AKAP12 A kinase (PRKA) anchor protein 12

2014-10-01   Irwin H Gelman 

Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA

Identity

HGNC
LOCATION
6q25.1
IMAGE
Atlas Image
LEGEND
AKAP12 (6q24-25.2) in normal cells probed with a about 5Kb human AKAP12 probe. Courtesy of Irwin Gelman; adapted from Xia et al., Cancer Res.61:5644-5651, 2001.
LOCUSID
ALIAS
AKAP250,SSeCKS
FUSION GENES

Abstract

Review on AKAP1, with data on DNA, on the protein encoded, and where the gene is implicated.

DNA/RNA

Note

The AKAP12 gene and promoter structure is strongly conserved from fish to humans, including syntenic regions conserved in the mouse (chrom. 10) and rat (chrom. 1). No family members (homologues) other than AKAP12 exist within a given species.
Atlas Image
Human and mouse cells have similar exon/intron usage and spacing. AKAP12 has three independent promoters, α, β, and γ. The gamma promoter is active only in the testes while the α and β are co-active in most cells and tissues studied. Exons 1A1 and 1A2 combine to then splice to a common splice acceptor on Exon 2 used by Exon 1B. Exons 1A1 and 1A2 produce the N-terminal 103 amino acids of "AKAP12 alpha" whereas Exon 1B encodes the N-terminal 8 amino acids of "AKAP12 β"; the remaining amino acids are encoded in Exon 2. "AKAP12 gamma" is encoded by a read-through transcript starting in the intron upstream of Exon 2, utilizing an in-frame ATG in Exon 2. Therefore, the α, β, and γ transcripts encode proteins that only differ in their N-termini.

Transcription

Human and rodent AKAP12 is expressed ubiquitously, with greater expression in mesenchymal cells, expression in specific epithelial types, such as breast and prostate, and little in most endothelial cells. Several highly specialized, differentiated cells type, such as parietal cells in the glomerulus, pericytes, astrocytes and Purkinje cells, show especially strong expression.

Pseudogene

None.

Proteins

Atlas Image
A: Except for testes, most cells express four major isoforms of AKAP12 protein. The 305 kDa isoforms is the myristylated AKAP12alpha whereas the 287 kDa isoforms is AKAP12beta. The 250 kDa and 43 kDa isoforms are proteolytic cleavage products common to the AKAP12alpha and beta isoforms.
B: Human AKAP12alpha encodes a 1,780 amino acids full-length protein. The first about 1,000 amino acids of human and rodent AKAP12 share 83% identity followed by about 600 amino acids with less than 20% identity. The N-terminal homology domain (green) shows about 40% identity to the Xgl (Xenopus gravin-like) gene in Xenopus. Both human and rodent AKAP12 share a shorter C-terminal domain containing the PKA-RII binding (AKAP) domain (green box in human AKAP12).

Expression

AKAP12 isoforms are expressed in most tissue and organ types, with high expression levels in the testes, ovary, brain, lung and heart. Most mesenchyme, smooth muscle and some epithelial cells (breast, prostate, lung, ovary) express significant AKAP12 levels. Lower levels of AKAP12 are found in endothelial cells, although express in these cells is usually associated with wounding and/or inflammation. High expression is also found in some specialized, differentiated cells such as the parietal cells of the glomerulus, pericytes, astrocytes and Purkinje cells.

Localisation

Most cell types display a cortical cytoskeletal staining pattern for AKAP12, with enrichment at the plasma membrane (associated with N-terminal myristylation and at least three N-terminal polybasic domains) and in the perinucleus. However, some staining has been observed in cell nuclei, probably directed by 4 SV40 Tag-like nuclear localization signals (NLS) found in the N-terminal third of the protein.

Function

1) Facilitates the sensitization/resensitization reaction of beta-adrenergic receptors.
2) Scaffolds protein kinase (PK) A, PKC, and SRC.
3) Autoantigen in some cases of myasthenia gravis.
4) Anti-angiogenic factor. The rodent orthologue has been shown to inhibit brain angiogenesis and induce the blood-brain barrier, and to inhibit VEGF-mediated metastasis.
5) Metastasis suppressor (see Gelman, 2012). Many studies show i) AKAP12 downregulation in metastases compared to primary-site tumors, associated with hypermethylation of promoter CpG islands, ii) AKAP12-/- mice are tumor- and metastasis-prone to skin carcinogens, iii) increased spontaneous lymph node metastasis of high-grade prostatic intraepithelial neoplasia in transgenic models, and iv) selective suppression of spontaneous and experimental metastasis following re-expression of rodent Akap12 in MAT-LyLu prostate cancer cells.
6) Effector of Polo-like kinase 1 to regulate cytokinesis.
7) Maintenance of barrier functions such as blood-brain, blood-retinal barriers and fibrotic scars in CNS repair.
8) Regulation of adhesion, chemotaxis and invasiveness through the control of actin cytoskeletal remodeling.
Atlas Image
AKAP12 is phosphorylated by PLK1 on T766 during metaphase, and shRNa knockdown of AKAP12 leads to cytokinesis failure (Canton et al., 2012).

Homology

Southern blotting analysis as well as analysis of sequenced genomes indicates that vertebrates encode single AKAP12 orthologues, with no gene family members. Thus, the protein diversity of this gene stems from promoter choice, alternative splicing, proteolytic maturation and post-translational modification. AKAP12 has limited sequence homology based on short domains. For example, the C-terminal AKAP domain is homologous to the analogous domain in AKP79. Also, AKAP12 shares some so-called MARCKS protein-like effector domains- positively charged stretches of amino acids involved in plasma membrane targeting.

Mutations

Note

141 mutations (mostly point mutants and some small deletions) have been described in COSMIC, the vast majority of which are found in the major protein-coding exon ("exon 2")
ESR1-AKAP12 fusions have been recently described (Ma et al., 2014) in clinical cases of breast cancer.
. There are at least 539 single nucleotide polymorphisms (SNP). 59 study datasets for AKAP12 SNPs described in GWAS analyses For example, HGVST779 identifies AKAP12 SNPs associated with chronic myelogenous leukemia. Another study identifies AKAP12 SNPs associated with psychiatric depression (Ripke et al., 2012).
A number of QTLs have been described associated with susceptibility for prostate cancer and insulin sensitivity, and for HDL cholesterol.

Implicated in

Entity name
Prostate cancer
Note
Tumor/metastasis suppressor.
Prognosis
AKAP12 downregulation correlates with earlier time-to-onset of metastasis (Ko et al., 2014).
Cytogenetics
gene deletion at 6q24-25.1.
Atlas Image
Left- Oncomine analysis (http://www.oncomine.org) of Lapointe prostate cancer expression data (Proc Natl Acad Sci U S A 2004;101:811-6) showing AKAP12 downregulation in human prostate cancer metastasis compared with levels in primary prostate cancer (1 prostate cancer) or normal prostate tissue. F, Kaplan-Meier plot analysis (http://www.cbioportal.org/public-portal/) of metastasis occurrence vs. time to onset in 37 prostate cancer metastasis cases from Taylor et al., 2010 in which 11 cases (29.7%) displayed AKAP12 downregulation compared with levels in primary prostate cancer lesions versus 26 cases with no change in AKAP12 levels.
Oncogenesis
Presumably, the loss of AKAP12s scaffolding activity for kinases such as Src and PKC result in hyperactivated pathways that contribute to oncogenic transformation, as shown in Su et al., 2010, and Su et al., 2013.
Entity name
Melanoma
Note
Tumor/metastasis suppressor/promoter.
Prognosis
Some papers identify AKAP12 downregulation as a marker human melanoma progression (Hacker et al., 2008; Bonazzi et al., 2009), or that its genetic loss in transgenic models leads to a metastasis-prone condition induced by skin carcinogens (Akakura et al., 2011). However, one paper suggests that hypoxia-induced expression of AKAP12-variant 2 (β isoform) enhances melanoma migration and invasiveness.
Oncogenesis
Presumably, the loss of AKAP12s scaffolding activity for kinases such as Src and PKC result in hyperactivated pathways that contribute to oncogenic transformation, as shown in Su et al., 2010 and Su et al., 2013.
Entity name
Ovarian cancer
Note
Possible mediator of drug-resistance.
Prognosis
AKAP12 upregulation associated with resistance to platins and taxanes (Chappell et al., 2012; Bateman et al., 2015).
Atlas Image
A majority of high-grade (HG) serous ovarian cancer (SOC) patients develop resistant disease despite high initial response rates to platinum/paclitaxel-based chemotherapy. We identified shed/secreted proteins in preclinical models of paclitaxel-resistant human HGSOC models and correlated these candidate proteins with patient outcomes using public data from HGSOC patients. Proteomic analyses of a HGSOC cell line secretome was compared to those from a syngeneic paclitaxel-resistant variant and from a line established from an intrinsically chemorefractory HGSOC patient. Associations between the identified candidate proteins and patient outcome were assessed in a discovery cohort of 545 patients and two validation cohorts totaling 795 independent SOC patients. Among the 81 differentially abundant proteins identified (q < 0.05) from paclitaxel-sensitive vs -resistant HGSOC cell secretomes, AKAP12 was verified to be elevated in all models of paclitaxel-resistant HGSOC. Furthermore, elevated AKAP12 transcript expression was associated with worse progression-free and overall survival. Associations with outcome were observed in three independent cohorts and remained significant after adjusted multivariate modeling. We further provide evidence to support that differential gene methylation status is associated with elevated expression of AKAP12 in taxol-resistant ovarian cancer cells and ovarian cancer patient subsets. Elevated expression and shedding/secretion of AKAP12 is characteristic of paclitaxel-resistant HGSOC cells, and elevated AKAP12 transcript expression is a poor prognostic and predictive marker for progression-free and overall survival in SOC patients. [from Bateman et al., 2015].
Oncogenesis
Presumably, the upregulation of AKAP12, and its ability to scaffold kinases such as Src and PKC, and to move them to cellular specific compartments, leads to pathways that facilitate drug resistance, as described in Le and Bast 2011.
Entity name
Pancreatic cancer
Note
Possible tumor/metastasis suppressor.
Prognosis
Whereas AKAP12 upregulation was noted in human pancreatic cells lines compared to immortalized human pancreatic ductal epithelial cells (Cao et al., Cancer Biol Ther. 2004, 3(11):1081-9), AKAP12 loss (due to promoter hypermethylation) correlated with increased liver metastasis in pancreatic ductal adenocarcinoma (Mardin et al., 2010).
Oncogenesis
Presumably, the loss of AKAP12s scaffolding activity for kinases such as Src and PKC result in hyperactivated pathways that contribute to oncogenic transformation, as shown in Su et al. 2010, and Su et al. 2013.
Entity name
Colon cancer
Note
Possible tumor/metastasis suppressor.
Prognosis
Downregulation of AKAP12 correlated with colon cancer progression (Zhu et al., J Cancer Res Ther. 2013, 9(3):467-70), especially due to promoter hypermethylation (Mori et al., 2006; Liu et al., 2010). Moreover, AKAP12 expression is positively regulated by TGFβ; in a SMAD4 -dependent manner in colon cancer cell lines (Ali et al., 2010), and given that SMAD4 is often mutated or deleted in colon cancer (Bellam and Pasche, 2010), the downregulation of AKAP12 might be related to SMAD4 loss. Re-expression of AKAP12 inhibits the metastatic potential of colorectal carcinoma cells (Liu et al., 2011).
Entity name
Lung cancer
Note
Possible tumor suppressor.
Prognosis
ownregulation of AKAP12 due to promoter hypermethylation in lung cancer as a susceptibility (Tessema et al., 2008) or progression marker (Jo et al., 2010). As in the case of ovarian cancer, AKAP12 expression increases in cisplatin-resistant variants of lung cancer cell lines (Lopez-Ayllon et al., 2014.
Oncogenesis
Presumably, the loss of AKAP12s scaffolding activity for kinases such as Src and PKC result in hyperactivated pathways that contribute to oncogenic transformation, as shown in Su et al., 2010 and Su et al., 2013.
Entity name
Gastric cancer
Note
Possible tumor suppressor.
Prognosis
Downregulation of AKAP12 due to promoter hypermethylation in gastric cancer (Choi et al., 2004) or in Barretts esophagus (Jin et al., 2008).
Oncogenesis
Presumably, the loss of AKAP12s scaffolding activity for kinases such as Src and PKC result in hyperactivated pathways that contribute to oncogenic transformation, as shown in Su et al., 2010 and Su et al., 2013.
Entity name
Liver cancer
Note
Possible tumor suppressor.
Prognosis
Downregulation of AKAP12 due to promoter hypermethylation in liver cancer (Goeppert et al., 2010; Hayashi et al., 2012).
Oncogenesis
Presumably, the loss of AKAP12s scaffolding activity for kinases such as Src and PKC result in hyperactivated pathways that contribute to oncogenic transformation, as shown in Su et al., 2010 and Su et al., 2013.

Bibliography

Pubmed IDLast YearTitleAuthors

Other Information

Locus ID:

NCBI: 9590
MIM: 604698
HGNC: 370
Ensembl: ENSG00000131016

Variants:

dbSNP: 9590
ClinVar: 9590
TCGA: ENSG00000131016
COSMIC: AKAP12

RNA/Proteins

Gene IDTranscript IDUniprot
ENSG00000131016ENST00000253332Q02952
ENSG00000131016ENST00000354675Q02952
ENSG00000131016ENST00000359755Q02952
ENSG00000131016ENST00000402676Q02952

Expression (GTEx)

0
50
100
150
200

Protein levels (Protein atlas)

Not detected
Low
Medium
High

References

Pubmed IDYearTitleCitations
166420352006An anchored PKA and PDE4 complex regulates subplasmalemmal cAMP dynamics.84
146570152003Protein kinase A regulates AKAP250 (gravin) scaffold binding to the beta2-adrenergic receptor.44
200188902010SSeCKS/Gravin/AKAP12 inhibits cancer cell invasiveness and chemotaxis by suppressing a protein kinase C- Raf/MEK/ERK pathway.41
152585662004AKAP12/Gravin is inactivated by epigenetic mechanism in human gastric carcinoma and shows growth suppressor activity.30
167629192006Regulation of AKAP-membrane interactions by calcium.27
181997172008Hypermethylation of the AKAP12 promoter is a biomarker of Barrett's-associated esophageal neoplastic progression.27
209790532010Down-regulation of tumor suppressor A kinase anchor protein 12 in human hepatocarcinogenesis by epigenetic mechanisms.27
174428322007AKAP12 regulates human blood-retinal barrier formation by downregulation of hypoxia-inducible factor-1alpha.21
192109882009Gravin dynamics regulates the subcellular distribution of PKA.21
264061182015A mitotic kinase scaffold depleted in testicular seminomas impacts spindle orientation in germ line stem cells.21

Citation

Irwin H Gelman

AKAP12 A kinase (PRKA) anchor protein 12

Atlas Genet Cytogenet Oncol Haematol. 2014-10-01

Online version: http://atlasgeneticsoncology.org/gene/607/case-report-explorer/

Historical Card

2006-11-01 AKAP12 A kinase (PRKA) anchor protein 12 by  Irwin H Gelman 

Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA