Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 267
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167022, 2024 03.
Article in English | MEDLINE | ID: mdl-38216068

ABSTRACT

BACKGROUND: CAMK1 has been shown to be involved in human disease progression via regulating mitochondrial dynamics. However, whether CAMK1 mediates mitochondrial dynamics to regulate diabetic nephropathy (DN) process remains unclear. METHODS: Mice were injected with streptozotocin (STZ) to mimic diabetic mice models in vivo, and mice with proximal tubule-specific knockout of CAMK1 (CAMK1-KO) were generated. HK-2 cells were treated with high-glucose (HG) to mimic DN cell model in vitro. Histopathological analysis was performed to confirm kidney injury in mice. ROS production and apoptosis were assessed by DHE staining and TUNEL staining. Mitochondria morphology was observed and analyzed by electron microscopy. Mitochondrial membrane potential was detected by JC-1 staining, and cell proliferation was measured by EdU assay. The mRNA and protein expression were examined by qRT-PCR, western blot and immunostaining. RNA interaction was confirmed by RIP assay and dual-luciferase reporter assay. The mRNA stability was tested by actinomycin D treatment, and m6A level was examined by MeRIP assay. RESULTS: CAMK1 was reduced in DN patients and STZ-induced diabetic mice. Conditional deletion of CAMK1 aggravated kidney injury and promoted mitochondrial fission in diabetic mice. CAMK1 overexpression inhibited mitochondrial fission to alleviate HG-induced HK-2 cell apoptosis. IGF2BP3 promoted the stability of CAMK1 mRNA by m6A modification. IGF2BP3 inhibited mitochondrial fission to repress cell apoptosis in vitro and kidney injury in vivo by increasing CAMK1 expression. CONCLUSION: IGF2BP3-mediated CAMK1 mRNA stability alleviated DN progression by inhibiting mitochondria fission.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Animals , Humans , Mice , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/pathology , Kidney Tubules/pathology , Mitochondrial Dynamics/physiology , RNA, Messenger/metabolism
2.
Nat Metab ; 5(6): 1045-1058, 2023 06.
Article in English | MEDLINE | ID: mdl-37277610

ABSTRACT

Hypothalamic AgRP/NPY neurons are key players in the control of feeding behaviour. Ghrelin, a major orexigenic hormone, activates AgRP/NPY neurons to stimulate food intake and adiposity. However, cell-autonomous ghrelin-dependent signalling mechanisms in AgRP/NPY neurons remain poorly defined. Here we show that calcium/calmodulin-dependent protein kinase ID (CaMK1D), a genetic hot spot in type 2 diabetes, is activated upon ghrelin stimulation and acts in AgRP/NPY neurons to mediate ghrelin-dependent food intake. Global Camk1d-knockout male mice are resistant to ghrelin, gain less body weight and are protected against high-fat-diet-induced obesity. Deletion of Camk1d in AgRP/NPY, but not in POMC, neurons is sufficient to recapitulate above phenotypes. In response to ghrelin, lack of CaMK1D attenuates phosphorylation of CREB and CREB-dependent expression of the orexigenic neuropeptides AgRP/NPY in fibre projections to the paraventricular nucleus (PVN). Hence, CaMK1D links ghrelin action to transcriptional control of orexigenic neuropeptide availability in AgRP neurons.


Subject(s)
Diabetes Mellitus, Type 2 , Ghrelin , Mice , Animals , Male , Ghrelin/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Diabetes Mellitus, Type 2/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Neurons/metabolism , Obesity/metabolism , Mice, Knockout , Eating , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism
4.
Mol Cell Biochem ; 478(4): 791-805, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36094721

ABSTRACT

Connexin 43 (Cx43, also known as Gja1) is the most abundant testicular gap junction protein. It has a crucial role in the support of spermatogenesis by Sertoli cells in the seminiferous tubules as well as in androgen synthesis by Leydig cells. The multifunctional family of Ca2+/calmodulin-dependent protein kinases (CaMK) is composed of CaMK I, II, and IV and each can serve as a mediator of nuclear Ca2+ signals. These kinases can control gene expression by phosphorylation of key regulatory sites on transcription factors. Among these, AP-1 members cFos and cJun are interesting candidates that seem to cooperate with CaMKs to regulate Cx43 expression in Leydig cells. In this study, the Cx43 promoter region important for CaMK-dependent activation is characterized using co-transfection of plasmid reporter-constructs with different plasmids coding for CaMKs and/or AP-1 members in MA-10 Leydig cells. Here we report that the activation of Cx43 expression by cFos and cJun is increased by CaMKI. Furthermore, results from chromatin immunoprecipitation suggest that the recruitment of AP-1 family members to the proximal region of the Cx43 promoter may involve another uncharacterized AP-1 DNA regulatory element and/or protein-protein interactions with other partners. Thus, our data provide new insights into the molecular regulatory mechanisms that control mouse Cx43 transcription in testicular Leydig cells.


Subject(s)
Leydig Cells , Neoplasms , Male , Mice , Animals , Leydig Cells/metabolism , Connexin 43/genetics , Connexin 43/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Neoplasms/metabolism
5.
Int J Mol Sci ; 23(21)2022 Oct 29.
Article in English | MEDLINE | ID: mdl-36361957

ABSTRACT

Hormone-induced Leydig cell steroidogenesis requires rapid changes in gene expression in response to various hormones, cytokines, and growth factors. These proteins act by binding to their receptors on the surface of Leydig cells leading to activation of multiple intracellular signaling cascades, downstream of which are several kinases, including protein kinase A (PKA), Ca2+/calmodulin-dependent protein kinase I (CAMKI), and extracellular signal-regulated protein kinase 1 and 2 (ERK1/2). These kinases participate in hormone-induced steroidogenesis by phosphorylating numerous proteins including transcription factors leading to increased steroidogenic gene expression. How these various kinases and transcription factors come together to appropriately induce steroidogenic gene expression in response to specific stimuli remains poorly understood. In the present work, we compared the effect of PKA, CAMKI and ERK1/2 on the transactivation potential of 15 transcription factors belonging to 5 distinct families on the activity of the Star gene promoter. We not only validated known cooperation between kinases and transcription factors, but we also identified novel cooperations that have not yet been before reported. Some transcription factors were found to respond to all three kinases, whereas others were only activated by one specific kinase. Differential responses were also observed within a family of transcription factors. The diverse response to kinases provides flexibility to ensure proper genomic response of steroidogenic cells to different stimuli.


Subject(s)
Phosphoproteins , Transcription Factors , Humans , Male , Cyclic AMP-Dependent Protein Kinases/metabolism , Hormones/metabolism , Leydig Cells/metabolism , Phosphoproteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 1/metabolism
6.
Int J Mol Sci ; 23(20)2022 Oct 14.
Article in English | MEDLINE | ID: mdl-36293185

ABSTRACT

The expression of the Calcium/Calmodulin-Dependent Protein Kinase I gamma (encoded by the Camk1g gene) depends on the activation of glucocorticoid receptors (GR) and is strongly regulated by stress. Since Camk1g is primarily expressed in neuronal cells of the limbic system in the brain, we hypothesized that it could be involved in signaling mechanisms that underlie the adaptive or maladaptive responses to stress. Here, we find that restraint-induced stress and the GR agonist dexamethasone robustly increase the expression of Camk1g in neurons of the amygdalar nuclei in the mouse brain. To assess the functional role of Camk1g expression, we performed a virally induced knock-down of the transcript. Mice with bilateral amygdala-specific Camk1g knock-down showed increased anxiety-like behaviors in the light-dark box, and an increase in freezing behavior after fear-conditioning, but normal spatial working memory during exploration of a Y-maze. Thus, we confirm that Camk1g is a neuron-specific GR-regulated transcript, and show that it is specifically involved in behaviors related to anxiety, as well as responses conditioned by aversive stimuli.


Subject(s)
Central Amygdaloid Nucleus , Glucocorticoids , Mice , Animals , Glucocorticoids/pharmacology , Central Amygdaloid Nucleus/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Calcium , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Anxiety/metabolism , Dexamethasone/pharmacology , Behavior, Animal
7.
Proc Natl Acad Sci U S A ; 119(16): e2117435119, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35412911

ABSTRACT

Elevation of intracellular Ca2+ concentration ([Ca2+]i) activates Ca2+/calmodulin-dependent kinases (CaMK) and promotes gene transcription. This signaling pathway is referred to as excitation­transcription (E-T) coupling. Although vascular myocytes can exhibit E-T coupling, the molecular mechanisms and physiological/pathological roles are unknown. Multiscale analysis spanning from single molecules to whole organisms has revealed essential steps in mouse vascular myocyte E-T coupling. Upon a depolarizing stimulus, Ca2+ influx through Cav1.2 voltage-dependent Ca2+ channels activates CaMKK2 and CaMK1a, resulting in intranuclear CREB phosphorylation. Within caveolae, the formation of a molecular complex of Cav1.2/CaMKK2/CaMK1a is promoted in vascular myocytes. Live imaging using a genetically encoded Ca2+ indicator revealed direct activation of CaMKK2 by Ca2+ influx through Cav1.2 localized to caveolae. CaMK1a is phosphorylated by CaMKK2 at caveolae and translocated to the nucleus upon membrane depolarization. In addition, sustained depolarization of a mesenteric artery preparation induced genes related to chemotaxis, leukocyte adhesion, and inflammation, and these changes were reversed by inhibitors of Cav1.2, CaMKK2, and CaMK, or disruption of caveolae. In the context of pathophysiology, when the mesenteric artery was loaded by high pressure in vivo, we observed CREB phosphorylation in myocytes, macrophage accumulation at adventitia, and an increase in thickness and cross-sectional area of the tunica media. These changes were reduced in caveolin1-knockout mice or in mice treated with the CaMKK2 inhibitor STO609. In summary, E-T coupling depends on Cav1.2/CaMKK2/CaMK1a localized to caveolae, and this complex converts [Ca2+]i changes into gene transcription. This ultimately leads to macrophage accumulation and media remodeling for adaptation to increased circumferential stretch.


Subject(s)
Calcium Channels, L-Type , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Calcium-Calmodulin-Dependent Protein Kinase Type 1 , Caveolae , Transcription, Genetic , Vascular Remodeling , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Caveolae/metabolism , Caveolin 1/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Excitation Contraction Coupling , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Neurons/metabolism , Phosphorylation
8.
Arch Biochem Biophys ; 720: 109170, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35276214

ABSTRACT

CaMK phosphatase (CaMKP/PPM1F/POPX2) is a Mn2+-dependent, calyculin A/okadaic acid-insensitive Ser/Thr protein phosphatase that belongs to the PPM family. CaMKP is thought to be involved in regulation of not only various protein kinases, such as CaM kinases and p21-activated protein kinase, but also of cellular proteins regulated by phosphorylation. A large-scale screening of a chemical library identified gallic acid and some of its alkyl esters as novel CaMKP inhibitors highly specific to CaMKP. Surprisingly, they caused specific carbonylation of CaMKP, leading to its inactivation. Under the same conditions, no carbonylation nor inactivation was observed when PPM1A, which is affiliated with the same family as CaMKP, and λ-phosphatase were used. The carbonylation reaction was inhibited by SH compounds such as cysteamine in a dose-dependent manner with a concomitant decrease in CaMKP inhibition by ethyl gallate. The pyrogallol structure of gallate was necessary for the gallate-mediated carbonylation of CaMKP. Point mutations of CaMKP leading to impairment of phosphatase activity did not significantly affect the gallate-mediated carbonylation. Ethyl gallate resulted in almost complete inhibition of CaMKP under the conditions where the carbonylation level was nearly identical to that of CaMKP carbonylation via metal-catalyzed oxidation with ascorbic acid/FeSO4, which resulted in only a partial inhibition of CaMKP. The gallate-mediated carbonylation of CaMKP absolutely required divalent cations such as Mn2+, Cu2+, Co2+ and Fe2+, and was markedly enhanced by a phosphopeptide substrate. When MDA-MB-231 cells transiently expressing CaM kinase I, a CaMKP substrate, were treated by ethyl gallate, significant enhancement of phosphorylation of CaM kinase I was observed, suggesting that ethyl gallate can penetrate into cells to inactivate cellular CaMKP. All the presented data strongly support the hypothesis that CaMKP undergoes carbonylation of its specific amino acid residues by incubation with alkyl gallates and the divalent metal cations, leading to inactivation specific to CaMKP.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 1 , Phosphoprotein Phosphatases , Calcium-Calmodulin-Dependent Protein Kinase Type 1/chemistry , Oxidation-Reduction , Phosphoprotein Phosphatases/chemistry , Phosphorylation , Protein Carbonylation , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/metabolism
9.
Commun Biol ; 5(1): 124, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35145192

ABSTRACT

Statins play a major role in reducing circulating cholesterol levels and are widely used to prevent coronary artery disease. Although they are recently confirmed to up-regulate mitophagy, little is known about the molecular mechanisms and its effect on endothelial progenitor cell (EPC). Here, we explore the role and mechanism underlying statin (pitavastatin, PTV)-activated mitophagy in EPC proliferation. ApoE-/- mice are fed a high-fat diet for 8 weeks to induce atherosclerosis. In these mice, EPC proliferation decreases and is accompanied by mitochondrial dysfunction and mitophagy impairment via the PINK1-PARK2 pathway. PTV reverses mitophagy and reduction in proliferation. Pink1 knockout or silencing Atg7 blocks PTV-induced proliferation improvement, suggesting that mitophagy contributes to the EPC proliferation increase. PTV elicits mitochondrial calcium release into the cytoplasm and further phosphorylates CAMK1. Phosphorylated CAMK1 contributes to PINK1 phosphorylation as well as mitophagy and mitochondrial function recover in EPCs. Together, our findings describe a molecular mechanism of mitophagy activation, where mitochondrial calcium release promotes CAMK1 phosphorylation of threonine177 before phosphorylation of PINK1 at serine228, which recruits PARK2 and phosphorylates its serine65 to activate mitophagy. Our results further account for the pleiotropic effects of statins on the cardiovascular system and provide a promising and potential therapeutic target for atherosclerosis.


Subject(s)
Atherosclerosis , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 1 , Endothelial Progenitor Cells , Protein Kinases , Quinolines , Animals , Mice , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Calcium/metabolism , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cell Proliferation/drug effects , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Mitophagy , Protein Kinases/genetics , Protein Kinases/metabolism , Quinolines/pharmacology , Ubiquitin-Protein Ligases/metabolism
10.
Article in English | MEDLINE | ID: mdl-34320404

ABSTRACT

Lithium is the first-line treatment for bipolar disorder (BD), but there is a large variation in response rate and adverse effects. Although the molecular effects of lithium have been studied extensively, the specific mechanisms of action remain unclear. In particular, the molecular changes underlying lithium adverse effects are little known. Multiple linear regression analyses of lithium serum concentrations and global gene expression levels in whole blood were carried out using a large case-control sample (n = 1450). Self-reported adverse effects of lithium were assessed with the "Udvalg for Kliniske Undersøgelser" (UKU) adverse effect rating scale, and regression analysis was used to identify significant associations between lithium-related genes and six of the most common adverse effects. Serum concentrations of lithium were significantly associated with the expression levels of 52 genes (FDR < 0.01), largely replicating previous results. We found 32 up-regulated genes and 20 down-regulated genes in lithium users compared to non-users. The down-regulated gene set was enriched for several processes related to the translational machinery. Two adverse effects were significantly associated (p < 0.01) with three or more lithium-associated genes: tremor (FAM13A-AS1, FAR2, ITGAX, RWDD1, and STARD10) and xerostomia (ANKRD13A, FAR2, RPS8, and RWDD1). The adverse effect association with the largest effect was between CAMK1D expression and nausea/vomiting. These results suggest putative transcriptional mechanisms that may predict lithium adverse effects, and could thus have a large potential for informing clinical practice.


Subject(s)
Antipsychotic Agents/adverse effects , Antipsychotic Agents/therapeutic use , Bipolar Disorder/drug therapy , Gene Expression/drug effects , Lithium/adverse effects , Lithium/therapeutic use , Antipsychotic Agents/blood , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Case-Control Studies , Cohort Studies , GTPase-Activating Proteins/genetics , Humans , Interviews as Topic , Lithium/blood
11.
Biochem Biophys Res Commun ; 587: 160-165, 2022 01 08.
Article in English | MEDLINE | ID: mdl-34875535

ABSTRACT

Ca2+/calmodulin-dependent protein kinase kinases (CaMKKα and ß) are regulatory kinases for multiple downstream kinases, including CaMKI, CaMKIV, PKB/Akt, and AMP-activated protein kinase (AMPK) through phosphorylation of each activation-loop Thr residue. In this report, we biochemically characterize the oligomeric structure of CaMKK isoforms through a heterologous expression system using COS-7 cells. Oligomerization of CaMKK isoforms was readily observed by treating CaMKK transfected cells with cell membrane permeable crosslinkers. In addition, His-tagged CaMKKα (His-CaMKKα) pulled down with FLAG-tagged CaMKKα (FLAG-CaMKKα) in transfected cells. The oligomerization of CaMKKα was confirmed by the fact that GST-CaMKKα/His-CaMKKα complex from transiently expressed COS-7 cells extracts was purified to near homogeneity by the sequential chromatography using glutathione-sepharose/Ni-sepharose and was observed in a Ca2+/CaM-independent manner by reciprocal pulldown assay, suggesting the direct interaction between monomeric CaMKKα. Furthermore, the His-CaMKKα kinase-dead mutant (D293A) complexed with FLAG-CaMKKα exhibited significant CaMKK activity, indicating the active CaMKKα multimeric complex. Collectively, these results suggest that CaMKKα can self-associate in the cells, constituting a catalytically active oligomer that might be important for the efficient activation of CaMKK-mediated intracellular signaling.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 1/chemistry , Glutathione Transferase/chemistry , Recombinant Fusion Proteins/chemistry , Animals , Binding Sites , COS Cells , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Chlorocebus aethiops , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Phosphorylation , Protein Binding , Protein Multimerization , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
12.
Molecules ; 26(24)2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34946752

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia worldwide. Despite extensive research and targeting of the main molecular components of the disease, beta-amyloid (Aß) and tau, there are currently no treatments that alter the progression of the disease. Here, we examine the effects of two specific kinase inhibitors for calcium/calmodulin-dependent protein kinase type 1D (CaMK1D) on Aß-mediated toxicity, using mouse primary cortical neurons. Tau hyperphosphorylation and cell death were used as AD indicators. These specific inhibitors were found to prevent Aß induced tau hyperphosphorylation in culture, but were not able to protect cells from Aß induced toxicity. While inhibitors were able to alter AD pathology in cell culture, they were insufficient to prevent cell death. With further research and development, these inhibitors could contribute to a multi-drug strategy to combat AD.


Subject(s)
Alzheimer Disease/drug therapy , Calcium-Calmodulin-Dependent Protein Kinase Type 1/antagonists & inhibitors , Disease Models, Animal , Neurons/drug effects , Protein Kinase Inhibitors/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cell Survival/drug effects , Cells, Cultured , Mice , Mice, Inbred BALB C , Models, Molecular , Neurons/metabolism , Neurons/pathology , Protein Kinase Inhibitors/chemistry
13.
Mol Brain ; 14(1): 148, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556164

ABSTRACT

The Cre/LoxP-based conditional knockout technology is a powerful tool for gene function analysis that allows region- and time-specific gene manipulation. However, inserting a pair of LoxP cassettes to generate conditional knockout can be technically challenging and thus time- and resource-consuming. This study proposes an efficient, low-cost method to generate floxed mice using in vitro fertilization and the CRISPR-Cas9 system over two consecutive generations. This method allowed us to produce floxed mice targeting exons 5 and 6 of CaMK1 in a short period of 125 days, using only 16 mice. In addition, we directly edited the genome of fertilized eggs of mice with our target genetic background, C57BL/6 N, to eliminate additional backcrossing steps. We confirmed that the genome of the generated floxed mice was responsive to the Cre protein. This low-cost, time-saving method for generating conditional knockout will facilitate comprehensive, tissue-specific genome analyses.


Subject(s)
CRISPR-Cas Systems , Electroporation/methods , Gene Editing/methods , Gene Targeting/methods , Mice, Knockout , Neurosciences/methods , Animals , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Embryo Transfer , Exons/genetics , Gene Editing/economics , Gene Targeting/economics , Integrases , Mice , Mice, Inbred C57BL , Neurosciences/economics , Transgenes
14.
Eur Rev Med Pharmacol Sci ; 25(4): 1899-1909, 2021 02.
Article in English | MEDLINE | ID: mdl-33660800

ABSTRACT

OBJECTIVE: Circular ribonucleic acids (circRNAs) are considered as the key regulatory factors for human malignancies in recent years, and lung adenocarcinoma (LUAD) is a common malignancy worldwide, but the molecular mechanism of circRNAs in LUAD has not been completely investigated. Therefore, the mechanism by which circRNA protein kinase C iota (circPRKCI) regulates LUAD cell migration proliferation, and cycle was preliminarily explored in this research, so as to provide new ideas for the treatment of LUAD. PATIENTS AND METHODS: First of all, the circPRKCI expression level in LUAD tissues was tested via quantitative reverse transcription-polymerase chain reaction (qRT-PCR) assay, and the relationship between circPRKCI and the patients' prognosis was analyzed. Then, circPRKCI expression was inhibited by small interfering RNA (siRNA), and the influence of circPRKCI on t LUAD cells' ability to proliferate was verified via 5-ethynyl-2'-deoxyuridine (EdU) and cell counting kit-8 (CCK-8) assays. Moreover, the influence of circPRKCI on LUAD cells' ability to migrate was testified by transwell assay, and the regulation of LUAD cell cycle by circPRKCI was confirmed by flow cytometry. The micro RNAs (miRNAs) with binding sites to the 3' untranslated region (UTR) of circPRKCI and the genes binding to miRNAs were discovered using bioinformatics websites, and their associative relation was further explored through Dual-Luciferase reporter gene assay, qRT-PCR assay, Pearson correlation analysis and reverse experiment. RESULTS: It was verified via qRT-PCR assay that circPRKCI was expressed at a remarkably higher level in LUAD tissues relative to that in paracancerous normal tissues. The highly expressed circPRKCI led to poor prognosis of patients. Besides, qRT-PCR assessment results indicated that circPRKCI expression level rose notably in LUAD cell lines, while it was lowered markedly in LUAD cells transfected with si-circPRKCI. According to CCK-8 and EdU assay results, the proliferative ability of LUAD cells was weakened clearly after knocking down circPRKCI. It was manifested in the results of transwell assay that the knockdown of circPRKCI significantly repressed the capacity of LUAD cells to migrate. Furthermore, the results of cell cycle test displayed that inhibiting circPRKCI could induce the arrest of LUAD cell cycle in the G1 phase. It was discovered through bioinformatics websites that miR-219a-5p had binding sites to circPRKCI 3'UTR, and the results of Dual-Luciferase reporter gene assay revealed that circPRKCI was able to bind to miR-219a-5p. It was uncovered by the qRT-PCR assay results that miR-219a-5p was lowly expressed in LUAD tissues, and its relative expression had an inverse relation with that of circPRKCI according to the Pearson correlation analysis. In addition, it was shown in the results of reverse experiment that miR-219a-5p could regulate the influence of circPRKCI on the malignant phenotype of LUAD. It was found by means of bioinformatics websites that calcium/calmodulin dependent protein kinase ID (CAMK1D) was a downstream target gene of miR-219a-5p and could the two conjugated with each other based on the results of Dual-Luciferase reporter gene assay. Moreover, qRT-PCR assay findings illustrated that CAMK1D was evidently highly expressed in LUAD tissues, and the results of Pearson correlation analysis revealed that CAMK1D expression exhibited a negative association with that of miR-219a-5p and a positive correlation with that of circPRKCI. CONCLUSIONS: CircPRKCI is significantly highly expressed in LUAD, and the highly expressed circPRKCI is capable of facilitating LUAD cell migration, proliferation and cycle. CircPRKCI may regulate the malignant phenotype of LUAD via the miR-219a-5p/CAMK1D axis.


Subject(s)
Adenocarcinoma of Lung/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Lung Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Circular/metabolism , Adenocarcinoma of Lung/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Cell Cycle , Cell Movement , Cell Proliferation , Cells, Cultured , Humans , Lung Neoplasms/pathology , MicroRNAs/genetics , RNA, Circular/genetics
15.
J Biochem ; 169(4): 445-458, 2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33417706

ABSTRACT

Ca2+/calmodulin (CaM)-dependent protein kinase Iδ (CaMKIδ) is a Ser/Thr kinase that plays pivotal roles in Ca2+ signalling. CaMKIδ is activated by Ca2+/CaM-binding and phosphorylation at Thr180 by CaMK kinase (CaMKK). In this study, we characterized four splice variants of mouse CaMKIδ (mCaMKIδs: a, b, c and d) found by in silico analysis. Recombinant mCaMKIδs expressed in Escherichia coli were phosphorylated by CaMKK; however, only mCaMKIδ-a and c showed protein kinase activities towards myelin basic protein in vitro, with mCaMKIδ-b and mCaMKIδ-d being inactive. Although mCaMKIδ-a and mCaMKIδ-c underwent autophosphorylation in vitro, only mCaMKIδ-c underwent autophosphorylation in 293T cells. Site-directed mutagenesis showed that the autophosphorylation site is Ser349, which is found in the C-terminal region of only variants c and b (Ser324). Furthermore, phosphorylation of these sites (Ser324 and Ser349) in mCaMKIδ-b and c was more efficiently catalyzed by cAMP-dependent protein kinase in vitro and in cellulo as compared to the autophosphorylation of mCaMKIδ-c. Thus, variants of mCaMKIδ possess distinct properties in terms of kinase activities, autophosphorylation and phosphorylation by another kinase, suggesting that they play physiologically different roles in murine cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 1 , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 1/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cell Line , Cyclic AMP/genetics , Cyclic AMP/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Phosphorylation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
16.
J Diabetes Complications ; 35(1): 107752, 2021 01.
Article in English | MEDLINE | ID: mdl-33127268

ABSTRACT

AIMS: Diabetes mellitus and hypertension are both complex diseases that are caused by interactions among multiple genetic and physiological factors. To investigate the association of common single-nucleotide polymorphisms (SNPs) of SUCNR1, GRK4 and CAMK1D genes with the susceptibility of the two diseases in a northern Chinese Han population. METHODS: 36 SNPs were genotyped in 2304 clinical patients (1152 type 2 diabetes mellitus, 1152 essential hypertension) and 1152 health controls by Sequenom Mass-ARRAY RS1000. RESULTS: In this study, we found that BMI, blood press, pulse pressure, FBG, total cholesterol and triglycerides were associated with an increased risk of type 2 diabetes mellitus (T2DM) and essential hypertension (EH). Three SNPs (SUCNR1: rs73168929; GRK4: rs1557213; CAMK1D: rs17151584) significantly associated with the susceptibility of T2DM and EH at the same time. Also, the susceptibility genotypes of 3 SNPs were significantly correlated with liver and renal function parameters. CONCLUSION: To the best of our knowledge, the present study is the first to report that three SNPs (SUCNR1: rs73168929; GRK4: rs1557213; CAMK1D: rs17151584) contributed to the risk of T2DM and EH in a northern Chinese Han population. These results provide a favourable evidence for better understand of the underlying common mechanism of these two diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Asian People/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1 , Case-Control Studies , China/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Essential Hypertension , G-Protein-Coupled Receptor Kinase 4 , Genetic Predisposition to Disease , Genotype , Humans , Polymorphism, Single Nucleotide , Receptors, G-Protein-Coupled
17.
J Cell Mol Med ; 25(2): 1198-1206, 2021 01.
Article in English | MEDLINE | ID: mdl-33342045

ABSTRACT

Calcium/calmodulin-dependent protein kinase (CAMKs) can control a wide range of cancer-related functions in multiple tumour types. Herein, we explore the expressions and clinical significances of calcium/calmodulin-dependent protein kinase 1 (CAMK1) in pancreatic cancer (PC). The expression of CAMK1 in PC was analysed by Gene Expression Profiling Interactive Analysis 2 (GEPIA 2) database and the Oncomine database. For further validation, the protein level of CAMK1 in PC tissues was also detected in the Human Protein Atlas (HPA) database and the tissue microarray (TMA)-based immunohistochemistry (IHC). GEPIA 2 and Kaplan-Meier Plotter (KM Plotter) databases were used to explore the prognostic significances of CAMK1 in overall survival (OS) and disease-free survival (DFS) of PC at mRNA level. The relationship between CAMK1 expression and the clinicopathological characteristics of PC was further explored. Additionally, the Search Tool for the Retrieval of Interacting Genes (STRING) database was used to analyse protein-protein interactions (PPI). We found CAMK1 was highly expressed in PC both in bioinformatics analyses and TMA-IHC results. The prognostic analyses from the public databases also showed consistent results with follow-up data. The PPI network suggested that CALM1, CALM3, CREB1, CALM2, SYN1, NOS3, ATF1, GAPDH, PPM1F and FBXL12 were important significant genes associated with CAMK1. Our finding revealed CAMK1 has prognostic value in PC patients, suggesting that CAMK1 may has a distinct role in PC patients and can be used as a candidate marker for investigating clinical prognosis of PC.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Databases, Genetic , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Protein Interaction Maps , Reproducibility of Results
18.
Vascul Pharmacol ; 136: 106820, 2021 02.
Article in English | MEDLINE | ID: mdl-33238205

ABSTRACT

AIM: Exsomes play a significant role in increasing pathophysiological processes by delivering their content. Recently, a variety of studies have showed exosomal microRNAs (miRNAs) are involved in pulmonary hypertension (PH) notably. In this study, we found that exosomal miR-211 was overexpressed in hypoxia-induced PH rats but its intrinsic regulation was unclear. Therefore, our aim was to reveal the underlying mechanism which overexpressed exosomal miR-211 targeted in the development of PH. METHODS: 18 male SD rats were randomly divided into normoxia and hypoxia group, housed in normal or hypoxic chamber for 3 weeks respectively. Then, mean pulmonary arterial pressure (mPAP), pulmonary vascular resistance(PVR), right ventricular hypertrophy index(RV/(LV + S)), the percentage of medial wall area (WA%) and the percentage of medial wall thickness (WT%) were measured. Expression of miR-211 in exosomes was detected by qRT-PCR. Expression of Ca2+/calmodulin-dependent kinase1(CaMK1)and peroxisome proliferator-activated receptors-γ(PPAR-γ)in lung tissue were detected by Western blot(WB); After miR-211 overexpressed exosomes were injected to rats through caudal vein, mPAP, PVR, RV/(LV + S), WA% and WT% were also measured. Sequentially, hypoxia rats were injected with lentivirus riched in miR-211 inhibitor via tail vein, and PH-related indicators were measured. In vitro, after miR-211 was positively or negatively regulated in pulmonary arterial smooth muscle cell (PASMC) by plasmid transfection, proliferation of PASMC was detected by CCK8, as well as the expression of CaMK1 and PPAR- γ. Further, the relationship between CaMK1 and miR-211 was verified by Dual-Luciferase assay. And the regulatory relationship of CaMK1/PPAR- γ aixs was demonstrated in PASMC. RESULTS: Evident increases of mPAP, PVR, RVHI, WT% and WA% were observed with hypoxia administration. And the concentration of plasma exosomes in hypoxia rats was increased and positively correlated with the above indexes. miR-211 in exosomes of PH was upregulated while the expression of CaMK1 and PPAR-γ decreased in lung tissues. Further, injection of exosomes overexpressed with miR-211 demonstrated that exosomal miR-211 aggravated PH while inhibition of miR-211 attenuated PH in rats. In vitro, overexpression of miR-211 promoted the proliferation of PASMC and inhibited expression of CaMK1 and PPAR-γ in PASMC. And Dual-luciferase assay demonstrated that CaMK1 was a downstream gene of miR-211. Plasmid transfection experiments indicated that CaMK1 can promote PPAR-γ expression. CONCLUSION: Exosomal miR-211 promoted PH via inhibiting CaMK1/PPAR-γ axis, promoting PASMC proliferation in rats.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Exosomes/metabolism , MicroRNAs/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , PPAR gamma/metabolism , Vascular Remodeling , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Exosomes/genetics , Exosomes/transplantation , Hypoxia/complications , Male , MicroRNAs/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , PPAR gamma/genetics , Pulmonary Arterial Hypertension/enzymology , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Rats, Sprague-Dawley , Signal Transduction
19.
Osteoarthritis Cartilage ; 29(2): 235-247, 2021 02.
Article in English | MEDLINE | ID: mdl-33248223

ABSTRACT

OBJECTIVE: Fibronectin is a matrix protein that is fragmented during cartilage degradation in osteoarthritis (OA). Treatment of chondrocytes with fibronectin fragments (FN-f) has been used to model OA in vitro, but the system has not been fully characterized. This study sought to define the transcriptional response of chondrocytes to FN-f, and directly compare it to responses traditionally observed in OA. DESIGN: Normal human femoral chondrocytes isolated from tissue donors were treated with either FN-f or PBS (control) for 3, 6, or 18 h. RNA-seq libraries were compared between time-matched FN-f and control samples in order to identify changes in gene expression over time. Differentially expressed genes were compared to a published OA gene set and used for pathway, transcription factor motif, and kinome analysis. RESULTS: FN-f treatment resulted in 3,914 differentially expressed genes over the time course. Genes that are up- or downregulated in OA were significantly up- (P < 0.00001) or downregulated (P < 0.0004) in response to FN-f. Early response genes were involved in proinflammatory pathways, whereas many late response genes were involved in ferroptosis. The promoters of upregulated genes were enriched for NF-κB, AP-1, and IRF motifs. Highly upregulated kinases included CAMK1G, IRAK2, and the uncharacterized kinase DYRK3, while growth factor receptors TGFBR2 and FGFR2 were downregulated. CONCLUSIONS: FN-f treatment of normal human articular chondrocytes recapitulated many key aspects of the OA chondrocyte phenotype. This in vitro model is promising for future OA studies, especially considering its compatibility with genomics and genome-editing techniques.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/drug effects , Fibronectins/pharmacology , Gene Expression/drug effects , Osteoarthritis/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Chondrocytes/metabolism , Femur , Gene Expression/genetics , Humans , In Vitro Techniques , Interferon Regulatory Factors/drug effects , Interferon Regulatory Factors/genetics , Interleukin-1 Receptor-Associated Kinases/drug effects , Interleukin-1 Receptor-Associated Kinases/genetics , NF-kappa B/drug effects , NF-kappa B/genetics , Osteoarthritis/metabolism , Peptide Fragments/pharmacology , Phenotype , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 2/drug effects , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Transforming Growth Factor-beta Type II/drug effects , Receptor, Transforming Growth Factor-beta Type II/genetics , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/genetics
20.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33228180

ABSTRACT

Reactive oxygen species (ROS) are not only harmful to cell survival but also essential to cell signaling through cysteine-based redox switches. In fact, ROS triggers the potential activation of mitogen-activated protein kinases (MAPKs). The 90 kDa ribosomal S6 kinase 1 (RSK1), one of the downstream mediators of the MAPK pathway, is implicated in various cellular processes through phosphorylating different substrates. As such, RSK1 associates with and phosphorylates neuronal nitric oxide (NO) synthase (nNOS) at Ser847, leading to a decrease in NO generation. In addition, the RSK1 activity is sensitive to inhibition by reversible cysteine-based redox modification of its Cys223 during oxidative stress. Aside from oxidative stress, nitrosative stress also contributes to cysteine-based redox modification. Thus, the protein kinases such as Ca2+/calmodulin (CaM)-dependent protein kinase I (CaMKI) and II (CaMKII) that phosphorylate nNOS could be potentially regulated by cysteine-based redox modification. In this review, we focus on the role of post-translational modifications in regulating nNOS and nNOS-phosphorylating protein kinases and communication among themselves.


Subject(s)
Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Oxidative Stress/genetics , Protein Processing, Post-Translational , Reactive Oxygen Species/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cysteine/metabolism , Glutathione/metabolism , Humans , Huntington Disease/enzymology , Huntington Disease/genetics , Huntington Disease/pathology , Nitric Oxide Synthase Type I/genetics , Oxidation-Reduction , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...