L, Yang Q, Kotlikoff MI, Chen J, Chien K, Gu H, Guan JL. Inactivation of focal adhesion kinase in cardiomyocytes promotes eccentric cardiac hypertrophy and fibrosis in mice. J Clin Invest. 2006;116 (1):217–227. doi: 10.1172/JCI24497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Peng X, Wu X, Druso JE, Wei H, Park AY, Kraus MS, Alcaraz A, Chen J, Chien S, Cerione RA, Guan JL. Cardiac developmental defects and eccentric right ventricular hypertrophy in cardiomyocyte focal adhesion kinase (FAK) conditional knockout mice. Proc Natl Acad Sci USA. 2008;105 (18):6638–6643. doi: 10.1073/pnas.0802319105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Perricone AJ, Bivona BJ, Jackson FR, Vander Heide RS. Conditional knockout of myocyte ocal adhesion kinase abrogates ischemic preconditioning in adult murine hearts. J Am Heart Assoc. 2013;2 (5):e000457. doi: 10.1161/JAHA.113.000457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Pham CG, Harpf AE, Keller RS, Vu HT, Shai SY, Loftus JC, Ross RS. Striated muscle-specific β1D-integrin and FAK are involved in cardiac myocyte hypertrophic response pathway. Am J Physiol Heart Circ Physiol. 2000;279 (6):H2916–2926. doi: 10.1152/ajpheart.2000.279.6.H2916. [DOI] [PubMed] [Google Scholar]
  • 64.Porter MJ, Bayer AL, Samarel AM. Regulation of PYK2 gene expression during cardiomyocyte hypertrophy. J Mol Cell Cardiol. 2004;34 (7):A11. (abstract) [Google Scholar]
  • 65.Sabri A, Rafiq K, Seqqat R, Kolpakov MA, Dillon R, Dell’italia LJ. Sympathetic activation causes focal adhesion signaling alteration in early compensated volume overload attributable to isolated mitral regurgitation in the dog. Circ Res. 2008;102 (9):1127–1136. doi: 10.1161/CIRCRESAHA.107.163642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Samarel AM. Costameres, focal adhesions, and cardiomyocyte mechanotransduction. Am J Physiol Heart Circ Physiol. 2005;289 (6):H2291–H2301. doi: 10.1152/ajpheart.00749.2005. [DOI] [PubMed] [Google Scholar]
  • 67.Sanger JW, Wang J, Fan Y, White J, Sanger JM. Assembly and dynamics of myofibrils. J Biomed Biotechnol. 2010;2010:858606. doi: 10.1155/2010/858606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Schaller MD, Hildebrand JD, Shannon JD, Fox JW, Vines RR, Parsons JT. Autophosphorylation of the focal adhesion kinase, pp125FAK, directs SH2-dependent binding of pp60src. Mol Cell Biol. 1994;14 (3):1680–1688. doi: 10.1128/mcb.14.3.1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Schlaepfer DD, Mitra SK, Ilic D. Control of motile and invasive cell phenotypes by focal adhesion kinase. Biochim Biophys Acta. 2004;1692 (2–3):77–102. doi: 10.1016/j.bbamcr.2004.04.008. [DOI] [PubMed] [Google Scholar]
  • 70.Schwartz MA. Cell biology. The force is with us. Science. 2009;323 (5914):588–589. doi: 10.1126/science.1169414. [DOI] [PubMed] [Google Scholar]
  • 71.Schwartzbauer G, Robbins J. The tumor suppressor gene PTEN can regulate cardiac hypertrophy and survival. J Biol Chem. 2001;276 (38):35786–35793. doi: 10.1074/jbc.M102479200. [DOI] [PubMed] [Google Scholar]
  • 72.Senetar MA, Moncman CL, McCann RO. Talin2 is induced during striated muscle differentiation and is targeted to stable adhesion complexes in mature muscle. Cell Motil Cytoskeleton. 2007;64 (3):157–173. doi: 10.1002/cm.20173. [DOI] [PubMed] [Google Scholar]
  • 73.Senyo SE, Koshman YE, Russell B. Stimulus interval, rate and direction differentially regulate phosphorylation for mechanotransduction in neonatal cardiac myocytes. FEBS Lett. 2007;581 (22):4241–4247. doi: 10.1016/j.febslet.2007.07.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Seqqat R, Guo X, Rafiq K, Kolpakov MA, Guo J, Koch WJ, Houser SR, Dell’italia LJ, Sabri A. Beta1-adrenergic receptors promote focal adhesion signaling downregulation and myocyte apoptosis in acute volume overload. J Mol Cell Cardiol. 2012;53 (2):240–249. doi: 10.1016/j.yjmcc.2012.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Shai SY, Harpf AE, Babbitt CJ, Jordan MC, Fishbein MC, Chen J, Omura M, Leil TA, Becker KD, Jiang M, Smith DJ, Cherry SR, Loftus JC, Ross RS. Cardiac myocyte-specific excision of the β1 integrin gene results in myocardial fibrosis and cardiac failure. Circ Res. 2002;90 (4):458–464. doi: 10.1161/hh0402.105790. [DOI] [PubMed] [Google Scholar]
  • 76.Sharp WW, Simpson DG, Borg TK, Samarel AM, Terracio L. Mechanical forces regulate focal adhesion and costamere assembly in cardiac myocytes. Am J Physiol. 1997;273 (2 Pt 2):H546–H556. doi: 10.1152/ajpheart.1997.273.2.H546. [DOI] [PubMed] [Google Scholar]
  • 77.Sieg DJ, Ilic D, Jones KC, Damsky CH, Hunter T, Schlaepfer DD. Pyk2 and Src-family protein-tyrosine kinases compensate for the loss of FAK in fibronectin-stimulated signaling events but Pyk2 does not fully function to enhance FAK-cell migration. EMBO J. 1998;17 (20):5933–5947. doi: 10.1093/emboj/17.20.5933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Simpson DG, Majeski M, Borg TK, Terracio L. Regulation of cardiac myocyte protein turnover and myofibrillar structure in vitro by specific directions of stretch. Circ Res. 1999;85 (10):e59–69. doi: 10.1161/01.res.85.10.e59. [DOI] [PubMed] [Google Scholar]
  • 79.Stout AL, Wang J, Sanger JM, Sanger JW. Tracking changes in Z-band organization during myofibrillogenesis with FRET imaging. Cell Motil Cytoskeleton. 2008;65 (5):353–367. doi: 10.1002/cm.20265. [DOI] [PubMed] [Google Scholar]
  • 80.Tadokoro S, Shattil SJ, Eto K, Tai V, Liddington RC, de Pereda JM, Ginsberg MH, Calderwood DA. Talin binding to integrin beta tails: a final common step in integrin activation. Science. 2003;302 (5642):103–106. doi: 10.1126/science.1086652. [DOI] [PubMed] [Google Scholar]
  • 81.Taniyama Y, Weber DS, Rocic P, Hilenski L, Akers ML, Park J, Hemmings BA, Alexander RW, Griendling KK. Pyk2- and Src-dependent tyrosine phosphorylation of PDK1 regulates focal adhesions. Mol Cell Biol. 2003;23 (22):8019–8029. doi: 10.1128/MCB.23.22.8019-8029.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Taylor JM, Rovin JD, Parsons JT. A role for focal adhesion kinase in phenylephrine-induced hypertrophy of rat ventricular cardiomyocytes. J Biol Chem. 2000;275 (25):19250–19257. doi: 10.1074/jbc.M909099199. [DOI] [PubMed] [Google Scholar]
  • 83.Tokiwa G, Dikic I, Lev S, Schlessinger J. Activation of Pyk2 by stress signals and coupling with JNK signaling pathway. Science. 1996;273 (5276):792–794. doi: 10.1126/science.273.5276.792. [DOI] [PubMed] [Google Scholar]
  • 84.Torsoni AS, Constancio SS, Nadruz W, Jr, Hanks SK, Franchini KG. Focal adhesion kinase is activated and mediates the early hypertrophic response to stretch in cardiac myocytes. Circ Res. 2003;93 (2):140–147. doi: 10.1161/01.RES.0000081595.25297.1B. [DOI] [PubMed] [Google Scholar]
  • 85.Torsoni AS, Fonseca PM, Crosara-Alberto DP, Franchini KG. Early activation of p160ROCK by pressure overload in rat heart. Am J Physiol Cell Physiol. 2003;284 (6):C1411–C1419. doi: 10.1152/ajpcell.00098.2002. [DOI] [PubMed] [Google Scholar]
  • 86.Wang J, Shaner N, Mittal B, Zhou Q, Chen J, Sanger JM, Sanger JW. Dynamics of Z-band based proteins in developing skeletal muscle cells. Cell Motil Cytoskeleton. 2005;61 (1):34–48. doi: 10.1002/cm.20063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wei H, Vander Heide RS. Ischemic preconditioning and heat shock activate Akt via a focal adhesion kinase-mediated pathway in Langendorff-perfused adult rat hearts. Am J Physiol Heart Circ Physiol. 2010;298 (1):H152–157. doi: 10.1152/ajpheart.00613.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Weis SM, Lim ST, Lutu-Fuga KM, Barnes LA, Chen XL, Gothert JR, Shen TL, Guan JL, Schlaepfer DD, Cheresh DA. Compensatory role for Pyk2 during angiogenesis in adult mice lacking endothelial cell FAK. J Cell Biol. 2008;181 (1):43–50. doi: 10.1083/jcb.200710038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Xu W, Baribault H, Adamson ED. Vinculin knockout results in heart and brain defects during embryonic development. Development. 1998;125 (2):327–337. doi: 10.1242/dev.125.2.327. [DOI] [PubMed] [Google Scholar]
  • 90.Ye F, Hu G, Taylor D, Ratnikov B, Bobkov AA, McLean MA, Sligar SG, Taylor KA, Ginsberg MH. Recreation of the terminal events in physiological integrin activation. J Cell Biol. 2010;188 (1):157–175. doi: 10.1083/jcb.200908045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Yi XP, Zhou J, Huber L, Qu J, Wang X, Gerdes AM, Li F. Nuclear compartmentalization of FAK and FRNK in cardiac myocytes. Am J Physiol Heart Circ Physiol. 2006;290 (6):H2509–2515. doi: 10.1152/ajpheart.00659.2005. [DOI] [PubMed] [Google Scholar]
  • 92.Zemljic-Harpf A, Manso AM, Ross RS. Vinculin and talin: focus on the myocardium. J Investig Med. 2009;57 (8):849–855. doi: 10.231/JIM.0b013e3181c5e074. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES