Abstract
Protein kinase A (PKA) is a basophilic kinase implicated in the modulation of many cell-signaling and physiological processes. PKA also contributes to cancer-relevant events such as growth factor action, cell cycle control, cell migration and tumor metabolism. Germline and somatic mutations in PKA, gene amplifications, and chromosome rearrangements that encode kinase fusions, are linked to a growing number of malignant neoplasms. Mislocalization of PKA by exclusion from A-Kinase Anchoring Protein (AKAP) signaling islands further underlies cancer progression. This article highlights the influence of AKAP signaling and local kinase action in selected hallmarks of cancer. We also feature the utility of kinase inhibitor drugs as frontline and future anti-cancer therapies.
Introduction
Protein kinases are critical mediators of cell division, oncogenesis and tumor progression [1]. This was first recognized by Hunter, Erickson, Brugge and others when they designated modified forms of the Src tyrosine kinase as an oncogene [2,3]. Soon after, Baltimore and colleagues discovered that most patients with chronic myeloid leukemia express a tyrosine kinase fusion called BCR-Abl [4,5]. This set the stage for Drucker, Sawyers and Lydon to develop imatinib (then called Gleevec) as the first targeted cancer kinase inhibitor drug [6,7]. Currently, there are ~ 185 orally effective protein kinase inhibitors that are prescribed or in clinical trials worldwide [8]. While most of these compounds are used to treat different cancer types, certain of these small molecules are therapies for other pathologies including rheumatoid arthritis, macular degeneration and certain forms of pulmonary fibrosis [9,10]. Kinase inhibitors are now a cornerstone of anti-cancer therapy. The use of these agents is exemplified in the first line approval of osimertinib, a third generation EGFR inhibitor that is used to treat non-small-cell lung carcinomas with specific mutations in this kinase ([11,12] Figure 1A).
Figure 1. Anchored kinases and pathological signaling.

(A) Receptor tyrosine kinases (RTKs) contain an intracellular kinase domain that anchors Growth factor receptor bound protein 2 (Grb2) via the Grb2 SH2 domain. The N terminal SH3 domain of Grb2 interacts with Son of Sevenless (SOS), a Ras-guanine exchange factor. This interaction results in the catalysis of Ras from an inactive to active form, resulting in the activation of downstream MAPK signaling (both indicated). Kinase inhibitor drug Osimertinib targets tumors with activating EGFR mutations through irreversibly binding to the Cys797 residue. This results in inhibition of Ras and downstream ERK signaling. (B) G protein coupled receptors (GPCRs) are localized to the plasma membrane and mobilize heterotrimeric G proteins to catalyze production of cAMP by adenylyl cyclases. A-kinase anchoring proteins (AKAPs) constrain PKA within local pools of cAMP to elicit downstream responses. (Inset) Several points of this cascade are implicated in pathological signaling in the context of cancer. (1) Overstimulation of GPCRs, (2) point mutations in the stimulatory G protein GNAS, (3) amplification of downstream kinase signaling through aberrant PKA signaling, (4) inactivation of the autoinhibitory domains of PKA regulatory subunits, and (5) mutations to PKAc that impact association with AKAPs to mis-localize the kinase. (C) In fibrolamellar carcinoma, a genetic deletion results in the fusion PKAc with the J domain of Hsp40, denoted DNAJ. This results in mis-localization from AKAPs and pathological signaling. (D) A gene fusion of exons 1–8 of AKAP9 with exons 9-18 BRAF results in elevated kinase activity and subsequent thyroid cancer. (Created in BioRender. Rosenthal, K. (2024) BioRender.com/b49b923).
While serine and threonine protein kinases are less often considered to be oncogenic, some of these enzymes are cancer drivers. Targetable oncogenic BRAF mutations are frequently seen in melanoma and in smaller subsets of RAS/MAPK-driven cancers [13]. Rare oncogenic alleles of AKT (also known as protein kinase B) are seen in tumors with activation of PI-3 kinase [14]. Serine/threonine kinases have broader tissue activities. Consequently, these enzymes often exhibit more challenging toxicity profiles for their inhibitors than drugs targeting tyrosine kinases. A subset of calcium/phospholipid dependent ‘protein kinase C’ (PKC) isoforms contribute to cancer progression [15,16]. Yet confoundingly, some inactivating mutations in atypical PKC’s are cancer-associated [17]. This latter finding argues a tumor suppressive role for certain PKC isoforms [17,18]. Thus, PKC’s role in oncogenesis are complex and context specific. The same is true for the kinase family featured in this article protein kinase A (PKA) [19].
Local PKA signaling
PKA is a broad specificity kinase that is activated by G protein-coupled receptor (GPCR) mediated synthesis of intracellular cAMP ([20], Figure 1B). Although classically considered a metabolic enzyme, PKA co-ordinates a host of vital physiological functions such as cardiac contractility, glycogenolysis, hormone release, and a variety of mitochondrial functions. The human genome contains three genes encoding catalytic subunits (Cα, Cβ, and Cγ), and four genes encoding the regulatory subunits (RIα, RIβ, RIIα and RIIβ) [21,22]. The dimeric regulatory subunits form the core of the intact PKA holoenzyme. A revised view of cAMP activation suggests that second messenger binding to R subunits loosens association with the C subunits to unleash kinase activity rather than liberating this enzyme from the tetrameric holoenzyme complex [23,24]. Hence, specificity in PKA action is fundamentally controlled though the spatial and temporal organization of the holoenzyme. This occurs via its association with A-kinase Anchoring proteins (AKAPs) ([25,26], Figure 1B). Genomic alterations in PKA subunits and AKAPs lead to constitutive activation or mis-localization of the kinase, particularly in cancers from tissues with endocrine or neuroendocrine histology [27].
As depicted in Figure 1B, oncogenic PKA action falls into five categories: (1) Overstimulation of GPCRs promotes tonic PKAc activity in thyroid cancer [28,29]. (2) Point mutations in the G protein α subunit GNAS are enriched in small cell lung cancer and other malignancies [30-33]. (3) Aberrant PKA signaling can phosphorylate downstream kinases such as Raf, GSK3 and FAK to modulate their activity in a variety of benign and malignant neoplasms. These events can have context dependent influence on the RAF-MEK-ERK mitogenic signaling cascade [34]. (4) Patients with germline inactivating mutations in the autoinhibitory regulatory subunit PKA-R1α are predisposed to myxomas, thyroid and gonadal tumors, collectively referred to as Carney Complex [35]. (5) Point mutations in PKAc that displace the active kinase from its usual subcellular sites of action underlie adrenocortical carcinoma [22,36], ACTH-producing pituitary tumors and adrenal Cushing’s syndrome [37-41].
Kinases that exist as gene fusions represent an important class of oncogenes [1]. These chimeric enzymes are produced by translocations or other chromosomal rearrangements. Their protein products represent ideal targets for the development of anti-cancer drugs [42]. Expression of a DNAJ-PKAc fusion kinase is a dominant oncogenic event in the adolescent liver cancer fibrolamellar carcinoma (FLC) ([40], Figure 1C). This gene fusion has been detected in >90% of FLC patients [40,43], with a few cases bearing deletions in the type Iα regulatory subunit (PKA-R1α) [44]. DNAJ-PKAc chimeras have also been described in oncocytic biliary tumors [45]. Genomic instability can also result in an in-frame fusion between AKAPs and active kinase domains. For example, an oncogenic AKAP9-BRAF fusion promotes abnormal MAPK pathway activation in thyroid cancer ([46], Figure 1D). Directing active BRAF to centrosomes via AKAP9 apportions undue influence on chromosomal segregation to impact mitosis [47,48]. Thus, aberrant partitioning of PKA supports a variety of hyperplasia’s and neoplasms.
Subcellular location is a critical determinant of PKA action. Originally thought of as a ‘cytoplasmic’ enzyme, a concerted research effort over the past 30 years has revealed that tethering to defined intracellular locations is a fundamental element of PKA action [49-51]. Three lines of inquiry have converged to show just how ‘local’ PKA action really is (Figure 1B). Elegant studies by Bock and colleagues have determined that intracellular cAMP accumulates within spherical nanodomains [52,53]. These tiny pools of second messenger form at their site of synthesis proximal to G-protein coupled receptor-Adenylyl cyclase complexes. Importantly, these receptor-associated independent cAMP nanodomains (RAINs) are insulated from second messenger pools generated in other intracellular compartments [52,53]. Von Zastrow, Irannejad and others have convincingly shown that GPCR-mediated cAMP generation not only occurs at the plasma membrane, but at intracellular endosomes, golgi, and other subcellular regions [54,55]. An implication of this latter discovery is that RAINs can be simultaneously generated at multiple intracellular sites in response to an individual extracellular stimulus. Thus, discrete islands of PKA activity transmit unique patterns of downstream signaling responses. Our group and others have been instrumental in defining this mechanism of local PKA action [23,24,56]. A collective of investigators have delineated a sophisticated molecular terrain where PKA holoenzymes are constrained via interaction with AKAPs ([57-62], Figure 1B).
Over 70 AKAPs have been identified in the human proteome [63,64]. Each anchoring protein contains a structurally conserved amphipathic helix that docks into a reciprocal binding furrow on the surface of the R subunit dimer [65,66]. Most of these anchoring proteins also constrain other protein kinases, phosphoprotein phosphatases, phosphodiesterases and GTPases to create multienzyme assemblies [67]. Unique targeting motifs confine AKAP complexes at defined subcellular sites to create autonomous signaling islands [21]. A central element of the ‘AKAP-signaling island’ model is that the active kinase remains bound to the AKAP-RII subcomplex and, hence, close to its site of action (Figure 1B). This led to a postulate that PKAc activity is constrained within AKAP-signaling islands rather than diffusing away from its site of activation within discrete RAINs [22,41]. Conversely, mislocalization of PKA via exclusion from AKAP signaling islands contributes to cancer progression.
Hallmarks of cancer
In 2002 and 2011 Hanahan and Weinberg published landmark reviews outlining basic characteristics of cancer cells [1,68]. The latest iteration published in 2022 lists fourteen criteria that encapsulate the complexity of tumorigenesis [69]. Whereas oncogenic tyrosine kinases typically promote hallmark cancer behaviors, the impact of PKA is more diverse across malignant phenotypes. This article highlights the contribution of local PKA action and AKAP signaling in selected hallmarks of cancer.
Proliferative signaling
Healthy cells adhere to a tightly controlled program of growth factor signaling to ensure fidelity in cell division. Cancer cells often gain proliferative advantage through autocrine production of growth factors or amplification of downstream pro-proliferation signals [68]. Paradoxically, cAMP can have direct and indirect effects on cell proliferation [70]. All intracellular effects of cAMP were originally thought to proceed solely through PKA phosphorylation [71]. It was subsequently recognized that EPAC guanine nucleotide exchange factors couple cAMP to the activation of Ras-like GTPases such as Rap1 [72]. Adding to this complexity, cAMP exerts pleotropic effects on the RAF-MEK-ERK kinase cascade. This triad of kinases regulates fundamental aspects of cell growth, survival and differentiation [73,74]. PKA phosphorylation and Rap1 GTPase activity attenuate BRAF to shut down ERK signaling [75]. Yet in other cellular contexts, cAMP mobilizes RAF-independent pathways that lead to MEK and ERK activation [76,77]. Thus, local production of cAMP can evoke context dependent effects based on cell type and intracellular location.
Several AKAPs sequester kinases to support cell proliferation [21]. AKAP12, also called Gravin, organizes PKA and PKC with the mitotic kinases Aurora A (AurA) and polo-like kinase 1 (Plk1) at centrosomes [48,78,79]. In glioblastoma, Gravin is phosphorylated at threonine 766 to prime the recruitment of Plk1 conferring its designation as an oncogene [80]. Depletion of Gravin or introduction of a T766A mutant suppresses cell proliferation and reduces aneuploidy [48]. Gravin has also been shown to direct G1 to S phase progression by regulating the stability and location of cyclin D [81]. In a more translational approach, the centrosomal targeting domain of Gravin was exploited to create genetically encoded platforms that restrain kinase inhibitor drugs [82]. Centrosomal delivery of clinical Plk1 and AurA inhibitors via this modified AKAP scaffold attenuates mitosis in a variety of cancer cell lines [82,83]. This versatile precision pharmacology tool has the capacity to enhance the investigation of local kinase biology in a variety of cellular models of cancer [25].
Other AKAPs co-ordinate cancer cell proliferation. AKAP4 is a potential cancer biomarker for colon, breast, cervical, non-small cell lung, and prostate cancers [84-87] . Gene silencing of AKAP4 results in decreased proliferation via down-regulation of cell cycle proteins [84]. In breast cancer cells, depletion of this anchoring protein causes cell cycle arrest at the G0/G1 transition with concomitant reductions in colony formation [88]. The protooncogene AKAP13/AKAP-Lbc brings together different protein kinases that propagate a range of proliferative responses [89-92]. This broadly distributed anchoring protein clusters PKA and protein kinase D (PKD) with the mitogen activated kinase scaffolding protein KSR-1 [93]. PKA phosphorylation of Serine 838 on KSR-1 promotes the assembly of the RAF-MEK-ERK cascade to favor cellular proliferation. This multikinase scaffold is also implicated as a proliferative factor in the rare adolescent liver cancer FLC [94].
Replicative immortality
Normal cells have a finite number of replications before they enter a non-proliferative state called senescence. Age induced telomere shortening of chromosomes induces senescence. This process is bypassed in cancer cells as telomerase, an enzyme that adds nucleotides to the ends of chromosomes, is abnormally active [95]. In parallel, cancer cells undergo genetic reprogramming that leads to a ‘transformed phenotype’ [96]. Oncogenic viruses, some of which stimulate kinases, drive cellular transformation allowing the formation of macroscopic tumors [69]. Combined, these factors give tumors a measure of immortality.
The multivalent anchoring protein AKAP12/Gravin has been implicated in control of replicative immortality. Disruption of Gravin signaling hubs is a common theme in many transformed cells as expression of this anchoring protein is suppressed by oncogenic src, ras, jun, myc, and fos [97-99]. The same investigators propose that loss of Gravin is postulated to contribute to a transformed phenotype in non-small cell lung cancer [100]. Other anchored kinases may contribute to cellular transformation as Gravin has the capacity to scaffold PKC and Plk-1 with src kinases [78,97,101]. These seemingly paradoxical findings emphasize the context specific and nature of AKAP signaling where secondary mutations may drive its effects on replicative immortality [57]. They also illustrate how combinatorial assembly of unique kinase-phosphatase combinations on the same AKAP can evoke distinct signaling outcomes [49].
Evading growth suppressors
Cancer cells often circumvent negative regulators of cell growth to achieve uncontrolled proliferation. This often proceeds through the deregulation of tumor suppressor genes such as p53, PTEN, Rb and Merlin [102,103]. Merlin participates in adherens junction assembly and mediates contact-dependent inhibition of EGFRs [104-106]. Merlin also preferentially anchors RIβ subunits of PKA in the brain [107-110]. Inactivating mutations in Merlin occur in patients with neurofibromatosis-type 2, a genetic condition that causes schwannomas, non-invasive glial cell tumors, to grow along nerves [105,111]. Strikingly, tissue-specific ablation of PKA RI in neural crest cells also produces schwannomas [112]. How anchored PKA participates in neurological disorder is unclear, but clustering of type I PKA at adherens junctions could favor cAMP responsive phosphorylation at sites of cell-cell contact.
In contrast, other AKAP signaling islands suppress cell growth. SSeCKS, a rodent ortholog of Gravin, is down-regulated in Ras transformed fibroblasts whereas SSeCKs knockout mice have enlarged prostates and focal dysplasia [113,114]. These effects may be tissue-specific, as subcutaneous implantation of human glioma cells expressing Gravin shRNA retards tumor growth [80]. Thus, paradoxically, this same anchoring protein has been reported to exert opposing effects on cancer cell growth. Again, this may be because multivalent anchoring proteins such as AKAP12/Gravin and AKAP79/150 anchor distinct kinase/phosphatase combinations in different cell or tumor types [78,115-118].
Promoting angiogenesis
Healthy cells recruit blood vessels to supply oxygen, nutrients, and export waste. Cancer cells must also acquire these same features. Consequently, tumors often induce angiogenesis to procure an expanded vasculature to facilitate growth. Pro-angiogenic factors such as VEGF and PDGF induce formation of new blood vessels by signaling to hypoxia inducible factors HIF-1α and HIF-1β. The scaffolding protein mAKAP binds to HIF-1α and components of the ubiquitin machinery to regulate the stability of this transcription factor in a bidirectional fashion [119]. Under normoxia, components of the mAKAP complex target HIF-1α for degradation. Yet under hypoxic conditions, mAKAP organizes factors that stabilize HIF-1α. The perinuclear location of this anchored kinase scaffold is required to position HIF-1α close to its target genes [119]. Thus, compartmentalization of oxygen-sensitive signaling components may influence the fidelity and magnitude of hypoxic responses [49,51].
The cytoskeletal anchoring protein AKAP2 interfaces with another transcriptional complex including steroid receptor co-activator 3 (Src3) and estrogen receptor alpha (ERα) to turn on pro-angiogenic genes [120]. It has been suggested that AKAP2 functions to localize PKA for phosphorylation of ERα to promote angiogenesis [120]. AKAP2 knockout mice exhibit decreased VEGF expression and impaired blood vessel formation. Conversely, AKAP2 is up-regulated in primary tumors suggesting that this signaling island boosts angiogenesis [121,122]. Likewise, AKAP4 complexes influence VEGF expression and angiogenesis in non-small cell lung cancer [123]. Collectively these findings suggest that several AKAP signaling islands are implicated in the molecular control of pro-angiogenic signaling pathways.
Resisting cell death
Another feature of tumors is circumventing cell death. Consequently many cancer therapeutics induce apoptosis, the Greek derivation of ‘falling off,’ as in leaves from a tree [124]. Apoptosis can be induced by chemotherapeutic agents that promote DNA damage [125]. Two major signaling pathways trigger apoptotic cell death: the mitochondrial (intrinsic) pathway and the death receptor (extrinsic) pathway [124]. PKA signaling to the BCL-2 family of proteins represses intrinsic cell death. The proapoptotic factor and PKA substrate Bcl-associated agonist of cell death (BAD) resides on the outer membranes of mitochondria [126]. Early studies implicated PKA phosphorylation via the mitochondrial anchoring protein dAKAP-1 as a means to inactivate BAD in response to survival factors [127].
Further evidence of an interface between anchored PKA and the apoptosis machinery comes from investigation of fibrolamellar hepatocellular carcinoma (FLC). This variant of hepatocellular carcinoma with distinctive fibrotic features typically afflicts patients between 15 to 35 years of age without previous history of liver disease [128,129]. Therapeutic intervention is challenging due to late-stage diagnosis and poor treatment responses. A defining feature of FLC is the expression of a fusion kinase denoted DNAJ-PKAc (Figure 2).
Figure 2. Oncogenic kinase signaling in fibrolamellar carcinoma.

(A) In normal livers, PKA is recruited to AKAP signaling islands. (B) In FLC, most of PKAc is fused with DNAJ, which facilitates the exclusion of the fusion kinase from AKAP signaling islands. The fusion recruits the co-chaperone BAG2 to the complex via Heat shock protein 70 (Hsp70). Recruitment of BAG2 to the complex activates Bcl2, which inhibits normal cell death pathways and contributes to aspects of FLC pathology including chemotherapeutic resistance. (Created in BioRender. Rosenthal, K. (2024) BioRender.com/s12j016).
This fusion protein results from a 400 kb deletion on chromosome 19 where the chaperone binding domain of heat shock protein 40 (Hsp40) is fused to exons 2–10 of the PKA catalytic subunit [40]. There are two plausible explanations as to how fusion of the J domain to the catalytic core of PKAc disseminates oncogenesis. It is possible that the substrate selectivity of DNAJ-PKAc fusions are ‘rewired’ thereby driving anomalous phosphorylation of oncogenic factors [40,130]. Alternatively, the chaperonin domain permits recruitment of chaperones such as heat shock protein 70 to augment the stability of the fusion kinase [20,94,131,132]. Unfortunately, direct targeting of DNAJ-PKAc with ATP analog inhibitors is not a therapeutic option. A recently reported inhibitor that achieves greater than >95% inhibition of PKAc is not tolerated in animals and failed phase 1 clinical trials [133,134]. Accordingly, potent and selective PKAc inhibitors such as BLU2864 have been relegated to the role of tool compounds rather than clinically viable drugs [25].
We have shown that release of DNAJ-PKAc from AKAP signaling islands is another contributing factor to FLC pathology [135]. Phosphoproteomic screening shows that mis-localization of the fusion kinase results in the detection of a distinct pattern of phosphoproteins including components of the protein synthesis machinery and several co-chaperones [131,135,136]. One prominent phosphoprotein is the Bcl-2 associated anthanogene 2 (BAG2), a co-chaperone recruited to scaffolds through association with its binding partner Hsp70 [135]. BAG2 is an athanogene that strengthens pro-survival phenotypes by reducing apoptosis in FLC tumors (Figure 2). This was supported by pharmacological evidence using the DNA damaging agent etoposide and the Bcl-2 inhibitor navitoclax. These drug studies showed that the DNAJ-PKAc/Hsp70/BAG2 axis contributes to chemotherapeutic resistance in a cellular model of FLC [135]. These studies not only implicate BAG2 as a marker for advanced FLC, but also suggest that recruitment of this athanogene may function as a chemotherapeutic resistance factor in DNAJ-PKAc signaling scaffolds (Figure 2).
This demonstrates that loss of AKAP signaling islands can have pathological consequences whereas, pharmacologically targeting Hsp70/BAG2 subcomplexes may be a key to unlock the challenge of overcoming chemotherapeutic resistance in FLC.
Evading immune surveillance
The immune system plays a central role in the recognition and destruction of neoplastic cells. T lymphocytes, helper T cells, and natural killer (NK) cells are key mediators of this process [137,138]. Tumor cells evade this process through a variety of intrinsic and local mechanisms of immune escape [139]. RIα has been implicated in suppressing the tumor surveillance machinery, in both innate and adaptive immunity, suggesting that anchoring of type 1 PKA holoenzymes can modulate immune cell function [140]. NK cells, including lymphokine activated killer (LAK) cells are members of the innate immune system that engage the lytic machinery to kill tumor cells [141-143]. Type I PKA activity is critical for adenosine or prostaglandin e2 induced inactivation cytotoxicity and cytokine release in these cells [144-146]. This phenomenon was also shown in the context of cancer, in which RIα inhibited the adenosine induced cytotoxic effect of NK cells against erythroleukemia and melanoma cells [147]. Other reports have implicated type I PKA in sphingosine 1 phosphate (S1P) and lysophosphatidic acid inhibition of NK cells in tumors [148,149]. These investigators utilized Rp-8-Br-cAMPS, a semi-selective type I PKA inhibitor, to attenuate immunosuppressive function in NK and LAK cells [145,148,149].
The tumor microenvironment (TME) is now recognized as a key mediator of tumor growth mechanisms and immune responses. This niche includes extracellular matrix components, cytokines, and various cell types that sustain tumor growth [150]. Macrophages populate the TME that and can have dual roles in the progression of tumors. This is due to the polarization of macrophages into M1 ( pro-inflammatory and anti-tumoral) and M2 (anti-inflammatory and tumor promoting) subtypes in the TME [151]. PKA signaling plays a central role in the switch from an M1 to an M2 phenotype. Tumors secrete signals that activate PKAc in tumor associated macrophages. PKAc acts on transcriptional reprogramming via CREB to promote an M2 phenotype to favor tumoral immune evasion [151]. Conversely, inhibition of PKA in tumor associated macrophages decreased growth and metastasis [152].
Like macrophages, neutrophils can have a context specific effect on tumor progression. Tumor associated neutrophils (TANs) are recruited to the TME and polarize into either type I or type 2 based on extracellular cues. Type I TANs (N1) have immune activating antitumoral activity, while type 2 TANs (N2) have a protumorigenic effect [153]. PKA signaling can be implicated in the recruitment of neutrophils to a tumor. For example, local PKA signaling has been shown to be important for neutrophil chemotaxis, which is necessary for the infiltration of neutrophils into the TME [154]. This is particularly relevant in colon cancer, where a cAMP-PKA-CREB signaling pathway results in carcinogenesis by enhancing inflammation in the TME and allowing infiltration of neutrophils [155].
CD4+ and CD8+ T cells are the principle effectors of adaptive anti-tumor machinery, and participate in cytokine induced recognition and lysis of tumor cells [156]. RIα has been shown to inhibit adenosine activated cytotoxicity and cytokine release in human antimelanoma CD4+ and CD8+ T cells [147]. Selectively blocking type I PKA with Rp-8-Br-cAMPS abrogated this affect [147]. In T cells the dual specificity AKAP ezrin clusters RIα with the C terminal srk kinase (Csk) at immunological synapses [157]. Disruption of PKA location via delivery of the anchoring disruptor peptide Ht31, or siRNA mediated depletion of Ezrin, inhibited T cell proliferation and IL-2 regulation [157]. These studies highlight the value of anchored PKA signaling in modulation of the immune response.
Epigenetic reprogramming
Epigenetic changes impact gene expression by influencing local chromatin structure and thus DNA accessibility to transcription factors. Hypermethylation, histone modification, accessibility of chromatin, or post translational modifications can impact the transcription of genes to favor tumorigenesis [158]. Thus, perhaps not surprisingly certain AKAPs support epigenetic changes that confer pathological advantage [64,159]. The histone deacetylase HDAC6 correlates with poor prognosis and high primary tumor grade in glioblastoma and ovarian cancers [160,161]. The altered acetylation of cytoskeletal components influences cell dynamics and motile phenotypes. HDAC6 is a component of the AKAP11/AKAP220 subcomplex at centrosomes and cilia [159,162]. Likewise, the protooncogene AKAP-Lbc couples activation of PKD with the phosphorylation-dependent nuclear export of the class II histone deacetylase HDAC5 [163]. This local phosphorylation event triggers 14-3-3 dependent nuclear export of this histone deacetylase and extensive chromatin remodeling [92,163].
AKAP8L, a component of the nuclear matrix, is hypermethylated in cholangiocarcinoma, kidney renal papillary and clear cell carcinomas and liver hepatocellular carcinoma [164]. Clinical studies correlate AKAP8L expression with reduced survival and progression free intervals [164]. Conversely, up-regulation of the AKAP8/ AKAP95 ortholog suppresses metastasis by antagonizing the epithelial–mesenchymal transition [165].
Genome instability and mutations
‘Survival of the fittest’ aptly describes cancer cells. Since genomic instability is emblematic of most cancers, it is hardly surprising that somatic mutations in cAMP signaling effectors underlie certain hyperplasias and neoplasms [26,56]. Allelic changes in the PRKACA gene are found in adrenocortical carcinomas and sporadic adenomas associated with Cushing’s syndrome [36,41,166-169]. Somatic mutations of the PRKAR1A gene are also implicated in the pathogenesis of cardiac myxomas, thyroid and adrenal cancers [170-173]. Mutations in PRKAR1A are also implicated in other tumors [22,174,175].
Adrenal Cushing’s syndrome is an autosomal dominant disease that emanates from single defective alleles of PRKACA resulting in overproduction of the stress hormone cortisol [176]. Malignant adrenal carcinomas that cause Cushing’s are rare [177,178]. More frequently, benign adenomas or micronodular hyperplasia drive adrenal Cushing’s syndrome [179]. Micronodules secrete cortisol from multiple locations in adrenal glands with adverse endocrine effects [180,181]. PKAc-L205R is the most prevalent mutant accounting for ~45% of cases [182]. Other disease-causing PKAc mutations and insertions occur at very low frequency [183]. A single mutation, S54L has been reported in the β isoform of PKAc [184,185]. These amino acid changes promote structural perturbations in PKAc that impair autoinhibition by type I or type II regulatory subunits, alter kinase substrate selectivity, and disrupt compartmentalization via recruitment to AKAPs [23,24,60,177]. One recently reported mutation PKAc-W196G is represented in ~20% of the population [169]. Structural and functional studies suggest that this subtle perturbation of the PKAc catalytic core skews R subunit selectivity and biases AKAP association. Compartmentalization of PKAcW196G is distorted in adrenal Cushing’s patients and increases off-target phosphorylation events that potentiate disease. This highlights the importance of genetic defects that impact kinase anchoring as a compounding factor in a number of pathological states [22].
Genomic reprogramming of cancer cells can also result in the generation of oncogenic AKAP fusions [28,131]. In thyroid papillary carcinoma, the MAPK pathway is commonly implicated in pathogenesis through sporadic mutations to the upstream BRAF [186]. In tumors, it has been found that a paracentric inversion on chromosome 7q results in an in-frame fusion of exons 1–8 of AKAP9 and exons 9–18 of BRAF [46]. This produces a fusion protein that lacks the autoinhibitory domain of BRAF, leading to a more active kinase with increased pathogenic potential. This fusion induces transformation in a focus forming assay and is likely responsible for the carcinogenesis in thyroid papillary carcinoma [46]. Likewise, a M463I AKAP9 mutation has been linked to increased risk for breast cancer, lung cancer, and colorectal cancer [187-189]. Other studies reported four mutations in AKAP9 linked to gastric cancer and over 20 mutations linked to colorectal cancer [190]. This burgeoning number of cancer causing mutations is indicative of instability within the AKAP9 gene [187-189]. However, it remains unclear if these point mutations change PKA anchoring or the recruitment of other signaling proteins that reside within AKAP9 signaling islands.
Invasion and metastasis
Metastasis is a process whereby the primary tumor releases metalloproteases that aid cancer cells to break away and travel through the blood or lymph system to form secondary metastatic tumors in other organs or tissues. Cells at the invasive front of a primary tumor undergo transcription factor-guided or epigenetic changes that engage genes related to invasion. Invasion and metastasis are often mediated by trans-differentiation through the epithelial to mesenchymal transition (EMT). This cellular process is triggered by signals from the microenvironment that mobilize pro-EMT transcription factors [191]. The concomitant reprogramming of genes confers a mesenchymal state that is more stem-like and migratory. Cells at the invasive front of a tumor often undergo EMT to gain invasive potential and enter the bloodstream. EMT is a reversible process [191]. Once at a distant site, migratory cells revert to an epithelial state to support the growth of a secondary tumor. This highly dynamic process involves the co-ordinated action of AKAP signaling islands.
AKAP9/AKAP350 has been shown to be involved in the pathogenesis of multiple cancer types, such as breast, lung, thyroid, melanoma, and colorectal cancers [47,187,188,192-195]. AKAP9 potentiates EMT in colorectal cancer cells through its interaction with cdc42 interacting protein 4. This protein-protein interaction regulates TGFβ1 induced EMT to potentiate the pathological phenotype and tumorigenesis of colorectal cancer cells in vivo [196]. AKAP95 supports an epithelial state through inhibiting alternative splicing of pro-EMT targets including CLSTN1 [165]. This phenotypic switch is thought to be predictive for breast cancer patient survival [165].
AKAP2 is a cytoskeletal family of six anchoring protein isoforms ranging in size from 95 to 160 kDa [197-199]. AKAP2 isoforms are enriched in the kidneys and lung, hence the colloquial name AKAP-KL [197]. In prostatic neuroendocrine carcinoma, AKAP2 was implicated in the invasiveness of prostate cancer cells through its anchoring of protein phosphatase 1 (PP1), [122]. PP1 targeting to cofilin permits its dephosphorylation to potentiate F-actin dynamics. Concomitant effects include enhanced cell migration and invasion ([122], Figure 3A). Consistent with this finding, AKAP2 is up-regulated in invasive prostatic neuroendocrine carcinoma compared with noncancerous prostate tissues. In ovarian cancer, AKAP2 activates the β-catenin/TCF signaling axis to promote invasion [121]. AKAP2 signaling islands play an important role in the pathogenesis of ovarian cancer. Anchored PKA activity is required for ovarian cancer invasion [200]. In contrast gene silencing of AKAP2 impairs invasiveness and metastatic capability [121].
Figure 3. AKAPs involved in cytoskeletal rearrangement and membrane dynamics that impact metastasis.

(A) AKAP2, a cytoskeletal AKAP, anchors PKA and protein phosphatase 1 (PP1) at the actin cytoskeleton. This brings PP1 in proximity to cofilin. PP1 mediated dephosphorylation promotes an invasive phenotype in prostate cancer cells through increased actin turnover. (B) Ezrin, a member of the ERM proteins, acts as a linker between the actin cytoskeleton and the cell membrane. Amplification of Ezrin promotes membrane protrusion to facilitate local invasion and subsequent metastasis. (Created in BioRender. Rosenthal, K. (2024) BioRender.com/t80z735).
The ERM proteins (ezrin, radixin and moesin) cross-link transmembrane proteins to the actin cytoskeleton [21]. Up-regulation of ezrin occurs at the onset of metastasis, a complex process involving EMT, actin reorganization, and colonization of metastatic cells [201]. Biochemical studies show that Ezrin interacts with both PKA and the AKT kinases, a family of three enzymes that phosphorylate proteins that participate in transcription and tumor cell proliferation and migration [202,203]. Anchoring of these kinases sites at the plasma membrane that interface with the cytoskeleton could have substantial implications for metastatic progression as both basophilic kinases display overlapping substrate specificities [204]. Overexpression of ezrin results in membrane protrusion and an increase in number of metastatic foci in cell and mouse models of prostate cancer [205]. Studies in breast and ovarian cancer cells propose these events proceed through activation of the ERK kinase cascade [206,207]. Hence metastatic cells expressing ezrin could be particularly sensitive to cAMP that unleashes PKA and phosphatidylinositol [3-5] trisphosphates that activate Akt’s [15]. Both of these local second messenger signaling events could impinge on ERK activation to augment cancer cell migration during tumor metastasis [208].
Deregulating cellular metabolism
Tumors require a continuous supply of energy in the form of ATP and nutrients to sustain their growth. Consequently, cancer cells are rewired to increase glucose uptake and fermentation to lactate. This energetic phenomenon known as the Warburg effect allows cancer cells to undergo glycolysis in the hypoxic TME [209]. Some of these metabolic changes involve PKA signaling on the surface of mitochondria [210]. Several investigators have focused on sAKAP84/dAKAP1 that is localized to the outer mitochondrial matrix [211-213]. Expression of dAKAP1 is reduced in mesenchymal breast cancer cell lines exhibiting a glycolytic phenotype [126]. Immunocytochemical analyses of patient samples reveals that dAKAP1 is reduced in metastatic lesions as compared with primary tumors [126]. This anchored pool of PKA promotes changes in mitochondrial morphology by driving phosphorylation of the dynamin related GTPase Drp1. During normal growth, Drp1 induces elongated mitochondria. However, upon nutrient deprivation, PKA phosphorylates Drp1 on Ser637 to inhibit Drp1 mediated mitochondrial fission in neurons ([214-216], Figure 4A). In the nutrient starved environment of breast cancer tumors, dAKAP1 signaling islands are reduced and PKA signaling becomes less directed in the vicinity of mitochondria [126,210]. This helps shift these organelles towards a more fragmented morphology that can favor mitophagy and global changes in cancer cell metabolism ([217], Figure 4B).
Figure 4. Depletion of dAKAP1 drives a glycolytic phenotype and invasive potential.

(A) In primary breast cancer, dAKAP1 is expressed at the outer mitochondrial membrane. Anchored PKA at dAKAP1 phosphorylates the GTPase effector protein Drp1, a mitochondrial fission regulator. (B) In invasive breast cancer cells, dAKAP1 is depleted, which results in decreased Drp1 phosphorylation and fragmented mitochondria through fission. This results in movement of mitochondria to the leading edge of the cell and increased glycolytic potential to produce local pools of ATP at the leading edge and increased motility. (Lower panels) Control and dAKAP1 siRNA treated cells showing changes in mitochondrial morphology (Mito tracker staining) upon depletion of the anchoring protein. (Created in BioRender. Rosenthal, K. (2024) BioRender.com/c07x768).
AKAP220 is involved in metabolic rewiring of cancer cells [218]. This vesicular AKAP complex sequesters kinases and phosphatases that participate in autophagy. This catabolic process degrades various cellular cargos through lysosomes activated in response to nutrient deprivation [218]. Recycling of nutrients liberated because of autophagy are used to create energy in the nutrient poor conditions of the TME. AKAP220 is a bivalent anchoring protein that can simultaneously engage type I and type II PKA holoenzymes [219,220]. Upon nutrient starvation, AKAP220 targets type I PKA to the autophagy adapter protein LC3 [221]. This macromolecular signaling complex checks cell death in serum deprived conditions as a protective measure to overcome nutrient deficiency in cancer cells [221].
An oncocytic phenotype consisting of many swollen mitochondria is a diagnostic feature of FLC [128]. Subsequent electron microscopy analyses reveal an accumulation of calcium deposits in tumor cells [222]. A molecular explanation for this observation can be provided by evidence that expression of DNAJ-PKAc correlates with up-regulation of the mitochondrial calcium uniporter, an outer mitochondrial membrane protein complex that co-ordinates calcium signaling, mitochondrial fatty acid oxidation and metabolic homeostasis [223]. The authors propose that increased mitochondrial calcium levels in FLC may represent an oncogenic adaptation through the transcriptional regulation of metabolism [223].
Conclusions and future perspectives
Dysregulated kinase signaling is a common feature of many cancers. Thus, gaining a holistic view of the molecular interactions that influence PKA and other kinases advances our understanding of cancer biology. To conclude, we consider three emerging concepts that encapsulate this view. First, liquid-phase separation into immobile membrane-less condensates has been an area of recent focus [224]. Formation of these sub-organellar structures is a plausible outcome of the scaffolding interactions that underlie the compartmentalized regulation of PKA [225]. The R1α subunit of PKA undergoes phase separation, an effect that is reversed by association of the DNAJ-PKAc fusion kinase [226]. While this is an intriguing observation, it remains unclear how release of R1α, which is autoinhibitory toward DNAJ-PKAc, can potentiate oncogenic properties of the fusion kinase in FLC [136]. While concentrating DNAJ-PKAc action in phase separated condensates is an appealing and contemporary concept, it is hard to reconcile how kinase signals generated in these molecularly crowded environments can be amplified or transmitted to rest of the cell.
Second, targeting kinases with ATP analogs (class I inhibitors) has transformed clinical management of cancer [227]. The current mode of action of class 1 inhibitors is to flood the cell with drugs that dampen phosphorylation by individual kinases. Thus, by mass action one should be able to reduce the prevalence of aberrant phosphorylation events that underlie disease. Gaining knowledge of which kinase drives which pathological event, such as DNAJ-PKAc and the phosphorylation of liver proteins in FLC, will help investigators make informed therapeutic decisions in cancer. Likewise, understanding the subcellular location of an enzyme such as PKA is an important element in discerning its mode of action. Recently, AKAP targeting motifs have been used as vectors to target class 1 inhibitors to the sites of action such as the centromere and kinetochores [25,82,83]. Current studies have used molecular biology approaches to target modified drugs conjugated to an AKAP. While the future is bright for this precision pharmacology strategy it will require the development of small molecules that are efficiently targeted to subcellular locations. Newer generations of class II inhibitors that target inactive kinase conformations and allosteric class III inhibitors that act outside the catalytic core of the enzyme extend the reach of kinase-directed therapeutics [228]. Allosteric MEK1/2 inhibitors bind to a cavity adjacent to the ATP-binding site to block mitogenic signaling via the ERK cascade [229]. Accordingly, dual inhibition of the ERK pathway with BRAF and allosteric MEK inhibitors is a standard treatment for metastatic melanoma patients harboring the BRAFV600E mutant [230]. Expanding this approach to include subcellular targeting of these compounds is an exciting future perspective.
Third, innovative proteomic, chemical genetics and RNA sequencing approaches have illuminated global changes to kinase signaling in several cancers [131,231-236]. The next step will be discerning how kinase inhibitors impact the TME. Profiling of the EGFR inhibitor crizotinib in non-small cell lung cancer was combined with scrutiny of the LINCS database [237]. This combined pharmacological/bioinformatic strategy charted systemic changes associated with drug tolerance upon persistent blocking of the c-MET and EGFR kinases [237]. Undoubtably, prudent use of artificial intelligence may open the door to advances in this burgeoning arena of kinase inhibitor drug action. Ultimately, precision targeting of kinase inhibitor drugs could offer a new means to modulate the subcellular localization of PKA in tumor initiation, progression and metastasis.
Acknowledgements
The authors wish to thank Katherine Forbush for editing and proof reading of this manuscript. K.J.R. was supported by NIH Pharmacological sciences training grant T32 GM7750-44. We recognize grant funding from CA279997, the Fibrolamellar Cancer Foundation and the Burroughs Wellcome Fund to J.D.G. and DK141129 and DK119192 and the Fibrolamellar Cancer Foundation to J.D.S.
Abbreviations
- AKAP
A-kinase Anchoring protein
- BAG2
Bcl-2 associated anthanogene 2
- EMT
epithelial to mesenchymal transition
- FLC
fibrolamellar carcinoma
- LAK
lymphokine activated killer
- NK
natural killer
- PKA
protein kinase A
- PKC
protein kinase C
- PKD
protein kinase D
- RAIN
receptor-associated independent cAMP nanodomain
- TAN
tumor associated neutrophils
- TME
tumor microenvironment
Footnotes
Competing Interests
The authors declare that there are no competing interests associated with the manuscript.
CRediT Author Contribution
John D. Scott: Conceptualization, Resources, Supervision, Funding acquisition, Writing — original draft, Writing — review and editing. Kacey Rosenthal: Conceptualization, Visualization, Writing — original draft, Writing — review and editing. John D. Gordan: Conceptualization, Funding acquisition, Visualization, Writing — original draft, Writing — review and editing.
References
- 1.Hanahan D and Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144, 646–674 10.1016/j.cell.2011.02.013 [DOI] [PubMed] [Google Scholar]
- 2.Hanks SK, Quinn AM and Hunter T (1988) The protein kinase family: conserved features and deduced phylogeny of the catalytic domains. Science 241, 42–52 10.1126/science.3291115 [DOI] [PubMed] [Google Scholar]
- 3.Hunter T. (1987) A thousand and one protein kinases. Cell 50, 823–829 10.1016/0092-8674(87)90509-5 [DOI] [PubMed] [Google Scholar]
- 4.Cohen GB, Ren R and Baltimore D (1995) Modular binding domains in signal transduction proteins. Cell 80, 237–248 10.1016/0092-8674(95)90406-9 [DOI] [PubMed] [Google Scholar]
- 5.Druker BJ, Mamon HJ and Roberts TM (1989) Oncogenes, growth factors, and signal transduction. N. Engl. J. Med 321, 1383–1391 10.1056/NEJM198911163212007 [DOI] [PubMed] [Google Scholar]
- 6.Druker BJ, Sawyers CL, Kantarjian H, Resta DJ, Reese SF, Ford JM et al. (2001) Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N. Engl. J. Med 344, 1038–1042 10.1056/NEJM200104053441402 [DOI] [PubMed] [Google Scholar]
- 7.Hochhaus A, Larson RA, Guilhot F, Radich JP, Branford S, Hughes TP et al. (2017) Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N. Engl. J. Med 376, 917–927 10.1056/NEJMoa1609324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Roskoski R. (2023) Small molecule protein kinase inhibitors approved by regulatory agencies outside of the United States. Pharmacol. Res 194, 106847 10.1016/j.phrs.2023.106847 [DOI] [PubMed] [Google Scholar]
- 9.Cohen P. (2002) Protein kinases–the major drug targets of the twenty-first century? Nat. Rev. Drug Discov 1, 309–315 10.1038/nrd773 [DOI] [PubMed] [Google Scholar]
- 10.Cohen P, Cross D and Jänne PA (2021) Kinase drug discovery 20 years after imatinib: progress and future directions. Nat. Rev. Drug Discov 20, 551–569 10.1038/s41573-021-00195-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gregorc V, Lazzari C, Karachaliou N, Rosell R and Santarpia M (2018) Osimertinib in untreated epidermal growth factor receptor (EGFR)-mutated advanced non-small cell lung cancer. Transl. Lung Cancer Res 7, S165–S170 10.21037/tlcr.2018.03.19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Planchard D, Janne PA, Cheng Y, Yang JC, Yanagitani N, Kim SW et al. (2023) Osimertinib with or without chemotherapy in EGFR-mutated advanced NSCLC. N. Engl. J. Med 389, 1935–1948 10.1056/NEJMoa2306434 [DOI] [PubMed] [Google Scholar]
- 13.Poulikakos PI, Sullivan RJ and Yaeger R (2022) Molecular pathways and mechanisms of BRAF in cancer therapy. Clin. Cancer Res 28, 4618–4628 10.1158/1078-0432.CCR-21-2138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kalinsky K, Hong F, McCourt CK, Sachdev JC, Mitchell EP, Zwiebel JA et al. (2021) Effect of capivasertib in patients with an AKT1 E17K-mutated tumor: NCI-MATCH subprotocol EAY131-Y nonrandomized trial. JAMA Oncol. 7, 271–278 10.1001/jamaoncol.2020.6741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Newton AC, Bootman MD and Scott JD (2016) Second messengers. Cold Spring Harb. Perspect. Biol 8, a005926 10.1101/cshperspect.a005926 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Baffi TR, Lordén G, Wozniak JM, Feichtner A, Yeung W, Kornev AP et al. (2021) mTORC2 controls the activity of PKC and Akt by phosphorylating a conserved TOR interaction motif. Sci. Signal 14, eabe4509 10.1126/scisignal.abe4509 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Antal CE, Hudson AM, Kang E, Zanca C, Wirth C, Stephenson NL et al. (2015) Cancer-associated protein kinase C mutations reveal kinase’s role as tumor suppressor. Cell 160, 489–502 10.1016/j.cell.2015.01.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Newton AC and Brognard J (2017) Reversing the paradigm: protein kinase C as a tumor suppressor. Trends Pharmacol. Sci 38, 438–447 10.1016/j.tips.2017.02.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Turnham RE and Scott JD (2016) Protein kinase A catalytic subunit isoform PRKACA; history, function and physiology. Gene 577, 101–108 10.1016/j.gene.2015.11.052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Riggle KM, Riehle KJ, Kenerson HL, Turnham R, Homma MK, Kazami M et al. (2016) Enhanced cAMP-stimulated protein kinase A activity in human fibrolamellar hepatocellular carcinoma. Pediatr. Res 80, 110–118 10.1038/pr.2016.36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Langeberg LK and Scott JD (2015) Signalling scaffolds and local organization of cellular behaviour. Nat. Rev. Mol. Cell Biol 16, 232–244 10.1038/nrm3966 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Omar MH and Scott JD (2020) AKAP signaling islands: venues for precision pharmacology. Trends Pharmacol. Sci 41, 933–946 10.1016/j.tips.2020.09.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Smith FD, Reichow SL, Esseltine JL, Shi D, Langeberg LK, Scott JD et al. (2013) Intrinsic disorder within an AKAP-protein kinase A complex guides local substrate phosphorylation. Elife 2, e01319 10.7554/eLife.01319 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Smith FD, Esseltine JL, Nygren PJ, Veesler D, Byrne DP, Vonderach M et al. (2017) Local protein kinase A action proceeds through intact holoenzymes. Science 356, 1288–1293 10.1126/science.aaj1669 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bucko PJ and Scott JD (2021) Drugs that regulate local cell signaling: AKAP targeting as a therapeutic option. Annu. Rev. Pharmacol. Toxicol 61, 361–379 10.1146/annurev-pharmtox-022420-112134 [DOI] [PubMed] [Google Scholar]
- 26.Bock A, Irannejad R and Scott JD (2024) cAMP signaling: a remarkably regional affair. Trends Biochem. Sci 49, 305–317 10.1016/j.tibs.2024.01.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Taylor SS, Ilouz R, Zhang P and Kornev AP (2012) Assembly of allosteric macromolecular switches: lessons from PKA. Nat. Rev. Mol. Cell Biol 13, 646–658 10.1038/nrm3432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Coles GL, Cristea S, Webber JT, Levin RS, Moss SM, He A et al. (2020) Unbiased proteomic profiling uncovers a targetable GNAS/PKA/PP2A axis in small cell lung cancer stem cells. Cancer Cell 38, 129–143 e7 10.1016/j.ccell.2020.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.McCudden CR, Hains MD, Kimple RJ, Siderovski DP and Willard FS (2005) G-protein signaling: back to the future. Cell. Mol. Life Sci 62, 551–577 10.1007/s00018-004-4462-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ramms DJ, Raimondi F, Arang N, Herberg FW, Taylor SS and Gutkind JS (2021) Gαs-protein kinase A (PKA) pathway signalopathies: the emerging genetic landscape and therapeutic potential of human diseases driven by aberrant Gαs-PKA signaling. Pharmacol. Rev 73, 155–197 10.1124/pharmrev.120.000269 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Iglesias-Bartolome R, Torres D, Marone R, Feng X, Martin D, Simaan M et al. (2015) Inactivation of a Galpha(s)-PKA tumour suppressor pathway in skin stem cells initiates basal-cell carcinogenesis. Nat. Cell Biol 17, 793–803 10.1038/ncb3164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Patra KC, Kato Y, Mizukami Y, Widholz S, Boukhali M, Revenco I et al. (2018) Mutant GNAS drives pancreatic tumourigenesis by inducing PKA-mediated SIK suppression and reprogramming lipid metabolism. Nat. Cell Biol 20, 811–822 10.1038/s41556-018-0122-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.O’Hayre M, Vazquez-Prado J, Kufareva I, Stawiski EW, Handel TM, Seshagiri S et al. (2013) The emerging mutational landscape of G proteins and G-protein-coupled receptors in cancer. Nat. Rev. Cancer 13, 412–424 10.1038/nrc3521 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhang H, Kong Q, Wang J, Jiang Y and Hua H (2020) Complex roles of cAMP–PKA–CREB signaling in cancer. Exp. Hematol. Oncol 9,32 10.1186/s40164-020-00191-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yin Z, Pringle DR, Jones GN, Kelly KM and Kirschner LS (2011) Differential role of PKA catalytic subunits in mediating phenotypes caused by knockout of the Carney complex gene Prkar1a. Mol. Endocrinol 25, 1786–1793 10.1210/me.2011-1008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Calebiro D, Hannawacker A, Lyga S, Bathon K, Zabel U, Ronchi C et al. (2014) PKA catalytic subunit mutations in adrenocortical Cushing’s adenoma impair association with the regulatory subunit. Nat. Commun 5, 5680 10.1038/ncomms6680 [DOI] [PubMed] [Google Scholar]
- 37.Cao Y, He M, Gao Z, Peng Y, Li Y, Li L et al. (2014) Activating hotspot L205R mutation in PRKACA and adrenal Cushing’s syndrome. Science 344, 913–917 10.1126/science.1249480 [DOI] [PubMed] [Google Scholar]
- 38.Salpea P and Stratakis CA (2014) Carney complex and McCune Albright syndrome: an overview of clinical manifestations and human molecular genetics. Mol. Cell. Endocrinol 386, 85–91 10.1016/j.mce.2013.08.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Berthon AS, Szarek E and Stratakis CA (2015) PRKACA: the catalytic subunit of protein kinase A and adrenocortical tumors. Front. Cell Dev. Biol 3, 26 10.3389/fcell.2015.00026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Honeyman JN, Simon EP, Robine N, Chiaroni-Clarke R, Darcy DG, Lim II et al. (2014) Detection of a recurrent DNAJB1-PRKACA chimeric transcript in fibrolamellar hepatocellular carcinoma. Science 343, 1010–1014 10.1126/science.1249484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Omar MH, Byrne DP, Jones KN, Lakey TM, Collins KB, Lee KS et al. (2022) Mislocalization of protein kinase A drives pathology in Cushing’s syndrome. Cell Rep. 40, 111073 10.1016/j.celrep.2022.111073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Stransky N, Cerami E, Schalm S, Kim JL and Lengauer C (2014) The landscape of kinase fusions in cancer. Nat. Commun 5, 4846 10.1038/ncomms5846 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Cornella H, Alsinet C, Sayols S, Zhang Z, Hao K, Cabellos L et al. (2015) Unique genomic profile of fibrolamellar hepatocellular carcinoma. Gastroenterology 148, 806–818 e10 10.1053/j.gastro.2014.12.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Graham RP, Lackner C, Terracciano L, Gonzalez-Cantu Y, Maleszewski JJ, Greipp PT et al. (2018) Fibrolamellar carcinoma in the Carney complex: PRKAR1A loss instead of the classic DNAJB1-PRKACA fusion. Hepatology 68, 1441–1447 10.1002/hep.29719 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Singhi AD, Wood LD, Parks E, Torbenson MS, Felsenstein M, Hruban RH et al. (2020) Recurrent rearrangements in PRKACA and PRKACB in intraductal oncocytic papillary neoplasms of the pancreas and bile duct. Gastroenterology 158, 573–582 e2 10.1053/j.gastro.2019.10.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ciampi R, Knauf JA, Kerler R, Gandhi M, Zhu Z, Nikiforova MN et al. (2005) Oncogenic AKAP9-BRAF fusion is a novel mechanism of MAPK pathway activation in thyroid cancer. J. Clin. Invest 115, 94–101 10.1172/JCI23237 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Lee J-H, Seok Lee E, Kim Y-S, Hee Won N and Chae Y-S (2006) BRAF mutation and AKAP9 expression in sporadic papillary thyroid carcinomas. Pathology 38, 201–204 10.1080/00313020600696264 [DOI] [PubMed] [Google Scholar]
- 48.Hehnly H, Canton D, Bucko P, Langeberg LK, Ogier L, Gelman I et al. (2015) A mitotic kinase scaffold depleted in testicular seminomas impacts spindle orientation in germ line stem cells. Elife 4, e09384 10.7554/eLife.09384 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wong W and Scott JD (2004) AKAP signalling complexes: focal points in space and time. Nat. Rev. Mol. Cell Biol 5, 959–970 10.1038/nrm1527 [DOI] [PubMed] [Google Scholar]
- 50.Scott JD, Dessauer CW and Tasken K (2013) Creating order from chaos: cellular regulation by kinase anchoring. Annu. Rev. Pharmacol. Toxicol 53, 187–210 10.1146/annurev-pharmtox-011112-140204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zaccolo M and Kovanich D (2024) Nanodomain cAMP signalling in cardiac pathophysiology: potential for developing targeted therapeutic interventions. Physiol. Rev, in press 10.1152/physrev.00013.2024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Anton SE, Kayser C, Maiellaro I, Nemec K, Moller J, Koschinski A et al. (2022) Receptor-associated independent cAMP nanodomains mediate spatiotemporal specificity of GPCR signaling. Cell 185, 1130–1142 e11 10.1016/j.cell.2022.02.011 [DOI] [PubMed] [Google Scholar]
- 53.Bock A, Annibale P, Konrad C, Hannawacker A, Anton SE, Maiellaro I et al. (2021) Optical mapping of cAMP signaling at the nanometer scale. Cell 184, 2793 10.1016/j.cell.2021.04.043 [DOI] [PubMed] [Google Scholar]
- 54.Irannejad R, Tsvetanova NG, Lobingier BT and von Zastrow M (2015) Effects of endocytosis on receptor-mediated signaling. Curr. Opin. Cell Biol 35, 137–143 10.1016/j.ceb.2015.05.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lin TY, Mai QN, Zhang H, Wilson E, Chien HC, Yee SW et al. (2024) Cardiac contraction and relaxation are regulated by distinct subcellular cAMP pools. Nat. Chem. Biol 20, 62–73 10.1038/s41589-023-01381-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Scott JD and Pawson T (2009) Cell signaling in space and time: where proteins come together and when they’re apart. Science 326, 1220–1224 10.1126/science.1175668 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Pawson T and Scott JD (1997) Signaling through scaffold, anchoring, and adaptor proteins. Science 278, 2075–2080 10.1126/science.278.5346.2075 [DOI] [PubMed] [Google Scholar]
- 58.Tasken K and Aandahl EM (2004) Localized effects of cAMP mediated by distinct routes of protein kinase A. Physiol. Rev 84, 137–167 10.1152/physrev.00021.2003 [DOI] [PubMed] [Google Scholar]
- 59.Lefkimmiatis K and Zaccolo M (2014) cAMP signaling in subcellular compartments. Pharmacol. Ther 143, 295–304 10.1016/j.pharmthera.2014.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Piggott LA, Bauman AL, Scott JD and Dessauer CW (2008) The A-kinase anchoring protein Yotiao binds and regulates adenylyl cyclase in brain. Proc. Natl Acad. Sci. U.S.A 105, 13835–13840 10.1073/pnas.0712100105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zhang J, Hupfeld CJ, Taylor SS, Olefsky JM and Tsien RY (2005) Insulin disrupts beta-adrenergic signalling to protein kinase A in adipocytes. Nature 437, 569–573 10.1038/nature04140 [DOI] [PubMed] [Google Scholar]
- 62.Zhang J, Ma Y, Taylor SS and Tsien RY (2001) Genetically encoded reporters of protein kinase A activity reveal impact of substrate tethering. Proc. Natl Acad. Sci. U.S.A 98, 14997–15002 10.1073/pnas.211566798 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hoshi N, Langeberg LK and Scott JD (2005) Distinct enzyme combinations in AKAP signalling complexes permit functional diversity. Nat. Cell Biol 7, 1066–1073 10.1038/ncb1315 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Smith FD, Omar MH, Nygren PJ, Soughayer J, Hoshi N, Lau H-T et al. (2018) Single nucleotide polymorphisms alter kinase anchoring and the subcellular targeting of A-kinase anchoring proteins. Proc. Natl Acad. Sci. U.S.A 115, E11465–E11474 10.1073/pnas.1816614115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Carr DW, Stofko-Hahn RE, Fraser ID, Bishop SM, Acott TS, Brennan RG et al. (1991) Interaction of the regulatory subunit (RII) of cAMP-dependent protein kinase with RII-anchoring proteins occurs through an amphipathic helix binding motif. J. Biol. Chem 266, 14188–14192 10.1016/S0021-9258(18)98665-5 [DOI] [PubMed] [Google Scholar]
- 66.Gold MG, Lygren B, Dokurno P, Hoshi N, McConnachie G, Tasken K et al. (2006) Molecular basis of AKAP specificity for PKA regulatory subunits. Mol. Cell 24, 383–395 10.1016/j.molcel.2006.09.006 [DOI] [PubMed] [Google Scholar]
- 67.Dodge-Kafka KL, Soughayer J, Pare GC, Carlisle Michel JJ, Langeberg LK, Kapiloff MS et al. (2005) The protein kinase A anchoring protein mAKAP coordinates two integrated cAMP effector pathways. Nature 437, 574–578 10.1038/nature03966 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hanahan D and Weinberg RA (2000) The hallmarks of cancer. Cell 100, 57–70 10.1016/S0092-8674(00)81683-9 [DOI] [PubMed] [Google Scholar]
- 69.Hanahan D. (2022) Hallmarks of cancer: new dimensions. Cancer Discov. 12, 31–46 10.1158/2159-8290.CD-21-1059 [DOI] [PubMed] [Google Scholar]
- 70.Stork PJ and Schmitt JM (2002) Crosstalk between cAMP and MAP kinase signaling in the regulation of cell proliferation. Trends Cell Biol. 12, 258–266 10.1016/S0962-8924(02)02294-8 [DOI] [PubMed] [Google Scholar]
- 71.Krebs EG (1985) The phosphorylation of proteins: a major mechanism for biological regulation. Biochem. Soc. Trans 13, 813–820 10.1042/bst0130813 [DOI] [PubMed] [Google Scholar]
- 72.Bos JL (2003) Epac: a new cAMP target and new avenues in cAMP research. Nat. Rev. Mol. Cell Biol 4, 733–738 10.1038/nrm1197 [DOI] [PubMed] [Google Scholar]
- 73.Rossomando AJ, Wu J, Michel H, Shabanowitz J, Hunt DF, Weber MJ et al. (1992) Identification of Tyr-185 as the site of tyrosine autophosphorylation of recombinant mitogen-activated protein kinase p42mapk. Proc. Natl Acad. Sci. U.S.A 89, 5779–5783 10.1073/pnas.89.13.5779 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Li Y, Dillon TJ, Takahashi M, Earley KT and Stork PJ (2016) Protein kinase A-independent Ras protein activation cooperates with Rap1 protein to mediate activation of the extracellular signal-regulated kinases (ERK) by cAMP. J. Biol. Chem 291, 21584–21595 10.1074/jbc.M116.730978 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Wu J, Dent P, Jelinek T, Wolfman A, Weber MJ and Sturgill TW (1993) Inhibition of the EGF-activated MAP kinase signaling pathway by adenosine 3′,5′-monophosphate. Science 262, 1065–1072 10.1126/science.7694366 [DOI] [PubMed] [Google Scholar]
- 76.Faure M and Bourne HR (1995) Differential effects on cAMP on the MAP kinase cascade: evidence for a cAMP-insensitive step that can bypass Raf-1. Mol. Biol. Cell 6, 1025–1035 10.1091/mbc.6.8.1025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Takahashi M, Li Y, Dillon TJ and Stork PJ (2017) Phosphorylation of Rap1 by cAMP-dependent protein kinase (PKA) creates a binding site for KSR to sustain ERK activation by cAMP. J. Biol. Chem 292, 1449–1461 10.1074/jbc.M116.768986 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Nauert JB, Klauck TM, Langeberg LK and Scott JD (1997) Gravin, an autoantigen recognized by serum from myasthenia gravis patients, is a kinase scaffold protein. Curr. Biol 7, 52–62 10.1016/S0960-9822(06)00027-3 [DOI] [PubMed] [Google Scholar]
- 79.Havekes R, Canton DA, Park AJ, Huang T, Nie T, Day JP et al. (2012) Gravin orchestrates protein kinase A and beta2-adrenergic receptor signaling critical for synaptic plasticity and memory. J. Neurosci 32, 18137–18149 10.1523/JNEUROSCI.3612-12.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Canton DA, Keene CD, Swinney K, Langeberg LK, Nguyen V, Pelletier L et al. (2012) Gravin is a transitory effector of polo-like kinase 1 during cell division. Mol. Cell 48, 547–559 10.1016/j.molcel.2012.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lin X, Nelson P and Gelman IH (2000) SSeCKS, a major protein kinase C substrate with tumor suppressor activity, regulates G(1)–>S progression by controlling the expression and cellular compartmentalization of cyclin D. Mol. Cell. Biol 20, 7259–7272 10.1128/MCB.20.19.7259-7272.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bucko PJ, Lombard CK, Rathbun L, Garcia I, Bhat A, Wordeman L et al. (2019) Subcellular drug targeting illuminates local kinase action. Elife 8, e52220 10.7554/eLife.52220 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Bucko PJ, Garcia I, Manocha R, Bhat A, Wordeman L and Scott JD (2020) Gravin-associated kinase signaling networks coordinate gamma-tubulin organization at mitotic spindle poles. J. Biol. Chem 295, 13784–13797 10.1074/jbc.RA120.014791 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Jagadish N, Parashar D, Gupta N, Agarwal S, Purohit S, Kumar V et al. (2015) A-kinase anchor protein 4 (AKAP4) a promising therapeutic target of colorectal cancer. J. Exp. Clin. Cancer Res 34, 142 10.1186/s13046-015-0258-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Agarwal S, Saini S, Parashar D, Verma A, Jagadish N, Batra A et al. (2013) Expression and humoral response of A-kinase anchor protein 4 in cervical cancer. Int. J. Gynecol. Cancer 23, 650–658 10.1097/IGC.0b013e31828a0698 [DOI] [PubMed] [Google Scholar]
- 86.Chiriva-Internati M, Yu Y, Mirandola L, D’Cunha N, Hardwicke F, Cannon MJ et al. (2012) Identification of AKAP-4 as a new cancer/testis antigen for detection and immunotherapy of prostate cancer. Prostate 72, 12–23 10.1002/pros.21400 [DOI] [PubMed] [Google Scholar]
- 87.Gumireddy K, Li A, Chang DH, Liu Q, Kossenkov AV, Yan J et al. (2015) AKAP4 is a circulating biomarker for non-small cell lung cancer. Oncotarget 6, 17637–17647 10.18632/oncotarget.3946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Jagadish N, Devi S, Gupta N, Suri V and Suri A (2020) Knockdown of A-kinase anchor protein 4 inhibits proliferation of triple-negative breast cancer cells in vitro and in vivo. Tumour Biol. 42, 1010428320914477 10.1177/1010428320914477 [DOI] [PubMed] [Google Scholar]
- 89.Diviani D and Scott JD (2001) AKAP signaling complexes at the cytoskeleton. J. Cell Sci 114, 1431–1437 10.1242/jcs.114.8.1431 [DOI] [PubMed] [Google Scholar]
- 90.Diviani D, Soderling J and Scott JD (2001) AKAP-Lbc anchors protein kinase A and nucleates Galpha 12-selective Rho-mediated stress fiber formation. J. Biol. Chem 276, 44247–44257 10.1074/jbc.M106629200 [DOI] [PubMed] [Google Scholar]
- 91.Diviani D, Baisamy L and Appert-Collin A (2006) AKAP-Lbc: a molecular scaffold for the integration of cyclic AMP and Rho transduction pathways. Eur. J. Cell Biol 85, 603–610 10.1016/j.ejcb.2006.01.001 [DOI] [PubMed] [Google Scholar]
- 92.Jin J, Smith FD, Stark C, Wells CD, Fawcett JP, Kulkarni S et al. (2004) Proteomic, functional, and domain-based analysis of in vivo 14-3-3 binding proteins involved in cytoskeletal regulation and cellular organization. Curr. Biol 14, 1436–1450 10.1016/j.cub.2004.07.051 [DOI] [PubMed] [Google Scholar]
- 93.Smith FD, Langeberg LK, Cellurale C, Pawson T, Morrison DK, Davis RJ et al. (2010) AKAP-Lbc enhances cyclic AMP control of the ERK1/2 cascade. Nat. Cell Biol 12, 1242–1249 10.1038/ncb2130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Turnham RE, Smith FD, Kenerson HL, Omar MH, Golkowski M, Garcia I et al. (2019) An acquired scaffolding function of the DNAJ-PKAc fusion contributes to oncogenic signaling in fibrolamellar carcinoma. eLife 8, e44187 10.7554/eLife.44187 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Shay JW and Wright WE (2011) Role of telomeres and telomerase in cancer. Semin. Cancer Biol 21, 349–353 10.1016/j.semcancer.2011.10.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Colacci A, Corvi R, Ohmori K, Paparella M, Serra S, Da Rocha Carrico I et al. (2023) The cell transformation assay: a historical assessment of current knowledge of applications in an integrated approach to testing and assessment for non-genotoxic carcinogens. Int. J. Mol. Sci 24, 5659 10.3390/ijms24065659 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lin X, Tombler E, Nelson PJ, Ross M and Gelman IH (1996) A novel src- and ras-suppressed protein kinase C substrate associated with cytoskeletal architecture. J. Biol. Chem 271, 28430–28438 10.1074/jbc.271.45.28430 [DOI] [PubMed] [Google Scholar]
- 98.Willoughby D, Wong W, Schaack J, Scott JD and Cooper DM (2006) An anchored PKA and PDE4 complex regulates subplasmalemmal cAMP dynamics. EMBO J. 25, 2051–2061 10.1038/sj.emboj.7601113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lin X and Gelman IH (2002) Calmodulin and cyclin D anchoring sites on the Src-suppressed C kinase substrate, SSeCKS. Biochem. Biophys. Res. Commun 290, 1368–1375 10.1006/bbrc.2002.6357 [DOI] [PubMed] [Google Scholar]
- 100.Lin X and Gelman IH (1997) Reexpression of the major protein kinase C substrate, SSeCKS, suppresses v-src-induced morphological transformation and tumorigenesis. Cancer Res. 57, 2304–2312 [PubMed] [Google Scholar]
- 101.Cohen SB, Waha A, Gelman IH and Vogt PK (2001) Expression of a down-regulated target, SSeCKS, reverses v-Jun-induced transformation of 10T1/2 murine fibroblasts. Oncogene 20, 141–146 10.1038/sj.onc.1204077 [DOI] [PubMed] [Google Scholar]
- 102.Sherr CJ (2004) Principles of tumor suppression. Cell 116, 235–246 10.1016/S0092-8674(03)01075-4 [DOI] [PubMed] [Google Scholar]
- 103.Petrilli AM and Fernández-Valle C (2016) Role of Merlin/NF2 inactivation in tumor biology. Oncogene 35, 537–548 10.1038/onc.2015.125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Curto M, Cole BK, Lallemand D, Liu CH and McClatchey AI (2007) Contact-dependent inhibition of EGFR signaling by Nf2/Merlin. J. Cell Biol 177, 893–903 10.1083/jcb.200703010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Rouleau GA, Merel P, Lutchman M, Sanson M, Zucman J, Marineau C et al. (1993) Alteration in a new gene encoding a putative membrane-organizing protein causes neuro-fibromatosis type 2. Nature 363, 515–521 10.1038/363515a0 [DOI] [PubMed] [Google Scholar]
- 106.Trofatter J. (1993) A novel moesin-, ezrin-, radixin-like gene is a candidate for the neurofibromatosis 2 tumor suppressor. Cell 72, 791–800 10.1016/0092-8674(93)90406-G [DOI] [PubMed] [Google Scholar]
- 107.Dukic AR, Haugen LH, Pidoux G, Leithe E, Bakke O and Tasken K (2017) A protein kinase A-ezrin complex regulates connexin 43 gap junction communication in liver epithelial cells. Cell Signal. 32, 1–11 10.1016/j.cellsig.2017.01.008 [DOI] [PubMed] [Google Scholar]
- 108.Gronholm M, Vossebein L, Carlson CR, Kuja-Panula J, Teesalu T, Alfthan K et al. (2003) Merlin links to the cAMP neuronal signaling pathway by anchoring the RIbeta subunit of protein kinase A. J. Biol. Chem 278, 41167–41172 10.1074/jbc.M306149200 [DOI] [PubMed] [Google Scholar]
- 109.Wang B, Means CK, Yang Y, Mamonova T, Bisello A, Altschuler DL et al. (2012) Ezrin-anchored protein kinase A coordinates phosphorylation-dependent disassembly of a NHERF1 ternary complex to regulate hormone-sensitive phosphate transport. J. Biol. Chem 287, 24148–24163 10.1074/jbc.M112.369405 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Song GJ, Leslie KL, Barrick S, Mamonova T, Fitzpatrick JM, Drombosky KW et al. (2015) Phosphorylation of ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50) by Akt promotes stability and mitogenic function of S-phase kinase-associated protein-2 (Skp2). J. Biol. Chem 290, 2879–2887 10.1074/jbc.M114.609768 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Sainz J, Huynh DP, Figueroa K, Ragge NK, Baser ME and Pulst S-M (1994) Mutations of the neurofibromatosis type 2 gene and lack of the gene product in vestibular schwannomas. Human Mol. Genet 3, 885–891 10.1093/hmg/3.6.885 [DOI] [PubMed] [Google Scholar]
- 112.Manchanda PK, Jones GN, Lee AA, Pringle DR, Zhang M, Yu L et al. (2013) Rac1 is required for Prkar1a-mediated Nf2 suppression in Schwann cell tumors. Oncogene 32, 3491–3499 10.1038/onc.2012.374 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Akakura S, Huang C, Nelson PJ, Foster B and Gelman IH (2008) Loss of the SSeCKS/Gravin/AKAP12 gene results in prostatic hyperplasia. Cancer Res. 68, 5096–5103 10.1158/0008-5472.CAN-07-5619 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Akakura S, Nochajski P, Gao L, Sotomayor P, Matsui S and Gelman IH (2010) Rb-dependent cellular senescence, multinucleation and susceptibility to oncogenic transformation through PKC scaffolding by SSeCKS/AKAP12. Cell Cycle 9, 4656–4665 10.4161/cc.9.23.13974 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Klauck TM, Faux MC, Labudda K, Langeberg LK, Jaken S and Scott JD (1996) Coordination of three signaling enzymes by AKAP79, a mammalian scaffold protein. Science 271, 1589–1592 10.1126/science.271.5255.1589 [DOI] [PubMed] [Google Scholar]
- 116.Tunquist BJ, Hoshi N, Guire ES, Zhang F, Mullendorff K, Langeberg LK et al. (2008) Loss of AKAP150 perturbs distinct neuronal processes in mice. Proc. Natl Acad. Sci. U.S.A 105, 12557–12562 10.1073/pnas.0805922105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Dessauer CW (2009) Adenylyl cyclase–A-kinase anchoring protein complexes: the next dimension in cAMP signaling. Mol. Pharmacol 76, 935–941 10.1124/mol.109.059345 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Bauman AL, Soughayer J, Nguyen BT, Willoughby D, Carnegie GK, Wong W et al. (2006) Dynamic regulation of cAMP synthesis through anchored PKA-adenylyl cyclase V/VI complexes. Mol. Cell 23, 925–931 10.1016/j.molcel.2006.07.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wong W, Goehring AS, Kapiloff MS, Langeberg LK and Scott JD (2008) mAKAP compartmentalizes oxygen-dependent control of HIF-1alpha. Sci. Signal 1, ra18 10.1126/scisignal.2000026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Maric D, Paterek A, Delaunay M, Lopez IP, Arambasic M and Diviani D (2021) A-kinase anchoring protein 2 promotes protection against myocardial infarction. Cells 10, 2861 10.3390/cells10112861 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Li X, Wang C, Zhang G, Liang M and Zhang B (2017) AKAP2 is upregulated in ovarian cancer, and promotes growth and migration of cancer cells. Mol. Med. Rep 16, 5151–5156 10.3892/mmr.2017.7286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Reggi E, Kaiser S, Sahnane N, Uccella S, La Rosa S and Diviani D (2024) AKAP2-anchored protein phosphatase 1 controls prostatic neuroendocrine carcinoma cell migration and invasion. Biochim. Biophys. Acta 1870, 166916 10.1016/j.bbadis.2023.166916 [DOI] [PubMed] [Google Scholar]
- 123.Zhang B, Hu Q, Zhang J, Jin Z, Ruan Y, Xia L et al. (2022) Silencing of A-kinase anchor protein 4 inhibits the metastasis and growth of non-small cell lung cancer. Bioengineered 13, 6895–6907 10.1080/21655979.2021.1977105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Elmore S. (2007) Apoptosis: a review of programmed cell death. Toxicol. Pathol 35, 495–516 10.1080/01926230701320337 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Carneiro BA and El-Deiry WS (2020) Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol 17, 395–417 10.1038/s41571-020-0341-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Aggarwal S, Gabrovsek L, Langeberg LK, Golkowski M, Ong SE, Smith FD et al. (2019) Depletion of dAKAP1-protein kinase A signaling islands from the outer mitochondrial membrane alters breast cancer cell metabolism and motility. J. Biol. Chem 294, 3152–3168 10.1074/jbc.RA118.006741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Harada H, Becknell B, Wilm M, Mann M, Huang LJ, Taylor SS et al. (1999) Phosphorylation and inactivation of BAD by mitochondria-anchored protein kinase A. Mol. Cell 3, 413–422 10.1016/S1097-2765(00)80469-4 [DOI] [PubMed] [Google Scholar]
- 128.Craig JR, Peters RL, Edmondson HA and Omata M (1980) Fibrolamellar carcinoma of the liver: a tumor of adolescents and young adults with distinctive clinico-pathologic features. Cancer 46, 372–379 [DOI] [PubMed] [Google Scholar]
- 129.Zack T LK, Maisel SM, Wild J, Yaqubie A, Herman M, Knox JJ et al. (2023) Defining incidence and complications of fibrolamellar liver cancer through tiered computational analysis of clinical data. Nat. Precis. Oncol 7, 29 10.1038/s41698-023-00371-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Simon EP, Freije CA, Farber BA, Lalazar G, Darcy DG, Honeyman JN et al. (2015) Transcriptomic characterization of fibrolamellar hepatocellular carcinoma. Proc. Natl Acad. Sci. U.S.A 112, E5916–E5925 10.1073/pnas.1424894112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Chan GKL, Maisel S, Hwang YC, Pascual BC, Wolber RRB, Vu P et al. (2023) Oncogenic PKA signaling increases c-MYC protein expression through multiple targetable mechanisms. Elife 12, e69521 10.7554/eLife.69521 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Dinh TA, Jewell ML, Kanke M, Francisco A, Sritharan R, Turnham RE et al. (2019) MicroRNA-375 suppresses the growth and invasion of fibrolamellar carcinoma. Cell. Mol. Gastroenterol. Hepatol 7, 803–817 10.1016/j.jcmgh.2019.01.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Schalm SS, O’Hearn E, Wilson K, LaBranche TP, Silva G, Zhang Z et al. (2022) Evaluation of PRKACA as a therapeutic target for fibrolamellar carcinoma. bioRxiv 10.1101/2022.01.31.477690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Wang X, Jiang L, Thao K, Sussman CR, LaBranche T, Palmer M et al. (2022) Protein kinase A downregulation delays the development and progression of polycystic kidney disease. J. Am. Soc. Nephrol 33, 1087–1104 10.1681/ASN.2021081125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Lauer SM, Omar MH, Golkowski MG, Kenerson HL, Lee KS, Pascual BC et al. (2024) Recruitment of BAG2 to DNAJ-PKAc scaffolds promotes cell survival and resistance to drug-induced apoptosis in fibrolamellar carcinoma. Cell Rep. 43, 113678 10.1016/j.celrep.2024.113678 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Dinh TA, Utria AF, Barry KC, Ma R, Abou-Alfa GK, Gordan JD et al. (2022) A framework for fibrolamellar carcinoma research and clinical trials. Nat. Rev. Gastroenterol. Hepatol 19, 328–342 10.1038/s41575-022-00580-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Teng MWL, Swann JB, Koebel CM, Schreiber RD and Smyth MJ (2008) Immune-mediated dormancy: an equilibrium with cancer. J. Leukoc. Biol 84, 988–993 10.1189/jlb.1107774 [DOI] [PubMed] [Google Scholar]
- 138.Dunn GP, Bruce AT, Ikeda H, Old LJ and Schreiber RD (2002) Cancer immunoediting: from immunosurveillance to tumor escape. Nat. Immunol 3, 991–998 10.1038/ni1102-991 [DOI] [PubMed] [Google Scholar]
- 139.Seliger B. (2005) Strategies of tumor immune evasion. BioDrugs 19, 347–354 10.2165/00063030-200519060-00002 [DOI] [PubMed] [Google Scholar]
- 140.Wehbi VL and Taskén K (2016) Molecular mechanisms for cAMP-mediated immunoregulation in T cells - role of anchored protein kinase A signaling units. Front. Immunol 7, 222 10.3389/fimmu.2016.00222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Zamai L, Ponti C, Mirandola P, Gobbi G, Papa S, Galeotti L et al. (2007) NK cells and cancer. J. Immunol 178, 4011–4016 10.4049/jimmunol.178.7.4011 [DOI] [PubMed] [Google Scholar]
- 142.Mosenden R and Tasken K (2011) Cyclic AMP-mediated immune regulation–overview of mechanisms of action in T cells. Cell Signal. 23, 1009–1016 10.1016/j.cellsig.2010.11.018 [DOI] [PubMed] [Google Scholar]
- 143.Grimm EA, Mazumder A, Zhang HZ and Rosenberg SA (1982) Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid tumor cells by interleukin 2-activated autologous human peripheral blood lymphocytes. J. Exp. Med 155, 1823–1841 10.1084/jem.155.6.1823 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Raskovalova T, Lokshin A, Huang X, Jackson EK and Gorelik E (2006) Adenosine-mediated inhibition of cytotoxic activity and cytokine production by IL-2/NKp46-activated NK cells: involvement of protein kinase A isozyme I (PKA I). Immunol. Res 36, 91–100 10.1385/IR:36:1:91 [DOI] [PubMed] [Google Scholar]
- 145.Su Y, Huang X, Raskovalova T, Zacharia L, Lokshin A, Jackson E et al. (2008) Cooperation of adenosine and prostaglandin E2 (PGE2) in amplification of cAMP–PKA signaling and immunosuppression. Cancer Immunol. Immunother 57, 1611–1623 10.1007/s00262-008-0494-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Lokshin A, Raskovalova T, Huang X, Zacharia LC, Jackson EK and Gorelik E (2006) Adenosine-mediated inhibition of the cytotoxic activity and cytokine production by activated natural killer cells. Cancer Res. 66, 7758–7765 10.1158/0008-5472.CAN-06-0478 [DOI] [PubMed] [Google Scholar]
- 147.Raskovalova T, Lokshin A, Huang X, Su Y, Mandic M, Zarour HM et al. (2007) Inhibition of cytokine production and cytotoxic activity of human antimelanoma specific CD8+ and CD4+ T lymphocytes by adenosine-protein kinase A type I signaling. Cancer Res. 67, 5949–5956 10.1158/0008-5472.CAN-06-4249 [DOI] [PubMed] [Google Scholar]
- 148.Lagadari M, Truta-Feles K, Lehmann K, Berod L, Ziemer M, Idzko M et al. (2009) Lysophosphatidic acid inhibits the cytotoxic activity of NK cells: involvement of Gs protein-mediated signaling. Int. Immunol 21, 667–677 10.1093/intimm/dxp035 [DOI] [PubMed] [Google Scholar]
- 149.Lagadari M, Lehmann K, Ziemer M, Truta-Feles K, Berod L, Idzko M et al. (1992) Sphingosine-1-phosphate inhibits the cytotoxic activity of NK cells via Gs protein-mediated signalling. Int. J. Oncol 34, 287–294 10.3892/ijo_00000151 [DOI] [PubMed] [Google Scholar]
- 150.de Visser KE and Joyce JA (2023) The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell 41, 374–403 10.1016/j.ccell.2023.02.016 [DOI] [PubMed] [Google Scholar]
- 151.Yunna C, Mengru H, Lei W and Weidong C (2020) Macrophage M1/M2 polarization. Eur. J. Pharmacol 877, 173090 10.1016/j.ejphar.2020.173090 [DOI] [PubMed] [Google Scholar]
- 152.Na YR, Kwon JW, Kim DY, Chung H, Song J, Jung D et al. Protein kinase A catalytic subunit is a molecular switch that promotes the pro-tumoral function of macrophages. Cell Rep. 2020;31:107643 10.1016/j.celrep.2020.107643 [DOI] [PubMed] [Google Scholar]
- 153.Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L et al. (2009) Polarization of tumor-associated neutrophil phenotype by TGF-β: “N1” versus “N2” TAN. Cancer Cell 16, 183–194 10.1016/j.ccr.2009.06.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Jones SL and Sharief Y (2005) Asymmetrical protein kinase A activity establishes neutrophil cytoskeletal polarity and enables chemotaxis. J. Leukoc. Biol 78, 248–258 10.1189/jlb.0804459 [DOI] [PubMed] [Google Scholar]
- 155.Pan P, Oshima K, Huang YW, Agle KA, Drobyski WR, Chen X et al. (2018) Loss of FFAR2 promotes colon cancer by epigenetic dysregulation of inflammation suppressors. Int. J. Cancer 143, 886–896 10.1002/ijc.31366 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Kim HJ and Cantor H (2014) CD4T-cell subsets and tumor immunity: the helpful and the not-so-helpful. Cancer Immunol. Res 2, 91–98 10.1158/2326-6066.CIR-13-0216 [DOI] [PubMed] [Google Scholar]
- 157.Ruppelt A, Mosenden R, Gronholm M, Aandahl EM, Tobin D, Carlson CR et al. (2007) Inhibition of T cell activation by cyclic adenosine 5’-monophosphate requires lipid raft targeting of protein kinase A type I by the A-kinase anchoring protein ezrin. J. Immunol 179, 5159–5168 10.4049/jimmunol.179.8.5159 [DOI] [PubMed] [Google Scholar]
- 158.Rothbart SB and Strahl BD (2014) Interpreting the language of histone and DNA modifications. Biochim. Biophys. Acta 1839, 627–643 10.1016/j.bbagrm.2014.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Gopalan J, Omar MH, Roy A, Cruz NM, Falcone J, Jones KN et al. (2021) Targeting an anchored phosphatase-deacetylase unit restores renal ciliary homeostasis. Elife 10, e67828 10.7554/eLife.67828 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Auzmendi-Iriarte J, Saenz-Antoñanzas A, Mikelez-Alonso I, Carrasco-Garcia E, Tellaetxe-Abete M, Lawrie CH et al. (2020) Characterization of a new small-molecule inhibitor of HDAC6 in glioblastoma. Cell Death Dis. 11, 417 10.1038/s41419-020-2586-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Ali A, Zhang F, Maguire A, Byrne T, Weiner-Gorzel K, Bridgett S et al. (2020) HDAC6 degradation inhibits the growth of high-grade serous ovarian cancer cells. Cancers (Basel) 12, 3734 10.3390/cancers12123734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Gopalan J, Wordeman L and Scott JD (2021) Kinase-anchoring proteins in ciliary signal transduction. Biochem. J 478, 1617–1629 10.1042/BCJ20200869 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Carnegie GK, Soughayer J, Smith FD, Pedroja BS, Zhang F, Diviani D et al. (2008) AKAP-Lbc mobilizes a cardiac hypertrophy signaling pathway. Mol. Cell 32, 169–179 10.1016/j.molcel.2008.08.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Zhou L, Mei J, Cao R, Liu X, Fu B, Ma M et al. (2023) Integrative analysis identifies AKAP8L as an immunological and prognostic biomarker of pan-cancer. Aging 15, 8851–8872 10.18632/aging.205003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Hu X, Harvey SE, Zheng R, Lyu J, Grzeskowiak CL, Powell E et al. (2020) The RNA-binding protein AKAP8 suppresses tumor metastasis by antagonizing EMT-associated alternative splicing. Nat. Commun 11, 486 10.1038/s41467-020-14304-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Di Dalmazi G, Kisker C, Calebiro D, Mannelli M, Canu L, Arnaldi G et al. (2014) Novel somatic mutations in the catalytic subunit of the protein kinase A as a cause of adrenal Cushing’s syndrome: a European Multicentric Study. J. Clin. Endocrinol. Metab 99, E2093–E2100 10.1210/jc.2014-2152 [DOI] [PubMed] [Google Scholar]
- 167.Beuschlein F, Fassnacht M, Assie G, Calebiro D, Stratakis CA, Osswald A et al. (2014) Constitutive activation of PKA catalytic subunit in adrenal Cushing’s syndrome. N. Engl. J. Med 370, 1019–1028 10.1056/NEJMoa1310359 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Broder MS, Neary MP, Chang E, Cherepanov D and Ludlam WH (2015) Incidence of Cushing’s syndrome and Cushing’s disease in commercially-insured patients <65 years old in the United States. Pituitary 18, 283–289 10.1007/s11102-014-0569-6 [DOI] [PubMed] [Google Scholar]
- 169.Omar MH, Byrne DP, Shrestha S, Lakey TM, Lee KS, Lauer SM et al. (2024) Discovery of a Cushing’s syndrome protein kinase A mutant that biases signaling through type I AKAPs. Sci. Adv 10, eadl1258 10.1126/sciadv.adl1258 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Tatsi C, Faucz FR, Blavakis E, Carneiro BA, Lyssikatos C, Belyavskaya E et al. (2019) Somatic PRKAR1A gene mutation in a nonsyndromic metastatic large cell calcifying Sertoli cell tumor. J. Endocr. Soc 3, 1375–1382 10.1210/js.2019-00022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Casey M, Vaughan CJ, He J, Hatcher CJ, Winter JM, Weremowicz S et al. (2000) Mutations in the protein kinase A R1alpha regulatory subunit cause familial cardiac myxomas and Carney complex. J. Clin. Invest 106, R31–R38 10.1172/JCI10841 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Guerra MS, Santos N, Neves F, Carlos Mota J, Miranda JA and Vouga L (2006) [Carney’s complex: familial cardiac myxoma]. Rev. Port. Cir. Cardiotorac. Vasc 13, 79–81 [PubMed] [Google Scholar]
- 173.Ma S, Liu W, Zhang A, Pan L, Tang W, Jiang B et al. (2019) Identification of a PRKAR1A mutation (c.491_492delTG) in familial cardiac myxoma: a case report. Medicine 98, e14866 10.1097/MD.0000000000014866 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Omar MH, Lauer SM, Lau HT, Golkowski M, Ong SE and Scott JD (2023) Proximity biotinylation to define the local environment of the protein kinase A catalytic subunit in adrenal cells. STAR Protoc. 4, 101992 10.1016/j.xpro.2022.101992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Veugelers M, Wilkes D, Burton K, McDermott DA, Song Y, Goldstein MM et al. (2004) Comparative PRKAR1A genotype-phenotype analyses in humans with Carney complex and prkar1a haploinsufficient mice. Proc. Natl Acad. Sci. U.S.A 101, 14222–14227 10.1073/pnas.0405535101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Golden SH, Robinson KA, Saldanha I, Anton B and Ladenson PW (2009) Clinical review: prevalence and incidence of endocrine and metabolic disorders in the United States: a comprehensive review. J. Clin. Endocrinol. Metab 94, 1853–1878 10.1210/jc.2008-2291 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Hernandez-Ramirez LC and Stratakis CA (2018) Genetics of Cushing’s syndrome. Endocrinol. Metab. Clin. North Am 47, 275–297 10.1016/j.ecl.2018.02.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Stratakis CA (2013) cAMP/PKA signaling defects in tumors: genetics and tissue-specific pluripotential cell-derived lesions in human and mouse. Mol. Cell. Endocrinol 371, 208–220 10.1016/j.mce.2013.01.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Stratakis CA (2018) Cyclic AMP-dependent protein kinase catalytic subunit A (PRKACA): the expected, the unexpected, and what might be next. J. Pathol 244, 257–259 10.1002/path.5014 [DOI] [PubMed] [Google Scholar]
- 180.Stratakis CA (2008) Cushing syndrome caused by adrenocortical tumors and hyperplasias (corticotropin- independent Cushing syndrome). Endocr. Dev 13, 117–132 10.1159/000134829 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Horvath A and Stratakis CA (2008) Unraveling the molecular basis of micronodular adrenal hyperplasia. Curr. Opin. Endocrinol. Diabetes Obes 15, 227–233 10.1097/MED.0b013e3282fe7416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Bathon K, Weigand I, Vanselow JT, Ronchi CL, Sbiera S, Schlosser A et al. (2019) Alterations in protein kinase A substrate specificity as a potential cause of Cushing syndrome. Endocrinology 160, 447–459 10.1210/en.2018-00775 [DOI] [PubMed] [Google Scholar]
- 183.Omar MH, Kihiu M, Byrne DP, Lee KS, Lakey TM, Butcher E et al. (2023) Classification of Cushing’s syndrome PKAc mutants based upon their ability to bind PKI. Biochem. J 480, 875–890 10.1042/BCJ20230183 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Espiard S and Bertherat J (2015) The genetics of adrenocortical tumors. Endocrinol. Metab. Clin. North Am 44, 311–334 10.1016/j.ecl.2015.02.004 [DOI] [PubMed] [Google Scholar]
- 185.Espiard S, Knape MJ, Bathon K, Assié G, Rizk-Rabin M, Faillot S et al. (2018) Activating PRKACB somatic mutation in cortisol-producing adenomas. JCI Insight 3, e98296 10.1172/jci.insight.98296 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Nikiforova MN, Kimura ET, Gandhi M, Biddinger PW, Knauf JA, Basolo F et al. (2003) BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas. J. Clin. Endocrinol. Metab 88, 5399–5404 10.1210/jc.2003-030838 [DOI] [PubMed] [Google Scholar]
- 187.Truong T, Sauter W, McKay JD, Hosgood HD III, Gallagher C, Amos CI et al. (2010) International Lung Cancer Consortium: coordinated association study of 10 potential lung cancer susceptibility variants. Carcinogenesis 31, 625–633 10.1093/carcin/bgq001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Webb EL, Rudd MF, Sellick GS, El Galta R, Bethke L, Wood W et al. (2006) Search for low penetrance alleles for colorectal cancer through a scan of 1467 non-synonymous SNPs in 2575 cases and 2707 controls with validation by kin-cohort analysis of 14 704 first-degree relatives. Hum. Mol. Genet 15, 3263–3271 10.1093/hmg/ddl401 [DOI] [PubMed] [Google Scholar]
- 189.Milne RL, Burwinkel B, Michailidou K, Arias-Perez JI, Zamora MP, Menendez-Rodriguez P et al. (2014) Common non-synonymous SNPs associated with breast cancer susceptibility: findings from the Breast Cancer Association Consortium. Hum. Mol. Genet 23, 6096–6111 10.1093/hmg/ddu311 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Jo YS, Kim MS, Yoo NJ and Lee SH (2016) Frameshift mutations of AKAP9 gene in gastric and colorectal cancers with high microsatellite instability. Pathol. Oncol. Res 22, 587–592 10.1007/s12253-016-0042-0 [DOI] [PubMed] [Google Scholar]
- 191.Kalluri R and Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J. Clin. Invest 119, 1420–1428 10.1172/JCI39104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Frank B, Wiestler M, Kropp S, Hemminki K, Spurdle AB, Sutter C et al. (2008) Association of a common AKAP9 variant with breast cancer risk: a collaborative analysis. J. Natl Cancer Inst 100, 437–442 10.1093/jnci/djn037 [DOI] [PubMed] [Google Scholar]
- 193.Caria P and Vanni R (2010) Cytogenetic and molecular events in adenoma and well-differentiated thyroid follicular-cell neoplasia. Cancer Genet. Cytogenet 203, 21–29 10.1016/j.cancergencyto.2010.08.025 [DOI] [PubMed] [Google Scholar]
- 194.Kabbarah O, Nogueira C, Feng B, Nazarian RM, Bosenberg M, Wu M et al. (2010) Integrative genome comparison of primary and metastatic melanomas. PLoS One 5, e10770 10.1371/journal.pone.0010770 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Yang MH, Hu ZY, Xu C, Xie LY, Wang XY, Chen SY et al. (2015) MALAT1 promotes colorectal cancer cell proliferation/migration/invasion via PRKA kinase anchor protein 9. Biochim. Biophys. Acta 1852, 166–174 10.1016/j.bbadis.2014.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Hu Z-Y, Liu Y-P, Xie L-Y, Wang X-Y, Yang F, Chen S-Y et al. (2016) AKAP-9 promotes colorectal cancer development by regulating Cdc42 interacting protein 4. Biochim. Biophys. Acta 1862, 1172–1181 10.1016/j.bbadis.2016.03.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Dong F, Felsmesser M, Casadevall A and Rubin CS (1998) Molecular characterization of a cDNA that encodes six isoforms of a novel murine A kinase anchor protein. J. Biol. Chem 273, 6533–6541 10.1074/jbc.273.11.6533 [DOI] [PubMed] [Google Scholar]
- 198.Gold MG, Reichow SL, O’Neill SE, Weisbrod CR, Langeberg LK, Bruce JE et al. (2012) AKAP2 anchors PKA with aquaporin-0 to support ocular lens transparency. EMBO Mol. Med 4, 15–26 10.1002/emmm.201100184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Gold MG, Fowler DM, Means CK, Pawson CT, Stephany JJ, Langeberg LK et al. (2013) Engineering A-kinase anchoring protein (AKAP)-selective regulatory subunits of protein kinase A (PKA) through structure-based phage selection. J. Biol. Chem 288, 17111–17121 10.1074/jbc.M112.447326 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.McKenzie AJ, Campbell SL and Howe AK (2011) Protein kinase A activity and anchoring are required for ovarian cancer cell migration and invasion. PLoS One 6, e26552 10.1371/journal.pone.0026552 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Li N, Kong J, Lin Z, Yang Y, Jin T, Xu M et al. (2019) Ezrin promotes breast cancer progression by modulating AKT signals. Br. J. Cancer 120, 703–713 10.1038/s41416-019-0383-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Dransfield DT, Bradford AJ, Smith J, Martin M, Roy C, Mangeat PH et al. (1997) Ezrin is a cyclic AMP-dependent protein kinase anchoring protein. EMBO J. 16, 101–109 10.1093/emboj/16.1.35 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PRJ, Reese CB et al. (1997) Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase B-alpha. Curr. Biol 7, 261–269 10.1016/S0960-9822(06)00122-9 [DOI] [PubMed] [Google Scholar]
- 204.Johnson JL, Yaron TM, Huntsman EM, Kerelsky A, Song J, Regev A et al. (2023) An atlas of substrate specificities for the human serine/ threonine kinome. Nature 613, 759–766 10.1038/s41586-022-05575-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Meng Y, Lu Z, Yu S, Zhang Q, Ma Y and Chen J (2010) Ezrin promotes invasion and metastasis of pancreatic cancer cells. J. Transl. Med 8, 61 10.1186/1479-5876-8-61 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Mak H, Naba A, Varma S, Schick C, Day A, Sengupta SK et al. (2012) Ezrin phosphorylation on tyrosine 477 regulates invasion and metastasis of breast cancer cells. BMC Cancer 12, 82 10.1186/1471-2407-12-82 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Horwitz V, Davidson B, Stern D, Trope CG, Tavor Re’em T and Reich R (2016) Ezrin is associated with disease progression in ovarian carcinoma. PLoS One 11, e0162502 10.1371/journal.pone.0162502 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Guo YJ, Pan WW, Liu SB, Shen ZF, Xu Y and Hu LL (2020) ERK/MAPK signalling pathway and tumorigenesis. Exp. Ther. Med 19, 1997–2007 10.3892/etm.2020.8454 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Liberti MV and Locasale JW (2016) The Warburg effect: how does it benefit cancer cells? Trends Biochem. Sci 41, 211–218 10.1016/j.tibs.2015.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Gabrovsek L, Collins KB, Aggarwal S, Saunders LM, Lau HT, Suh D et al. (2020) A-kinase-anchoring protein 1 (dAKAP1)-based signaling complexes coordinate local protein synthesis at the mitochondrial surface. J. Biol. Chem 295, 10749–10765 10.1074/jbc.RA120.013454 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Merrill RA and Strack S (2014) Mitochondria: a kinase anchoring protein 1, a signaling platform for mitochondrial form and function. Int. J. Biochem. Cell Biol 48, 92–96 10.1016/j.biocel.2013.12.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Livigni A, Scorziello A, Agnese S, Adornetto A, Carlucci A, Garbi C et al. (2006) Mitochondrial AKAP121 links cAMP and src signaling to oxidative metabolism. Mol. Biol. Cell 17, 263–271 10.1091/mbc.e05-09-0827 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Means CK, Lygren B, Langeberg LK, Jain A, Dixon RE, Vega AL et al. (2011) An entirely specific type I A-kinase anchoring protein that can sequester two molecules of protein kinase A at mitochondria. Proc. Natl Acad. Sci. U.S.A 108, E1227–E1235 10.1073/pnas.1107182108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Chang CR and Blackstone C (2007) Cyclic AMP-dependent protein kinase phosphorylation of Drp1 regulates its GTPase activity and mitochondrial morphology. J. Biol. Chem 282, 21583–21587 10.1074/jbc.C700083200 [DOI] [PubMed] [Google Scholar]
- 215.Cribbs JT and Strack S (2007) Reversible phosphorylation of Drp1 by cyclic AMP-dependent protein kinase and calcineurin regulates mitochondrial fission and cell death. EMBO Rep. 8, 939–944 10.1038/sj.embor.7401062 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Smirnova E, Griparic L, Shurland DL and van der Bliek AM (2001) Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Mol. Biol. Cell 12, 2245–2256 10.1091/mbc.12.8.2245 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Sprenger H-G and Langer T (2019) The good and the bad of mitochondrial breakups. Trends Cell Biol. 29, 888–900 10.1016/j.tcb.2019.08.003 [DOI] [PubMed] [Google Scholar]
- 218.Levine B and Kroemer G (2019) Biological functions of autophagy genes: a disease perspective. Cell 176, 11–42 10.1016/j.cell.2018.09.048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Whiting JL, Nygren PJ, Tunquist BJ, Langeberg LK, Seternes OM and Scott JD (2015) Protein kinase A opposes the phosphorylation-dependent recruitment of glycogen synthase kinase 3beta to A-kinase anchoring protein 220. J. Biol. Chem 290, 19445–19457 10.1074/jbc.M115.654822 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Whiting JL, Ogier L, Forbush KA, Bucko P, Gopalan J, Seternes OM et al. (2016) AKAP220 manages apical actin networks that coordinate aquaporin-2 location and renal water reabsorption. Proc. Natl Acad. Sci. U.S.A 113, E4328–E4337 10.1073/pnas.1607745113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Deng Z, Li X, Blanca Ramirez M, Purtell K, Choi I, Lu JH et al. (2021) Selective autophagy of AKAP11 activates cAMP/PKA to fuel mitochondrial metabolism and tumor cell growth. Proc. Natl Acad. Sci. U.S.A 118, e2020215118 10.1073/pnas.2020215118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Marsh NM, MacEwen MJS, Chea J, Kenerson HL, Kwong AA, Locke TM et al. (2024) Mitochondrial calcium signaling regulates branched-chain amino acid catabolism in fibrolamellar carcinoma. bioRxiv 10.1101/2024.05.27.596106 [DOI] [Google Scholar]
- 223.Sancak Y, Markhard AL, Kitami T, Kovacs-Bogdan E, Kamer KJ, Udeshi ND et al. (2013) EMRE is an essential component of the mitochondrial calcium uniporter complex. Science 342, 1379–1382 10.1126/science.1242993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Wang B, Zhang L, Dai T, Qin Z, Lu H, Zhang L et al. (2021) Liquid–liquid phase separation in human health and diseases. Signal Transduct. Target. Ther 6, 290 10.1038/s41392-021-00678-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.McSwiggen DT, Mir M, Darzacq X and Tjian R (2019) Evaluating phase separation in live cells: diagnosis, caveats, and functional consequences. Genes Dev. 33, 1619–1634 10.1101/gad.331520.119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Zhang JZ, Lu TW, Stolerman LM, Tenner B, Yang JR, Zhang JF et al. (2020) Phase separation of a PKA regulatory subunit controls cAMP compartmentation and oncogenic signaling. Cell 182, 1531–1544.e15 10.1016/j.cell.2020.07.043 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Bhullar KS, Lagarón NO, McGowan EM, Parmar I, Jha A, Hubbard BP et al. (2018) Kinase-targeted cancer therapies: progress, challenges and future directions. Mol. Cancer 17, 48 10.1186/s12943-018-0804-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Dar AC and Shokat KM (2011) The evolution of protein kinase inhibitors from antagonists to agonists of cellular signaling. Annu. Rev. Biochem 80, 769–795 10.1146/annurev-biochem-090308-173656 [DOI] [PubMed] [Google Scholar]
- 229.Caunt CJ, Sale MJ, Smith PD and Cook SJ (2015) MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road. Nat. Rev. Cancer 15, 577–592 10.1038/nrc4000 [DOI] [PubMed] [Google Scholar]
- 230.Eroglu Z and Ribas A (2016) Combination therapy with BRAF and MEK inhibitors for melanoma: latest evidence and place in therapy. Ther. Adv. Med. Oncol 8, 48–56 10.1177/1758834015616934 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Bischoff P, Trinks A, Obermayer B, Pett JP, Wiederspahn J, Uhlitz F et al. (2021) Single-cell RNA sequencing reveals distinct tumor microenvironmental patterns in lung adenocarcinoma. Oncogene 40, 6748–6758 10.1038/s41388-021-02054-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Dinh TA, Sritharan R, Smith FD, Francisco AB, Ma RK, Bunaciu RP et al. (2020) Hotspots of aberrant enhancer activity in fibrolamellar carcinoma reveal candidate oncogenic pathways and therapeutic vulnerabilities. Cell Rep. 31, 107509 10.1016/j.celrep.2020.03.073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Dinh TA, Vitucci EC, Wauthier E, Graham RP, Pitman WA, Oikawa T et al. (2017) Comprehensive analysis of The cancer Genome Atlas reveals a unique gene and non-coding RNA signature of fibrolamellar carcinoma. Sci. Rep 7, 44653 10.1038/srep44653 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Banko MR, Allen JJ, Schaffer BE, Wilker EW, Tsou P, White JL et al. (2011) Chemical genetic screen for AMPKalpha2 substrates uncovers a network of proteins involved in mitosis. Mol. Cell 44, 878–892 10.1016/j.molcel.2011.11.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Lawrence RT, Perez EM, Hernandez D, Miller CP, Haas KM, Irie HY et al. (2015) The proteomic landscape of triple-negative breast cancer. Cell Rep. 11, 990 10.1016/j.celrep.2015.04.059 [DOI] [PubMed] [Google Scholar]
- 236.Golkowski M, Lau HT, Chan M, Kenerson H, Vidadala VN, Shoemaker A et al. (2020) Pharmacoproteomics identifies kinase pathways that drive the epithelial-mesenchymal transition and drug resistance in hepatocellular carcinoma. Cell Syst. 11, 196–207.e7 10.1016/j.cels.2020.07.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Aissa AF, Islam A, Ariss MM, Go CC, Rader AE, Conrardy RD et al. (2021) Single-cell transcriptional changes associated with drug tolerance and response to combination therapies in cancer. Nat. Commun 12, 1628 10.1038/s41467-021-21884-z [DOI] [PMC free article] [PubMed] [Google Scholar]
