Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
EMBO J ; 43(13): 2661-2684, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38811851

ABSTRACT

The molecular mechanisms governing the response of hematopoietic stem cells (HSCs) to stress insults remain poorly defined. Here, we investigated effects of conditional knock-out or overexpression of Hmga2 (High mobility group AT-hook 2), a transcriptional activator of stem cell genes in fetal HSCs. While Hmga2 overexpression did not affect adult hematopoiesis under homeostasis, it accelerated HSC expansion in response to injection with 5-fluorouracil (5-FU) or in vitro treatment with TNF-α. In contrast, HSC and megakaryocyte progenitor cell numbers were decreased in Hmga2 KO animals. Transcription of inflammatory genes was repressed in Hmga2-overexpressing mice injected with 5-FU, and Hmga2 bound to distinct regions and chromatin accessibility was decreased in HSCs upon stress. Mechanistically, we found that casein kinase 2 (CK2) phosphorylates the Hmga2 acidic domain, promoting its access and binding to chromatin, transcription of anti-inflammatory target genes, and the expansion of HSCs under stress conditions. Notably, the identified stress-regulated Hmga2 gene signature is activated in hematopoietic stem progenitor cells of human myelodysplastic syndrome patients. In sum, these results reveal a TNF-α/CK2/phospho-Hmga2 axis controlling adult stress hematopoiesis.


Subject(s)
Casein Kinase II , Chromatin , HMGA2 Protein , Hematopoietic Stem Cells , Mice, Knockout , HMGA2 Protein/metabolism , HMGA2 Protein/genetics , Animals , Hematopoietic Stem Cells/metabolism , Mice , Humans , Casein Kinase II/metabolism , Casein Kinase II/genetics , Chromatin/metabolism , Chromatin/genetics , Tumor Necrosis Factor-alpha/metabolism , Hematopoiesis , Stress, Physiological , Fluorouracil/pharmacology , Regeneration , Phosphorylation , Myelodysplastic Syndromes/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Mice, Inbred C57BL
2.
Proc Natl Acad Sci U S A ; 121(24): e2404383121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38843184

ABSTRACT

Transcription is extremely important for cellular processes but can be hindered by RNA polymerase II (RNAPII) pausing and stalling. Cockayne syndrome protein B (CSB) promotes the progression of paused RNAPII or initiates transcription-coupled nucleotide excision repair (TC-NER) to remove stalled RNAPII. However, the specific mechanism by which CSB initiates TC-NER upon damage remains unclear. In this study, we identified the indispensable role of the ARK2N-CK2 complex in the CSB-mediated initiation of TC-NER. The ARK2N-CK2 complex is recruited to damage sites through CSB and then phosphorylates CSB. Phosphorylation of CSB enhances its binding to stalled RNAPII, prolonging the association of CSB with chromatin and promoting CSA-mediated ubiquitination of stalled RNAPII. Consistent with this finding, Ark2n-/- mice exhibit a phenotype resembling Cockayne syndrome. These findings shed light on the pivotal role of the ARK2N-CK2 complex in governing the fate of RNAPII through CSB, bridging a critical gap necessary for initiating TC-NER.


Subject(s)
Cockayne Syndrome , DNA Helicases , DNA Repair Enzymes , DNA Repair , Poly-ADP-Ribose Binding Proteins , RNA Polymerase II , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , Humans , Animals , Mice , DNA Helicases/metabolism , DNA Helicases/genetics , Cockayne Syndrome/genetics , Cockayne Syndrome/metabolism , Transcription, Genetic , Phosphorylation , Casein Kinase II/metabolism , Casein Kinase II/genetics , Mice, Knockout , DNA Damage , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Chromatin/metabolism , Ubiquitination , Excision Repair
3.
J Neurosci ; 44(29)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38886057

ABSTRACT

Calcineurin inhibitors, such as cyclosporine and tacrolimus (FK506), are commonly used immunosuppressants for preserving transplanted organs and tissues. However, these drugs can cause severe and persistent pain. GluA2-lacking, calcium-permeable AMPA receptors (CP-AMPARs) are implicated in various neurological disorders, including neuropathic pain. It is unclear whether and how constitutive calcineurin, a Ca2+/calmodulin protein phosphatase, controls synaptic CP-AMPARs. In this study, we found that blocking CP-AMPARs with IEM-1460 markedly reduced the amplitude of AMPAR-EPSCs in excitatory neurons expressing vesicular glutamate transporter-2 (VGluT2), but not in inhibitory neurons expressing vesicular GABA transporter, in the spinal cord of FK506-treated male and female mice. FK506 treatment also caused an inward rectification in the current-voltage relationship of AMPAR-EPSCs specifically in VGluT2 neurons. Intrathecal injection of IEM-1460 rapidly alleviated pain hypersensitivity in FK506-treated mice. Furthermore, FK506 treatment substantially increased physical interaction of α2δ-1 with GluA1 and GluA2 in the spinal cord and reduced GluA1/GluA2 heteromers in endoplasmic reticulum-enriched fractions of spinal cords. Correspondingly, inhibiting α2δ-1 with pregabalin, Cacna2d1 genetic knock-out, or disrupting α2δ-1-AMPAR interactions with an α2δ-1 C terminus peptide reversed inward rectification of AMPAR-EPSCs in spinal VGluT2 neurons caused by FK506 treatment. In addition, CK2 inhibition reversed FK506 treatment-induced pain hypersensitivity, α2δ-1 interactions with GluA1 and GluA2, and inward rectification of AMPAR-EPSCs in spinal VGluT2 neurons. Thus, the increased prevalence of synaptic CP-AMPARs in spinal excitatory neurons plays a major role in calcineurin inhibitor-induced pain hypersensitivity. Calcineurin and CK2 antagonistically regulate postsynaptic CP-AMPARs through α2δ-1-mediated GluA1/GluA2 heteromeric assembly in the spinal dorsal horn.


Subject(s)
Calcineurin , Casein Kinase II , Receptors, AMPA , Spinal Cord , Tacrolimus , Animals , Receptors, AMPA/metabolism , Mice , Calcineurin/metabolism , Male , Female , Tacrolimus/pharmacology , Spinal Cord/metabolism , Spinal Cord/drug effects , Casein Kinase II/metabolism , Neurons/metabolism , Neurons/drug effects , Mice, Inbred C57BL , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Synapses/drug effects , Synapses/metabolism , Synapses/physiology , Calcineurin Inhibitors/pharmacology , Phenotype , Calcium Channels
4.
J Biol Chem ; 300(6): 107377, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38762174

ABSTRACT

Homologous recombination (HR) plays a key role in maintaining genomic stability, and the efficiency of the HR system is closely associated with tumor response to chemotherapy. Our previous work reported that CK2 kinase phosphorylates HIV Tat-specific factor 1 (HTATSF1) Ser748 to facilitate HTATSF1 interaction with TOPBP1, which in turn, promotes RAD51 recruitment and HR repair. However, the clinical implication of the CK2-HTATSF1-TOPBP1 pathway in tumorigenesis and chemotherapeutic response remains to be elucidated. Here, we report that the CK2-HTATSF1-TOPBP1 axis is generally hyperactivated in multiple malignancies and renders breast tumors less responsive to chemotherapy. In contrast, deletion mutations of each gene in this axis, which also occur in breast and lung tumor samples, predict higher HR deficiency scores, and tumor cells bearing a loss-of-function mutation of HTATSF1 are vulnerable to poly(ADP-ribose) polymerase inhibitors or platinum drugs. Taken together, our study suggests that the integrity of the CK2-HTATSF1-TOPBP1 axis is closely linked to tumorigenesis and serves as an indicator of tumor HR status and modulates chemotherapy response.


Subject(s)
Carrier Proteins , Casein Kinase II , DNA-Binding Proteins , Signal Transduction , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Signal Transduction/drug effects , Casein Kinase II/metabolism , Casein Kinase II/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Animals , Female , Mice , Cell Line, Tumor , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology
5.
Diabetologia ; 67(7): 1368-1385, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38503901

ABSTRACT

AIMS/HYPOTHESIS: Protein kinase CK2 acts as a negative regulator of insulin expression in pancreatic beta cells. This action is mainly mediated by phosphorylation of the transcription factor pancreatic and duodenal homeobox protein 1 (PDX1). In pancreatic alpha cells, PDX1 acts in a reciprocal fashion on glucagon (GCG) expression. Therefore, we hypothesised that CK2 might positively regulate GCG expression in pancreatic alpha cells. METHODS: We suppressed CK2 kinase activity in αTC1 cells by two pharmacological inhibitors and by the CRISPR/Cas9 technique. Subsequently, we analysed GCG expression and secretion by real-time quantitative RT-PCR, western blot, luciferase assay, ELISA and DNA pull-down assays. We additionally studied paracrine effects on GCG secretion in pseudoislets, isolated murine islets and human islets. In vivo, we examined the effect of CK2 inhibition on blood glucose levels by systemic and alpha cell-specific CK2 inhibition. RESULTS: We found that CK2 downregulation reduces GCG secretion in the murine alpha cell line αTC1 (e.g. from 1094±124 ng/l to 459±110 ng/l) by the use of the CK2-inhibitor SGC-CK2-1. This was due to a marked decrease in Gcg gene expression through alteration of the binding of paired box protein 6 (PAX6) and transcription factor MafB to the Gcg promoter. The analysis of the underlying mechanisms revealed that both transcription factors are displaced by PDX1. Ex vivo experiments in isolated murine islets and pseudoislets further demonstrated that CK2-mediated reduction in GCG secretion was only slightly affected by the higher insulin secretion after CK2 inhibition. The kidney capsule transplantation model showed the significance of CK2 for GCG expression and secretion in vivo. Finally, CK2 downregulation also reduced the GCG secretion in islets isolated from humans. CONCLUSIONS/INTERPRETATION: These novel findings not only indicate an important function of protein kinase CK2 for proper GCG expression but also demonstrate that CK2 may be a promising target for the development of novel glucose-lowering drugs.


Subject(s)
Casein Kinase II , Glucagon-Secreting Cells , Glucagon , Homeodomain Proteins , Casein Kinase II/metabolism , Casein Kinase II/genetics , Animals , Glucagon/metabolism , Mice , Humans , Glucagon-Secreting Cells/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Male , Cell Line , Insulin/metabolism
6.
Am J Med Genet A ; 194(9): e63642, 2024 09.
Article in English | MEDLINE | ID: mdl-38711237

ABSTRACT

The autosomal dominant Okur-Chung neurodevelopmental syndrome (OCNDS: OMIM #617062) is a rare neurodevelopmental disorder first described in 2016. Features include developmental delay (DD), intellectual disability (ID), behavioral problems, hypotonia, language deficits, congenital heart abnormalities, and non-specific dysmorphic facial features. OCNDS is caused by heterozygous pathogenic variants in CSNK2A1 (OMIM *115440; NM_177559.3). To date, 160 patients have been diagnosed worldwide. The number will likely increase due to the growing use of exome sequencing (ES) and genome sequencing (GS). Here, we describe a novel OCNDS patient carrying a CSNK2A1 variant (NM_177559.3:c.140G>A; NP_808227.1:p.Arg47Gln). Phenotypically, he presented with DD, ID, generalized hypotonia, speech delay, short stature, microcephaly, and dysmorphic features such as low-set ears, hypertelorism, thin upper lip, and a round face. The patient showed several signs not yet described that may extend the phenotypic spectrum of OCNDS. These include prenatal bilateral clubfeet, exotropia, and peg lateral incisors. However, unlike the majority of descriptions, he did not present sleep disturbance, seizures or gait difficulties. A literature review shows phenotypic heterogeneity for OCNDS, whether these patients have the same variant or not. This case report is an opportunity to refine the phenotype of this syndrome and raise the question of the genotype-phenotype correlation.


Subject(s)
Casein Kinase II , Neurodevelopmental Disorders , Child , Humans , Male , Casein Kinase II/genetics , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Exome Sequencing , Genetic Predisposition to Disease , Heterozygote , Intellectual Disability/genetics , Intellectual Disability/pathology , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Mutation , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Neurodevelopmental Disorders/diagnosis , Phenotype
7.
Neurochem Res ; 49(5): 1254-1267, 2024 May.
Article in English | MEDLINE | ID: mdl-38381246

ABSTRACT

Fibrotic scars play important roles in tissue reconstruction and functional recovery in the late stage of nervous system injury. However, the mechanisms underlying fibrotic scar formation and regulation remain unclear. Casein kinase II (CK2) is a protein kinase that regulates a variety of cellular functions through the phosphorylation of proteins, including bromodomain-containing protein 4 (BRD4). CK2 and BRD4 participate in fibrosis formation in a variety of tissues. However, whether CK2 affects fibrotic scar formation remains unclear, as do the mechanisms of signal regulation after cerebral ischemic injury. In this study, we assessed whether CK2 could modulate fibrotic scar formation after cerebral ischemic injury through BRD4. Primary meningeal fibroblasts were isolated from neonatal rats and treated with transforming growth factor-ß1 (TGF-ß1), SB431542 (a TGF-ß1 receptor kinase inhibitor) or TBB (a highly potent CK2 inhibitor). Adult SD rats were intraperitoneally injected with TBB to inhibit CK2 after MCAO/R. We found that CK2 expression was increased in vitro in the TGF-ß1-induced fibrosis model and in vivo in the MCAO/R injury model. The TGF-ß1 receptor kinase inhibitor SB431542 decreased CK2 expression in fibroblasts. The CK2 inhibitor TBB reduced the increases in proliferation, migration and activation of fibroblasts caused by TGF-ß1 in vitro, and it inhibited fibrotic scar formation, ameliorated histopathological damage, protected Nissl bodies, decreased infarct volume and alleviated neurological deficits after MCAO/R injury in vivo. Furthermore, CK2 inhibition decreased BRD4 phosphorylation both in vitro and in vivo. The findings of the present study suggested that CK2 may control BRD4 phosphorylation to regulate fibrotic scar formation, to affecting outcomes after ischemic stroke.


Subject(s)
Benzamides , Bromodomain Containing Proteins , Casein Kinase II , Cicatrix , Dioxoles , Ischemic Stroke , Animals , Rats , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Cicatrix/metabolism , Cicatrix/pathology , Fibroblasts/metabolism , Fibrosis , Ischemic Stroke/complications , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Nuclear Proteins , Phosphorylation , Rats, Sprague-Dawley , Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Bromodomain Containing Proteins/drug effects , Bromodomain Containing Proteins/metabolism
8.
J Biochem Mol Toxicol ; 38(8): e23780, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39056188

ABSTRACT

Sodium and potassium channels, especially Nav1.5 and Kir2.1, play key roles in the formation of action potentials in cardiomyocytes. These channels interact with, and are regulated by, synapse-associated protein 97 (SAP97). However, the regulatory role of SAP97 in myocyte remains incompletely understood. Here, we investigate the function of SAP97 phosphorylation in the regulation of Nav1.5 and Kir2.1 channel complexes and the upstream regulation of SAP97. We found that SAP97 is phosphorylated by casein kinase II (CK2) in vitro. In addition, transfection of casein kinase 2 interacting protein-1 (CKIP-1) into cardiomyocytes to drive CK2 from the nucleus to the cytoplasm, increased SAP97 phosphorylation and Nav1.5 and Kir2.1 current activity. These findings demonstrated that CKIP-1 modulates the subcellular translocation of CK2, which regulates Nav1.5 and Kir2.1 channel complex formation and activity in cardiomyocytes.


Subject(s)
Casein Kinase II , Myocytes, Cardiac , NAV1.5 Voltage-Gated Sodium Channel , Potassium Channels, Inwardly Rectifying , Myocytes, Cardiac/metabolism , Casein Kinase II/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Inwardly Rectifying/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , Animals , Rats , Phosphorylation , Protein Transport , Humans , Carrier Proteins/metabolism , Rats, Sprague-Dawley
9.
Parasitol Res ; 123(1): 80, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38163833

ABSTRACT

Chagas disease, endemic from Latin America, is caused by Trypanosoma cruzi and is transmitted by triatomine feces. This parasite undergoes complex morphological changes through its life cycle, promoted by significant changes in signal transduction pathways. The activity of protein kinase CK2 has been described in trypanosomatids. Using a specific peptide and radioactive ATP, we identified CK2 activity on the cellular surface and the cytoplasmic content in Trypanosoma cruzi, apart from the secreted form. Dephosphorylated casein promoted an increase of 48% in the secreted CK2 activity. Total extract of peritoneal macrophages from BALB/c and inactivated human serum promoted an increase of 67% and 36%, respectively, in this activity. The protein secreted by parasites was purified by HPLC and had shown compatibility with the catalytic subunit of mammalian CK2. Incubation of the parasites with CK2 inhibitors, added to the culture medium, prevented their growth. The opposite was observed when CK2 activators were used. Results of interaction between Trypanosoma cruzi and the gut of the vector have revealed that, in the presence of CK2 inhibitors, there is a reduction in the association rate. A similar inhibition profile was seen in the Trypanosoma cruzi-macrophages interaction, confirming the importance of this enzyme in the life cycle of this protozoan.


Subject(s)
Chagas Disease , Trypanosoma cruzi , Animals , Humans , Trypanosoma cruzi/metabolism , Casein Kinase II/metabolism , Chagas Disease/parasitology , Invertebrates , Mammals
10.
Cell Syst ; 15(6): 544-562.e8, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38861992

ABSTRACT

Most biological processes are regulated by signaling modules that bind to short linear motifs. For protein kinases, substrates may have full or only partial matches to the kinase recognition motif, a property known as "substrate quality." However, it is not clear whether differences in substrate quality represent neutral variation or if they have functional consequences. We examine this question for the kinase CK2, which has many fundamental functions. We show that optimal CK2 sites are phosphorylated at maximal stoichiometries and found in many conditions, whereas minimal substrates are more weakly phosphorylated and have regulatory functions. Optimal CK2 sites tend to be more conserved, and substrate quality is often tuned by selection. For intermediate sites, increases or decreases in substrate quality may be deleterious, as we demonstrate for a CK2 substrate at the kinetochore. The results together suggest a strong role for substrate quality in phosphosite function and evolution. A record of this paper's transparent peer review process is included in the supplemental information.


Subject(s)
Casein Kinase II , Casein Kinase II/metabolism , Phosphorylation , Humans , Substrate Specificity , Kinetochores/metabolism , Evolution, Molecular , Binding Sites
11.
Sci Adv ; 10(21): eadk8908, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781342

ABSTRACT

DNA replication is a vulnerable cellular process, and its deregulation leads to genomic instability. Here, we demonstrate that chromobox protein homolog 3 (CBX3) binds replication protein A 32-kDa subunit (RPA2) and regulates RPA2 retention at stalled replication forks. CBX3 is recruited to stalled replication forks by RPA2 and inhibits ring finger and WD repeat domain 3 (RFWD3)-facilitated replication restart. Phosphorylation of CBX3 at serine-95 by casein kinase 2 (CK2) kinase augments cadherin 1 (CDH1)-mediated CBX3 degradation and RPA2 dynamics at stalled replication forks, which permits replication fork restart. Increased expression of CBX3 due to gene amplification or CK2 inhibitor treatment sensitizes prostate cancer cells to poly(ADP-ribose) polymerase (PARP) inhibitors while inducing replication stress and DNA damage. Our work reveals CBX3 as a key regulator of RPA2 function and DNA replication, suggesting that CBX3 could serve as an indicator for targeted therapy of cancer using PARP inhibitors.


Subject(s)
Casein Kinase II , DNA Replication , Poly(ADP-ribose) Polymerase Inhibitors , Replication Protein A , Humans , Casein Kinase II/metabolism , Casein Kinase II/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Replication Protein A/metabolism , Replication Protein A/genetics , Cell Line, Tumor , Proteolysis , DNA Damage , Phosphorylation , Chromosomal Proteins, Non-Histone
12.
Eur J Med Chem ; 276: 116672, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39067440

ABSTRACT

Casein kinase-2 (CK2) are serine/threonine kinases with dual co-factor (ATP and GTP) specificity, that are involved in the regulation of a wide variety of cellular functions. Small molecules targeting CK2 have been described in the literature targeting different binding pockets of the kinase with a focus on type I inhibitors such as the recently published chemical probe SGC-CK2-1. In this study, we investigated whether known allosteric inhibitors binding to a pocket adjacent to helix αD could be combined with ATP mimetic moieties defining a novel class of ATP competitive compounds with a unique binding mode. Linking both binding sites requires a chemical linking moiety that would introduce a 90-degree angle between the ATP mimetic ring system and the αD targeting moiety, which was realized using a sulfonamide. The synthesized inhibitors were highly selective for CK2 with binding constants in the nM range and low micromolar activity. While these inhibitors need to be further improved, the present work provides a structure-based design strategy for highly selective CK2 inhibitors.


Subject(s)
Casein Kinase II , Protein Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Humans , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Structure-Activity Relationship , Molecular Structure , Dose-Response Relationship, Drug , Models, Molecular , Adenosine Triphosphate/metabolism , Binding Sites
13.
Sci Rep ; 14(1): 1463, 2024 01 17.
Article in English | MEDLINE | ID: mdl-38233478

ABSTRACT

Histidine residues contribute to numerous molecular interactions, owing to their structure with the ionizable aromatic side chain with pKa close to the physiological pH. Herein, we studied how the two histidine residues, His115 and His160 of the catalytic subunit of human protein kinase CK2, affect the binding of the halogenated heterocyclic ligands at the ATP-binding site. Thermodynamic studies on the interaction between five variants of hCK2α (WT protein and four histidine mutants) and three ionizable bromo-benzotriazoles and their conditionally non-ionizable benzimidazole counterparts were performed with nanoDSF, MST, and ITC. The results allowed us to identify the contribution of interactions involving the particular histidine residues to ligand binding. We showed that despite the well-documented hydrogen bonding/salt bridge formation dragging the anionic ligands towards Lys68, the protonated His160 also contributes to the binding of such ligands by long-range electrostatic interactions. Simultaneously, His 115 indirectly affects ligand binding, placing the hinge region in open/closed conformations.


Subject(s)
Casein Kinase II , Histidine , Humans , Histidine/metabolism , Protein Binding , Casein Kinase II/metabolism , Ligands , Binding Sites , Catalytic Domain , Adenosine Triphosphate/metabolism , Hydrogen-Ion Concentration
14.
Cell Rep ; 43(7): 114405, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38923463

ABSTRACT

The RNA cap methyltransferase CMTR1 methylates the first transcribed nucleotide of RNA polymerase II transcripts, impacting gene expression mechanisms, including during innate immune responses. Using mass spectrometry, we identify a multiply phosphorylated region of CMTR1 (phospho-patch [P-Patch]), which is a substrate for the kinase CK2 (casein kinase II). CMTR1 phosphorylation alters intramolecular interactions, increases recruitment to RNA polymerase II, and promotes RNA cap methylation. P-Patch phosphorylation occurs during the G1 phase of the cell cycle, recruiting CMTR1 to RNA polymerase II during a period of rapid transcription and RNA cap formation. CMTR1 phosphorylation is required for the expression of specific RNAs, including ribosomal protein gene transcripts, and promotes cell proliferation. CMTR1 phosphorylation is also required for interferon-stimulated gene expression. The cap-snatching virus, influenza A, utilizes host CMTR1 phosphorylation to produce the caps required for virus production and infection. We present an RNA cap methylation control mechanism whereby CK2 controls CMTR1, enhancing co-transcriptional capping.


Subject(s)
Casein Kinase II , Methyltransferases , RNA Caps , Animals , Humans , Casein Kinase II/metabolism , Casein Kinase II/genetics , HEK293 Cells , Influenza A virus , Influenza, Human/virology , Influenza, Human/metabolism , Influenza, Human/genetics , Methylation , Methyltransferases/metabolism , Phosphorylation , RNA Caps/metabolism , RNA Polymerase II/metabolism
15.
Biofactors ; 50(4): 624-633, 2024.
Article in English | MEDLINE | ID: mdl-38158592

ABSTRACT

The protein kinase casein kinase 2 (CK2) exerts its influence on the metabolism of three major cellular substances by phosphorylating essential protein molecules involved in various cellular metabolic pathways. These substances include hormones, especially insulin, rate-limiting enzymes, transcription factors of key genes, and cytokines. This regulatory role of CK2 is closely tied to important cellular processes such as cell proliferation and apoptosis. Additionally, tumor cells undergo metabolic reprogramming characterized by aerobic glycolysis, accelerated lipid ß-oxidation, and abnormally active glutamine metabolism. In this context, CK2, which is overexpressed in various tumors, also plays a pivotal role. Hence, this review aims to summarize the regulatory mechanisms of CK2 in diverse metabolic pathways and tumor development, providing novel insights for the diagnosis, treatment, and prognosis of metabolism-related diseases and cancers.


Subject(s)
Casein Kinase II , Neoplasms , Casein Kinase II/metabolism , Casein Kinase II/genetics , Humans , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Cell Proliferation , Apoptosis , Animals , Glycolysis , Phosphorylation , Metabolic Networks and Pathways/genetics , Gene Expression Regulation, Neoplastic
16.
Nat Commun ; 15(1): 2727, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38548791

ABSTRACT

Piwi-interacting RNAs (piRNAs) are genomically encoded small RNAs that engage Piwi Argonaute proteins to direct mRNA surveillance and transposon silencing. Despite advances in understanding piRNA pathways and functions, how the production of piRNA is regulated remains elusive. Here, using a genetic screen, we identify casein kinase II (CK2) as a factor required for piRNA pathway function. We show that CK2 is required for the localization of PRG-1 and for the proper localization of several factors that comprise the 'upstream sequence transcription complex' (USTC), which is required for piRNA transcription. Loss of CK2 impairs piRNA levels suggesting that CK2 promotes USTC function. We identify the USTC component twenty-one-U fouled-up 4 (TOFU-4) as a direct substrate for CK2. Our findings suggest that phosphorylation of TOFU-4 by CK2 promotes the assembly of USTC and piRNA transcription. Notably, during the aging process, CK2 activity declines, resulting in the disassembly of USTC, decreased piRNA production, and defects in piRNA-mediated gene silencing, including transposons silencing. These findings highlight the significance of posttranslational modification in regulating piRNA biogenesis and its implications for the aging process. Overall, our study provides compelling evidence for the involvement of a posttranslational modification mechanism in the regulation of piRNA biogenesis.


Subject(s)
Drosophila Proteins , Soy Foods , Animals , Piwi-Interacting RNA , RNA, Small Interfering/metabolism , Casein Kinase II/genetics , Casein Kinase II/metabolism , Phosphorylation , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics
17.
Autophagy ; 20(7): 1694-1695, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38511596

ABSTRACT

Tuning and assimilation of endoplasmic reticulum (ER) content in each cell of the human body is an essential part of organismal homeostasis and adaptation to stress. As such, the lysosomal turnover of ER (reticulophagy) needs to be regulated in a spatio-temporal as well as cell-type specific manner. We recently identified CSNK2/CK2 (casein kinase 2) as the enzyme that phosphorylates the reticulophagy receptors RETREG1/FAM134B and RETREG3/FAM134C and regulates their activity. Phosphorylation of the receptors is a prerequisite for their subsequent functional ubiquitination and the formation of high-density clusters, presumably representing active macroautophagy/autophagy sites at the ER membrane. Consistently, treatment with kinase inhibitor SGC-CK2-1, knockdown of endogenous CSNK2, or mutation of respective phospho-sites prevents ubiquitination, the formation of high-density clusters as well as reticulophagy flux. We hypothesize that CSNK2 has a broader impact on ER and Golgi content in a cell-type and context-specific manner by orchestrating the activity of several autophagy receptors and potentially also factors of the ER-associated protein degradation pathway.


Subject(s)
Autophagy , Casein Kinase II , Endoplasmic Reticulum , Animals , Humans , Autophagy/physiology , Casein Kinase II/metabolism , Endoplasmic Reticulum/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Phosphorylation , Ubiquitination , Mice
18.
Front Immunol ; 15: 1393485, 2024.
Article in English | MEDLINE | ID: mdl-38807597

ABSTRACT

Introduction: In classical Hodgkin lymphoma (cHL), the survival of neoplastic cells is mediated by the activation of NF-κB, JAK/STAT and PI3K/Akt signaling pathways. CK2 is a highly conserved serine/threonine kinase, consisting of two catalytic (α) and two regulatory (ß) subunits, which is involved in several cellular processes and both subunits were found overexpressed in solid tumors and hematologic malignancies. Methods and results: Biochemical analyses and in vitro assays showed an impaired expression of CK2 subunits in cHL, with CK2α being overexpressed and a decreased expression of CK2ß compared to normal B lymphocytes. Mechanistically, CK2ß was found to be ubiquitinated in all HL cell lines and consequently degraded by the proteasome pathway. Furthermore, at basal condition STAT3, NF-kB and AKT are phosphorylated in CK2-related targets, resulting in constitutive pathways activation. The inhibition of CK2 with CX-4945/silmitasertib triggered the de-phosphorylation of NF-κB-S529, STAT3-S727, AKT-S129 and -S473, leading to cHL cell lines apoptosis. Moreover, CX-4945/silmitasertib was able to decrease the expression of the immuno-checkpoint CD274/PD-L1 but not of CD30, and to synergize with monomethyl auristatin E (MMAE), the microtubule inhibitor of brentuximab vedotin. Conclusions: Our data point out a pivotal role of CK2 in the survival and the activation of key signaling pathways in cHL. The skewed expression between CK2α and CK2ß has never been reported in other lymphomas and might be specific for cHL. The effects of CK2 inhibition on PD-L1 expression and the synergistic combination of CX-4945/silmitasertib with MMAE pinpoints CK2 as a high-impact target for the development of new therapies for cHL.


Subject(s)
B7-H1 Antigen , Casein Kinase II , Hodgkin Disease , Signal Transduction , Humans , Hodgkin Disease/metabolism , Hodgkin Disease/drug therapy , Hodgkin Disease/genetics , Hodgkin Disease/pathology , Casein Kinase II/metabolism , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Cell Line, Tumor , Phenazines , Naphthyridines/pharmacology , Apoptosis , Gene Expression Regulation, Neoplastic , Phosphorylation
19.
Med Oncol ; 41(5): 94, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38526625

ABSTRACT

Casein kinase II (CK2) is an enzyme with pleiotropic kinase activity that catalyzes the phosphorylation of lots of substrates, including STAT3, p53, JAK2, PTEN, RELA, and AKT, leading to the regulation of diabetes, cardiovascular diseases, angiogenesis, and tumor progression. CK2 is observed to have high expression in multiple types of cancer, which is associated with poor prognosis. CK2 holds significant importance in the intricate network of pathways involved in promoting cell proliferation, invasion, migration, apoptosis, and tumor growth by multiple pathways such as JAK2/STAT3, PI3K/AKT, ATF4/p21, and HSP90/Cdc37. In addition to the regulation of cancer progression, increasing evidence suggests that CK2 could regulate tumor immune responses by affecting immune cell activity in the tumor microenvironment resulting in the promotion of tumor immune escape. Therefore, inhibition of CK2 is initially proposed as a pivotal candidate for cancer treatment. In this review, we discussed the role of CK2 in cancer progression and tumor therapy.


Subject(s)
Casein Kinase II , Neoplasms , Humans , Casein Kinase II/metabolism , Signal Transduction/physiology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Neoplasms/drug therapy , Tumor Microenvironment
20.
mBio ; 15(2): e0327523, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38193728

ABSTRACT

The casein kinase 2 (CK2) complex has garnered extensive attention over the past decades as a potential therapeutic target for diverse human diseases, including cancer, diabetes, and obesity, due to its pivotal roles in eukaryotic growth, differentiation, and metabolic homeostasis. While CK2 is also considered a promising antifungal target, its role in fungal pathogens remains unexplored. In this study, we investigated the functions and regulatory mechanisms of the CK2 complex in Cryptococcus neoformans, a major cause of fungal meningitis. The cryptococcal CK2 complex consists of a single catalytic subunit, Cka1, and two regulatory subunits, Ckb1 and Ckb2. Our findings show that Cka1 plays a primary role as a protein kinase, while Ckb1 and Ckb2 have major and minor regulatory functions, respectively, in growth, cell cycle control, morphogenesis, stress response, antifungal drug resistance, and virulence factor production. Interestingly, triple mutants lacking all three subunits (cka1Δ ckb1Δ ckb2Δ) exhibited more severe phenotypic defects than the cka1Δ mutant alone, suggesting that Ckb1/2 may have Cka1-independent functions. In a murine model of systemic cryptococcosis, cka1Δ and cka1Δ ckb1Δ ckb2Δ mutants showed severely reduced virulence. Transcriptomic, proteomic, and phosphoproteomic analyses further revealed that the CK2 complex controls a wide array of effector proteins involved in transcriptional regulation, cell cycle control, nutrient metabolisms, and stress responses. Most notably, CK2 disruption led to dysregulation of key signaling cascades central to C. neoformans pathogenicity, including the Hog1, Mpk1 MAPKs, cAMP/PKA, and calcium/calcineurin signaling pathways. In summary, our study provides novel insights into the multifaceted roles of the fungal CK2 complex and presents a compelling case for targeting it in the development of new antifungal drugs.IMPORTANCEThe casein kinase 2 (CK2) complex, crucial for eukaryotic growth, differentiation, and metabolic regulation, presents a promising therapeutic target for various human diseases, including cancer, diabetes, and obesity. Its potential as an antifungal target is further highlighted in this study, which explores CK2's functions in C. neoformans, a key fungal meningitis pathogen. The CK2 complex in C. neoformans, comprising the Cka1 catalytic subunit and Ckb1/2 regulatory subunits, is integral to processes like growth, cell cycle, morphogenesis, stress response, drug resistance, and virulence. Our findings of CK2's role in regulating critical signaling pathways, including Hog1, Mpk1 MAPKs, cAMP/PKA, and calcium/calcineurin, underscore its importance in C. neoformans pathogenicity. This study provides valuable insights into the fungal CK2 complex, reinforcing its potential as a target for novel antifungal drug development and pointing out a promising direction for creating new antifungal agents.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Diabetes Mellitus , Meningitis, Fungal , Neoplasms , Animals , Mice , Humans , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cryptococcus neoformans/metabolism , Antifungal Agents/metabolism , Calcium/metabolism , Calcineurin/metabolism , Proteomics , Signal Transduction , Cryptococcosis/microbiology , Obesity
SELECTION OF CITATIONS
SEARCH DETAIL