Abstract
While current treatment regimens for acute leukemia can dramatically improve patient survival, there remains room for improvement. Due to its roles in cell differentiation, cell survival, and apoptotic signaling, modulation of the cyclic AMP (cAMP) pathway has provided a meaningful target in hematological malignancies. Several studies have demonstrated that gene expression profiles associated with increased pro-survival cAMP activity or downregulation of various pro-apoptotic factors associated with the cAMP pathway are apparent in acute leukemia patients. Previous work to increase leukemia cell intracellular cAMP focused on the use of cAMP analogs, stimulating cAMP production via transmembrane-associated adenylyl cyclases, or decreasing cAMP degradation by inhibiting phosphodiesterase activity. However, targeting cyclic nucleotide efflux by ATP-binding cassette (ABC) transporters represents an unexplored approach for modulation of intracellular cyclic nucleotide levels. Preliminary studies have shown that inhibition of cAMP efflux can stimulate leukemia cell differentiation, cell growth arrest, and apoptosis, indicating that targeting cAMP efflux may show promise for future therapeutic development. Furthermore, inhibition of cyclic nucleotide transporter activity may also contribute multiple anticancer benefits by reducing extracellular pro-survival signaling in malignant cells. Hence, several opportunities for drug repurposing may exist for targeting cyclic nucleotide transporters.
Keywords: Acute myelogenous leukemia (AML); Acute lymphoblastic leukemia (ALL); 3’,5’-cyclic adenosine monophosphate (cAMP); ATP-binding cassette (ABC) transporters; Efflux, Inhibitors of cAMP efflux (ICE)
Acute leukemias — Novel therapeutics are urgently required
1.1. Biology and etiology of acute leukemias
While not the most prevalent malignancy in the United States, acute leukemia ranks among the top ten cancers in terms of both morbidity and mortality [1]. These hematological malignancies involve aberrant proliferation of blood cells with immature phenotypes. Acute leukemias are typically identified by the presence of > 20% blast cells in the peripheral blood or bone marrow [2]. Due to inherent characteristics related to a reduced differentiation state, and the ability to rapidly propagate, acute leukemias may have worse prognoses than chronic leukemias.
Acute myelogenous leukemia (AML) is most prominent in elderly adults over 65 years of age. Annually, 21,450 cases are diagnosed in the United States, and 10,920 cases succumb to the disease [1]. The five year overall survival for this disease is a disheartening 28.3% for adults [1] and about 60% for children [3]. T-cell lineage and B-cell precursor acute lymphoblastic leukemia (T-ALL and B-ALL) primarily affects children, adolescents and young adults, with about 6,000 new cases diagnosed and 1,500 deaths each year [4, 5]. This disease is considered > 80% curable, however there is much room for improvement, specifically for high-risk subtypes such as Ph-like ALL, leukemias harboring rearrangements of KMT2A gene at 11q23 or BCR-ABL1 translocations, etc. [6–8] Therefore, there is a strong need for the development of novel approaches to treat these malignancies and improve patient outcome.
The primary classification of acute leukemia is based on the presence of chromosomal abnormalities and translocations along with immunophenotyping. For example, 25–30% of B-ALL have hyperdiploidy [9], while a surprising 40–50% of AML are cytogenetically normal [2]. An analysis of primary AML samples determined an average of 13 mutated genes per sample [10]. It has also been reported that > 80% of B-ALL cases contain deletions within genes related to B cell development [11]. De novo B-ALL genomes also contain 10–20 gene coding mutations [12].
Most commonly, the etiology of acute leukemias is attributed to the combination of mutations that govern pro-survival signaling and reduce tumor suppressor genes [13]. Many factors are potentially implicated in leukemogenesis, and most include aberrancies that increase cell proliferation and induce differential arrest. Due to potential mutations, rapid growth, and deviant signaling, leukemia cells have the capacity to overcome normal mechanisms that would typically result in cell death.
1.2. Treatment paradigms in acute leukemias
Treatment for acute leukemia begins with induction therapy, wherein chemotherapeutic agents are used to induce remission of the disease. In this approach, the goals are to reduce the population of blast cells and/or induce them to differentiate. Most commonly, this stage of treatment involves drugs that are used in many other cancers and diseases, and hence their mechanisms of action are nonspecific for leukemia. These drugs are aimed at killing all cells that are proliferating by inducing DNA damage or interfering with cell replication mechanisms. For some AML and ALL patients, allogeneic stem cell transplants are a treatment option as well.
The standard of care for AML is the use of cytosine arabinoside (cytarabine; AraC) with an anthracycline (e.g., daunorubicin, idarubicin, doxorubicin) for 7–10 days [14]. Over 80% of patients can achieve a complete response with this treatment, but often relapse will occur over time [15]. This is followed by consolidation therapy, which is typically high dose AraC [16]. Refractory or relapsed AML is also treated with high dose AraC, or the FLAG (fludarabine, AraC, granulocyte colony-stimulating factor (G-CSF)) regimen, but these result in modest complete remission rates of 32% and 47.5%, respectively [17].
B-ALL induction therapy involves 4–6 weeks of a glucocorticoid (e.g., dexamethasone or prednisone), asparaginase, vincristine, and an anthracycline. This is followed by several months of consolidation and maintenance therapies, which consist of some induction agents, as well as 6-mercaptopurine or 6-thioguanine, or methotrexate [9]. Like AML, the time elapsed between remission and relapse was associated with better overall survival. In one longitudinal study of pediatric B-ALL patients, only 8.8% of participants relapsed within 5 years [18], although other reports have indicated relapse rates of about 15–20% [23]. Nonetheless, a small percentage of B-ALL patients are vulnerable to the development of extramedullary disease in the central nervous system [19].
For both AML and B-ALL, remission is often determined by the minimal residual disease in the bone marrow. Most commonly, this is classified as < 1 leukemic blast in 10,000–100,000 cells [9, 19]. The length of remission varies by patient, although some mutations and translocations have been associated with longer event free survival. For example, AML cases with NPM1 mutations are considered to have favorable risk, while those containing an internal tandem duplication of fms-related tyrosine kinase 3 (FLT3-ITD) have adverse risk [20]. In B-ALL, the ETV6-RUNX1 fusion protein is associated with better treatment response and overall good outcome [21]. Rearrangements of the mixed-lineage leukemia (KMT2A) gene are considered prognostically less favorable [7, 8, 21]. Despite the fact that these and other cytogenetic factors are characteristically associated with prognosis, many patient responses to treatment are individual, and even those with similar cytogenetic factors could have opposite responses to the same therapy.
The treatment paradigms for acute leukemias are not without side effects. Primarily due to their lack of selectivity for leukemic cells, leukemia chemotherapeutic agents (LCA) can cause systemic damage to the patient. Most notably, anthracyclines and other chemotherapeutics exhibit cardiotoxicity. This is probably because, like cancer cells, the cardiac muscle is highly metabolically active. Survivors of pediatric ALL are at increased risk for the long-term development of heart disease [22]. LCA are associated with damage to the liver and kidneys, as these are the primary organs for drug metabolism. Patients with ALL are also at risk for osteonecrosis [23, 24]. Furthermore, leukemia treatments have been related to long-term cognitive impairment, including memory, attention, and executive function deficits [25, 26]. Hence, the development of drugs that are more selective for malignant cells could seriously reduce the incidence of adverse side effects suffered by patients.
cAMP-dependent pathway - A meaningful target in hematological malignancies
2.1. The cellular roles of cAMP and its regulation
In 1958, the first second messenger in cells, 3’,5’-cyclic adenosine monophosphate (cAMP) [27], was identified. This molecule is critically important for all cells and is central to many cellular processes that regulate growth, survival, differentiation, and the transcription of a myriad of genes. cAMP signal transduction can result in activation of pro-survival or death pathways, depending upon cell type and conditions [28]. The primary downstream effectors of cAMP are protein kinase A (PKA) and exchange proteins activated by cAMP (EPAC). While these two effectors can activate multiple transcription factors, the one most commonly associated with cAMP pathway signaling is the cAMP response element binding protein (CREB). CREB activity is associated with transcription of many pro-survival genes, including cyclins A, D1, and D2 [29].
Canonical cAMP synthesis involves activation of any of nine transmembrane-associated adenylyl cyclases (tmAC), which are stimulated by Gαs-coupled receptors. These activated receptors are capable of stimulating cAMP synthesis from within endosomes [30]. Activation of Gαi-coupled receptors inhibits tmAC activity, and hence reduces intracellular cAMP (icAMP). tmACs are envisioned to be responsible for the regulation of cAMP level in the vicinity of the cell membrane. Additionally, the cAMP signaling is shown to be spatially segregated throughout the cytosolic compartment, where the second messenger can be generated by a different enzyme, a non-membrane associated soluble adenylyl cyclase (sAC; ADCY10). This enzyme is responsible for cAMP production in cytosolic microdomains, such as the cytoplasm, or within the nucleus or mitochondria [31–33]. Unlike tmACs, sAC is activated by bicarbonate (HCO3−) or oxidative stress [32, 34–37]. Elevated icAMP triggers intrinsic apoptosis by PKA signaling. PKA can activate the transcription factor cAMP response element binding (CREB) protein that triggers up-regulation of the expression of the pro-apoptotic protein Bim [38]. Additionally, sAC stimulation of PKA can cause translocation of pro-apoptotic Bax into mitochondria [39, 40]. All these signaling events can play a role in the activation of the mitochondria-related intrinsic apoptotic pathway.
icAMP concentrations can be modulated by two principal mechanisms. The most commonly studied cAMP regulators are phosphodiesterases (PDEs). These enzymes hydrolyze cyclic nucleotides. PDEs 1–4, 7, 8, 10, and 11 are capable of degrading cAMP to 5’-AMP [41]. The second mechanism implies active removal of cAMP from cytosol. The fact that cAMP can be released outside the cell (ecAMP) was first described in 1963 [42]. Decades later, it was determined that icAMP can be actively removed from cytosol via the ATP-binding cassette (ABC) transporters. The members of the multidrug resistance (MRP) family of transporters, ABCC4 (MRP4), ABCC5 (MRP5), and ABCC11 (MRP8) are reported to efflux cAMP [41]. In this review, we focus on the modulation of this process by small molecules.
Because cAMP activity occurs in discrete locations (microdomains), many cAMP-dependent regulatory proteins can be complexed to facilitate signal transduction. A-kinase anchoring proteins (AKAPs) are scaffolding proteins that allow adenylyl cyclases, PKA, and PDEs to be localized in close proximity to one another [43]. Hence, the compartmentalization of icAMP into microdomains near the plasma membrane, mitochondria, nucleus, or regulatory proteins, is a critical determinant of its concentration and activity [44–46] (Figure 1).
Figure 1. Basic schematic of cAMP compartmentalization and effectors.

cAMP (green) is produced by transmembrane (tmAC) or soluble (sAC) adenylyl cyclases. Intracellular cAMP (icAMP) is downregulated by PDEs or efflux by ABC transporters to the extracellular space (ecAMP). icAMP can activate the downstream effectors EPAC and PKA. ecAMP can activate ePKA, or it can be hydrolyzed by the enzymes CD39 and CD73 into adenosine, which can then stimulate its receptors. EPAC, exchange proteins activated by cAMP. Gαs-GPCR, stimulatory G-protein coupled receptor. PKA, protein kinase A. ROS, reactive oxygen species.
2.2. cAMP targets in acute leukemias
Considering that icAMP regulation is altered in hematopoietic malignancies, and that increased icAMP reduces white blood cell survival, the cAMP pathway has long been of interest as a target for leukemia therapeutics [28, 41, 47–49]. Many cAMP-associated proteins are dysregulated in acute leukemias. The primary tmAC expressed in lymphoid cells is the adenylyl cyclase type 7 (ADCY7) [50], and the expression of this protein inversely correlates with the overall survival of AML patients [51]. The overall PDE activity was reported to be 10–20-fold higher in certain leukemia and lymphoma cells as compared to normal blood cells [52]. It should also be noted that glucocorticoids, which are used to treat ALL, have been shown to downregulate PDE activity [53]. Of note, genomic studies on primary AML and adult ALL samples have reported overexpression of CREB and/or its active, phosphorylated form [54–59]. Another study identified downregulation of ICER (inducible cAMP early repressor), the physiological antagonist of CREB that is associated with tumor suppression activity [60]. In B-ALL samples, there is an increased incidence of mutations in transcripts for CREBBP, the binding protein for CREB. These CREBBP mutations were associated with dominant-negative or deleted activity [61, 62]. To summarize, acute leukemia cells demonstrate both increased pro-survival cAMP activity through increased CREB signaling, as well as downregulation of the pro-apoptotic factor ICER.
2.3. History of cAMP targeting in cancer with a focus on hematological malignancies
Most previous attempts to modulate cAMP for cancer therapeutics have focused on the use of cAMP analogs, or by targeting canonical proteins of the pathway through: 1) stimulation of cAMP production by Gαs-coupled receptors, or 2) inhibition of cAMP degradation by PDEs. These efforts have had modest success, and are briefly summarized here.
Few cAMP modulating agents targeting cancer have been tested in clinical trials. Despite demonstrating anticancer effects in vitro and in vivo, phase I clinical trials with the analog 8-Cl-cAMP in patients with refractory solid tumors resulted in hypocalcemia and toxicity to normal tissues [63–65]. A phase II clinical trial evaluated the PDE-inhibitor theophylline for chronic lymphoblastic leukemia (CLL) patients. Of the 25 CLL patients treated with theophylline, only one achieved a complete response, and 18 patients maintained a stable disease state [66].
Nonetheless, cAMP pathway modulation has been well investigated as a target in hematopoietic malignancies in vitro and in vivo. Elevation of icAMP using cAMP analogs has demonstrated the ability to induce cell cycle arrest (G1 or G2 phase), differentiation, and/or intrinsic apoptosis in leukemia, lymphoma, myeloma, and normal B cells in vitro, most often through PKA-mediated mechanisms [38, 67–73]. Similar results have been reported from studies where cAMP production was stimulated via activation of tmAC [74–77]. PDE inhibitors have also been employed to increase cAMP signaling in hematopoietic cells. Meyers, et al. provided evidence that PDE4 inhibition induced cell death in B-CLL cell lines, but not in normal B and T cells [50]. This selectivity is plausible, since leukemia and lymphoma cells exhibit elevated PDE expression and activity [49, 52, 78]. Mitton, et al. used small molecule CREB inhibitors to reduce the expression of pro-survival factors in AML cells in vitro and in vivo. They showed that this approach induced cell cycle arrest and apoptosis [59]. Nevertheless, it is important to note that elevated icAMP can also rescue blood cancer cells from apoptosis, although those studies involved activation of cAMP signaling after substantial DNA damage had occurred in the treated cells [79–84]. Consequently, there is still substantial evidence to support cAMP pathway targeting in hematological malignancies.
Targeting cAMP transporters/efflux – an unexplored approach to modulation of the cyclic nucleotide pathway
3.1. Cyclic nucleotide efflux as a previously overlooked target
While the intracellular concentrations of cyclic nucleotides are canonically regulated by degradation by PDE in microdomains [46], efflux mechanisms by ABC transporters are less explored. This class of proteins consists of 7 families (ABC-A through ABC-G), in which two molecules of ATP are used to translocate substrates into the extracellular space. As previously mentioned, cAMP and cGMP are transported by members of the ABCC (MRP) family, ABCC4 (MRP4), ABCC5 (MRP5), and ABCC11 (MRP8). These transporters are distributed variably throughout the body, with ABCC4 having the highest expression in the bladder, kidney, and prostate [85]. The affinity of these transporters varies for cAMP or cGMP. ABCC5 has a greater affinity for cGMP, whereas ABCC4 is preferential for cAMP [86]. ABCC4, ABCC5, and ABCC11 regulate xenobiotic metabolism, and they can efflux antiviral drugs (PMEA, ganciclovir) as well as chemotherapeutic agents (fluorouracil, mercaptopurine, thioguanine, campothecins, methotrexate). Importantly, these proteins also serve a role in the transport of natural, endogenous substrates. These include eicosanoids (e.g., prostaglandins E1 and E2, leukotriene C4), estradiol-17β-glucuronide, folic acid, bile acids (taurocholate, glycocholate), and some steroids (e.g., DHEAS) [87]. Furthermore, the cyclic nucleotide transporters also play a major role in relieving oxidative stress, as they remove glutathione conjugates from the cell [88].
Past work in which cAMP efflux was inhibited by small molecules was performed in epithelial cell lines, and primarily focused on cAMP concentration changes and the activity of the pathway-associated proteins [89, 90]. The efflux capacity of leukemia cells has been reported as an important factor for predicting patient outcomes [91]. However, the analysis of cAMP-specific efflux activity has not been extensive. The expression of ABCC4 is inversely correlated with hematopoietic cell differentiation [92]. Previous studies relied on the use of indiscriminate ABC transporter inhibitors (e.g., probenecid) or silencing RNA to block expression of cAMP effluxing proteins [93–95]. These approaches, however, lacked the efficiency and breadth necessary for resolving the utility of targeting cAMP efflux for cancer therapeutics.
Recently, our group initiated studies to further explore the role of cAMP efflux targeting in acute leukemias [96, 97]. We hypothesized that malignant cells produce and remove the excess icAMP from mitochondria-containing microdomains to evade intrinsic apoptosis and to promote cell survival [96]. Consequently, inhibition of cAMP efflux should increase icAMP and selectively trigger leukemia cell death. We proposed that small molecule cAMP efflux inhibition, if achieved using existing drugs through a repurposing strategy, has the potential to expedite the translation of cAMP efflux-targeting therapeutics [96]. Because elevated cAMP efflux activity could be a specific adaptation of malignant cells that is not apparent in normal cells, drugs developed using this approach have the potential to selectively target leukemia or other tumor cells.
To monitor cAMP efflux from living cells and primary patient samples, we designed and patented a novel assay that rely on cellular retention of a fluorescent cAMP analog (F-cAMP) [98]. This assay was later adapted for use in a high throughput flow cytometry platform [97]. After screening two chemical libraries consisting of off-patent FDA-approved drugs and biologically active compounds, we identified and validated six compounds termed “inhibitors of cAMP efflux” (ICE) [96, 97].
Using the F-cAMP-based approach, our group confirmed that cAMP efflux was undetectable in normal peripheral blood mononuclear cells (PBMCs) but was active in leukemia cell lines [96]. Several interpretations for this phenomenon can be considered. As mentioned before, the cAMP efflux could be a malignancy-specific adaptation directly related to the evasion of apoptosis in cancer. It can be also related to a metabolic specificity of leukemic cells (Warburg effect). cAMP is a well known regulator of cellular respiration, metabolism and an accumulation of cAMP in cancer cells can trigger metabolic reprogramming, mitochondrial biogenesis and anti-Warburg effect [99]. The fact that normal hematopoietic progenitors exhibit an increased expression of ABC transporters capable of cAMP efflux [92], a phenotype that is shared by blasts associated with acute leukemias [100], can indirectly support this idea.
Previously, Copsel et al., demonstrated that blocking cAMP efflux in an AML-relevant model system not only increased icAMP accumulation, but also triggered a series of downstream signaling events relevant to the pathway activation [93, 94]. We also validated the ability of newly identified ICE compounds to increase cAMP pathway activity by demonstrating its ability to modulate cellular downstream signaling. This included CREB (pS133) / ATF-1 (pS63) phosphorylation and CD49d/CD29 (VLA-4) integrin deactivation [96]. Active VLA-4 integrin is responsible for homing and retention of hematopoietic blasts and other cells in the bone marrow. Deactivation of this adhesion molecule by cyclic nucleotide-dependent signaling mechanism is crucial for mobilization of white blood cells into the peripheral blood [101, 102], and activation of VLA-4 integrin is known to be impaired in a subset of CLL patients [103]. Our experiments further demonstrated that the ability of ICE to reduce cell viability was partially dependent on the activity of sAC, a source for cytosolic, mitochondrial, and nuclear cAMP production [36, 104]. Furthermore, ICE reduced the viability of leukemia cell lines and ex vivo primary patient samples at much lower concentrations than required for PBMCs, indicating that cAMP efflux inhibition could be a feasible target for malignant cells. Because several identified ICE are FDA approved drugs, these studies provide a potential path for drug repurposing against leukemias [96].
We also compared ABCC4 expression in several leukemic cell lines and determined that there was no apparent relationship between antibody binding and the F-cAMP efflux ability [96]. The simplest explanation here is the possibility that transporter activity is regulated by protein modification, such as phosphorylation, for example. The best-studied member of ABCC subfamily, ABCC7, was shown to be phosphorylated by PKA and this directly affects its activity [105]. Another possibility is that ICE may act by some alternative mechanism(s) unrelated to transporter inhibition per se. Finally, the work by Guo, et al. previously showed that, of the known cAMP efflux transporters, only the expression of ABCC11 was prognostically relevant in leukemia [106]. Other studies have indicated the possibility for the ABCC1 and ABCG2 transporters to efflux cyclic nucleotides [95, 107]. It is therefore plausible that acute leukemia cells may reduce intracellular cAMP using ABCC5, ABCC11, or other MRP transporters.
It should be emphasized that the cross-competition between transporter substrates can theoretically perturb transport activity resulting in the inhibition of one with another. Hence, it is possible that some molecules identified as inhibitors of a given transporter may instead reduce the efflux of other substrates, without actually blocking the activity of that transporter. Extensive mechanistic studies are needed to validate the mechanism of action of efflux inhibitors on transporters. It should be noted that all substrates of a transporter have the potential to inhibit the transport other substrates of the same transporter. Therefore, molecules that affect similar pathways could initially be identified as efflux inhibitors, but may in fact potentially have mechanism(s) of action that are unrelated to antagonism of transport proteins.
3.2. ICE and distinct intracellular cAMP compartments. Design of cAMP signalosomes
cAMP signaling is highly compartmentalized and responses to cAMP are spatially and temporary restricted. Thus, it is necessary to understand the effects of ICE on major cellular functions, such as cell proliferation, differentiation and cell death. This should be done in the context of cAMP signalosomes, large multi-protein complexes that consist of key players of the cAMP signaling pathway. A representative cAMP signalosome includes several components. Scaffold AKAP proteins provide binding sites for all other signalosome components, and organize as well as target the complex to a specific cellular location. For example, AKAPs that target signalosomes to the plasma membrane are AKAP7, the small-membrane AKAP (smAKAP, C2orf88) [108]. AKAP1 (D-AKAP1, S-AKAP84, AKAP121) target signalosomes to the outer mitochondrial membrane or the endoplasmic reticulum [109, 110] and AKAP6 (mAKAP), one of the proteins that targets signalosomes to the nuclear membrane [111]. Other components of the signalosome include cAMP-dependent protein kinase PKA, and the PDE responsible for the degradation of cAMP and other proteins. This specific signalosome organization that places proteins in close proximity to one another is envisioned to provide tight regulation of cAMP/PKA signaling in specific cellular compartment. In resting cells, PDEs maintain a low concentration of localized cAMP that is insufficient to activate PKA. Upon activation of AC, increased cAMP production triggers PKA activity. The subsequent phosphorylation of PDE by PKA stimulates cAMP hydrolysis and returns cAMP levels to the resting state. Disruption of the interaction between PKA and AKAP blocks PKA induced PDE activation [112]. This suggests that spatial organization of the signalosome, where proteins are anchored in proximity to one another is essential for proper functioning of the signaling mechanism.
To decipher how ICE may affect distinct cellular compartments, here we focus on known functions of the PKA/cAMP pathways in mitochondrial, nuclear, and plasma membrane as well as cytosolic signalosomes.
3.2.1. Mitochondrial cAMP-PKA signaling
Several excellent reviews that describe cAMP signaling in mitochondrial compartment were recently published [113, 114]. Here we will briefly focus on main functions of cAMP-dependent processes that are important for understanding the potential effects of ICE on mitochondria. The outer mitochondrial membrane is permeable for small molecules [115], and therefore, cAMP produced by tmACs and sAC in the cytosol can reach the intermembrane mitochondrial space. However, the inner mitochondrial membrane is impermeable to cAMP. As mentioned above sAC could be a source of cAMP in the mitochondrial matrix [31, 32].
In mitochondria, oxidative phosphorylation (OXPHOS) [32], protein import into mitochondria [116–118], mitophagy, mitochondrial fission and fusion [113], metabolic reprogramming and the anti-Warburg effect [99], as well as the intrinsic apoptotic pathway [38], can all be modulated by the cAMP-PKA-related signaling pathway. However, it seems that the majority of these processes can be affected by cAMP diffusing to the outer mitochondrial membrane. Only OXPHOS and metabolic reprogramming/anti-Warburg effect require cAMP level changes in the mitochondrial matrix. As ABCC transporters that represent a potential target for ICE are expressed in the plasma membrane, it is expected that blocking cAMP efflux can have direct effects on protein import, mitophagy and mitochondria-induced apoptosis. Our recent data suggest that in leukemic cells ICE can trigger four apoptotic endpoints including mitochondrial membrane depolarization and activation of effector caspases 3 and 7 [96]. Thus, it is plausible that cAMP produced by tmAC can reach the outer mitochondrial membrane and trigger AML cells apoptosis.
We also found an unexpected connection to potential dysregulation of protein import in adult AML patient samples. Our analysis of several related gene expression clusters associated with poor outcome, worse overall survival and highest rates of resistant disease revealed enrichment for genes related to transport across membranes (transporters, carriers and channels). One of the identified genes was TOMM7, the mitochondrial import receptor subunit translocase of outer membrane (TOM) homolog [119]. This protein regulates assembly and stability of the main translocase complex [120]. The up-regulation of TOM7 in AML cells may be related to aberrant protein import in cancer. It is envisioned that defects in protein translocation into mitochondria can be devastating for organelle function, since the TOM complex provides an entry point for 99% of all precursor proteins in mitochondria [121, 122]. While cAMP-PKA signaling can diminish import of proteins through TOM and triggers the switch from OXPHOS to glycolysis [114], it is not known whether it can be directly related to the defects of the TOM system in leukemia. Does cytosolic sAC, activated by HCO3−, low pH or oxidative stress [32, 34–37], produce cAMP that triggers mitochondrial protein import dysfunction in cancer? How these changes are related to the Warburg effect? All these questions should be examined in the future.
3.2.1. Plasma membrane and cytosolic compartments
The plasma membrane and cytosolic compartments are the most studied among all cAMP-PKA-related sites. Before the concept of cAMP signaling compartmentalization was introduced, it was assumed that the main site of cAMP-PKA signaling was the plasma membrane and cytosol. Numerous reviews of the field exist. Thus, here we discuss a few novel and previously unrecognized players.
As mentioned above, tmAC activity is controlled by two types of GPCRs, Gαs-coupled (stimulating cAMP production) and Gαi-coupled (inhibiting) receptors, the majority of which are localized to the plasma membrane. cAMP that is produced by tmACs diffuses in cytosol. CXCR4 (Gαi-coupled) and CXCR7 (a non-classical GPCR that can form heterodimers with CXCR4, acting as scavengers for the CXCR4 ligand CXCL12) are both relevant to leukemia and other cancers [123]. The idea that elevated signaling thorough Gαs-coupled H2-histamine receptor can be beneficial for treatment of AML [124], supports the notion that cAMP elevation in the cytosolic compartment using ICE is also beneficial [96]. Unfortunately, in some myeloid cells despite the elevation of the cAMP level that resulted from H2-histamine receptor stimulation, cell differentiation failed to occur. To explain this discrepancy, the authors suggested that cAMP efflux through MRP transporters could be modulating the cAMP level [124]. This notion evokes the suggestion that combining ICE with H2-histamine agonists could be an option to overcome this problem. Another report that shows an improvement in immune checkpoint therapy by activating the Gαs-coupled G protein-coupled estrogen receptor (GPER) indirectly supports this idea [125].
Hematological malignancies are characterized by the presence of a substantial number of cells exhibiting immature phenotypes. In acute leukemias up to one fifth or more of total cells can show blast morphology in the peripheral blood [2]. This reinforces the idea that leukemic cells possess a defect in their adhesion molecules, resulting in a premature release from the bone marrow niche into the peripheral blood. VLA-4 integrin is critical for homing and retention of hematopoietic blasts in the bone marrow niche, and it can be inactivated by a cyclic nucleotide-dependent signaling mechanism [101, 102]. The finding that the cytoplasmic domain of the VLA-4 subunit (alpha4, CD49d) serves as a type I PKA-specific AKAP puts VLA-4 integrins right at the heart of the plasma membrane cAMP signalosome. Sequestering type I PKA away from VLA-4 dramatically reduced VLA-4 subunit phosphorylation and inhibited VLA-4 dependent migration toward CXCL12 [126].
The role of the VLA-4-PKA protein complex in leukemia pathogenesis is not known. Similarly to VLA-4, the cAMP efflux transporters (a likely target for ICE compounds) are localized in the plasma membrane. Thus, they can also play the roles in the regulation of plasma membrane cAMP signalosome. The fact that the effect of ICE on VLA-4 functional activity can be detected within seconds after compounds addition suggests a very close association between these signaling complexes (see Figure 4 in [96]). It is also possible that blocking cAMP efflux at the plasma membrane results in cAMP accumulation and subsequent diffusion toward the mitochondrial outer membrane, where it can affect PKA activity, modulate protein import and trigger apoptosis.
3.2.3. Nuclear compartment
CREB and activating transcription factor-1 (ATF-1) are the classical cAMP effectors that activate target genes through cAMP response elements (CRE). This pathway is directly implicated in cAMP-induced apoptosis in leukemia [38]. Because the mitochondrial proteome largely originates from nuclear DNA, CREB together with downstream transcription factors, peroxisome proliferator-activated receptor γ coactivator-1 (PGC-1α) and nuclear respiratory factors (NRF), activates transcription of numerous mitochondrial genes and promotes biogenesis of mitochondria [127, 128].
There are multiple indications of aberrant CREB function in leukemia. CREB is shown to be overexpressed in primary samples obtained from AML and ALL patients [29, 129]. CREB expression was associated with increased risk of relapse and lower rate of the event-free survival [130]. CREB inactivation inhibited AML cell proliferation and triggered apoptosis, while had no significant effect on normal hematopoietic stem cells [54], prompting the development of small molecule modulators of CREB [131]. A recent report shows that a small molecule inhibitor of CREB, XX-650–23 blocked interaction between CREB and its co-activator CBP, and was capable of triggering apoptosis, and cell-cycle arrest in AML cells. Moreover, it increased mice survival in human AML xenograft models, suggesting that targeting CBP–CREB interaction may represent a novel approach for AML therapy [59].
In our experiments, ICE were capable of increasing CREB (pS133) phosphorylation [96], which is known to induce the formation of a complex between CREB and CBP [132]. These findings seem to tilt the signaling balance towards CREB pathway activation, which in AML cells is interpreted as pro-survival signal. ICE were capable of inducing the loss of mitochondrial membrane potential within 2 h after treatment [96]. We proposed that ICE-induced the elevation of cAMP targeted primarily the mitochondrial cAMP-PKA compartment resulting in initiation of the intrinsic apoptotic pathway [38]. CREB (pS133) phosphorylation was simply an indicator of activation in another cAMP-dependent pathway.
To summarize, ABCC4, ABCC5 and ABCC11 are localized in the plasma membrane. Therefore, targeting cAMP efflux by ICE is expected to affect intracellular compartments that are spatially adjacent to this compartment. Also, it is anticipated that the cAMP compartment that provides easy access to cytosolically produced cAMP would be primarily affected by diminished cAMP efflux. Thus, ICE are expected to target processes that can be modulated by plasma membrane and cytosol, outer mitochondrial membrane and nuclear signalosomes.
3.3. Cyclic nucleotide transporter inhibition potentially provides multiple anticancer benefits
Because the inhibition of cyclic nucleotide efflux obviously results in increased intracellular concentrations of transporter substrates, it can be expected that signaling processes will be affected accordingly. As previously mentioned, elevated icAMP signaling can result in the activation of the intrinsic apoptotic pathway. Conversely, extracellular cAMP (ecAMP) can result in autocrine and paracrine signaling. ecAMP has been shown to reduce immune cell activation [133], and thus cAMP efflux may provide another survival advantage for malignant cells by allowing them to avoid clearance by a normal immune response. Furthermore, Bhang, et al. have demonstrated that extracellular PKA, a downstream effector of ecAMP, was significantly elevated in the sera of dogs with cancer in comparison to healthy dogs or those with other diseases [134]. The authors do not speculate as to the purpose of extracellular PKA, although others have indicated that it may aid in the activation of prostaglandin H synthase [135], a factor related to cancer angiogenesis [136].
Another interesting aspect of ecAMP signaling relates to the existence of an ecAMP/adenosine pathway. Here, extracellular cAMP, which presumably has exited the cell through an efflux transporter (or leaked passively), is converted into adenosine by the activity of the ectonucleoside triphosphate diphosphohydrolase-1 (ENTPDase1) and ecto-5’-nucleotidase (NT5E), CD39 and CD73, respectively [137]. Some researchers have suggested that this pathway exists because free adenosine has a very short half-life, whereas ecAMP is stable in plasma, allowing it to participate in regulated signaling processes [138].
The adenosine produced by the hydrolysis of ecAMP can play many roles. The adenosine generated by CD73 activity is associated with increased tumor growth and suppression of normal immune responses [139–142]. Leukocytes express the adenosine receptor A2A (A2AR), a Gαs-coupled receptor [143]. The activation of A2AR can induce several downstream signaling cascades. Importantly, A2AR can stimulate a positive feedback mechanism where cAMP production, cAMP efflux through ABCC4, and ecAMP hydrolysis can promote more A2A stimulation [144, 145]. As such, inhibition of A2A receptors is capable of inhibiting the growth and metastasis of tumor cells [146–150].
It is conceivable that leukemia cells exploit this ecAMP/adenosine pathway activity, though the linkage is not straightforward. Expression of ectonucleotidases may be triggered by hypoxia [151], which is a key characteristic of the bone marrow microenvironment. An analysis of whole blood from patients indicated that CD39 was expressed on > 90% of both normal and malignant B-cells, and 8% of normal T cells. Elevated CD39 activity was associated with earlier stages of CLL, whereas decreased CD39 was associated with worse disease [152]. Inhibition of CD39 activity has been proposed as a potential anticancer target [153–155]. Another study has shown that 32% of CLL patients expressed CD73, and that this enzyme was associated with reduced response to therapy [156]. Similarly, CD73 expression on ALL cells was significantly related to expression of CD10, a protein associated with progenitor phenotypes [157]. Moreover, gene expression analysis of a drug resistant T-ALL cell line identified increased CD73 expression and resistance to receptor-mediated apoptosis, although the authors hypothesized that a direct interaction between CD73 and the cell death receptor, rather than CD73 enzymatic activity, may have been the mechanism of action [158]. Furthermore, CD73 appeared to have no prognostic value in predicting pediatric ALL response to therapy [159].
Another substrate of cyclic nucleotide transporters merits mention here, prostaglandin E2 (PGE2). PGE2 itself is associated with pro-survival signaling, through its binding to the Gαs-coupled receptors EP2 (PTGER2) and EP4 (PTGER4). Here, the downstream signaling is analogous to A2AR. Acute leukemia blast cells express EP2 [160], hypothetically in response to PGE2 secreted by bone marrow mesenchymal cells [161]. In principle, PGE2 could inhibit bone formation via increased icAMP signaling through EPAC, allowing for remodeling of the bone marrow microenvironment [162]. We propose that PGE2-mediated signaling may occur in an autocrine manner, by the PGE2 effluxed by cyclic nucleotide transporters.
Conclusions
This review has highlighted the need for new treatment regimens for acute leukemias. We have described the vital roles that cAMP signaling plays in the regulation of cell proliferation, survival, and apoptosis. We provided justification for targeting the cAMP pathway, since pathway-associated proteins are dysregulated in these malignancies. We also introduced the concept that the reduction of cyclic nucleotide efflux activity could potentially provide multiple anticancer benefits.
Because chemotherapy resistance can occur by cAMP efflux transporters [106, 163], there is a possibility of using cAMP in combination with current therapeutics to produce synergistic effects against leukemias. Synergism produced by these drug combinations would be beneficial for decreasing leukemia cell burden, and potentially to reduce the likelihood of resistance and/or relapse. Because several FDA approved drugs showed ICE activity, an opportunity for an accelerated introduction of novel therapeutics thorough a drug repurposing mechanism represents an exciting possibility [96].
In sum, this review could provide support for a new class of antileukemia drugs, cAMP efflux inhibiting compounds, to be tested in clinical trials. This new approach would be significant because it could substantiate inhibition of efflux as a paradigm shift in cAMP pathway targeting for cancer therapeutics.
Acknowledgments
This work was supported by the National Institutes of Health (USA) grants: NCI 1R01CA237165-01A1 (KMW), UNM Comprehensive Cancer Center CCSG P30 CA118100 grant, the UNM Autophagy Inflammation and Metabolism CoBRE P20 GM121176 grant, the UNM Clinical and Translational Science Center grant UL1TR001449 and NIH Minority Institutional Research Training Program Award T32 HL007736.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest statement
The authors declare that there are no conflicts of interest.
References
- [1].Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA, SEER Cancer Statistics Review, 1975–2016, National Cancer Institute, Bethesda, MD, 2019. [Google Scholar]
- [2].Kansal R, Acute myeloid leukemia in the era of precision medicine: recent advances in diagnostic classification and risk stratification, Cancer Biol Med 13(1) (2016) 41–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Perry AM, Attar EC, New insights in AML biology from genomic analysis, Semin Hematol 51(4) (2014) 282–97. [DOI] [PubMed] [Google Scholar]
- [4].Cancer Facts & Figures 2019, in: A.C. Society (Ed.) American Cancer Society, Atlanta, 2019. [Google Scholar]
- [5].Roberts KG, Mullighan CG, Genomics in acute lymphoblastic leukaemia: insights and treatment implications, Nat Rev Clin Oncol 12(6) (2015) 344–57. [DOI] [PubMed] [Google Scholar]
- [6].Hunger SP, Mullighan CG, Redefining ALL classification: toward detecting high-risk ALL and implementing precision medicine, Blood 125(26) (2015) 3977–3987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Kang H, Wilson CS, Harvey RC, Chen I-M, Murphy MH, Atlas SR, Bedrick EJ, Devidas M, Carroll AJ, Robinson BW, Stam RW, Valsecchi MG, Pieters R, Heerema NA, Hilden JM, Felix CA, Reaman GH, Camitta B, Winick N, Carroll WL, Dreyer ZE, Hunger SP, Willman CL, Gene expression profiles predictive of outcome and age in infant acute lymphoblastic leukemia: a Children’s Oncology Group study, Blood 119(8) (2012) 1872–1881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Matlawska-Wasowska K, Kang H, Devidas M, Wen J, Harvey RC, Nickl CK, Ness SA, Rusch M, Li Y, Onozawa M, Martinez C, Wood BL, Asselin BL, Chen IM, Roberts KG, Baruchel A, Soulier J, Dombret H, Zhang J, Larson RS, Raetz EA, Carroll WL, Winick NJ, Aplan PD, Loh ML, Mullighan CG, Hunger SP, Heerema NA, Carroll AJ, Dunsmore KP, Winter SS, MLL rearrangements impact outcome in HOXA-deregulated T-lineage acute lymphoblastic leukemia: a Children’s Oncology Group Study, Leukemia 30 (2016) 1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Hunger SP, Mullighan CG, Acute Lymphoblastic Leukemia in Children, N Engl J Med 373(16) (2015) 1541–52. [DOI] [PubMed] [Google Scholar]
- [10].Genomic and Epigenomic Landscapes of Adult De Novo Acute Myeloid Leukemia, New England Journal of Medicine 368(22) (2013) 2059–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Fiegl M, Epidemiology, pathogenesis, and etiology of acute leukemia, (2016) 3–13. [Google Scholar]
- [12].Ma X, Edmonson M, Yergeau D, Muzny DM, Hampton OA, Rusch M, Song G, Easton J, Harvey RC, Wheeler DA, Ma J, Doddapaneni H, Vadodaria B, Wu G, Nagahawatte P, Carroll WL, Chen IM, Gastier-Foster JM, Relling MV, Smith MA, Devidas M, Guidry Auvil JM, Downing JR, Loh ML, Willman CL, Gerhard DS, Mullighan CG, Hunger SP, Zhang J, Rise and fall of subclones from diagnosis to relapse in pediatric B-acute lymphoblastic leukaemia, Nat Commun 6 (2015) 6604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Kelly LM, Gilliland DG, Genetics of myeloid leukemias, Annual Review of Genomics and Human Genetics 3(1) (2002) 179–198. [DOI] [PubMed] [Google Scholar]
- [14].Emadi A, Karp JE, The clinically relevant pharmacogenomic changes in acute myelogenous leukemia, Pharmacogenomics 13(11) (2012) 1257–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Walter RB, Othus M, Burnett AK, Löwenberg B, Kantarjian HM, Ossenkoppele GJ, Hills RK, Ravandi F, Pabst T, Evans A, Pierce SR, Vekemans MC, Appelbaum FR, Estey EH, Resistance prediction in AML: analysis of 4601 patients from MRC/NCRI, HOVON/SAKK, SWOG and MD Anderson Cancer Center, Leukemia 29 (2014) 312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].De Kouchkovsky I, Abdul-Hay M, ‘Acute myeloid leukemia: a comprehensive review and 2016 update’, Blood Cancer J 6(7) (2016) e441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Ramos NR, Mo CC, Karp JE, Hourigan CS, Current Approaches in the Treatment of Relapsed and Refractory Acute Myeloid Leukemia, J Clin Med 4(4) (2015) 665–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Vora A, Goulden N, Wade R, Mitchell C, Hancock J, Hough R, Rowntree C, Richards S, Treatment reduction for children and young adults with low-risk acute lymphoblastic leukaemia defined by minimal residual disease (UKALL 2003): a randomised controlled trial, The Lancet Oncology 14(3) (2013) 199–209. [DOI] [PubMed] [Google Scholar]
- [19].Goto H, Childhood relapsed acute lymphoblastic leukemia: Biology and recent treatment progress, Pediatr Int 57(6) (2015) 1059–66. [DOI] [PubMed] [Google Scholar]
- [20].Grimwade D, Knapper S, Mrózek K, Acute Myeloid Leukemia, (2016) 527–559. [Google Scholar]
- [21].Moorman AV, Ensor HM, Richards SM, Chilton L, Schwab C, Kinsey SE, Vora A, Mitchell CD, Harrison CJ, Prognostic effect of chromosomal abnormalities in childhood B-cell precursor acute lymphoblastic leukaemia: results from the UK Medical Research Council ALL97/99 randomised trial, The Lancet Oncology 11(5) (2010) 429–438. [DOI] [PubMed] [Google Scholar]
- [22].Printz C, Adult survivors of childhood and adolescent cancer have more heart disease, Cancer 116(11) (2010) 2507–2507. [DOI] [PubMed] [Google Scholar]
- [23].Kadan-Lottick NS, Dinu I, Wasilewski-Masker K, Kaste S, Meacham LR, Mahajan A, Stovall M, Yasui Y, Robison LL, Sklar CA, Osteonecrosis in Adult Survivors of Childhood Cancer: A Report From the Childhood Cancer Survivor Study, Journal of Clinical Oncology 26(18) (2008) 3038–3045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Te Winkel ML, Pieters R, Wind EJ, Bessems JH, van den Heuvel-Eibrink MM, Management and treatment of osteonecrosis in children and adolescents with acute lymphoblastic leukemia, Haematologica 99(3) (2014) 430–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Krull KR, Brinkman TM, Li C, Armstrong GT, Ness KK, Srivastava DK, Gurney JG, Kimberg C, Krasin MJ, Pui CH, Robison LL, Hudson MM, Neurocognitive outcomes decades after treatment for childhood acute lymphoblastic leukemia: a report from the St Jude lifetime cohort study, J Clin Oncol 31(35) (2013) 4407–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Duffner PK, Armstrong FD, Chen L, Helton KJ, Brecher ML, Bell B, Chauvenet AR, Neurocognitive and neuroradiologic central nervous system late effects in children treated on Pediatric Oncology Group (POG) P9605 (standard risk) and P9201 (lesser risk) acute lymphoblastic leukemia protocols (ACCL0131): a methotrexate consequence? A report from the Children’s Oncology Group, J Pediatr Hematol Oncol 36(1) (2014) 8–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Rall TW, Sutherland EW, Formation of a cyclic adenine ribonucleotide by tissue particles, J Biol Chem 232(2) (1958) 1065–76. [PubMed] [Google Scholar]
- [28].Insel PA, Zhang L, Murray F, Yokouchi H, Zambon AC, Cyclic AMP is both a pro-apoptotic and anti-apoptotic second messenger, Acta Physiol (Oxf) 204(2) (2012) 277–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Cho EC, Mitton B, Sakamoto KM, CREB and leukemogenesis, Crit Rev Oncog 16(1–2) (2011) 37–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Antoni FA, New paradigms in cAMP signalling, Mol Cell Endocrinol 353(1–2) (2012) 3–9. [DOI] [PubMed] [Google Scholar]
- [31].Tresguerres M, Levin LR, Buck J, Intracellular cAMP signaling by soluble adenylyl cyclase, Kidney Int 79(12) (2011) 1277–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Acin-Perez R, Salazar E, Kamenetsky M, Buck J, Levin LR, Manfredi G, Cyclic AMP produced inside mitochondria regulates oxidative phosphorylation, Cell Metab 9(3) (2009) 265–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Zippin JH, Farrell J, Huron D, Kamenetsky M, Hess KC, Fischman DA, Levin LR, Buck J, Bicarbonate-responsive “soluble” adenylyl cyclase defines a nuclear cAMP microdomain, J Cell Biol 164(4) (2004) 527–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Steegborn C, Structure, mechanism, and regulation of soluble adenylyl cyclases - similarities and differences to transmembrane adenylyl cyclases, Biochim Biophys Acta 1842(12 Pt B) (2014) 2535–47. [DOI] [PubMed] [Google Scholar]
- [35].Chen YQ, Cann MJ, Litvin TN, Iourgenko V, Sinclair ML, Levin LR, Buck J, Soluble adenylyl cyclase as an evolutionarily conserved bicarbonate sensor, Science 289(5479) (2000) 625–628. [DOI] [PubMed] [Google Scholar]
- [36].Ladilov Y, Appukuttan A, Role of soluble adenylyl cyclase in cell death and growth, Biochim Biophys Acta 1842(12 Pt B) (2014) 2646–55. [DOI] [PubMed] [Google Scholar]
- [37].Kumar S, Appukuttan A, Maghnouj A, Hahn S, Peter Reusch H, Ladilov Y, Suppression of soluble adenylyl cyclase protects smooth muscle cells against oxidative stress-induced apoptosis, Apoptosis 19(7) (2014) 1069–79. [DOI] [PubMed] [Google Scholar]
- [38].Huseby S, Gausdal G, Keen TJ, Kjaerland E, Krakstad C, Myhren L, Bronstad K, Kunick C, Schwede F, Genieser HG, Kleppe R, Doskeland SO, Cyclic AMP induces IPC leukemia cell apoptosis via CRE-and CDK-dependent Bim transcription, Cell Death Dis 2 (2011) e237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Kumar S, Kostin S, Flacke JP, Reusch HP, Ladilov Y, Soluble adenylyl cyclase controls mitochondria-dependent apoptosis in coronary endothelial cells, J Biol Chem 284(22) (2009) 14760–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Appukuttan A, Kasseckert SA, Kumar S, Reusch HP, Ladilov Y, Oxysterol-induced apoptosis of smooth muscle cells is under the control of a soluble adenylyl cyclase, Cardiovasc Res 99(4) (2013) 734–42. [DOI] [PubMed] [Google Scholar]
- [41].Fajardo AM, Piazza GA, Tinsley HN, The role of cyclic nucleotide signaling pathways in cancer: targets for prevention and treatment, Cancers (Basel) 6(1) (2014) 436–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Davoren PR, Sutherland EW, The Effect of L-Epinephrine and Other Agents on the Synthesis and Release of Adenosine 3’,5’-Phosphate by Whole Pigeon Erythrocytes, J Biol Chem 238 (1963) 3009–15. [PubMed] [Google Scholar]
- [43].Carnegie GK, Means CK, Scott JD, A-kinase anchoring proteins: from protein complexes to physiology and disease, IUBMB Life 61(4) (2009) 394–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Lefkimmiatis K, Zaccolo M, cAMP signaling in subcellular compartments, Pharmacol Ther 143(3) (2014) 295–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Desman G, Waintraub C, Zippin JH, Investigation of cAMP microdomains as a path to novel cancer diagnostics, Biochim Biophys Acta 1842(12 Pt B) (2014) 2636–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Arora K, Sinha C, Zhang W, Ren A, Moon CS, Yarlagadda S, Naren AP, Compartmentalization of cyclic nucleotide signaling: a question of when, where, and why?, Pflugers Arch 465(10) (2013) 1397–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Murray F, Insel PA, Targeting cAMP in chronic lymphocytic leukemia: a pathway-dependent approach for the treatment of leukemia and lymphoma, Expert Opin Ther Targets 17(8) (2013) 937–49. [DOI] [PubMed] [Google Scholar]
- [48].Sapio L, Di Maiolo F, Illiano M, Esposito A, Chiosi E, Spina A, Naviglio S, Targeting protein kinase A in cancer therapy: an update, EXCLI J 13 (2014) 843–55. [PMC free article] [PubMed] [Google Scholar]
- [49].Lerner A, Kim DH, Lee R, The cAMP signaling pathway as a therapeutic target in lymphoid malignancies, Leukemia & Lymphoma 37(1–2) (2000) 39.-+. [DOI] [PubMed] [Google Scholar]
- [50].Meyers JA, Su DW, Lerner A, Chronic lymphocytic leukemia and B and T cells differ in their response to cyclic nucleotide phosphodiesterase inhibitors, J Immunol 182(9) (2009) 5400–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Li C, Xie J, Lu Z, Chen C, Yin Y, Zhan R, Fang Y, Hu X, Zhang CC, ADCY7 supports development of acute myeloid leukemia, Biochem Biophys Res Commun 465(1) (2015) 47–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Hait WN, Weiss B, Increased cyclic nucleotide phosphodiesterase activity in leukaemic lymphocytes, Nature 259(5541) (1976) 321–323. [DOI] [PubMed] [Google Scholar]
- [53].Baus E, Van Laethem F, Andris F, Rolin S, Urbain J, Leo O, Dexamethasone increases intracellular cyclic AMP concentration in murine T lymphocyte cell lines, Steroids 66(1) (2001) 39–47. [DOI] [PubMed] [Google Scholar]
- [54].Cheng JC, Kinjo K, Judelson DR, Chang J, Wu WS, Schmid I, Shankar DB, Kasahara N, Stripecke R, Bhatia R, Landaw EM, Sakamoto KM, CREB is a critical regulator of normal hematopoiesis and leukemogenesis, Blood 111(3) (2008) 1182–1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Shankar DB, Cheng JC, Sakamoto KM, Role of cyclic AMP response element binding protein in human leukemias, Cancer 104(9) (2005) 1819–24. [DOI] [PubMed] [Google Scholar]
- [56].van der Sligte NE, Kampen KR, ter Elst A, Scherpen FJ, Meeuwsen-de Boer TG, Guryev V, van Leeuwen FN, Kornblau SM, de Bont ES, Essential role for cyclic-AMP responsive element binding protein 1 (CREB) in the survival of acute lymphoblastic leukemia, Oncotarget 6(17) (2015) 14970–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Crans-Vargas HN, Landaw EM, Bhatia S, Sandusky G, Moore TB, Sakamoto KM, Expression of cyclic adenosine monophosphate response-element binding protein in acute leukemia, Blood 99(7) (2002) 2617–9. [DOI] [PubMed] [Google Scholar]
- [58].Petrov I, Suntsova M, Mutorova O, Sorokin M, Garazha A, Ilnitskaya E, Spirin P, Larin S, Zhavoronkov A, Kovalchuk O, Prassolov V, Roumiantsev A, Buzdin A, Molecular pathway activation features of pediatric acute myeloid leukemia (AML) and acute lymphoblast leukemia (ALL) cells, Aging-Us 8(11) (2016) 2936–2947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Mitton B, Chae HD, Hsu K, Dutta R, Aldana-Masangkay G, Ferrari R, Davis K, Tiu BC, Kaul A, Lacayo N, Dahl G, Xie F, Li BX, Breese MR, Landaw EM, Nolan G, Pellegrini M, Romanov S, Xiao X, Sakamoto KM, Small molecule inhibition of cAMP response element binding protein in human acute myeloid leukemia cells, Leukemia 30(12) (2016) 2302–2311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Pigazzi M, Manara E, Baron E, Basso G, ICER expression inhibits leukemia phenotype and controls tumor progression, Leukemia 22(12) (2008) 2217–25. [DOI] [PubMed] [Google Scholar]
- [61].Mullighan CG, Genomic profiling of B-progenitor acute lymphoblastic leukemia, Best Pract Res Clin Haematol 24(4) (2011) 489–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Ding LW, Sun QY, Tan KT, Chien W, Mayakonda A, Yeoh AEJ, Kawamata N, Nagata Y, Xiao JF, Loh XY, Lin DC, Garg M, Jiang YY, Xu L, Lim SL, Liu LZ, Madan V, Sanada M, Fernandez LT, Hema Preethi SS, Lill M, Kantarjian HM, Kornblau SM, Miyano S, Liang DC, Ogawa S, Shih LY, Yang H, Koeffler HP, Mutational Landscape of Pediatric Acute Lymphoblastic Leukemia, Cancer Res 77(2) (2017) 390–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Propper DJ, Saunders MP, Salisbury AJ, Long L, O’Byrne KJ, Braybrooke JP, Dowsett M, Taylor M, Talbot DC, Ganesan TS, Harris AL, Phase I study of the novel cyclic AMP (cAMP) analogue 8-chloro-cAMP in patients with cancer: toxicity, hormonal, and immunological effects, Clin Cancer Res 5(7) (1999) 1682–9. [PubMed] [Google Scholar]
- [64].Saunders MP, Salisbury AJ, O’Byrne KJ, Long L, Whitehouse RM, Talbot DC, Mawer EB, Harris AL, A novel cyclic adenosine monophosphate analog induces hypercalcemia via production of 1,25-dihydroxyvitamin D in patients with solid tumors, J Clin Endocrinol Metab 82(12) (1997) 4044–8. [DOI] [PubMed] [Google Scholar]
- [65].Schwede F, Maronde E, Genieser HG, Jastorff B, Cyclic nucleotide analogs as biochemical tools and prospective drugs, Pharmacology & Therapeutics 87(2–3) (2000) 199–226. [DOI] [PubMed] [Google Scholar]
- [66].Wiernik PH, Paietta E, Goloubeva O, Lee SJ, Makower D, Bennett JM, Wade JL, Ghosh C, Kaminer LS, Pizzolo J, Tallman MS, Eastern Cooperative Oncology G, Phase II study of theophylline in chronic lymphocytic leukemia: a study of the Eastern Cooperative Oncology Group (E4998), Leukemia 18(10) (2004) 1605–10. [DOI] [PubMed] [Google Scholar]
- [67].Coffino P, Bourne HR, Tomkins GM, Mechanism of lymphoma cell death induced by cyclic AMP, Am J Pathol 81(1) (1975) 199–204. [PMC free article] [PubMed] [Google Scholar]
- [68].Lomo J, Blomhoff HK, Beiske K, Stokke T, Smeland EB, TGF-beta 1 and cyclic AMP promote apoptosis in resting human B lymphocytes, J Immunol 154(4) (1995) 1634–43. [PubMed] [Google Scholar]
- [69].Myklebust JH, Josefsen D, Blomhoff HK, Levy FO, Naderi S, Reed JC, Smeland EB, Activation of the cAMP signaling pathway increases apoptosis in human B-precursor cells and is associated with downregulation of Mcl-1 expression, Journal of Cellular Physiology 180(1) (1999) 71–80. [DOI] [PubMed] [Google Scholar]
- [70].Cheng YM, Zhu Q, Yao YY, Tang Y, Wang MM, Zou LF, 8-Chloroadenosine 3’,5’-monophosphate induces cell cycle arrest and apoptosis in multiple myeloma cells through multiple mechanisms, Oncol Lett 4(6) (2012) 1384–1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Krett NL, Zell JL, Halgren RG, Pillay S, Traynor AE, Rosen ST, Cyclic adenosine-3’,5’-monophosphate-mediated cytotoxicity in steroid sensitive and resistant myeloma, Clin Cancer Res 3(10) (1997) 1781–7. [PubMed] [Google Scholar]
- [72].Zambon AC, Wilderman A, Ho A, Insel PA, Increased expression of the pro-apoptotic protein BIM, a mechanism for cAMP/protein kinase A (PKA)-induced apoptosis of immature T cells, J Biol Chem 286(38) (2011) 33260–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Zhang L, Insel PA, The pro-apoptotic protein Bim is a convergence point for cAMP/protein kinase A- and glucocorticoid-promoted apoptosis of lymphoid cells, J Biol Chem 279(20) (2004) 20858–65. [DOI] [PubMed] [Google Scholar]
- [74].Yin Y, Allen PD, Jia L, MacEy MG, Kelsey SM, Newland AC, Constitutive levels of cAMP-dependent protein kinase activity determine sensitivity of human multidrug-resistant leukaemic cell lines to growth inhibition and apoptosis by forskolin and tumour necrosis factor alpha, Br J Haematol 108(3) (2000) 565–73. [DOI] [PubMed] [Google Scholar]
- [75].Naderi S, Blomhoff HK, Activation of cAMP signaling enhances Fas-mediated apoptosis and activation-induced cell death through potentiation of caspase 8 activation, Hum Immunol 69(12) (2008) 833–6. [DOI] [PubMed] [Google Scholar]
- [76].Shayo C, Legnazzi BL, Monczor F, Fernández N, Riveiro M.a.E., Baldi A, Davio C, The time-course of cyclic AMP signaling is critical for leukemia U-937 cell differentiation, Biochemical and Biophysical Research Communications 314(3) (2004) 798–804. [DOI] [PubMed] [Google Scholar]
- [77].Yan L, Herrmann V, Hofer JK, Insel PA, beta-adrenergic receptor/cAMP-mediated signaling and apoptosis of S49 lymphoma cells, Am J Physiol Cell Physiol 279(5) (2000) C1665–74. [DOI] [PubMed] [Google Scholar]
- [78].Zhang L, Murray F, Zahno A, Kanter JR, Chou D, Suda R, Fenlon M, Rassenti L, Cottam H, Kipps TJ, Insel PA, Cyclic nucleotide phosphodiesterase profiling reveals increased expression of phosphodiesterase 7B in chronic lymphocytic leukemia, Proc Natl Acad Sci U S A 105(49) (2008) 19532–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Garcia-Bermejo L, Perez C, Vilaboa NE, de Blas E, Aller P, cAMP increasing agents attenuate the generation of apoptosis by etoposide in promonocytic leukemia cells, J Cell Sci 111 ( Pt 5) (1998) 637–44. [DOI] [PubMed] [Google Scholar]
- [80].Gausdal G, Wergeland A, Skavland J, Nguyen E, Pendino F, Rouhee N, McCormack E, Herfindal L, Kleppe R, Havemann U, Schwede F, Bruserud O, Gjertsen BT, Lanotte M, Segal-Bendirdjian E, Doskeland SO, Cyclic AMP can promote APL progression and protect myeloid leukemia cells against anthracycline-induced apoptosis, Cell Death Dis 4 (2013) e516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Kloster MM, Naderi EH, Carlsen H, Blomhoff HK, Naderi S, Hyperactivation of NF-kappaB via the MEK signaling is indispensable for the inhibitory effect of cAMP on DNA damage-induced cell death, Mol Cancer 10 (2011) 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Naderi EH, Jochemsen AG, Blomhoff HK, Naderi S, Activation of cAMP Signaling Interferes with Stress-Induced p53 Accumulation in ALL-Derived Cells by Promoting the Interaction between p53 and HDM2, Neoplasia 13(7) (2011) 653–IN14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Shabestari RM, Safa M, Banan M, Kazemi A, Cyclic AMP-Induced p53 Destabilization is Independent of CREB in Pre-B Acute Lymphoblastic Leukemia Cells, International Journal of Molecular and Cellular Medicine 5(4) (2016) 220–228. [PMC free article] [PubMed] [Google Scholar]
- [84].Xiao LY, Kan WM, Cyclic AMP (cAMP) confers drug resistance against DNA damaging agents via PKAIA in CML cells, Eur J Pharmacol 794 (2017) 201–208. [DOI] [PubMed] [Google Scholar]
- [85].Lee K, Belinsky MG, Bell DW, Testa JR, Kruh GD, Isolation of MOAT-B, a Widely Expressed Multidrug Resistance-associated Protein/Canalicular Multispecific Organic Anion Transporter-related Transporter, Cancer Research 58(13) (1998) 2741–2747. [PubMed] [Google Scholar]
- [86].Morrissey KM, Wen CC, Johns SJ, Zhang L, Huang SM, Giacomini KM, The UCSF-FDA TransPortal: a public drug transporter database, Clin Pharmacol Ther 92(5) (2012) 545–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Kock K, Grube M, Jedlitschky G, Oevermann L, Siegmund W, Ritter CA, Kroemer HK, Expression of adenosine triphosphate-binding cassette (ABC) drug transporters in peripheral blood cells: relevance for physiology and pharmacotherapy, Clin Pharmacokinet 46(6) (2007) 449–70. [DOI] [PubMed] [Google Scholar]
- [88].Lai L, Tan TM, Role of glutathione in the multidrug resistance protein 4 (MRP4/ABCC4)-mediated efflux of cAMP and resistance to purine analogues, Biochem J 361(Pt 3) (2002) 497–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Xie M, Rich TC, Scheitrum C, Conti M, Richter W, Inactivation of multidrug resistance proteins disrupts both cellular extrusion and intracellular degradation of cAMP, Mol Pharmacol 80(2) (2011) 281–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Pavan B, Capuzzo A, Forlani G, Quercetin and quercetin-3-O-glucoside interact with different components of the cAMP signaling cascade in human retinal pigment epithelial cells, Life Sci 121 (2015) 166–73. [DOI] [PubMed] [Google Scholar]
- [91].Kim HP, Bernard L, Berkowitz J, Nitta J, Hogge DE, Flow cytometry-based assessment of mitoxantrone efflux from leukemic blasts varies with response to induction chemotherapy in acute myeloid leukemia, Cytometry B Clin Cytom 82(5) (2012) 283–94. [DOI] [PubMed] [Google Scholar]
- [92].Oevermann L, Scheitz J, Starke K, Kock K, Kiefer T, Dolken G, Niessen J, Greinacher A, Siegmund W, Zygmunt M, Kroemer HK, Jedlitschky G, Ritter CA, Hematopoietic stem cell differentiation affects expression and function of MRP4 (ABCC4), a transport protein for signaling molecules and drugs, Int J Cancer 124(10) (2009) 2303–11. [DOI] [PubMed] [Google Scholar]
- [93].Copsel S, Garcia C, Diez F, Vermeulem M, Baldi A, Bianciotti LG, Russel FG, Shayo C, Davio C, Multidrug resistance protein 4 (MRP4/ABCC4) regulates cAMP cellular levels and controls human leukemia cell proliferation and differentiation, J Biol Chem 286(9) (2011) 6979–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Copsel S, Bruzzone A, May M, Beyrath J, Wargon V, Cany J, Russel FGM, Shayo C, Davio C, Multidrug resistance protein 4/ATP binding cassette transporter 4: a new potential therapeutic target for acute myeloid leukemia, Oncotarget 5(19) (2014) 9308–9321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Wang W, Li Y, Zhu JY, Fang DD, Ding HF, Dong Z, Jing Q, Su SB, Huang S, Triple negative breast cancer development can be selectively suppressed by sustaining an elevated level of cellular cyclic AMP through simultaneously blocking its efflux and decomposition, Oncotarget 7(52) (2016) 87232–87245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Perez DR, Smagley Y, Garcia M, Carter MB, Evangelisti A, Matlawska-Wasowska K, Winter SS, Sklar LA, Chigaev A, Cyclic AMP efflux inhibitors as potential therapeutic agents for leukemia, Oncotarget 7(23) (2016) 33960–33982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Perez D, Simons PC, Smagley Y, Sklar LA, Chigaev A, A High-Throughput Flow Cytometry Assay for Identification of Inhibitors of 3′,5′-Cyclic Adenosine Monophosphate Efflux, in: Janzen WP (Ed.), High Throughput Screening: Methods and Protocols, Springer; New York, New York, NY, 2016, pp. 227–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Chigaev A, Perez DR, Sklar LA, Method for Cancer Cell Reprogramming, STC.UNM, US, 2016. [Google Scholar]
- [99].Xing F, Luan Y, Cai J, Wu S, Mai J, Gu J, Zhang H, Li K, Lin Y, Xiao X, Liang J, Li Y, Chen W, Tan Y, Sheng L, Lu B, Lu W, Gao M, Qiu P, Su X, Yin W, Hu J, Chen Z, Sai K, Wang J, Chen F, Chen Y, Zhu S, Liu D, Cheng S, Xie Z, Zhu W, Yan G, The Anti-Warburg Effect Elicited by the cAMP-PGC1alpha Pathway Drives Differentiation of Glioblastoma Cells into Astrocytes, Cell Reports 18(2) (2017) 468–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Fukuda Y, Lian S, Schuetz JD, Leukemia and ABC transporters, Adv Cancer Res 125 (2015) 171–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Chigaev A, Waller A, Amit O, Sklar LA, Galphas-coupled receptor signaling actively down-regulates alpha4beta1-integrin affinity: a possible mechanism for cell de-adhesion, BMC Immunol 9 (2008) 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Chigaev A, Smagley Y, Sklar LA, Nitric oxide/cGMP pathway signaling actively down-regulates alpha4beta1-integrin affinity: an unexpected mechanism for inducing cell de-adhesion, BMC Immunol 12 (2011) 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Ganghammer S, Hutterer E, Hinterseer E, Brachtl G, Asslaber D, Krenn PW, Girbl T, Berghammer P, Geisberger R, Egle A, Zucchetto A, Kruschinski A, Gattei V, Chigaev A, Greil R, Hartmann TN, CXCL12-induced VLA-4 activation is impaired in trisomy 12 chronic lymphocytic leukemia cells: a role for CCL21, Oncotarget 6(14) (2015) 12048–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Zippin JH, Chen Y, Nahirney P, Kamenetsky M, Wuttke MS, Fischman DA, Levin LR, Buck J, Compartmentalization of bicarbonate-sensitive adenylyl cyclase in distinct signaling microdomains, FASEB J 17(1) (2003) 82–4. [DOI] [PubMed] [Google Scholar]
- [105].Zwick M, Esposito C, Hellstern M, Seelig A, How Phosphorylation and ATPase Activity Regulate Anion Flux though the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR), Journal of Biological Chemistry 291(28) (2016) 14483–14498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Guo Y, Kock K, Ritter CA, Chen ZS, Grube M, Jedlitschky G, Illmer T, Ayres M, Beck JF, Siegmund W, Ehninger G, Gandhi V, Kroemer HK, Kruh GD, Schaich M, Expression of ABCC-type nucleotide exporters in blasts of adult acute myeloid leukemia: relation to long-term survival, Clin Cancer Res 15(5) (2009) 1762–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Cheepala S, Hulot JS, Morgan JA, Sassi Y, Zhang W, Naren AP, Schuetz JD, Cyclic nucleotide compartmentalization: contributions of phosphodiesterases and ATP-binding cassette transporters, Annu Rev Pharmacol Toxicol 53 (2013) 231–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Burgers PP, Ma Y, Margarucci L, Mackey M, van der Heyden MA, Ellisman M, Scholten A, Taylor SS, Heck AJ, A small novel A-kinase anchoring protein (AKAP) that localizes specifically protein kinase A-regulatory subunit I (PKA-RI) to the plasma membrane, J Biol Chem 287(52) (2012) 43789–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Merrill RA, Strack S, Mitochondria: a kinase anchoring protein 1, a signaling platform for mitochondrial form and function, Int J Biochem Cell Biol 48 (2014) 92–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Huang LJ, Wang L, Ma Y, Durick K, Perkins G, Deerinck TJ, Ellisman MH, Taylor SS, NH2-Terminal targeting motifs direct dual specificity A-kinase-anchoring protein 1 (D-AKAP1) to either mitochondria or endoplasmic reticulum, J Cell Biol 145(5) (1999) 951–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Kapiloff MS, Schillace RV, Westphal AM, Scott JD, mAKAP: an A-kinase anchoring protein targeted to the nuclear membrane of differentiated myocytes, J Cell Sci 112 ( Pt 16) (1999) 2725–36. [DOI] [PubMed] [Google Scholar]
- [112].Dodge KL, Khouangsathiene S, Kapiloff MS, Mouton R, Hill EV, Houslay MD, Langeberg LK, Scott JD, mAKAP assembles a protein kinase A/PDE4 phosphodiesterase cAMP signaling module, EMBO J 20(8) (2001) 1921–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Ould Amer Y, Hebert-Chatelain E, Mitochondrial cAMP-PKA signaling: What do we really know?, Biochim Biophys Acta Bioenerg 1859(9) (2018) 868–877. [DOI] [PubMed] [Google Scholar]
- [114].Zhang F, Zhang L, Qi Y, Xu H, Mitochondrial cAMP signaling, Cell Mol Life Sci 73(24) (2016) 4577–4590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Zalman LS, Nikaido H, Kagawa Y, Mitochondrial outer membrane contains a protein producing nonspecific diffusion channels, J Biol Chem 255(5) (1980) 1771–4. [PubMed] [Google Scholar]
- [116].Schmidt O, Harbauer AB, Rao S, Eyrich B, Zahedi RP, Stojanovski D, Schonfisch B, Guiard B, Sickmann A, Pfanner N, Meisinger C, Regulation of mitochondrial protein import by cytosolic kinases, Cell 144(2) (2011) 227–39. [DOI] [PubMed] [Google Scholar]
- [117].Gerbeth C, Schmidt O, Rao S, Harbauer AB, Mikropoulou D, Opalinska M, Guiard B, Pfanner N, Meisinger C, Glucose-induced regulation of protein import receptor Tom22 by cytosolic and mitochondria-bound kinases, Cell Metab 18(4) (2013) 578–87. [DOI] [PubMed] [Google Scholar]
- [118].Rao S, Schmidt O, Harbauer AB, Schonfisch B, Guiard B, Pfanner N, Meisinger C, Biogenesis of the preprotein translocase of the outer mitochondrial membrane: protein kinase A phosphorylates the precursor of Tom40 and impairs its import, Mol Biol Cell 23(9) (2012) 1618–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Chigaev A, Does aberrant membrane transport contribute to poor outcome in adult acute myeloid leukemia?, Front Pharmacol 6 (2015) 134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Yamano K, Tanaka-Yamano S, Endo T, Tom7 regulates Mdm10-mediated assembly of the mitochondrial import channel protein Tom40, J Biol Chem 285(53) (2010) 41222–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Harbauer AB, Zahedi RP, Sickmann A, Pfanner N, Meisinger C, The protein import machinery of mitochondria-a regulatory hub in metabolism, stress, and disease, Cell Metab 19(3) (2014) 357–72. [DOI] [PubMed] [Google Scholar]
- [122].Opalinska M, Meisinger C, Metabolic control via the mitochondrial protein import machinery, Curr Opin Cell Biol 33 (2015) 42–8. [DOI] [PubMed] [Google Scholar]
- [123].Xu D, Li R, Wu J, Jiang L, Zhong HA, Drug Design Targeting the CXCR4/CXCR7/CXCL12 Pathway, Curr Top Med Chem 16(13) (2016) 1441–51. [DOI] [PubMed] [Google Scholar]
- [124].Monczor F, Copsel S, Fernandez N, Davio C, Shayo C, Histamine H2 Receptor in Blood Cells: A Suitable Target for the Treatment of Acute Myeloid Leukemia, Handb Exp Pharmacol 241 (2017) 141–160. [DOI] [PubMed] [Google Scholar]
- [125].Natale CA, Li J, Zhang J, Dahal A, Dentchev T, Stanger BZ, Ridky TW, Activation of G protein-coupled estrogen receptor signaling inhibits melanoma and improves response to immune checkpoint blockade, Elife 7 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Lim CJ, Han J, Yousefi N, Ma Y, Amieux PS, McKnight GS, Taylor SS, Ginsberg MH, Alpha4 integrins are type I cAMP-dependent protein kinase-anchoring proteins, Nat Cell Biol 9(4) (2007) 415–21. [DOI] [PubMed] [Google Scholar]
- [127].Ventura-Clapier R, Garnier A, Veksler V, Transcriptional control of mitochondrial biogenesis: the central role of PGC-1alpha, Cardiovasc Res 79(2) (2008) 208–17. [DOI] [PubMed] [Google Scholar]
- [128].Scarpulla RC, Vega RB, Kelly DP, Transcriptional integration of mitochondrial biogenesis, Trends Endocrinol Metab 23(9) (2012) 459–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Esparza SD, Chang J, Shankar DB, Zhang B, Nelson SF, Sakamoto KM, CREB regulates Meis1 expression in normal and malignant hematopoietic cells, Leukemia 22(3) (2008) 665–7. [DOI] [PubMed] [Google Scholar]
- [130].Shankar DB, Cheng JC, Kinjo K, Federman N, Moore TB, Gill A, Rao NP, Landaw EM, Sakamoto KM, The role of CREB as a proto-oncogene in hematopoiesis and in acute myeloid leukemia, Cancer Cell 7(4) (2005) 351–62. [DOI] [PubMed] [Google Scholar]
- [131].Wiedemann B, Weisner J, Rauh D, Chemical modulation of transcription factors, Medchemcomm 9(8) (2018) 1249–1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Parker D, Ferreri K, Nakajima T, LaMorte VJ, Evans R, Koerber SC, Hoeger C, Montminy MR, Phosphorylation of CREB at Ser-133 induces complex formation with CREB-binding protein via a direct mechanism, Mol Cell Biol 16(2) (1996) 694–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Sciaraffia E, Riccomi A, Lindstedt R, Gesa V, Cirelli E, Patrizio M, De Magistris MT, Vendetti S, Human monocytes respond to extracellular cAMP through A2A and A2B adenosine receptors, J Leukoc Biol 96(1) (2014) 113–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Bhang DH, Choi US, Kim BG, Lee SN, Lee S, Roh HS, Chung WJ, Jeon KO, Song WJ, Youn HY, Baek KH, Characteristics of extracellular cyclic AMP-dependent protein kinase as a biomarker of cancer in dogs, Vet Comp Oncol 15(4) (2017) 1585–1589. [DOI] [PubMed] [Google Scholar]
- [135].Elalamy I, Said FA, Singer M, Couetil JP, Hatmi M, Inhibition by extracellular cAMP of phorbol 12-myristate 13-acetate-induced prostaglandin H synthase-2 expression in human pulmonary microvascular endothelial cells. Involvement of an ecto-protein kinase A activity, J Biol Chem 275(18) (2000) 13662–7. [DOI] [PubMed] [Google Scholar]
- [136].Sheehan KM, Sheahan K, O’Donoghue DP, MacSweeney F, Conroy RM, Fitzgerald DJ, Murray FE, The Relationship Between Cyclooxygenase-2 Expression and Colorectal Cancer, JAMA 282(13) (1999) 1254–1257. [DOI] [PubMed] [Google Scholar]
- [137].Godinho RO, Duarte T, Pacini ES, New perspectives in signaling mediated by receptors coupled to stimulatory G protein: the emerging significance of cAMP e ffl ux and extracellular cAMP-adenosine pathway, Front Pharmacol 6 (2015) 58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Hofer AM, Lefkimmiatis K, Extracellular calcium and cAMP: second messengers as “third messengers”?, Physiology (Bethesda) 22 (2007) 320–7. [DOI] [PubMed] [Google Scholar]
- [139].Allard B, Beavis PA, Darcy PK, Stagg J, Immunosuppressive activities of adenosine in cancer, Curr Opin Pharmacol 29 (2016) 7–16. [DOI] [PubMed] [Google Scholar]
- [140].Young A, Ngiow SF, Barkauskas DS, Sult E, Hay C, Blake SJ, Huang Q, Liu J, Takeda K, Teng MWL, Sachsenmeier K, Smyth MJ, Co-inhibition of CD73 and A2AR Adenosine Signaling Improves Anti-tumor Immune Responses, Cancer Cell 30(3) (2016) 391–403. [DOI] [PubMed] [Google Scholar]
- [141].Jin D, Fan J, Wang L, Thompson LF, Liu A, Daniel BJ, Shin T, Curiel TJ, Zhang B, CD73 on tumor cells impairs antitumor T-cell responses: a novel mechanism of tumor-induced immune suppression, Cancer Res 70(6) (2010) 2245–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Hausler SF, Montalban del Barrio I, Strohschein J, Chandran PA, Engel JB, Honig A, Ossadnik M, Horn E, Fischer B, Krockenberger M, Heuer S, Seida AA, Junker M, Kneitz H, Kloor D, Klotz KN, Dietl J, Wischhusen J, Ectonucleotidases CD39 and CD73 on OvCA cells are potent adenosine-generating enzymes responsible for adenosine receptor 2A-dependent suppression of T cell function and NK cell cytotoxicity, Cancer Immunol Immunother 60(10) (2011) 1405–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Thiel M, Caldwell CC, Sitkovsky MV, The critical role of adenosine A2A receptors in downregulation of inflammation and immunity in the pathogenesis of infectious diseases, Microbes and Infection 5(6) (2003) 515–526. [DOI] [PubMed] [Google Scholar]
- [144].Pleli T, Mondorf A, Ferreiros N, Thomas D, Dvorak K, Biondi RM, Heringdorf DMZ, Zeuzem S, Geisslinger G, Zimmermann H, Waidmann O, Piiper A, Activation of Adenylyl Cyclase Causes Stimulation of Adenosine Receptors, Cell Physiol Biochem 45(6) (2018) 2516–2528. [DOI] [PubMed] [Google Scholar]
- [145].Fehr TF, Dickinson ES, Goldman SJ, Slakey LL, Cyclic-Amp Efflux Is Regulated by Occupancy of the Adenosine Receptor in Pig Aortic Smooth-Muscle Cells, Journal of Biological Chemistry 265(19) (1990) 10974–10980. [PubMed] [Google Scholar]
- [146].Beavis PA, Divisekera U, Paget C, Chow MT, John LB, Devaud C, Dwyer K, Stagg J, Smyth MJ, Darcy PK, Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors, Proc Natl Acad Sci U S A 110(36) (2013) 14711–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Braganhol E, Tamajusuku AS, Bernardi A, Wink MR, Battastini AM, Ecto-5’-nucleotidase/CD73 inhibition by quercetin in the human U138MG glioma cell line, Biochim Biophys Acta 1770(9) (2007) 1352–9. [DOI] [PubMed] [Google Scholar]
- [148].Leone RD, Lo YC, Powell JD, A2aR antagonists: Next generation checkpoint blockade for cancer immunotherapy, Comput Struct Biotechnol J 13 (2015) 265–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Wang L, Zhou X, Zhou T, Ma D, Chen S, Zhi X, Yin L, Shao Z, Ou Z, Zhou P, Ecto-5’-nucleotidase promotes invasion, migration and adhesion of human breast cancer cells, J Cancer Res Clin Oncol 134(3) (2008) 365–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Stagg J, Divisekera U, McLaughlin N, Sharkey J, Pommey S, Denoyer D, Dwyer KM, Smyth MJ, Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis, Proc Natl Acad Sci U S A 107(4) (2010) 1547–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Li X, Zhou T, Zhi X, Zhao F, Yin L, Zhou P, Effect of hypoxia/reoxygenation on CD73 (ecto-5’-nucleotidase) in mouse microvessel endothelial cell lines, Microvasc Res 72(1–2) (2006) 48–53. [DOI] [PubMed] [Google Scholar]
- [152].Pulte D, Olson KE, Broekman MJ, Islam N, Ballard HS, Furman RR, Olson AE, Marcus AJ, CD39 activity correlates with stage and inhibits platelet reactivity in chronic lymphocytic leukemia, J Transl Med 5 (2007) 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Bastid J, Cottalorda-Regairaz A, Alberici G, Bonnefoy N, Eliaou JF, Bensussan A, ENTPD1/CD39 is a promising therapeutic target in oncology, Oncogene 32(14) (2013) 1743–51. [DOI] [PubMed] [Google Scholar]
- [154].Feng L, Sun X, Csizmadia E, Han L, Bian S, Murakami T, Wang X, Robson SC, Wu Y, Vascular CD39/ENTPD1 Directly Promotes Tumor Cell Growth by Scavenging Extracellular Adenosine Triphosphate, Neoplasia 13(3) (2011) 206–IN2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Li L, Wang L, Li J, Fan Z, Yang L, Zhang Z, Zhang C, Yue D, Qin G, Zhang T, Li F, Chen X, Ping Y, Wang D, Gao Q, He Q, Huang L, Li H, Huang J, Zhao X, Xue W, Sun Z, Lu J, Yu JJ, Zhao J, Zhang B, Zhang Y, Metformin-Induced Reduction of CD39 and CD73 Blocks Myeloid-Derived Suppressor Cell Activity in Patients with Ovarian Cancer, Cancer Res 78(7) (2018) 1779–1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Serra S, Horenstein AL, Vaisitti T, Brusa D, Rossi D, Laurenti L, D’Arena G, Coscia M, Tripodo C, Inghirami G, Robson SC, Gaidano G, Malavasi F, Deaglio S, CD73-generated extracellular adenosine in chronic lymphocytic leukemia creates local conditions counteracting drug-induced cell death, Blood 118(23) (2011) 6141–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Pieters R, Thompson LF, Broekema GJ, Huismans DR, Peters GJ, Pals ST, Horst E, Hahlen K, Veerman AJ, Expression of 5’-nucleotidase (CD73) related to other differentiation antigens in leukemias of B-cell lineage, Blood 78(2) (1991) 488–92. [PubMed] [Google Scholar]
- [158].Mikhailov A, Sokolovskaya A, Yegutkin GG, Amdahl H, West A, Yagita H, Lahesmaa R, Thompson LF, Jalkanen S, Blokhin D, Eriksson JE, CD73 Participates in Cellular Multiresistance Program and Protects against TRAIL-Induced Apoptosis, The Journal of Immunology 181(1) (2008) 464–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Wieten E, van der Linden-Schrever BE, Sonneveld E, Veerman AJ, Pieters R, CD73 (5’-nucleotidase) expression has no prognostic value in children with acute lymphoblastic leukemia, Leukemia 25(8) (2011) 1374–6. [DOI] [PubMed] [Google Scholar]
- [160].Denizot Y, Donnard M, Truffinet V, Malissein E, Faucher JL, Turlure P, Bordessoule D, Trimoreau F, Functional EP2 receptors on blast cells of patients with acute leukemia, Int J Cancer 115(3) (2005) 499–501. [DOI] [PubMed] [Google Scholar]
- [161].Li H-J, Reinhardt F, Herschman HR, Weinberg RA, Cancer-Stimulated Mesenchymal Stem Cells Create a Carcinoma Stem Cell Niche via Prostaglandin E2 Signaling, Cancer Discovery 2(9) (2012) 840–855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [162].Mirsaidi A, Tiaden AN, Richards PJ, Prostaglandin E2 inhibits matrix mineralization by human bone marrow stromal cell-derived osteoblasts via Epac-dependent cAMP signaling, Sci Rep 7(1) (2017) 2243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Fukuda Y, Schuetz JD, ABC transporters and their role in nucleoside and nucleotide drug resistance, Biochem Pharmacol 83(8) (2012) 1073–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
