Open Access

Functional characteristics and research trends of PDE11A in human diseases (Review)

  • Authors:
    • Gyeyeong Kong
    • Hyunji Lee
    • Thuy-Trang T. Vo
    • Uijin Juang
    • So Hee Kwon
    • Jisoo Park
    • Jongsun Park
    • Seon-Hwan Kim
  • View Affiliations

  • Published online on: August 4, 2022     https://doi.org/10.3892/mmr.2022.12814
  • Article Number: 298
  • Copyright: © Kong et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

cAMP and cGMP are important secondary messengers involved in cell regulation and metabolism driven by the G protein‑coupled receptor. cAMP is converted via adenylyl cyclase (AC) and activates protein kinase A to phosphorylate intracellular proteins that mediate specific responses. cAMP signaling serves a role at multiple steps in tumorigenesis. The level of cAMP is increased in association with cancer cell formation through activation of AC‑stimulatory G protein by mutation. Phosphodiesterases (PDEs) hydrolyze cAMP and cGMP to AMP and GMP. PDEs are composed of 11 families, and each can hydrolyze cAMP and cGMP or both cAMP and cGMP. PDEs perform various roles depending on their location and expression site, and are involved in several diseases, including male erectile dysfunction, pulmonary hypertension, Alzheimer's disease and schizophrenia. PDE11A is the 11th member of the PDE family and is characterized by four splice variants with varying tissue expression and N‑terminal regulatory regions. Among tissues, the expression of PDE11A was highest in the prostate, and it was also expressed in hepatic skeletal muscle, pituitary, pancreas and kidney. PDE11A is the first PDE associated with an adrenocortical tumor associated genetic condition. In several studies, three PDE11A mutations have been reported in patients with Cushing syndrome with primary pigmented nodular adrenocortical disease or isolated micronodular adrenocortical disease without other genetic defects. It has been reported that an increase in PDE11A expression affects the proliferation of glioblastoma and worsens patient prognosis. The present mini‑review summarizes the location of PDE11A expression, the impact of structural differences and disease relevance.

Introduction

Molecular and genetic studies of the endocrine system have progressed rapidly over the past few decades. Cyclic adenosine monophosphate (cAMP) is the most important secondary messenger involved in endocrine system development and function. Dysregulation of cAMP expression and signaling perturbs the endocrine physiology and causes disease. cAMP production and degradation is mediated by ACs and phosphodiesterases (PDEs) respectively (15). PDEs hydrolyze the phosphate bonds of cyclic nucleotides; 11 PDE gene families have been identified based on amino acid sequences, biochemical properties, and inhibitor profiles (6,7). PDEs may share a catalytic function but differ in subcellular localization and tissue expression status (7). PDEs hydrolyze cAMP and cyclic guanosine monophosphate (cGMP) to AMP and GMP. PDEs may degrade cAMP (PDE4, 7, 8), cGMP (PDE5, 6, 9), or both (PDE1, 2, 3, 10, 11) (1,810). Thus, PDEs perform various roles depending on their location and expression status. For example, inhibitors of PDE5 serve as therapeutic agents for male erectile dysfunction and pulmonary hypertension (11,12). PDE9A and PDE10A are widely distributed throughout the central nervous system (CNS); modulation of their expression usefully treats Alzheimer's disease (13) and schizophrenia (1416). PDE11A degrades both cAMP and cGMP (1721). PDE11A features four splice variants (14) varying in terms of tissue expression and the N-terminal regulatory regions. The N-terminal domain is regulatory in nature and the C-terminal domain catalytic. The longest isoforms of PDE11A in the mouse and human share ~95% protein sequence homology. The PDE11A level is highest in the prostate (22) of the various splice variants, PDE11A1 and PDE11A3 are found in the spleen (23,24) and PDE11A4 in the hippocampus (25). PDE11A is also expressed in the liver, skeletal muscle, pituitary gland, pancreas, and kidneys (18,19,22). Thus, PDE11A expression and structural characteristics vary by tissue location. This mini-review summarizes the locations of PDE11A expression, the effects of structural differences, and disease involvement.

cAMP/cGMP-dependent signaling

cAMP and cGMP are important secondary messengers involved in cell regulation and metabolism (6) driven by the GPCR. AC catalyzes conversion of ATP (Adenosine triphosphate) to cAMP and inorganic pyrophosphate; cAMP activates protein kinase A (PKA), which in turn phosphorylates intracellular proteins that mediate specific responses (26). cAMP activation is triggered by adrenocorticotropic hormone bound to the adrenocorticotropic hormone receptor; this in turn induces dissociation of the Gsα subunit (encoded by the GNAS gene) from G-protein, AC activation, cAMP generation, and PKA activation. PKA is a tetrameric complex of two regulatory subunits (PRKACA and PRKACB). The latter is responsible for phosphorylation of various enzymes and transcription factors, including the cAMP response element-binding protein (CREB) (Fig. 1). cAMP signaling plays roles during several steps of tumorigenesis. Inactivation of germline mutations in the alpha regulatory subunit gene of PKA induces the Carney complex (2732) (an autosomal-dominant disease characterized by cardiac myxoma, schwannoma, and endocrine tumors; Carney complex is one of the most common types of primary pigmentary crystalline adrenocortical disease associated hyperplasia) (33). Such mutations are also implicated in cancer cell formation via activation of the stimulatory G protein of AC, increasing the cAMP level (34). Similar to cAMP, cGMP is degraded by class I phosphodiesterase in metazoans, some of which are activated by cGMP binding to the GAF domain (35). Cyclic GMP signaling is not observed in eubacteria, plants, and yeast but is found in vertebrates. Besides that, It was also found in Drosophila and Caenorhabditis elegans with cGMP signaling, which is mediated by cGMP regulatory protein kinase G, possibly Ras guanine nucleotide exchange factor, and ion channels, is similar to that of vertebrates. Furthermore, these regulators contain the cyclic nucleotide-binding domain instead of the GAF domain (36,37). In metazoans, cGMP is synthesized by two guanylyl cyclases, one membrane-bound and the other soluble, and has a common phylogenetic precursor. Although no close homologs of this protein have been found in Dictyostelium, Dictyostelium guanylyl cyclases (38), guanylyl cyclases A and soluble guanylyl cyclases are similar to AC. guanylyl cyclases A has a dozen ubiquitous topologies on metazoan AC, whereas soluble guanylyl cyclases is just homologous to a small family of soluble AC present in vertebrates and bacteria (39,40). Upregulation of cGMP levels by PDEs induces activation of PKG, which promotes vasodilation and increases blood flow, particularly in the brain (41). cAMP also contributes to tumorigenesis via PDE (6), which increases cAMP and cGMP levels (1,810), resulting in sustained activation of the cAMP/PKA cascade. PDE is expressed in many different cancer cells, which may also host PDE mutations (examples PDE11A R804H, and R867G (6,42,43). An association between PDE genetic changes and tumorigenesis has been noted, particularly in the prostate, testis, and adrenal cortex (44,45). Hence, mutations in PDE and circulation of cAMP and cGMP are essential not only for human development but also for cancer and many diseases.

Phosphodiesterase family

PDEs regulate cAMP and cGMP production and are essential enzymes. PDEs are found in various tissues where they perform different roles. PDE features 11 different isoforms (Fig. 2). The four PDE1 isoforms (PDE1A, PDE1B, PDE1B1-2, and PDE1C1-2) are found in the brain, sperm, kidney, liver, pancreas, and thyroid gland (4648); the heart (49); immune cells (50); and the olfactory epithelium (51) respectively, and regulate both cAMP and cGMP action. The PDEs play roles in vascular smooth muscle contraction and proliferation, sperm function, dopamine signaling, and immune cell activation. The common PDE1 subtype inhibitors include Vinpocetine, IC224 (PDE1A), SCH51866, 8-MeoM-IBMX (PDE1B), Zaprinast (PDE1B1-2), and Sildenafil (PDE1C1-2). PDE2A1-3 is expressed in the adrenal glomerulosa (52). The PDE2 proteins regulate both cAMP and cGMP actions and control aldosterone and ACTH secretion and long-term memory. Common PDE2A inhibitors include EHNA, BAY60-7550, PDP, and IC933. PDE3 includes PDE3A1-3 and PDE3B. The former is expressed in the heart (53), adipocytes, oocytes, cardiac and vascular smooth muscle, myocardium, and platelets (54). PDE3B is expressed in heart muscle (55), the immune system (56), endothelial cells (mediating permeability and cell proliferation) (57), the brain (58) and the liver (59). PDE3A and PDE3B regulate both cAMP and cGMP production; PDE3A controls cardiac contraction, platelet aggregation, vascular smooth muscle contraction, cell maturation, and renin release. PDE3B modulates lipolysis, glycogenolysis, insulin secretion, and heart function. Common PDE3 inhibitors include amrinone, cilostazol, milrinone, and enoximone. In addition, many inhibitors have been reported to modulate PDEs (Table I). PDE4 includes PDE4A, PDE4B, PDE4C, and PDE4D. PDE4 is expressed in the heart and small intestine (60), immune cells (61), and the brain (62). Unlike PDEs 1, 2, and 3, PDE4 exhibits a higher affinity for cAMP than cGMP and controls brain function, monocyte and macrophage activation, neutrophil infiltration, vascular smooth muscle proliferation, fertility, and heart β-adrenergic signaling and excitatory/contract coupling. PDE5 includes PDE5A1-3 expressed in the lung, penis, smooth muscle (28), platelets (63), brain (64) and cardiac muscle (65). Both PDE5 enzymes regulate cGMP; the nitrous oxide (NO)/cGMP effects in vascular smooth muscle, platelets, and the lower urinary tract; and the cardiac stress response. PDE6 includes PDE6A, PDE6B, and PDE6C expressed in photoreceptors (66) and the pineal gland (67). PDE6 regulates cGMP action, controls the cGMP concentrations of rod and cone photoreceptors, and is the primary effector enzyme of the phototransduction cascade. PDE7 features PDE7A1-2 and PDE7B1-3 found in immune cells (68), skeletal and cardiac muscles (69) and the brain (70). PDE7 modulates the cAMP activity and plays an important role in the regulation of human T cell function. PDE8 includes PDE8A1-5 and PDE8B1-3 found in immune cells (71), the heart (72), the ovary and testes (73), the thyroid gland (74), placenta, brain (75) and the adrenal gland (76). Both PDE8s regulate cAMP activation and TSH levels, adrenal steroid production, luteinizing hormone signaling, and steroidogenesis in Leydig cells, and activate T cells. PDE9 includes PDE9A1-6 expressed in the kidney, spleen, gut, and prostate (77). PDE9 regulates cGMP activation to play a role in energy balance. PDE10 includes PDE10A1-2 expressed in the brain, testis, and thyroid (78). PDE10 regulates both cAMP and cGMP actions and plays roles in striatal activation and behavioral activity. PDE11 includes PDE11A1-4 of the testis, pituitary gland, heart, kidney, liver (18,19), prostate, adrenal gland, and colon (22), but only the A4 splice mutant is expressed in adrenal tissue. PDE11A regulates both cAMP and cGMP actions and is involved in spermatogenesis. PDE11A4 was recently found in the hippocampus (23,79). Besides that, all PDEs are expressed somewhere in the CNS and hydrolyze cAMP and cGMP to perform their respective roles (Fig. 3A) (80). However, when the central nervous system is damaged, the increase of PDEs expression activates immune cells and decreases the regeneration of neuronal cells, resulting in the death of neural cells (Fig. 3B) (81). Therefore, all PDE families can perform their respective roles depending on the expression site, and all PDEs also can contribute to the growth and development of nerve cells and cancer cells.

Table I.

Inhibitors commonly used for PDEs.

Table I.

Inhibitors commonly used for PDEs.

FamilyTypeCommonly used inhibitors
PDE1PDE1AVinpocetine, IC224, SCH51866, 8-MeoM-IBMX
PDE1B
PDE1B1-2Zaprinast
PDE1C1-2Sildenafil
PDE2PDE2A1-3EHNA, BAY60-7550, PDP, IC933
PDE3PDE3A1-3Milrinone, Tolafentrine, Cilostazol, Cilostamide, Trequinsin, OPC-33540, Dihydropyridazinone, Lixazinone
PDE3B
PDE4PDE4ACilomilast, Rolipram, Ro20-1724, Roflumilast, AWD12281, V11294A, SCH35159, Denbufylline, Arofylline, Tolafentrine, Zardaverine
PDE4B
PDE4C
PDE4D
PDE5PDE5A1-3Sildenafil, Tadalafil, DA8159, E402, Vardenafil, Zaprinast, DMPPO, Dipyridamole
PDE6PDE6AZaprinast, Dypyridamole, Sildenafil, Verdenafil, Tadalafil
PDE6B
PDE6C
PDE7PDE7A1-2BRL 50481, IC242, Dipyridamole, BMS-586353, Thiadiazoles
PDE7B1-3
PDE8PDE8A1-5Dipyridamole
PDE8B1-3
PDE9PDE9A1-6BAY 73–669, SCH51866, Zaprinast
PDE10PDE10A1-2Papaverine, Zaprinast, Dipyridamole, PQ-10
PDE11PDE11A1-4Dipyridamole, Zaprinast

[i] PDE, phosphodiesterase.

PDE11A and tumors

cAMP and cGMP are important GPCR-driven secondary messengers controlling cellular regulation and metabolism. cAMP is formed by the actions of AC and PDE and mediates cellular responses by activating PKA to phosphorylate intracellular proteins (4,5). In addition, the cAMP has been implicated in various tumorigenesis due to either increasing expression levels by activating the stimulatory G protein of AC or degraded by PDE11A. PDE11A encoded on chromosome 2q31.2 is highly polymorphic and was also the first PDE associated with an adrenocortical tumor-associated genetic condition. Furthermore, PDE11A degrades not only cAMP but also cGMP (82). Besides, several previous studies found that PDE11A mutations were mainly expressed in abnormal adrenal glands (19,83). Three PDE11A mutations have been reported in Cushing syndrome patients with a primary pigmented nodular adrenocortical disease or isolated micronodular adrenocortical disease without other genetic defects. An association between the GWA single-nucleotide polymorphism (SNP) of PDE11 and adrenocortical tumors has been also confirmed (43). Mutations and relationships of PDE11A have been reported in numerous types of cancer, as well as the most studied adrenal cortical tumors (https://www.cbioportal.org/). The heterozygous inactivation strains of PDE11A in patients were identified in non-secreting adrenal cortical adenoma, and heterozygous missense strains were more common in Primary bilateral macronodular adrenal hyperplasia (24%) and adrenocortical carcinomas (19%) compared to control group (5.7%) (84). Furthermore, the p.R867G PDE11A mutation was found in one patient with familial Primary bilateral macronodular adrenal hyperplasia (85). In a Primary bilateral macronodular adrenal hyperplasia cohort, the frequency of all PDE11A variants was significantly higher in Primary bilateral macronodular adrenal hyperplasia patients (28%) than in controls (7.2%). The inactivating PDE11A mutation (p.R307) was also found in adrenocortical cancer-associated genetic condition patients (19). Not only that, in the New York Cancer Project, Horvath et al (43,86) studied 745 patients with adrenocortical tumors and found PDE11A sequence changes including three truncation mutations (c.171Tdel/fs41X, c.919C>T/p.R307X and c.1655_1657TCTdelCCins/fs15X) and two missense substitutions [c.2411G>A(R804H) and c.2599C>(R867G)] (Fig. 4). Mutations in PDE11A have also been reported in testicular germ cell tumors. In 259 patients with testicular German cell tumors, 55 PDE11A strains (20 missense, 4 splice sites, 2 non-sense sites, 7 synonyms, 22 introductions, 10 missense strains, 9 transcriptions) were identified. Among them, rare mutations (p.F258Y, p.G291R, p.V820M, p.R545X, p.K568R) were found. Mutations in PDE11A testicular germ cell tumors degrade PDE11A function, ultimately increasing the cAMP/cGMP level (87). Mutation of PDE11A was also observed in Carney complex, somatic dystrophy and various endocrine tumors (kidney, prostate, colon, lung and breast) (22,44,82,88,89). In addition, mutations in Y727C and E840K of PDE11A have been reported to be extremely high expressed in prostate cancer (90). Moreover, the role of PDE11A in brain tumors has recently been studied and reported due to the brain belongs to an essential part of the human body and is responsible for a critical part of the CNS (91). All brain cancers are graded from 1 to 4 based on how the cancer cells look under the microscope and how well they reproduce. The most aggressive and fast-growing malignant Grade 4 tumor is called glioblastoma (92). Currently open surgery, radiation therapy (93), and chemotherapy (94) are all using for the treatment of glioblastoma. However, they are ineffective or have a very high probability of side effects. Therefore, to develop an effective treatment strategy for glioblastoma, the studies of analytical methods using molecular targeting are necessary. Besides, with the increase of studies on the association between PDE11A and the brain, it was found in the brain's hippocampus (62), and the deletion of PDE11A in the brain has been shown to increase microglial activation (25). More specially, recently, PDE11A was also found to be highly expressed in glioblastoma. Lee et al (95) found that the PDE11A expression level in glioblastomas was higher than in normal brains and PDE11A knockdown reduced cancer cell proliferation. This suggests that the expression of PDE11A can regulate the development of glioblastoma in patients. Other PDEs (PDE5, PDE8, and PDE10) are also involved in cancer cell proliferation; various mutations have been described (9698). Therefore, PDE11A and other PDEs fail to act as regulators of cAMP and cGMP, affecting the growth and development of cancer cells. Furthermore, it is thought that alternatives to PDEs related to such mutation can play a clear role in CNS and testicular cancer, where PDEs are highly expressed.

Other diseases cause by PDE11A

PDE11A is expressed in the human testis, pituitary gland, heart, kidney, liver, prostate, adrenal gland, colon, and hippocampus (18,19,22,23,79), and is associated with tumors and other diseases. Adrenocorticotropin independent macronodular adrenocortical hyperplasia (a bilateral tumor) is a rare cause of Cushing's syndrome (less than 1% of all cases). Several types of adrenocortical tumors that cause the Cushing's syndrome were found to be caused by abnormal cAMP and could be caused by mutations in PDE11A (86). The bilaterality of such benign tumors suggests that a genetic factor is in play; Adrenocorticotropin independent macronodular adrenocortical hyperplasia has been associated with PDE11A mutations (99,100). This mutation was also observed in patients with acromegaly. Acromegaly is a condition in which the body produces excessive growth hormones, causing body tissues and bones to grow faster. Mutations of PDE11A (Y727C, R804H, R867G, M878V, FS41X) were reported in patients with acromegaly (101). PDE11A also affects brain expression and development. The previous paper reported that it might be related to a bipolar disorder associated with lithium reactivity (102). Moreover, two rare PDE11A pentasensory mutations were found in patients with Alzheimer's disease, and PDE11A levels were reduced in brain samples (79,103). Expression of PDE11A4 is 3–10 fold higher in the ventral hippocampus than in the eastern hippocampus. This means that in brain development, it has the potential to modulate behavior by regulating cytokine and hippocampal glutamate signaling and protein translation. Therefore, it is speculated that the expression level of PDE11A4 in the brain may affect schizophrenia and neurodevelopment (23,104). In reality, PDE11A knockout can impair protein translation required in abdominal hippocampus formation in the brian hippocampus, inhibiting memory integration, and demonstrating reduced expression of RSK2 and lower phosphorylation of S6 compared to WT mice. Based on these results, it is suggested that PDE11A can affect perception and association (105). Besides, PDE11A is also implicated in sperm physiology and is primarily described in the prostate. More specifically, PDE11A3 localizes to the testis (106), and PDE11A4 is highly expressed in the prostate and developing sperm. In addition, fertilization is also related to sperm concentration and motility and the percentage of live sperm (107). It is regulated in part by cAMP and cGMP (7). Ejaculated sperm from PDE11A knockout mice that could regulate both cAMP and cGMP showed reduced sperm concentration and rate of progression. This suggests that the expression of PDE11A may have physiological effects on tissues (107).

Further focus for PDE11A

PDE11A hydrolyzes cAMP and cGMP to AMP and GMP (82). cAMP and cGMP are essential secondary messengers involved in the development and function of the immune and endocrine systems (82). cAMP signaling is mediated by AC activity triggered by the GPCR; PKA is activated and phosphorylates intracellular proteins (4,5), controlling cellular responses, development, and function. cAMP and cGMP signaling pathways are closely associated with tumorigenesis (31,32). Germline mutational inactivation of the alpha regulatory subunit gene of Protein Kinase CAMP-Dependent Type I Regulatory Subunit Alpha triggers Carney complex (27,28) (an autosomal-dominant disease characterized by cardiac myxomas and endocrine tumors). cAMP and cGMP contribute to tumorigenesis by affecting PDE action. PDE increases the intracellular levels of cAMP and cGMP, resulting in sustained activation of the cAMP and cGMP/PKA cascade. PDE expression is high in several types of cancer cells; PDE mutations were observed. The PDE isoforms fall into 11 families that share catalytic functions but differ in terms of subcellular localization and tissue expression. For example, PDEs that degrade both cAMP and cGMP include PDE1, 2, 3, 10, and 11; PDEs that degrade only cAMP include PDE4, 7, and 8; and PDEs that degrade only cGMP include PDE5, 6, and 9 (1,810) The roles of the various PDE isoforms vary in different human tissues. PDE11A features four splice variants. PDE11A expression is highest in the prostate, but the enzyme is also expressed in the spleen, hippocampus, liver, skeletal muscle, pituitary, pancreas, and kidney (18,19). PDE11A regulates both cAMP and cGMP levels (82), is expressed in many cancer cells, and several mutations have been defined. Three PDE11A truncations and two substitution mutations were identified in 745 patients (43,86). In addition, the PDE11A level was enhanced in glioblastomas and PDE11A knockdown inhibited cancer proliferation. High-level PDE11A expression (and mutations) have been observed in various endocrine tumors in the kidney, prostate, colon, lung, and breast. The relationships between genetic PDE11A changes and tumors of the prostate, testis, and adrenal glands are under study. It is already clear that PDE11A and other PDEs are expressed by many cancers and are essential for cancer cell growth. Thus, the identification of PDE11A and other PDE-related targets may aid the treatment of refractory cancers. Moreover, the expression of PDE11A in sperm affects sperm production, motility, and survival. This suggests that PDE11A and all other PDE families may be involved in physiology and development in various tissues by regulating cAMP and cGMP depending on the location and intensity of expression. Besides, the effect of PDE11A on the development of cancer cells also has sufficient potential as a new therapeutic strategy in brain tumors and other cancers.

Acknowledgements

The authors would like to thank Prof. Guang-Ho Cha (Chungnam National University, South Korea) and Dr Sung-Jin Park (National Institute of Health, USA) for fruitful discussions.

Funding

This work was financially supported by a research fund of Chungnam National University (2020) (grant no. CNU-2020SHK) and by the National Research Foundation of Korea (NRF) grant funded by the Korea Government (MEST) (grant nos. NRF-2021R1A2C1008492, NRF-2020R1F1A1049801 and NRF-2021R1C1C2008456).

Availability of data and materials

Not applicable.

Authors' contributions

GK and JoP conceived the present study. GK and HL analyzed the PDE11A literature and made substantial contributions to finalization of this manuscript. TTV, UJ, JiP and SHK assessed and analyzed the oncogene database (https://www.cbioportal.org/). SHK, JiP, JoP and SK commented on previous versions of the manuscript. JoP and SK were involved in data interpretation and writing the discussion. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Hannah-Shmouni F, Faucz FR and Stratakis CA: Alterations of phosphodiesterases in adrenocortical tumors. Front Endocrinol (Lausanne). 7:1112016. View Article : Google Scholar : PubMed/NCBI

2 

Rall TW and Sutherland EW: Formation of a cyclic adenine ribonucleotide by tissue particles. J Biol Chem. 232:1065–1076. 1958. View Article : Google Scholar : PubMed/NCBI

3 

Butcher RW and Sutherland EW: Adenosine 3′,5′-phosphate in biological materials. I. purification and properties of cyclic 3′,5′-Nucleotide phosphodiesterase and use of this enzyme to characterize adenosine 3′,5′-phosphate in human urine. J Biol Chem. 237:1244–1250. 1962. View Article : Google Scholar : PubMed/NCBI

4 

Liu Y, Chen J, Fontes SK, Bautista EN and Cheng Z: Physiological and pathological roles of protein kinase a in the heart. Cardiovasc Res. 118:386–398. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Calamera G, Moltzau LR, Levy FO and Andressen KW: Phosphodiesterases and compartmentation of camp and cgmp signaling in regulation of cardiac contractility in normal and failing hearts. Int J Mol Sci. 23:21452022. View Article : Google Scholar

6 

Levy I, Horvath A, Azevedo M, de Alexandre RB and Stratakis CA: Phosphodiesterase function and endocrine cells: Links to human disease and roles in tumor development and treatment. Curr Opin Pharmacol. 11:689–697. 2011. View Article : Google Scholar

7 

Makhlouf A, Kshirsagar A and Niederberger C: Phosphodiesterase 11: A brief review of structure, expression and function. Int J Impot Res. 18:501–519. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Huang J, Hu B, Xu Z, Ye Y, Wang H, Wang S, Liu Z and Wang J: Selectivity mechanism of phosphodiesterase isoform inhibitor through in silico investigations. J Mol Model. 28:92021. View Article : Google Scholar

9 

Omori K and Kotera J: Overview of PDEs and their regulation. Circ Res. 100:309–327. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Ke H and Wang H: Crystal structures of phosphodiesterases and implications on substrate specificity and inhibitor selectivity. Curr Top Med Chem. 7:391–403. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Rotella DP: Phosphodiesterase 5 inhibitors: Current status and potential applications. Nat Rev Drug Discov. 1:674–682. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Galie N, Ghofrani HA, Torbicki A, Barst RJ, Rubin LJ, Badesch D, Fleming T, Parpia T, Burgess G, Branzi A, et al: Sildenafil citrate therapy for pulmonary arterial hypertension. N Engl J Med. 353:2148–2157. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Kleiman RJ, Chapin DS, Christoffersen C, Freeman J, Fonseca KR, Geoghegan KF, Grimwood S, Guanowsky V, Hajós M, Harms JF, et al: Phosphodiesterase 9a regulates central cgmp and modulates responses to cholinergic and monoaminergic perturbation in vivo. J Pharmacol Exp Ther. 341:396–409. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Schmidt CJ: Phosphodiesterase inhibitors as potential cognition enhancing agents. Curr Top Med Chem. 10:222–230. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Blokland A, Schreiber R and Prickaerts J: Improving memory: A role for phosphodiesterases. Curr Pharm Des. 12:2511–2523. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Menniti FS, Faraci WS and Schmidt CJ: Phosphodiesterases in the Cns: Targets for drug development. Nat Rev Drug Discov. 5:660–670. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Hetman JM, Robas N, Baxendale R, Fidock M, Phillips SC, Soderling SH and Beavo JA: Cloning and characterization of two splice variants of human phosphodiesterase 11A. Proc Natl Acad Sci USA. 97:12891–12895. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Fawcett L, Baxendale R, Stacey P, McGrouther C, Harrow I, Soderling S, Hetman J, Beavo JA and Phillips SC: Molecular Cloning and characterization of a distinct human phosphodiesterase gene family: PDE11A. Proc Natl Acad Sci USA. 97:3702–3707. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Yuasa K, Kotera J, Fujishige K, Michibata H, Sasaki T and Omori K: Isolation and characterization of two novel phosphodiesterase PDE11A variants showing unique structure and tissue-specific expression. J Biol Chem. 275:31469–31479. 2000. View Article : Google Scholar : PubMed/NCBI

20 

Yuasa K, Ohgaru T, Asahina M and Omori K: Identification of rat cyclic nucleotide phosphodiesterase 11A (PDE11A): Comparison of rat and human PDE11A Splicing variants. Eur J Biochem. 268:4440–4448. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Weeks JL II, Zoraghi R, Francis SH and Corbin JD: N-Terminal domain of phosphodiesterase-11A4 (PDE11A4) decreases affinity of the catalytic site for substrates and tadalafil, and is involved in oligomerization. Biochemistry. 46:10353–10364. 2007. View Article : Google Scholar : PubMed/NCBI

22 

D'Andrea MR, Qiu Y, Haynes-Johnson D, Bhattacharjee S, Kraft P and Lundeen S: Expression of PDE11A in normal and malignant human tissues. J Histochem Cytochem. 53:895–903. 2005. View Article : Google Scholar

23 

Kelly MP: A role for phosphodiesterase 11A (PDE11A) in the formation of social memories and the stabilization of mood. Adv Neurobiol. 17:201–230. 2017. View Article : Google Scholar

24 

Kelly MP: Does phosphodiesterase 11A (PDE11A) hold promise as a future therapeutic target? Curr Pharm Des. 21:389–416. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Pilarzyk K, Farmer R, Porcher L and Kelly MP: The role of PDE11A4 in social isolation-induced changes in intracellular signaling and neuroinflammation. Front Pharmacol. 12:7496282021. View Article : Google Scholar

26 

Wettschureck N and Offermanns S: Mammalian G proteins and their cell type specific functions. Physiol Rev. 85:1159–1204. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Stratakis CA: Mutations of the gene encoding the protein kinase a type I-Alpha regulatory subunit (PRKAR1A) in patients with the ‘complex of spotty skin pigmentation, myxomas, endocrine overactivity, and schwannomas’ (Carney Complex). Ann N Y Acad Sci. 968:3–21. 2002. View Article : Google Scholar

28 

Bertherat J, Horvath A, Groussin L, Grabar S, Boikos S, Cazabat L, Libe R, René-Corail F, Stergiopoulos S, Bourdeau I, et al: Mutations in regulatory subunit type 1A of cyclic adenosine 5′-Monophosphate-dependent protein kinase (PRKAR1A): Phenotype analysis in 353 patients and 80 different genotypes. J Clin Endocrinol Metab. 94:2085–2091. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Greene EL, Horvath AD, Nesterova M, Giatzakis C, Bossis I and Stratakis CA: In vitro functional studies of naturally occurring pathogenic PRKAR1A mutations that are not subject to nonsense mRNA decay. Hum Mutat. 29:633–639. 2008. View Article : Google Scholar

30 

Groussin L, Kirschner LS, Vincent-Dejean C, Perlemoine K, Jullian E, Delemer B, Zacharieva S, Pignatelli D, Carney JA, Luton JP, et al: Molecular analysis of the cyclic AMP-Dependent Protein Kinase A (PKA) regulatory subunit 1A (PRKAR1A) gene in patients with carney complex and primary pigmented nodular adrenocortical disease (PPNAD) reveals novel mutations and clues for pathophysiology: Augmented PKA signaling is associated with adrenal tumorigenesis in PPNAD. Am J Hum Genet. 71:1433–1442. 2002. View Article : Google Scholar

31 

Horvath A, Bertherat J, Groussin L, Guillaud-Bataille M, Tsang K, Cazabat L, Libé R, Remmers E, René-Corail F, Faucz FR, et al: Mutations and polymorphisms in the gene encoding regulatory subunit type 1-alpha of protein kinase A (PRKAR1A): An update. Hum Mutat. 31:369–379. 2010. View Article : Google Scholar

32 

Sandrini F and Stratakis C: Clinical and molecular genetics of carney complex. Mol Genet Metab. 78:83–92. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, Cho-Chung YS and Stratakis CA: Mutations of the gene encoding the protein kinase a type I-alpha regulatory subunit in patients with the carney complex. Nat Genet. 26:89–92. 2000. View Article : Google Scholar

34 

Weinstein LS, Shenker A, Gejman PV, Merino MJ, Friedman E and Spiegel AM: Activating mutations of the stimulatory G protein in the McCune-albright syndrome. N Engl J Med. 325:1688–1695. 1991. View Article : Google Scholar : PubMed/NCBI

35 

Stewart V and Yanofsky C: Role of leader peptide synthesis in tryptophanase operon expression in Escherichia Coli K-12. J Bacteriol. 167:383–386. 1986. View Article : Google Scholar

36 

Velterop JS, Sellink E, Meulenberg JJ, David S, Bulder I and Postma PW: Synthesis of pyrroloquinoline quinone in vivo and in vitro and detection of an intermediate in the biosynthetic pathway. J Bacteriol. 177:5088–5098. 1995. View Article : Google Scholar

37 

Meulenberg JJ, Sellink E, Riegman NH and Postma PW: Nucleotide sequence and structure of the klebsiella pneumoniae Pqq operon. Mol Gen Genet. 232:284–294. 1992. View Article : Google Scholar

38 

Roelofs J, Smith JL and Van Haastert PJ: Cgmp signalling: Different ways to create a pathway. Trends Genet. 19:132–134. 2003. View Article : Google Scholar

39 

Ochman H: Distinguishing the ORFs from the ELFs: Short bacterial genes and the annotation of genomes. Trends Genet. 18:335–337. 2002. View Article : Google Scholar

40 

Yanofsky C: Transcription Attenuation. J Biol Chem. 263:609–612. 1988. View Article : Google Scholar : PubMed/NCBI

41 

You JY, Liu XW, Bao YX, Shen ZN, Wang Q, He GY, Lu J, Zhang JG, Chen JW and Liu PQ: A novel phosphodiesterase 9A inhibitor LW33 protects against ischemic stroke through the cGMP/PKG/CREB Pathway. Eur J Pharmacol. 925:1749872022. View Article : Google Scholar

42 

Libe R, Fratticci A, Coste J, Tissier F, Horvath A, Ragazzon B, Rene-Corail F, Groussin L, Bertagna X, Raffin-Sanson ML, et al: Phosphodiesterase 11A (PDE11A) and genetic predisposition to adrenocortical tumors. Clin Cancer Res. 14:4016–4024. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Horvath A, Boikos S, Giatzakis C, Robinson-White A, Groussin L, Griffin KJ, Stein E, Levine E, Delimpasi G, Hsiao HP, et al: A genome-wide scan identifies mutations in the gene encoding phosphodiesterase 11A4 (PDE11A) in individuals with adrenocortical hyperplasia. Nat Genet. 38:794–800. 2006. View Article : Google Scholar

44 

Horvath A, Korde L, Greene MH, Libe R, Osorio P, Faucz FR, Raffin-Sanson ML, Tsang KM, Drori-Herishanu L, Patronas Y, et al: Functional phosphodiesterase 11A mutations may modify the risk of familial and bilateral testicular germ cell tumors. Cancer Res. 69:5301–5306. 2009. View Article : Google Scholar : PubMed/NCBI

45 

de Alexandre RB, Horvath AD, Szarek E, Manning AD, Leal LF, Kardauke F, Epstein JA, Carraro DM, Soares FA, Apanasovich TV, et al: Phosphodiesterase Sequence variants may predispose to prostate cancer. Endocr Relat Cancer. 22:519–530. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Lefievre L, de Lamirande E and Gagnon C: Presence of Cyclic nucleotide phosphodiesterases PDE1A, existing as a stable complex with calmodulin, and PDE3A in human spermatozoa. Biol Reprod. 67:423–430. 2002. View Article : Google Scholar : PubMed/NCBI

47 

Fidock M, Miller M and Lanfear J: Isolation and differential tissue distribution of two human cDNAs encoding PDE1 splice variants. Cell Signal. 14:53–60. 2002. View Article : Google Scholar

48 

Michibata H, Yanaka N, Kanoh Y, Okumura K and Omori K: Human Ca2+/Calmodulin-dependent phosphodiesterase PDE1A: Novel splice variants, their specific expression, genomic organization, and chromosomal localization. Biochim Biophys Acta. 1517:278–287. 2001. View Article : Google Scholar

49 

Loughney K, Martins TJ, Harris EA, Sadhu K, Hicks JB, Sonnenburg WK, Beavo JA and Ferguson K: Isolation and Characterization of CDNAs corresponding to two human calcium, calmodulin-regulated, 3′,5′-cyclic nucleotide phosphodiesterases. J Biol Chem. 271:796–806. 1996. View Article : Google Scholar : PubMed/NCBI

50 

Kanda N and Watanabe S: Regulatory roles of adenylate cyclase and cyclic nucleotide phosphodiesterases 1 and 4 in interleukin-13 production by activated human T cells. Biochem Pharmacol. 62:495–507. 2001. View Article : Google Scholar

51 

Yan C, Zhao AZ, Bentley JK, Loughney K, Ferguson K and Beavo JA: Molecular cloning and characterization of a calmodulin-dependent phosphodiesterase enriched in olfactory sensory neurons. Proc Natl Acad Sci USA. 92:9677–9681. 1995. View Article : Google Scholar : PubMed/NCBI

52 

Nikolaev VO, Gambaryan S, Engelhardt S, Walter U and Lohse MJ: Real-Time Monitoring of the PDE2 activity of live cells: Hormone-stimulated camp hydrolysis is faster than hormone-stimulated camp synthesis. J Biol Chem. 280:1716–1719. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Maurice DH, Palmer D, Tilley DG, Dunkerley HA, Netherton SJ, Raymond DR, Elbatarny HS and Jimmo SL: Cyclic nucleotide phosphodiesterase activity, expression, and targeting in cells of the cardiovascular system. Mol Pharmacol. 64:533–546. 2003. View Article : Google Scholar

54 

Degerman E, Belfrage P and Manganiello VC: Structure, localization, and regulation of cgmp-inhibited phosphodiesterase (PDE3). J Biol Chem. 272:6823–6826. 1997. View Article : Google Scholar : PubMed/NCBI

55 

Mongillo M, Tocchetti CG, Terrin A, Lissandron V, Cheung YF, Dostmann WR, Pozzan T, Kass DA, Paolocci N, Houslay MD and Zaccolo M: Compartmentalized phosphodiesterase-2 activity blunts beta-adrenergic cardiac inotropy via an No/cGMP-dependent pathway. Circ Res. 98:226–234. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Bender AT, Ostenson CL, Giordano D and Beavo JA: Differentiation of human monocytes in vitro with granulocyte-macrophage colony-stimulating factor and macrophage colony-stimulating factor produces distinct changes in cGMP phosphodiesterase expression. Cell Signal. 16:365–374. 2004. View Article : Google Scholar

57 

Seybold J, Thomas D, Witzenrath M, Boral S, Hocke AC, Burger A, Hatzelmann A, Tenor H, Schudt C, Krüll M, et al: Tumor necrosis factor-alpha-dependent expression of phosphodiesterase 2: Role in endothelial hyperpermeability. Blood. 105:3569–3576. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Domek-Lopacinska K and Strosznajder JB: The effect of selective inhibition of cyclic GMP hydrolyzing phosphodiesterases 2 and 5 on learning and memory processes and nitric oxide synthase activity in brain during aging. Brain Res. 1216:68–77. 2008. View Article : Google Scholar : PubMed/NCBI

59 

de Oliveira SK and Smolenski A: Phosphodiesterases link the aryl hydrocarbon receptor complex to cyclic nucleotide signaling. Biochem Pharmacol. 77:723–733. 2009. View Article : Google Scholar

60 

Rena G, Begg F, Ross A, MacKenzie C, McPhee I, Campbell L, Huston E, Sullivan M and Houslay MD: Molecular cloning, genomic positioning, promoter identification, and characterization of the novel cyclic amp-specific phosphodiesterase PDE4A10. Mol Pharmacol. 59:996–1011. 2001. View Article : Google Scholar

61 

Wang P, Wu P, Ohleth KM, Egan RW and Billah MM: Phosphodiesterase 4B2 is the predominant phosphodiesterase species and undergoes differential regulation of gene expression in human monocytes and neutrophils. Mol Pharmacol. 56:170–174. 1999. View Article : Google Scholar

62 

Bolger G, Michaeli T, Martins T, St John T, Steiner B, Rodgers L, Riggs M, Wigler M and Ferguson K: A family of human phosphodiesterases homologous to the dunce learning and memory gene product of drosophila melanogaster are potential targets for antidepressant drugs. Mol Cell Biol. 13:6558–6571. 1993. View Article : Google Scholar

63 

Dunkern TR and Hatzelmann A: The effect of sildenafil on human platelet secretory function is controlled by a complex interplay between phosphodiesterases 2, 3 and 5. Cell Signal. 17:331–339. 2005. View Article : Google Scholar

64 

Prickaerts J, Sik A, van Staveren WC, Koopmans G, Steinbusch HW, van der Staay FJ, de Vente J and Blokland A: Phosphodiesterase type 5 inhibition improves early memory consolidation of object information. Neurochem Int. 45:915–928. 2004. View Article : Google Scholar

65 

Miller CL and Yan C: Targeting cyclic nucleotide phosphodiesterase in the heart: Therapeutic implications. J Cardiovasc Transl Res. 3:507–515. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Ridge KD, Abdulaev NG, Sousa M and Palczewski K: Phototransduction: Crystal clear. Trends Biochem Sci. 28:479–487. 2003. View Article : Google Scholar

67 

Morin F, Lugnier C, Kameni J and Voisin P: Expression and role of phosphodiesterase 6 in the chicken pineal gland. J Neurochem. 78:88–99. 2001. View Article : Google Scholar : PubMed/NCBI

68 

Bloom TJ and Beavo JA: Identification and tissue-specific expression of PDE7 phosphodiesterase splice variants. Proc Natl Acad Sci USA. 93:14188–14192. 1996. View Article : Google Scholar : PubMed/NCBI

69 

Han P, Zhu X and Michaeli T: Alternative splicing of the high affinity cAMP-specific phosphodiesterase (PDE7A) mRNA in human skeletal muscle and heart. J Biol Chem. 272:16152–16157. 1997. View Article : Google Scholar : PubMed/NCBI

70 

Sasaki T, Kotera J and Omori K: Transcriptional activation of phosphodiesterase 7B1 by dopamine d1 receptor stimulation through the cyclic AMP/Cyclic AMP-dependent protein kinase/cyclic AMP-response element binding protein pathway in primary striatal neurons. J Neurochem. 89:474–483. 2004. View Article : Google Scholar : PubMed/NCBI

71 

Glavas NA, Ostenson C, Schaefer JB, Vasta V and Beavo JA: T cell activation up-regulates cyclic nucleotide phosphodiesterases 8A1 and 7A3. Proc Natl Acad Sci USA. 98:6319–6324. 2001. View Article : Google Scholar : PubMed/NCBI

72 

Patrucco E, Albergine MS, Santana LF and Beavo JA: Phosphodiesterase 8A (PDE8A) regulates excitation-contraction coupling in ventricular myocytes. J Mol Cell Cardiol. 49:330–333. 2010. View Article : Google Scholar

73 

Mehats C, Andersen CB, Filopanti M, Jin SL and Conti M: Cyclic nucleotide phosphodiesterases and their role in endocrine cell signaling. Trends Endocrinol Metab. 13:29–35. 2002. View Article : Google Scholar : PubMed/NCBI

74 

Hayashi M, Matsushima K, Ohashi H, Tsunoda H, Murase S, Kawarada Y and Tanaka T: Molecular cloning and characterization of human PDE8B, a novel thyroid-specific isozyme of 3′,5′-cyclic nucleotide phosphodiesterase. Biochem Biophys Res Commun. 250:751–756. 1998. View Article : Google Scholar : PubMed/NCBI

75 

Hayashi M, Shimada Y, Nishimura Y, Hama T and Tanaka T: Genomic organization, chromosomal localization, and alternative splicing of the human phosphodiesterase 8B gene. Biochem Biophys Res Commun. 297:1253–1258. 2002. View Article : Google Scholar : PubMed/NCBI

76 

Horvath A, Giatzakis C, Tsang K, Greene E, Osorio P, Boikos S, Libè R, Patronas Y, Robinson-White A, Remmers E, et al: A cAMP-specific phosphodiesterase (PDE8B) that is mutated in adrenal hyperplasia is expressed widely in human and mouse tissues: A novel PDE8B isoform in human adrenal cortex. Eur J Hum Genet. 16:1245–1253. 2008. View Article : Google Scholar

77 

Rentero C, Monfort A and Puigdomenech P: Identification and distribution of different mRNA variants produced by differential splicing in the human phosphodiesterase 9A gene. Biochem Biophys Res Commun. 301:686–692. 2003. View Article : Google Scholar : PubMed/NCBI

78 

Furukawa T, Youssef EM, Yatsuoka T, Yokoyama T, Makino N, Inoue H, Fukushige S, Hoshi M, Hayashi Y, Sunamura M and Horii A: Cloning and characterization of the human Udp-N-Acetylglucosamine: Alpha-1,3-D-mannoside beta-1,4-N-acetylglucosaminyltransferase IV-Homologue (hGnT-IV-H) gene. J Hum Genet. 44:397–401. 1999. View Article : Google Scholar

79 

Kelly MP, Logue SF, Brennan J, Day JP, Lakkaraju S, Jiang L, Zhong X, Tam M, Sukoff Rizzo SJ, Platt BJ, et al: Phosphodiesterase 11A in brain is enriched in ventral hippocampus and deletion causes psychiatric disease-related phenotypes. Proc Natl Acad Sci USA. 107:8457–8462. 2010. View Article : Google Scholar : PubMed/NCBI

80 

Kleppisch T: Phosphodiesterases in the central nervous system. Handb Exp Pharmacol. 71–92. 2009. View Article : Google Scholar

81 

Knott EP, Assi M, Rao SN, Ghosh M and Pearse DD: Phosphodiesterase inhibitors as a therapeutic approach to neuroprotection and repair. Int J Mol Sci. 18:6962017. View Article : Google Scholar

82 

Libe R, Horvath A, Vezzosi D, Fratticci A, Coste J, Perlemoine K, Ragazzon B, Guillaud-Bataille M, Groussin L, Clauser E, et al: Frequent phosphodiesterase 11A gene (PDE11A) defects in patients with carney complex (CNC) Caused by PRKAR1A Mutations: PDE11A may contribute to adrenal and testicular tumors in CNC as a modifier of the phenotype. J Clin Endocrinol Metab. 96:E208–E214. 2011. View Article : Google Scholar : PubMed/NCBI

83 

Jager R, Russwurm C, Schwede F, Genieser HG, Koesling D and Russwurm M: Activation of PDE10 and PDE11 phosphodiesterases. J Biol Chem. 287:1210–1219. 2012. View Article : Google Scholar : PubMed/NCBI

84 

Pitsava G and Stratakis CA: Genetic alterations in benign adrenal tumors. Biomedicines. 10:10412022. View Article : Google Scholar : PubMed/NCBI

85 

Hsiao HP, Kirschner LS, Bourdeau I, Keil MF, Boikos SA, Verma S, Robinson-White AJ, Nesterova M, Lacroix A and Stratakis CA: Clinical and genetic heterogeneity, overlap with other tumor syndromes, and atypical glucocorticoid hormone secretion in adrenocorticotropin-independent macronodular adrenal hyperplasia compared with other adrenocortical tumors. J Clin Endocrinol Metab. 94:2930–2937. 2009. View Article : Google Scholar : PubMed/NCBI

86 

Horvath A, Giatzakis C, Robinson-White A, Boikos S, Levine E, Griffin K, Stein E, Kamvissi V, Soni P, Bossis I, et al: Adrenal hyperplasia and adenomas are associated with inhibition of phosphodiesterase 11A in carriers of PDE11A sequence variants that are frequent in the population. Cancer Res. 66:11571–11575. 2006. View Article : Google Scholar : PubMed/NCBI

87 

Pathak A, Stewart DR, Faucz FR, Xekouki P, Bass S, Vogt A, Zhang X, Boland J, Yeager M, Loud JT, et al: Rare inactivating PDE11A variants associated with testicular germ cell tumors. Endocr Relat Cancer. 22:909–917. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Dal J, Nielsen EH, Klose M, Feldt-Rasmussen U, Andersen M, Vang S, Korbonits M and Jørgensen JOL: Phenotypic and genotypic features of a large kindred with a germline AIP variant. Clin Endocrinol (Oxf). 93:146–153. 2020. View Article : Google Scholar : PubMed/NCBI

89 

Pinto EM, Faucz FR, Paza LZ, Wu G, Fernandes ES, Bertherat J, Stratakis CA, Lalli E, Ribeiro RC, Rodriguez-Galindo C, et al: Germline variants in phosphodiesterase genes and genetic predisposition to pediatric adrenocortical tumors. Cancers (Basel). 12:5062020. View Article : Google Scholar

90 

Faucz FR, Horvath A, Rothenbuhler A, Almeida MQ, Libe R, Raffin-Sanson ML, Bertherat J, Carraro DM, Soares FA, Molina Gde C, et al: Phosphodiesterase 11A (PDE11A) genetic variants may increase susceptibility to prostatic cancer. J Clin Endocrinol Metab. 96:E135–E140. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Dono A, Nickles J, Rodriguez-Armendariz AG, McFarland BC, Ajami NJ, Ballester LY, Wargo JA and Esquenazi Y: Glioma and the gut-brain axis: Opportunities and future perspectives. Neurooncol Adv. 4:vdac0542022.PubMed/NCBI

92 

Schwartz KA, Noel M, Nikolai M, Olson LK, Hord NG, Zakem M, Clark J, Elnabtity M, Figueroa B and Chang HT: Long term survivals in aggressive primary brain malignancies treated with an adjuvant ketogenic diet. Front Nutr. 9:7707962022. View Article : Google Scholar : PubMed/NCBI

93 

Burns TC, Awad AJ, Li MD and Grant GA: Radiation-induced brain injury: Low-hanging fruit for neuroregeneration. Neurosurg Focus. 40:E32016. View Article : Google Scholar : PubMed/NCBI

94 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

95 

Lee H, Park S, Kong G, Kwon SH, Park J, Park J and Kim SH: Phosphodiesterase 11A (PDE11A), a potential biomarker for glioblastoma. Toxicol Res. 2022. View Article : Google Scholar

96 

Rothenbuhler A, Horvath A, Libe R, Faucz FR, Fratticci A, Raffin Sanson ML, Vezzosi D, Azevedo M, Levy I, Almeida MQ, et al: Identification of novel genetic variants in phosphodiesterase 8B (PDE8B), a cAMP-specific phosphodiesterase highly expressed in the adrenal cortex, in a cohort of patients with adrenal tumours. Clin Endocrinol (Oxf). 77:195–199. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Hou Y, Wren A, Mylarapu N, Browning K, Islam BN, Wang R, Vega KJ and Browning DD: Inhibition of colon cancer cell growth by phosphodiesterase inhibitors is independent of cGMP Signaling. J Pharmacol Exp Ther. 381:42–53. 2022. View Article : Google Scholar : PubMed/NCBI

98 

Di Iorio P, Ronci M, Giuliani P, Caciagli F, Ciccarelli R, Caruso V, Beggiato S and Zuccarini M: Pros and cons of pharmacological manipulation of cGMP-PDEs in the prevention and treatment of breast cancer. Int J Mol Sci. 23:2622021. View Article : Google Scholar

99 

Vezzosi D, Cartier D, Regnier C, Otal P, Bennet A, Parmentier F, Plantavid M, Lacroix A, Lefebvre H and Caron P: Familial adrenocorticotropin-independent macronodular adrenal hyperplasia with aberrant serotonin and vasopressin adrenal receptors. Eur J Endocrinol. 156:21–31. 2007. View Article : Google Scholar

100 

Vezzosi D, Libe R, Baudry C, Rizk-Rabin M, Horvath A, Levy I, René-Corail F, Ragazzon B, Stratakis CA, Vandecasteele G and Bertherat J: Phosphodiesterase 11A (PDE11A) gene defects in patients with acth-independent macronodular adrenal hyperplasia (AIMAH): Functional variants may contribute to genetic susceptibility of bilateral adrenal tumors. J Clin Endocrinol Metab. 97:E2063–E2069. 2012. View Article : Google Scholar : PubMed/NCBI

101 

Peverelli E, Ermetici F, Filopanti M, Elli FM, Ronchi CL, Mantovani G, Ferrero S, Bosari S, Beck-Peccoz P, Lania A and Spada A: Analysis of genetic variants of phosphodiesterase 11A in acromegalic patients. Eur J Endocrinol. 161:687–694. 2009. View Article : Google Scholar

102 

Pathak G, Agostino MJ, Bishara K, Capell WR, Fisher JL, Hegde S, Ibrahim BA, Pilarzyk K, Sabin C, Tuczkewycz T, et al: PDE11A negatively regulates lithium responsivity. Mol Psychiatry. 22:1714–1724. 2017. View Article : Google Scholar : PubMed/NCBI

103 

Qin W, Zhou A, Zuo X, Jia L, Li F, Wang Q, Li Y, Wei Y, Jin H, Cruchaga C, et al: Exome Sequencing Revealed PDE11A as a novel candidate gene for early-onset Alzheimer's disease. Hum Mol Genet. 30:811–822. 2021. View Article : Google Scholar

104 

Pilarzyk K, Klett J, Pena EA, Porcher L, Smith AJ and Kelly MP: Loss of function of phosphodiesterase 11A4 shows that recent and remote long-term memories can be uncoupled. Curr Biol. 29:2307–2321. e52019. View Article : Google Scholar

105 

Hegde S, Capell WR, Ibrahim BA, Klett J, Patel NS, Sougiannis AT and Kelly MP: Phosphodiesterase 11A (PDE11A), enriched in ventral hippocampus neurons, is required for consolidation of social but not nonsocial memories in mice. Neuropsychopharmacology. 41:2920–2931. 2016. View Article : Google Scholar : PubMed/NCBI

106 

Loughney K, Taylor J and Florio VA: 3′,5′-cyclic nucleotide phosphodiesterase 11A: Localization in human tissues. Int J Impot Res. 17:320–325. 2005. View Article : Google Scholar : PubMed/NCBI

107 

Wayman C, Phillips S, Lunny C, Webb T, Fawcett L, Baxendale R and Burgess G: Phosphodiesterase 11 (PDE11) regulation of spermatozoa physiology. Int J Impot Res. 17:216–223. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2022
Volume 26 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kong G, Lee H, Vo TT, Juang U, Kwon SH, Park J, Park J and Kim S: Functional characteristics and research trends of PDE11A in human diseases (Review). Mol Med Rep 26: 298, 2022
APA
Kong, G., Lee, H., Vo, T.T., Juang, U., Kwon, S.H., Park, J. ... Kim, S. (2022). Functional characteristics and research trends of PDE11A in human diseases (Review). Molecular Medicine Reports, 26, 298. https://doi.org/10.3892/mmr.2022.12814
MLA
Kong, G., Lee, H., Vo, T. T., Juang, U., Kwon, S. H., Park, J., Park, J., Kim, S."Functional characteristics and research trends of PDE11A in human diseases (Review)". Molecular Medicine Reports 26.4 (2022): 298.
Chicago
Kong, G., Lee, H., Vo, T. T., Juang, U., Kwon, S. H., Park, J., Park, J., Kim, S."Functional characteristics and research trends of PDE11A in human diseases (Review)". Molecular Medicine Reports 26, no. 4 (2022): 298. https://doi.org/10.3892/mmr.2022.12814