Entry - *115441 - CASEIN KINASE II, BETA; CSNK2B - OMIM
 
* 115441

CASEIN KINASE II, BETA; CSNK2B


Alternative titles; symbols

CASEIN KINASE II, BETA SUBUNIT; CK2B
PHOSVITIN


HGNC Approved Gene Symbol: CSNK2B

Cytogenetic location: 6p21.33     Genomic coordinates (GRCh38): 6:31,666,080-31,670,067 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
6p21.33 Poirier-Bienvenu neurodevelopmental syndrome 618732 AD 3

TEXT

Description

The CSNK2B gene encodes a regulatory subunit of casein kinase II (CK2), a highly conserved ubiquitous enzyme consisting of subunits alpha (CSNK2A1; 115440), alpha-prime (CSNK2A2; 115442), and beta. It is present in high levels in the brain and appears to be constitutively active. Animal models suggest that CK2 may play a role in dopamine signaling (summary by Poirier et al., 2017; Yang et al., 2018).


Cloning and Expression

CK2 is a ubiquitous serine/threonine kinase, localized in both the cytoplasm and the nucleus. Jakobi et al. (1989) prepared subunit beta from human placenta and determined the amino acid sequence of a protease digestion peptide. The deduced nucleotide sequence was used for the synthesis of a mixture of 20-mers as a hybridization probe to screen a lambda-gt10 HeLa cell cDNA library for clones encoding the beta subunit. The beta subunit presumably serves regulatory functions. Heller-Harrison et al. (1989) found evidence of a single gene. They described a cDNA of 2.57 kb containing 96 bp of 5-prime untranslated sequence, 645 bp of open reading frame, and 1,832 bp of 3-prime untranslated sequence.

Li et al. (2019) noted that the CSNK2B gene is highly expressed in the developing prefrontal cortex, with lesser expression in childhood and adulthood.


Gene Structure

Voss et al. (1991) analyzed the structure of the gene encoding human casein kinase II subunit beta and Boldyreff and Issinger (1995) determined the structure of the mouse counterpart. The latter is composed of 7 exons contained within 7,874 bp. The lengths of the mouse coding exons correspond exactly to the lengths of the exons in the human CK2B gene. Both genes contain a first untranslated exon. Despite common features, a striking difference concerned the human CK2A subunit binding domain at position -170 to -239 of the human gene. This domain has no counterpart in the mouse gene.


Mapping

By hybridization to spot-blotting filters of flow-sorted human chromosomes followed by in situ hybridization, Yang-Feng et al. (1990) mapped the CSNK2B gene to 6p21.1.

Albertella et al. (1996) characterized the genes in the central 1,100-kb class III region of the major histocompatibility complex. One of the genes found in this region was identified as CSNK2B. This would suggest that CSNK2B is located in the 6p21.3 region rather than the 6p21.1 region.


Gene Function

Sarno et al. (2000) reported that a C-terminally truncated form of CK2-beta lacking residues 170 to 215 could not stably associate with the catalytic CK2 subunits. This CK2-beta mutant retained its central homodimerization domain and still existed as a dimer. However, the mutant was defective in a number of other properties mediated by elements still present in its N-terminal half, notably downregulation of catalytic activity, autophosphorylation, and responsiveness to polycationic effectors. All these functions were restored by simultaneous addition of a synthetic peptide reproducing the CK2-beta deleted region, which was able to associate with the catalytic subunits and to stimulate catalytic activity. This peptide includes a segment that shares similarity with a region of cyclin A (see 604036) involved in activation of CDK2 (116953), and Sarno et al. (2000) found that a peptide reproducing this sequence (residues 181 to 203) interacted with the CK2-alpha subunit and stimulated its catalytic activity. This smaller peptide also partially restored the ability of truncated CK2-beta to autophosphorylate. Sarno et al. (2000) concluded that residues 181 to 203 are essential for the regulatory properties of CK2-beta.

Phosphorylation of the human p53 protein (191170) at ser392 is responsive to ultraviolet (UV) but not gamma irradiation. Keller et al. (2001) identified and purified a mammalian UV-activated protein kinase complex that phosphorylates ser392 in vitro. This kinase complex contains CK2 and the chromatin transcriptional elongation factor FACT, a heterodimer of SPT16 (605012) and SSRP1 (604328). In vitro studies showed that FACT alters the specificity of CK2 in the complex such that it selectively phosphorylates p53 over other substrates, including casein. In addition, phosphorylation by the kinase complex was found to enhance p53 activity. These results provided a potential mechanism for p53 activation by UV irradiation.

Doray et al. (2002) demonstrated that the Golgi-localized, gamma-ear-containing adenosine diphosphate ribosylation factor-binding proteins (GGA1, 606004 and GGA3, 606006) and the coat protein adaptor protein-1 (AP-1) complex (see AP1G2, 603534) colocalize in clathrin-coated buds of the trans-Golgi networks of mouse L cells and human HeLa cells. Binding studies revealed a direct interaction between the hinge domains of the GGAs and the gamma-ear domain of AP-1. Further, AP-1 contained bound casein kinase-2 that phosphorylated GGA1 and GGA3, thereby causing autoinhibition. Doray et al. (2002) demonstrated that this autoinhibition could induce the directed transfer of mannose 6-phosphate receptors (see 154540) from the GGAs to AP-1. Mannose 6-phosphate receptors that were defective in binding to GGAs were poorly incorporated into adaptor protein complex containing clathrin coated vesicles. Thus, Doray et al. (2002) concluded that GGAs and the AP-1 complex interact to package mannose 6-phosphate receptors into AP-1-containing coated vesicles.

Rodriguez et al. (2008) stated that, in addition to cytoplasm, nuclei, and other organelles, CK2 localizes to the external side of the cell membrane, where it acts as an ectokinase and phosphorylates extracellular proteins and external domains of proteins. By mutation analysis, they showed that an N-terminal region of Xenopus Ck2-beta containing 2 phenylalanines and an acidic cluster was necessary but not sufficient to allow Ck2-alpha to function as an ectokinase in transfected HEK293 cells.

In vitro cellular studies by Yang et al. (2018) showed that knockdown of the Csnk2b gene in mouse embryonic neural stem cells increased proliferation, impaired cell differentiation, and reduced the dendritic length and branch points compared to controls. Knockdown of Csnk2b also altered synaptic transmission compared to controls. The authors suggested that variation in the CSNK2B gene may contribute to the risk of schizophrenia (SCZD; see 181500), which is believed to be a disorder related to altered neurodevelopment.

Excessive erythrocytosis is a major hallmark of chronic mountain sickness (CMS; see 616182), or Monge disease, a clinical syndrome caused by years of exposure to high-altitude hypoxia. Using RNA-sequencing analysis, Azad et al. (2023) identified HIKER (LINC02228; 620525) as a differentially expressed long noncoding RNA in individuals with CMS compared with non-CMS controls under hypoxic conditions. HIKER was upregulated in CMS cells, but not in non-CMS cells. HIKER regulated erythropoiesis in CMS individuals under hypoxia via the downstream factor CSNK2B. Further analysis confirmed that CSNK2B was an erythropoietic regulator in CMS and non-CMS cells under hypoxia and showed that CSNK2B regulated erythropoiesis at high altitude partially through GATA1 (305371). Furthermore, csnk2b knockdown induced severe hemoglobinization defects in zebrafish embryos, confirming an evolutionarily conserved role of CSNK2B in erythropoiesis.


Molecular Genetics

In 2 unrelated patients with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Poirier et al. (2017) identified de novo heterozygous splice site mutations in the CSNK2B gene (115441.0001 and 115441.0002). The mutations, which were found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, were not found in the 1000 Genomes Project and ExAC databases. Analysis of patient cells showed decreased mRNA levels compared to controls, and RT-PCR showed that the mutations resulted in exon skipping and premature termination, consistent with haploinsufficiency and a loss of function. However, the authors noted that the mutations may induce the production of an aberrant truncated protein. Poirier et al. (2017) postulated that the mutations may cause abnormal dopamine signaling.

In a 21-month-old Japanese boy with POBINDS, Sakaguchi et al. (2017) identified a de novo heterozygous frameshift mutation in the CSNK2B gene (115441.0003). The mutation was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing. Functional studies of the variant and studies of patient cells were not performed, but the variant was predicted to result in haploinsufficiency.

In a 15-year-old Malaysian girl (patient 3) with POBINDS, Nakashima et al. (2019) identified a de novo heterozygous frameshift mutation in the CSNK2B gene (115441.0004). The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was classified as pathogenic according to ACMG guidelines. Transfection of the mutation into HEK293 cells showed that the mutant protein was expressed, but was unable to bind with CSNK2A1 (115440), which may induce instability of the CK2 holoenzyme.

In 9 unrelated patients with POBINDS, Li et al. (2019) identified de novo heterozygous mutations in the CSNK2B gene (see, e.g., 115441.0005-115441.0007). The mutations, which were found by trio-based whole-exome sequencing of a cohort of 816 probands with epilepsy, were confirmed by Sanger sequencing. The mutations occurred throughout the gene and comprised 4 missense variants, 3 frameshifts, and 1 splice site mutation. None of the variants were found in the 1000 Genomes Project or gnomAD databases; all were predicted to be pathogenic (8) or likely pathogenic (1) by ACMG criteria, but only 4 had predictive evidence of 'very strong' pathogenicity. Functional studies of the variants and studies of patient cells were not performed. Five variants occurred in the zinc-binding domain, suggesting a hotspot; all of these patients responded to antiepileptic treatment. However, patients with missense mutations could have a severe phenotype and those with frameshift mutations could have a mild phenotype, precluding establishment of a definitive genotype/phenotype correlation.


Animal Model

Blond et al. (2005) found that complete knockout of the Csnk2b gene in mice was embryonic lethal. Heterozygous knockout mice did not exhibit any abnormalities, although the number of heterozygous offspring was lower than expected, suggesting that some heterozygous mice do not survive.

Huillard et al. (2010) found that mice with Ck2b deletion in neural stem/progenitor cells (NSCs) of developing brain were born at the expected mendelian ratio, but they did not feed and died shortly after birth. Loss of Ck2b in embryonic NSCs compromised forebrain NSC proliferation and impaired NSC differentiation to develop oligodendrocyte precursor cells (OPCs), resulting in defects in telencephalon development in brain. In vitro analyses identified Olig2 (606386), a critical modulator of OPC development, as a Ck2b-dependent substrate. Ck2b interacted directly with the bHLH domain of Olig2, and Ck2 phosphorylated Olig2 on its serine/threonine-rich (STR) domain. The phosphorylated STR domain was involved in the oligodendroglial function of Olig2.


ALLELIC VARIANTS ( 7 Selected Examples):

.0001 POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, IVS5DS, T-C, +2
  
RCV001003349

In a 10-year-old boy (patient 1) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Poirier et al. (2017) identified a de novo heterozygous T-to-C transition (c.367+2T-C, NM_001320.5) affecting a splice site in intron 5 of the CSNK2B gene. The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was not found in the 1000 Genomes Project or ExAC database. Analysis of patient cells showed decreased mRNA levels compared to controls, and RT-PCR showed that the mutation resulted in the skipping of exon 5 and premature termination (Leu98AlafsTer11), consistent with haploinsufficiency and a loss of function. However, the authors noted that the mutation may induce the production of an aberrant truncated protein.

Li et al. (2019) noted that this mutation affects the zinc-binding domain and that the patient reported by Poirier et al. (2017) did not have seizures.


.0002 POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, IVS3DS, T-G, +2
  
RCV000993565

In a 19-year-old man (patient 2) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Poirier et al. (2017) identified a de novo heterozygous T-to-G transversion (c.175+2T-G, NM_001320.5) affecting a splice site in intron 3 of the CSNK2B gene. The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was not found in the 1000 Genomes Project or ExAC databases. Analysis of patient cells showed decreased mRNA levels compared to controls, and RT-PCR showed that the mutation resulted in the skipping of exon 3 and caused premature termination (Val25MetfsTer13), consistent with haploinsufficiency and a loss of function. However, the authors noted that the mutation may induce the production of an aberrant truncated protein.


.0003 POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, 1-BP DUP, NT108
  
RCV000495848...

In a 21-month-old Japanese boy with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Sakaguchi et al. (2017) identified a de novo heterozygous 1-bp duplication (c.108dup, NM_001320.6) in exon 3 of the CSNK2B gene, predicted to result in a frameshift and premature termination (Thr37Tyrfs). The mutation was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing. Functional studies of the variant and studies of patient cells were not performed.


.0004 POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, 2-BP INS, 533GT
  
RCV000993567

In a 15-year-old Malaysian girl (patient 3) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Nakashima et al. (2019) identified a de novo heterozygous 2-bp insertion (c.533_534insGT, NM_001320.5) in the CSNK2B gene, resulting in a frameshift and premature termination (Pro179TyrfsTer49). The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was classified as pathogenic according to ACMG guidelines. Transfection of the mutation into HEK293 cells showed that the mutant protein was expressed, but was unable to bind with CSNK2A1 (115440), which may induce instability of the CK2 holoenzyme.


.0005 POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, 1-BP INS, 620C
  
RCV000993568

In a 3-year-old Chinese girl (P2) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Li et al. (2019) identified a de novo heterozygous 1-bp insertion (c.620_621insC, NM_001320) in exon 7 of the CSNK2B gene, predicted to result in a frameshift and premature termination (Phe207PhefsTer39). The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was not found in several public databases, including gnomAD. Functional studies of the variant and studies of patient cells were not performed, but the variant was predicted to result in haploinsufficiency and was classified as strongly pathogenic according to ACMG guidelines.


.0006 RECLASSIFIED - VARIANT OF UNKNOWN SIGNIFICANCE

CSNK2B, 1-BP DEL, 264C
  
RCV000993569

This variant, formerly designated POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME, has been reclassified because the numbering of the variant in the article by Li et al. (2019) does not correspond with the cited reference sequence.

In a 2-year-old Chinese boy (P5) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Li et al. (2019) identified a de novo heterozygous 1-bp deletion (c.264delC, NM_001320) in exon 4 of the CSNK2B gene, predicted to result in a frameshift and premature termination (Ile88IlefsTer46). The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was not found in several public databases, including gnomAD. Functional studies of the variant and studies of patient cells were not performed, but the variant was predicted to result in haploinsufficiency and was classified as strongly pathogenic according to ACMG guidelines.


.0007 POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, IVS5AS, A-G, -2
  
RCV000993570

In a 6-month-old Chinese girl (P9) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Li et al. (2019) identified a de novo heterozygous A-to-G transition in intron 5 of the CSNK2B gene (c.368-2A-G), predicted to result in a splice site alteration. The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was not found in several public databases, including gnomAD. Functional studies of the variant and studies of patient cells were not performed, but it was predicted to result in haploinsufficiency and was classified as strongly pathogenic according to ACMG guidelines. The mutation affected the zinc-binding domain, and the patient had controlled seizures and mildly impaired development.


REFERENCES

  1. Albertella, M. R., Jones, H., Thomson, W., Olavesen, M. G., Campbell, R. D. Localization of eight additional genes in the human major histocompatibility complex, including the gene encoding the casein kinase II beta subunit (CSNK2B). Genomics 36: 240-251, 1996. [PubMed: 8812450, related citations] [Full Text]

  2. Azad, P., Zhou, D., Tu, H. C., Villafuerte, F. C., Traver, D., Rana, T. M., Haddad, G. G. Long noncoding RNA HIKER regulates erythropoiesis in Monge's disease via CSNK2B. J. Clin. Invest. 133: e165831, 2023. [PubMed: 37022795, images, related citations] [Full Text]

  3. Blond, O., Jensen, H. H., Buchou, T., Cochet, C., Issinger, O.-G., Boldyreff, B. Knocking out the regulatory beta subunit of protein kinase CK2 in mice: gene dosage effects in ES cells and embryos. Molec. Cell Biochem. 274: 31-37, 2005. [PubMed: 16335526, related citations] [Full Text]

  4. Boldyreff, B., Issinger, O.-G. Structure of the gene encoding the murine protein kinase CK2-beta subunit. Genomics 29: 253-256, 1995. [PubMed: 8530080, related citations] [Full Text]

  5. Doray, B., Ghosh, P., Griffith, J., Geuze, H. J., Kornfeld, S. Cooperation of GGAs and AP-1 in packaging MPRs at the trans-Golgi network. Science 297: 1700-1703, 2002. [PubMed: 12215646, related citations] [Full Text]

  6. Heller-Harrison, R. A., Meisner, H., Czech, M. P. Cloning and characterization of a cDNA encoding the beta subunit of human casein kinase II. Biochemistry 28: 9053-9058, 1989. [PubMed: 2513884, related citations] [Full Text]

  7. Huillard, E., Ziercher, L., Blond, O., Wong, M., Deloulme, J.-C., Souchelnytsky, S., Baudier, J., Cochet, C., Buchou, T. Disruption of CK2-beta in embryonic neural stem cells compromises proliferation and oligodendrogenesis in the mouse telencephalon. Molec. Cell Biol. 30: 2737-2749, 2010. [PubMed: 20368359, images, related citations] [Full Text]

  8. Jakobi, R., Voss, H., Pyerin, W. Human phosvitin/casein kinase type II: molecular cloning and sequencing of full-length cDNA encoding subunit beta. Europ. J. Biochem. 183: 227-233, 1989. [PubMed: 2666134, related citations] [Full Text]

  9. Keller, D. M., Zeng, X., Wang, Y., Zhang, Q. H., Kapoor, M., Shu, H., Goodman, R., Lozano, G., Zhao, Y., Lu, H. A DNA damage-induced p53 serine 392 kinase complex contains CK2, hSpt16, and SSRP1. Molec. Cell 7: 283-292, 2001. [PubMed: 11239457, related citations] [Full Text]

  10. Li, J., Gao, K., Cai, S., Liu, Y., Wang, Y., Huang, S., Zha, J., Hu, W., Yu, S., Yang, Z., Xie, H., Yan, H., Wang, J., Wu, Y., Jiang, Y. Germline de novo variants in CSNK2B in Chinese patients with epilepsy. Sci. Rep. 9: 17909, 2019. Note: Electronic Article. [PubMed: 31784560, images, related citations] [Full Text]

  11. Nakashima, M., Tohyama, J., Nakagawa, E., Watanabe, Y., Siew, C. G., Kwong, C. S., Yamoto, K., Hiraide, T., Fukuda, T., Kaname, T., Nakabayashi, K., Hata, K., Ogata, T., Saitsu, H., Matsumoto, N. Identification of de novo CSNK2A1 and CSNK2B variants in cases of global developmental delay with seizures. J. Hum. Genet. 64: 313-322, 2019. [PubMed: 30655572, related citations] [Full Text]

  12. Poirier, K., Hubert, L., Viot, G., Rio, M., Billuart, P., Besmond, C., Bienvenu, T. CSNK2B splice site mutations in patients cause intellectual disability with or without myoclonic epilepsy. Hum. Mutat. 38: 932-941, 2017. [PubMed: 28585349, related citations] [Full Text]

  13. Rodriguez, F. A., Contreras, C., Bolanos-Garcia, V., Allende, J. E. Protein kinase CK2 as an ectokinase: the role of the regulatory CK2-beta subunit. Proc. Nat. Acad. Sci. 105: 5693-5698, 2008. [PubMed: 18391191, images, related citations] [Full Text]

  14. Sakaguchi, Y., Uehara, T., Suzuki, H., Kosaki, K., Takenouchi, T. Truncating mutation in CSNK2B and myoclonic epilepsy. (Letter) Hum. Mutat. 38: 1611-1612, 2017. [PubMed: 28762608, related citations] [Full Text]

  15. Sarno, S., Marin, O., Boschetti, M., Pagano, M. A., Meggio, F., Pinna, L. A. Cooperative modulation of protein kinase CK2 by separate domains of its regulatory beta-subunit. Biochemistry 39: 12324-12329, 2000. [PubMed: 11015211, related citations] [Full Text]

  16. Voss, H., Wirkner, U., Jacoki, R., Hewitt, N. A., Schwager, C., Zimmermann, J., Ansorge, W., Pyerin, W. Structure of the gene encoding human casein kinase II subunit beta. J. Biol. Chem. 266: 13706-13711, 1991. [PubMed: 1856204, related citations]

  17. Yang, C.-P., Li, X., Wu, Y., Shen, Q., Zeng, Y., Xiong, Q., Wei, M., Chen, C., Liu, J., Huo, Y., Li, K., Xue, G., Yao, Y.-G., Zhang, C., Li, M., Chen, Y., Luo, X.-J. Comprehensive integrative analyses identify GLT8D1 and CSNK2B as schizophrenia risk genes. Nature Commun. 9: 838, 2018. Note: Electronic Article. Erratum: Nature Commun. 9: 4905 only, 2018. [PubMed: 29483533, images, related citations] [Full Text]

  18. Yang-Feng, T. L., Teitz, T., Cheung, M. C., Kan, Y. W., Canaani, D. Assignment of the human casein kinase II beta-subunit gene to 6p12-p21. Genomics 8: 741-742, 1990. [PubMed: 2276748, related citations] [Full Text]


Bao Lige - updated : 09/26/2023
Bao Lige - updated : 03/06/2020
Cassandra L. Kniffin - updated : 01/13/2020
Matthew B. Gross - updated : 06/11/2008
Patricia A. Hartz - updated : 6/6/2008
Ada Hamosh - updated : 10/23/2002
Stylianos E. Antonarakis - updated : 3/12/2001
Creation Date:
Victor A. McKusick : 11/22/1989
alopez : 02/14/2024
mgross : 09/26/2023
carol : 01/04/2022
carol : 03/03/2021
carol : 03/18/2020
mgross : 03/06/2020
mgross : 03/06/2020
carol : 01/15/2020
ckniffin : 01/13/2020
mgross : 06/11/2008
terry : 6/6/2008
mgross : 9/18/2003
alopez : 10/23/2002
alopez : 10/23/2002
mgross : 3/12/2001
mgross : 3/12/2001
psherman : 10/22/1999
mark : 1/19/1998
mark : 10/9/1996
terry : 10/9/1996
terry : 10/30/1995
mark : 10/2/1995
supermim : 3/16/1992
carol : 1/2/1991
carol : 12/14/1990
carol : 10/26/1990

* 115441

CASEIN KINASE II, BETA; CSNK2B


Alternative titles; symbols

CASEIN KINASE II, BETA SUBUNIT; CK2B
PHOSVITIN


HGNC Approved Gene Symbol: CSNK2B

Cytogenetic location: 6p21.33     Genomic coordinates (GRCh38): 6:31,666,080-31,670,067 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
6p21.33 Poirier-Bienvenu neurodevelopmental syndrome 618732 Autosomal dominant 3

TEXT

Description

The CSNK2B gene encodes a regulatory subunit of casein kinase II (CK2), a highly conserved ubiquitous enzyme consisting of subunits alpha (CSNK2A1; 115440), alpha-prime (CSNK2A2; 115442), and beta. It is present in high levels in the brain and appears to be constitutively active. Animal models suggest that CK2 may play a role in dopamine signaling (summary by Poirier et al., 2017; Yang et al., 2018).


Cloning and Expression

CK2 is a ubiquitous serine/threonine kinase, localized in both the cytoplasm and the nucleus. Jakobi et al. (1989) prepared subunit beta from human placenta and determined the amino acid sequence of a protease digestion peptide. The deduced nucleotide sequence was used for the synthesis of a mixture of 20-mers as a hybridization probe to screen a lambda-gt10 HeLa cell cDNA library for clones encoding the beta subunit. The beta subunit presumably serves regulatory functions. Heller-Harrison et al. (1989) found evidence of a single gene. They described a cDNA of 2.57 kb containing 96 bp of 5-prime untranslated sequence, 645 bp of open reading frame, and 1,832 bp of 3-prime untranslated sequence.

Li et al. (2019) noted that the CSNK2B gene is highly expressed in the developing prefrontal cortex, with lesser expression in childhood and adulthood.


Gene Structure

Voss et al. (1991) analyzed the structure of the gene encoding human casein kinase II subunit beta and Boldyreff and Issinger (1995) determined the structure of the mouse counterpart. The latter is composed of 7 exons contained within 7,874 bp. The lengths of the mouse coding exons correspond exactly to the lengths of the exons in the human CK2B gene. Both genes contain a first untranslated exon. Despite common features, a striking difference concerned the human CK2A subunit binding domain at position -170 to -239 of the human gene. This domain has no counterpart in the mouse gene.


Mapping

By hybridization to spot-blotting filters of flow-sorted human chromosomes followed by in situ hybridization, Yang-Feng et al. (1990) mapped the CSNK2B gene to 6p21.1.

Albertella et al. (1996) characterized the genes in the central 1,100-kb class III region of the major histocompatibility complex. One of the genes found in this region was identified as CSNK2B. This would suggest that CSNK2B is located in the 6p21.3 region rather than the 6p21.1 region.


Gene Function

Sarno et al. (2000) reported that a C-terminally truncated form of CK2-beta lacking residues 170 to 215 could not stably associate with the catalytic CK2 subunits. This CK2-beta mutant retained its central homodimerization domain and still existed as a dimer. However, the mutant was defective in a number of other properties mediated by elements still present in its N-terminal half, notably downregulation of catalytic activity, autophosphorylation, and responsiveness to polycationic effectors. All these functions were restored by simultaneous addition of a synthetic peptide reproducing the CK2-beta deleted region, which was able to associate with the catalytic subunits and to stimulate catalytic activity. This peptide includes a segment that shares similarity with a region of cyclin A (see 604036) involved in activation of CDK2 (116953), and Sarno et al. (2000) found that a peptide reproducing this sequence (residues 181 to 203) interacted with the CK2-alpha subunit and stimulated its catalytic activity. This smaller peptide also partially restored the ability of truncated CK2-beta to autophosphorylate. Sarno et al. (2000) concluded that residues 181 to 203 are essential for the regulatory properties of CK2-beta.

Phosphorylation of the human p53 protein (191170) at ser392 is responsive to ultraviolet (UV) but not gamma irradiation. Keller et al. (2001) identified and purified a mammalian UV-activated protein kinase complex that phosphorylates ser392 in vitro. This kinase complex contains CK2 and the chromatin transcriptional elongation factor FACT, a heterodimer of SPT16 (605012) and SSRP1 (604328). In vitro studies showed that FACT alters the specificity of CK2 in the complex such that it selectively phosphorylates p53 over other substrates, including casein. In addition, phosphorylation by the kinase complex was found to enhance p53 activity. These results provided a potential mechanism for p53 activation by UV irradiation.

Doray et al. (2002) demonstrated that the Golgi-localized, gamma-ear-containing adenosine diphosphate ribosylation factor-binding proteins (GGA1, 606004 and GGA3, 606006) and the coat protein adaptor protein-1 (AP-1) complex (see AP1G2, 603534) colocalize in clathrin-coated buds of the trans-Golgi networks of mouse L cells and human HeLa cells. Binding studies revealed a direct interaction between the hinge domains of the GGAs and the gamma-ear domain of AP-1. Further, AP-1 contained bound casein kinase-2 that phosphorylated GGA1 and GGA3, thereby causing autoinhibition. Doray et al. (2002) demonstrated that this autoinhibition could induce the directed transfer of mannose 6-phosphate receptors (see 154540) from the GGAs to AP-1. Mannose 6-phosphate receptors that were defective in binding to GGAs were poorly incorporated into adaptor protein complex containing clathrin coated vesicles. Thus, Doray et al. (2002) concluded that GGAs and the AP-1 complex interact to package mannose 6-phosphate receptors into AP-1-containing coated vesicles.

Rodriguez et al. (2008) stated that, in addition to cytoplasm, nuclei, and other organelles, CK2 localizes to the external side of the cell membrane, where it acts as an ectokinase and phosphorylates extracellular proteins and external domains of proteins. By mutation analysis, they showed that an N-terminal region of Xenopus Ck2-beta containing 2 phenylalanines and an acidic cluster was necessary but not sufficient to allow Ck2-alpha to function as an ectokinase in transfected HEK293 cells.

In vitro cellular studies by Yang et al. (2018) showed that knockdown of the Csnk2b gene in mouse embryonic neural stem cells increased proliferation, impaired cell differentiation, and reduced the dendritic length and branch points compared to controls. Knockdown of Csnk2b also altered synaptic transmission compared to controls. The authors suggested that variation in the CSNK2B gene may contribute to the risk of schizophrenia (SCZD; see 181500), which is believed to be a disorder related to altered neurodevelopment.

Excessive erythrocytosis is a major hallmark of chronic mountain sickness (CMS; see 616182), or Monge disease, a clinical syndrome caused by years of exposure to high-altitude hypoxia. Using RNA-sequencing analysis, Azad et al. (2023) identified HIKER (LINC02228; 620525) as a differentially expressed long noncoding RNA in individuals with CMS compared with non-CMS controls under hypoxic conditions. HIKER was upregulated in CMS cells, but not in non-CMS cells. HIKER regulated erythropoiesis in CMS individuals under hypoxia via the downstream factor CSNK2B. Further analysis confirmed that CSNK2B was an erythropoietic regulator in CMS and non-CMS cells under hypoxia and showed that CSNK2B regulated erythropoiesis at high altitude partially through GATA1 (305371). Furthermore, csnk2b knockdown induced severe hemoglobinization defects in zebrafish embryos, confirming an evolutionarily conserved role of CSNK2B in erythropoiesis.


Molecular Genetics

In 2 unrelated patients with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Poirier et al. (2017) identified de novo heterozygous splice site mutations in the CSNK2B gene (115441.0001 and 115441.0002). The mutations, which were found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, were not found in the 1000 Genomes Project and ExAC databases. Analysis of patient cells showed decreased mRNA levels compared to controls, and RT-PCR showed that the mutations resulted in exon skipping and premature termination, consistent with haploinsufficiency and a loss of function. However, the authors noted that the mutations may induce the production of an aberrant truncated protein. Poirier et al. (2017) postulated that the mutations may cause abnormal dopamine signaling.

In a 21-month-old Japanese boy with POBINDS, Sakaguchi et al. (2017) identified a de novo heterozygous frameshift mutation in the CSNK2B gene (115441.0003). The mutation was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing. Functional studies of the variant and studies of patient cells were not performed, but the variant was predicted to result in haploinsufficiency.

In a 15-year-old Malaysian girl (patient 3) with POBINDS, Nakashima et al. (2019) identified a de novo heterozygous frameshift mutation in the CSNK2B gene (115441.0004). The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was classified as pathogenic according to ACMG guidelines. Transfection of the mutation into HEK293 cells showed that the mutant protein was expressed, but was unable to bind with CSNK2A1 (115440), which may induce instability of the CK2 holoenzyme.

In 9 unrelated patients with POBINDS, Li et al. (2019) identified de novo heterozygous mutations in the CSNK2B gene (see, e.g., 115441.0005-115441.0007). The mutations, which were found by trio-based whole-exome sequencing of a cohort of 816 probands with epilepsy, were confirmed by Sanger sequencing. The mutations occurred throughout the gene and comprised 4 missense variants, 3 frameshifts, and 1 splice site mutation. None of the variants were found in the 1000 Genomes Project or gnomAD databases; all were predicted to be pathogenic (8) or likely pathogenic (1) by ACMG criteria, but only 4 had predictive evidence of 'very strong' pathogenicity. Functional studies of the variants and studies of patient cells were not performed. Five variants occurred in the zinc-binding domain, suggesting a hotspot; all of these patients responded to antiepileptic treatment. However, patients with missense mutations could have a severe phenotype and those with frameshift mutations could have a mild phenotype, precluding establishment of a definitive genotype/phenotype correlation.


Animal Model

Blond et al. (2005) found that complete knockout of the Csnk2b gene in mice was embryonic lethal. Heterozygous knockout mice did not exhibit any abnormalities, although the number of heterozygous offspring was lower than expected, suggesting that some heterozygous mice do not survive.

Huillard et al. (2010) found that mice with Ck2b deletion in neural stem/progenitor cells (NSCs) of developing brain were born at the expected mendelian ratio, but they did not feed and died shortly after birth. Loss of Ck2b in embryonic NSCs compromised forebrain NSC proliferation and impaired NSC differentiation to develop oligodendrocyte precursor cells (OPCs), resulting in defects in telencephalon development in brain. In vitro analyses identified Olig2 (606386), a critical modulator of OPC development, as a Ck2b-dependent substrate. Ck2b interacted directly with the bHLH domain of Olig2, and Ck2 phosphorylated Olig2 on its serine/threonine-rich (STR) domain. The phosphorylated STR domain was involved in the oligodendroglial function of Olig2.


ALLELIC VARIANTS 7 Selected Examples):

.0001   POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, IVS5DS, T-C, +2
SNP: rs1583610610, ClinVar: RCV001003349

In a 10-year-old boy (patient 1) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Poirier et al. (2017) identified a de novo heterozygous T-to-C transition (c.367+2T-C, NM_001320.5) affecting a splice site in intron 5 of the CSNK2B gene. The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was not found in the 1000 Genomes Project or ExAC database. Analysis of patient cells showed decreased mRNA levels compared to controls, and RT-PCR showed that the mutation resulted in the skipping of exon 5 and premature termination (Leu98AlafsTer11), consistent with haploinsufficiency and a loss of function. However, the authors noted that the mutation may induce the production of an aberrant truncated protein.

Li et al. (2019) noted that this mutation affects the zinc-binding domain and that the patient reported by Poirier et al. (2017) did not have seizures.


.0002   POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, IVS3DS, T-G, +2
SNP: rs1583605716, ClinVar: RCV000993565

In a 19-year-old man (patient 2) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Poirier et al. (2017) identified a de novo heterozygous T-to-G transversion (c.175+2T-G, NM_001320.5) affecting a splice site in intron 3 of the CSNK2B gene. The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was not found in the 1000 Genomes Project or ExAC databases. Analysis of patient cells showed decreased mRNA levels compared to controls, and RT-PCR showed that the mutation resulted in the skipping of exon 3 and caused premature termination (Val25MetfsTer13), consistent with haploinsufficiency and a loss of function. However, the authors noted that the mutation may induce the production of an aberrant truncated protein.


.0003   POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, 1-BP DUP, NT108
SNP: rs1131692161, ClinVar: RCV000495848, RCV000993566

In a 21-month-old Japanese boy with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Sakaguchi et al. (2017) identified a de novo heterozygous 1-bp duplication (c.108dup, NM_001320.6) in exon 3 of the CSNK2B gene, predicted to result in a frameshift and premature termination (Thr37Tyrfs). The mutation was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing. Functional studies of the variant and studies of patient cells were not performed.


.0004   POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, 2-BP INS, 533GT
SNP: rs1583611843, ClinVar: RCV000993567

In a 15-year-old Malaysian girl (patient 3) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Nakashima et al. (2019) identified a de novo heterozygous 2-bp insertion (c.533_534insGT, NM_001320.5) in the CSNK2B gene, resulting in a frameshift and premature termination (Pro179TyrfsTer49). The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was classified as pathogenic according to ACMG guidelines. Transfection of the mutation into HEK293 cells showed that the mutant protein was expressed, but was unable to bind with CSNK2A1 (115440), which may induce instability of the CK2 holoenzyme.


.0005   POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, 1-BP INS, 620C
SNP: rs1583613268, ClinVar: RCV000993568

In a 3-year-old Chinese girl (P2) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Li et al. (2019) identified a de novo heterozygous 1-bp insertion (c.620_621insC, NM_001320) in exon 7 of the CSNK2B gene, predicted to result in a frameshift and premature termination (Phe207PhefsTer39). The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was not found in several public databases, including gnomAD. Functional studies of the variant and studies of patient cells were not performed, but the variant was predicted to result in haploinsufficiency and was classified as strongly pathogenic according to ACMG guidelines.


.0006   RECLASSIFIED - VARIANT OF UNKNOWN SIGNIFICANCE

CSNK2B, 1-BP DEL, 264C
SNP: rs1583608557, ClinVar: RCV000993569

This variant, formerly designated POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME, has been reclassified because the numbering of the variant in the article by Li et al. (2019) does not correspond with the cited reference sequence.

In a 2-year-old Chinese boy (P5) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Li et al. (2019) identified a de novo heterozygous 1-bp deletion (c.264delC, NM_001320) in exon 4 of the CSNK2B gene, predicted to result in a frameshift and premature termination (Ile88IlefsTer46). The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was not found in several public databases, including gnomAD. Functional studies of the variant and studies of patient cells were not performed, but the variant was predicted to result in haploinsufficiency and was classified as strongly pathogenic according to ACMG guidelines.


.0007   POIRIER-BIENVENU NEURODEVELOPMENTAL SYNDROME

CSNK2B, IVS5AS, A-G, -2
SNP: rs1583611290, ClinVar: RCV000993570

In a 6-month-old Chinese girl (P9) with Poirier-Bienvenu neurodevelopmental syndrome (POBINDS; 618732), Li et al. (2019) identified a de novo heterozygous A-to-G transition in intron 5 of the CSNK2B gene (c.368-2A-G), predicted to result in a splice site alteration. The mutation, which was found by trio-based whole-exome sequencing and confirmed by Sanger sequencing, was not found in several public databases, including gnomAD. Functional studies of the variant and studies of patient cells were not performed, but it was predicted to result in haploinsufficiency and was classified as strongly pathogenic according to ACMG guidelines. The mutation affected the zinc-binding domain, and the patient had controlled seizures and mildly impaired development.


REFERENCES

  1. Albertella, M. R., Jones, H., Thomson, W., Olavesen, M. G., Campbell, R. D. Localization of eight additional genes in the human major histocompatibility complex, including the gene encoding the casein kinase II beta subunit (CSNK2B). Genomics 36: 240-251, 1996. [PubMed: 8812450] [Full Text: https://doi.org/10.1006/geno.1996.0459]

  2. Azad, P., Zhou, D., Tu, H. C., Villafuerte, F. C., Traver, D., Rana, T. M., Haddad, G. G. Long noncoding RNA HIKER regulates erythropoiesis in Monge's disease via CSNK2B. J. Clin. Invest. 133: e165831, 2023. [PubMed: 37022795] [Full Text: https://doi.org/10.1172/JCI165831]

  3. Blond, O., Jensen, H. H., Buchou, T., Cochet, C., Issinger, O.-G., Boldyreff, B. Knocking out the regulatory beta subunit of protein kinase CK2 in mice: gene dosage effects in ES cells and embryos. Molec. Cell Biochem. 274: 31-37, 2005. [PubMed: 16335526] [Full Text: https://doi.org/10.1007/s11010-005-3117-x]

  4. Boldyreff, B., Issinger, O.-G. Structure of the gene encoding the murine protein kinase CK2-beta subunit. Genomics 29: 253-256, 1995. [PubMed: 8530080] [Full Text: https://doi.org/10.1006/geno.1995.1239]

  5. Doray, B., Ghosh, P., Griffith, J., Geuze, H. J., Kornfeld, S. Cooperation of GGAs and AP-1 in packaging MPRs at the trans-Golgi network. Science 297: 1700-1703, 2002. [PubMed: 12215646] [Full Text: https://doi.org/10.1126/science.1075327]

  6. Heller-Harrison, R. A., Meisner, H., Czech, M. P. Cloning and characterization of a cDNA encoding the beta subunit of human casein kinase II. Biochemistry 28: 9053-9058, 1989. [PubMed: 2513884] [Full Text: https://doi.org/10.1021/bi00449a014]

  7. Huillard, E., Ziercher, L., Blond, O., Wong, M., Deloulme, J.-C., Souchelnytsky, S., Baudier, J., Cochet, C., Buchou, T. Disruption of CK2-beta in embryonic neural stem cells compromises proliferation and oligodendrogenesis in the mouse telencephalon. Molec. Cell Biol. 30: 2737-2749, 2010. [PubMed: 20368359] [Full Text: https://doi.org/10.1128/MCB.01566-09]

  8. Jakobi, R., Voss, H., Pyerin, W. Human phosvitin/casein kinase type II: molecular cloning and sequencing of full-length cDNA encoding subunit beta. Europ. J. Biochem. 183: 227-233, 1989. [PubMed: 2666134] [Full Text: https://doi.org/10.1111/j.1432-1033.1989.tb14917.x]

  9. Keller, D. M., Zeng, X., Wang, Y., Zhang, Q. H., Kapoor, M., Shu, H., Goodman, R., Lozano, G., Zhao, Y., Lu, H. A DNA damage-induced p53 serine 392 kinase complex contains CK2, hSpt16, and SSRP1. Molec. Cell 7: 283-292, 2001. [PubMed: 11239457] [Full Text: https://doi.org/10.1016/s1097-2765(01)00176-9]

  10. Li, J., Gao, K., Cai, S., Liu, Y., Wang, Y., Huang, S., Zha, J., Hu, W., Yu, S., Yang, Z., Xie, H., Yan, H., Wang, J., Wu, Y., Jiang, Y. Germline de novo variants in CSNK2B in Chinese patients with epilepsy. Sci. Rep. 9: 17909, 2019. Note: Electronic Article. [PubMed: 31784560] [Full Text: https://doi.org/10.1038/s41598-019-53484-9]

  11. Nakashima, M., Tohyama, J., Nakagawa, E., Watanabe, Y., Siew, C. G., Kwong, C. S., Yamoto, K., Hiraide, T., Fukuda, T., Kaname, T., Nakabayashi, K., Hata, K., Ogata, T., Saitsu, H., Matsumoto, N. Identification of de novo CSNK2A1 and CSNK2B variants in cases of global developmental delay with seizures. J. Hum. Genet. 64: 313-322, 2019. [PubMed: 30655572] [Full Text: https://doi.org/10.1038/s10038-018-0559-z]

  12. Poirier, K., Hubert, L., Viot, G., Rio, M., Billuart, P., Besmond, C., Bienvenu, T. CSNK2B splice site mutations in patients cause intellectual disability with or without myoclonic epilepsy. Hum. Mutat. 38: 932-941, 2017. [PubMed: 28585349] [Full Text: https://doi.org/10.1002/humu.23270]

  13. Rodriguez, F. A., Contreras, C., Bolanos-Garcia, V., Allende, J. E. Protein kinase CK2 as an ectokinase: the role of the regulatory CK2-beta subunit. Proc. Nat. Acad. Sci. 105: 5693-5698, 2008. [PubMed: 18391191] [Full Text: https://doi.org/10.1073/pnas.0802065105]

  14. Sakaguchi, Y., Uehara, T., Suzuki, H., Kosaki, K., Takenouchi, T. Truncating mutation in CSNK2B and myoclonic epilepsy. (Letter) Hum. Mutat. 38: 1611-1612, 2017. [PubMed: 28762608] [Full Text: https://doi.org/10.1002/humu.23307]

  15. Sarno, S., Marin, O., Boschetti, M., Pagano, M. A., Meggio, F., Pinna, L. A. Cooperative modulation of protein kinase CK2 by separate domains of its regulatory beta-subunit. Biochemistry 39: 12324-12329, 2000. [PubMed: 11015211] [Full Text: https://doi.org/10.1021/bi0011431]

  16. Voss, H., Wirkner, U., Jacoki, R., Hewitt, N. A., Schwager, C., Zimmermann, J., Ansorge, W., Pyerin, W. Structure of the gene encoding human casein kinase II subunit beta. J. Biol. Chem. 266: 13706-13711, 1991. [PubMed: 1856204]

  17. Yang, C.-P., Li, X., Wu, Y., Shen, Q., Zeng, Y., Xiong, Q., Wei, M., Chen, C., Liu, J., Huo, Y., Li, K., Xue, G., Yao, Y.-G., Zhang, C., Li, M., Chen, Y., Luo, X.-J. Comprehensive integrative analyses identify GLT8D1 and CSNK2B as schizophrenia risk genes. Nature Commun. 9: 838, 2018. Note: Electronic Article. Erratum: Nature Commun. 9: 4905 only, 2018. [PubMed: 29483533] [Full Text: https://doi.org/10.1038/s41467-018-03247-3]

  18. Yang-Feng, T. L., Teitz, T., Cheung, M. C., Kan, Y. W., Canaani, D. Assignment of the human casein kinase II beta-subunit gene to 6p12-p21. Genomics 8: 741-742, 1990. [PubMed: 2276748] [Full Text: https://doi.org/10.1016/0888-7543(90)90266-w]


Contributors:
Bao Lige - updated : 09/26/2023
Bao Lige - updated : 03/06/2020
Cassandra L. Kniffin - updated : 01/13/2020
Matthew B. Gross - updated : 06/11/2008
Patricia A. Hartz - updated : 6/6/2008
Ada Hamosh - updated : 10/23/2002
Stylianos E. Antonarakis - updated : 3/12/2001

Creation Date:
Victor A. McKusick : 11/22/1989

Edit History:
alopez : 02/14/2024
mgross : 09/26/2023
carol : 01/04/2022
carol : 03/03/2021
carol : 03/18/2020
mgross : 03/06/2020
mgross : 03/06/2020
carol : 01/15/2020
ckniffin : 01/13/2020
mgross : 06/11/2008
terry : 6/6/2008
mgross : 9/18/2003
alopez : 10/23/2002
alopez : 10/23/2002
mgross : 3/12/2001
mgross : 3/12/2001
psherman : 10/22/1999
mark : 1/19/1998
mark : 10/9/1996
terry : 10/9/1996
terry : 10/30/1995
mark : 10/2/1995
supermim : 3/16/1992
carol : 1/2/1991
carol : 12/14/1990
carol : 10/26/1990