Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167022, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38216068

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Animales , Humanos , Ratones , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/patología , Túbulos Renales/patología , Dinámicas Mitocondriales/fisiología , ARN Mensajero/metabolismo
2.
Nat Metab ; 5(6): 1045-1058, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37277610

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 2 , Ghrelina , Ratones , Animales , Masculino , Ghrelina/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Ratones Noqueados , Ingestión de Alimentos , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo
3.
Mol Cell Biochem ; 478(4): 791-805, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36094721

RESUMEN

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.


Asunto(s)
Células Intersticiales del Testículo , Neoplasias , Masculino , Ratones , Animales , Células Intersticiales del Testículo/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Neoplasias/metabolismo
4.
Int J Mol Sci ; 23(21)2022 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-36361957

RESUMEN

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.


Asunto(s)
Fosfoproteínas , Factores de Transcripción , Humanos , Masculino , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hormonas/metabolismo , Células Intersticiales del Testículo/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo
5.
Int J Mol Sci ; 23(20)2022 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-36293185

RESUMEN

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.


Asunto(s)
Núcleo Amigdalino Central , Glucocorticoides , Ratones , Animales , Glucocorticoides/farmacología , Núcleo Amigdalino Central/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Calcio , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Ansiedad/metabolismo , Dexametasona/farmacología , Conducta Animal
6.
Proc Natl Acad Sci U S A ; 119(16): e2117435119, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35412911

RESUMEN

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.


Asunto(s)
Canales de Calcio Tipo L , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina , Caveolas , Transcripción Genética , Remodelación Vascular , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Caveolas/metabolismo , Caveolina 1/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Acoplamiento Excitación-Contracción , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Neuronas/metabolismo , Fosforilación
7.
Commun Biol ; 5(1): 124, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35145192

RESUMEN

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.


Asunto(s)
Aterosclerosis , Señalización del Calcio , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina , Células Progenitoras Endoteliales , Proteínas Quinasas , Quinolinas , Animales , Ratones , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Proliferación Celular/efectos de los fármacos , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Mitofagia , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Quinolinas/farmacología , Ubiquitina-Proteína Ligasas/metabolismo
8.
Biochem Biophys Res Commun ; 587: 160-165, 2022 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-34875535

RESUMEN

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.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/química , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/química , Glutatión Transferasa/química , Proteínas Recombinantes de Fusión/química , Animales , Sitios de Unión , Células COS , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Chlorocebus aethiops , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Fosforilación , Unión Proteica , Multimerización de Proteína , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal
9.
Molecules ; 26(24)2021 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-34946752

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Modelos Animales de Enfermedad , Neuronas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Neuronas/metabolismo , Neuronas/patología , Inhibidores de Proteínas Quinasas/química
10.
Eur Rev Med Pharmacol Sci ; 25(4): 1899-1909, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33660800

RESUMEN

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.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Neoplasias Pulmonares/metabolismo , MicroARNs/metabolismo , ARN Circular/metabolismo , Adenocarcinoma del Pulmón/patología , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/genética , Ciclo Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Humanos , Neoplasias Pulmonares/patología , MicroARNs/genética , ARN Circular/genética
11.
J Biochem ; 169(4): 445-458, 2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33417706

RESUMEN

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.


Asunto(s)
Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina , Animales , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/química , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Línea Celular , AMP Cíclico/genética , AMP Cíclico/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
J Cell Mol Med ; 25(2): 1198-1206, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33342045

RESUMEN

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.


Asunto(s)
Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Bases de Datos Genéticas , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Mapas de Interacción de Proteínas , Reproducibilidad de los Resultados
13.
Vascul Pharmacol ; 136: 106820, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33238205

RESUMEN

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.


Asunto(s)
Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Exosomas/metabolismo , MicroARNs/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , PPAR gamma/metabolismo , Remodelación Vascular , Animales , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/genética , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Exosomas/genética , Exosomas/trasplante , Hipoxia/complicaciones , Masculino , MicroARNs/genética , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , PPAR gamma/genética , Hipertensión Arterial Pulmonar/enzimología , Hipertensión Arterial Pulmonar/etiología , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/patología , Arteria Pulmonar/enzimología , Arteria Pulmonar/patología , Ratas Sprague-Dawley , Transducción de Señal
14.
Int J Mol Sci ; 21(22)2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33228180

RESUMEN

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.


Asunto(s)
Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo/genética , Procesamiento Proteico-Postraduccional , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Transducción de Señal , Animales , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Cisteína/metabolismo , Glutatión/metabolismo , Humanos , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Óxido Nítrico Sintasa de Tipo I/genética , Oxidación-Reducción , Fosforilación , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética
15.
Cancer Genomics Proteomics ; 17(6): 747-755, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33099476

RESUMEN

BACKGROUND/AIM: Pregnancy up-regulated non-ubiquitous calmodulin kinase (PNCK) is a member of calmodulin kinase, and overexpression of PNCK with involvement in carcinogenesis have been reported in HER-2 amplified breast cancer, clear cell renal cell carcinoma and nasopharygeal carcinoma. However, the expression of PNCK and its clinical implication have not been elucidated in hepatocellular carcinoma (HCC). MATERIALS AND METHODS: We investigated PNCK expression at both the protein and mRNA level using immunohistochemistry (IHC) and microarray gene expression profiling in HCC tissue samples, and evaluated its association with clinicopathological parameters and their potential prognostic significance. RESULTS: High PNCK protein expression and high PNCK mRNA level was observed in 61.7% and 34.7% of total HCC cases, respectively. PNCK mRNA level was higher in tumor tissues than in background non-tumor tissues, and significantly correlated with protein expression by IHC. High PNCK expression was associated with higher Edmondson grade, intrahepatic metastasis, microvascular invasion and higher AFP levels. Patients with high PNCK expression showed shorter recurrence-free survival and disease-specific survival, and high mRNA expression of PNCK was an independent prognostic factor in disease-specific survival. CONCLUSION: Up-regulation of PNCK expression as well as its association with poor prognosis was demonstrated in HCC. PNCK might be a prognostic biomarker of HCC, and could be a potential candidate therapeutic target.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Carcinoma Hepatocelular/secundario , Neoplasias Hepáticas/patología , Recurrencia Local de Neoplasia/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/cirugía , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/cirugía , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/cirugía , Pronóstico , Tasa de Supervivencia
16.
Cell Rep ; 32(9): 108104, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32877683

RESUMEN

Rare variants in GRIN genes, which encode NMDAR subunits, are strongly associated with neurodevelopmental disorders. Among these, GRIN2A, which encodes the GluN2A subunit of NMDARs, is widely accepted as an epilepsy-causative gene. Here, we functionally characterize the de novo GluN2A-S1459G mutation identified in an epilepsy patient. We show that S1459 is a CaMKIIα phosphorylation site, and that endogenous phosphorylation is regulated during development and in response to synaptic activity in a dark rearing model. GluN2A-S1459 phosphorylation results in preferential binding of NMDARs to SNX27 and a corresponding decrease in PSD-95 binding, which consequently regulates NMDAR trafficking. Furthermore, the epilepsy-associated GluN2A-S1459G variant displays defects in interactions with both SNX27 and PSD-95, resulting in trafficking deficits, reduced spine density, and decreased excitatory synaptic transmission. These data demonstrate a role for CaMKIIα phosphorylation of GluN2A in receptor targeting and implicate NMDAR trafficking defects as a link to epilepsy.


Asunto(s)
Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Epilepsia/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/genética , Epilepsia/genética , Femenino , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética
17.
Biochem Biophys Res Commun ; 530(3): 513-519, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32600616

RESUMEN

Protein phosphatase PPM1H is known to participate in various biological or pathophysiological mechanisms. However, little is known about the molecular mechanisms of its regulation. In this study, we investigated the protein kinases that directly phosphorylate PPM1H, identifying them as cAMP-dependent protein kinase (PKA) and Ca2+/calmodulin-dependent protein kinase I (CaMKI). In vitro and in silico analyses showed that the phosphorylation sites of PPM1H by PKA and CaMKI were Ser-123 and Ser-210, respectively. The phosphorylation state of PPM1H in cells exhibited the kinase activator- and inhibitor-dependent changes. In mouse neuroblastoma Neuro2a cells, phosphorylation of Ser-210 was much higher in the phospho-mimetic mutant (S123D) than in the non-phosphorylatable mutant (S123A) when they were treated with ionomycin. This suggests that a hierarchical phosphorylation, with initial phosphorylation of Ser-123 promoting subsequent phosphorylation of Ser-210, occurs in these neuron-like cells. Moreover, in cell-based assay a PPM1H(S123A/S210A) double mutant barely dephosphorylated Smad1, a transcription factor known as an endogenous substrate of PPM1H. These results suggest that cAMP and Ca2+/calmodulin regulate dephosphorylation of Smad1 through the dual phosphorylation of PPM1H at Ser-123 and Ser-210.


Asunto(s)
Proteína Smad1/metabolismo , Animales , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células HEK293 , Humanos , Ratones , Fosforilación
18.
Cell ; 181(7): 1547-1565.e15, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32492405

RESUMEN

Homeostasis of neural firing properties is important in stabilizing neuronal circuitry, but how such plasticity might depend on alternative splicing is not known. Here we report that chronic inactivity homeostatically increases action potential duration by changing alternative splicing of BK channels; this requires nuclear export of the splicing factor Nova-2. Inactivity and Nova-2 relocation were connected by a novel synapto-nuclear signaling pathway that surprisingly invoked mechanisms akin to Hebbian plasticity: Ca2+-permeable AMPA receptor upregulation, L-type Ca2+ channel activation, enhanced spine Ca2+ transients, nuclear translocation of a CaM shuttle, and nuclear CaMKIV activation. These findings not only uncover commonalities between homeostatic and Hebbian plasticity but also connect homeostatic regulation of synaptic transmission and neuronal excitability. The signaling cascade provides a full-loop mechanism for a classic autoregulatory feedback loop proposed ∼25 years ago. Each element of the loop has been implicated previously in neuropsychiatric disease.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Potenciación a Largo Plazo/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Potenciales de Acción/fisiología , Empalme Alternativo/genética , Empalme Alternativo/fisiología , Animales , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Femenino , Células HEK293 , Homeostasis/fisiología , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/fisiología , Antígeno Ventral Neuro-Oncológico , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Proteínas de Unión al ARN/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
19.
Ann Rheum Dis ; 79(9): 1234-1242, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32546599

RESUMEN

OBJECTIVES: Myositis is a heterogeneous family of diseases that includes dermatomyositis (DM), antisynthetase syndrome (AS), immune-mediated necrotising myopathy (IMNM), inclusion body myositis (IBM), polymyositis and overlap myositis. Additional subtypes of myositis can be defined by the presence of myositis-specific autoantibodies (MSAs). The purpose of this study was to define unique gene expression profiles in muscle biopsies from patients with MSA-positive DM, AS and IMNM as well as IBM. METHODS: RNA-seq was performed on muscle biopsies from 119 myositis patients with IBM or defined MSAs and 20 controls. Machine learning algorithms were trained on transcriptomic data and recursive feature elimination was used to determine which genes were most useful for classifying muscle biopsies into each type and MSA-defined subtype of myositis. RESULTS: The support vector machine learning algorithm classified the muscle biopsies with >90% accuracy. Recursive feature elimination identified genes that are most useful to the machine learning algorithm and that are only overexpressed in one type of myositis. For example, CAMK1G (calcium/calmodulin-dependent protein kinase IG), EGR4 (early growth response protein 4) and CXCL8 (interleukin 8) are highly expressed in AS but not in DM or other types of myositis. Using the same computational approach, we also identified genes that are uniquely overexpressed in different MSA-defined subtypes. These included apolipoprotein A4 (APOA4), which is only expressed in anti-3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) myopathy, and MADCAM1 (mucosal vascular addressin cell adhesion molecule 1), which is only expressed in anti-Mi2-positive DM. CONCLUSIONS: Unique gene expression profiles in muscle biopsies from patients with MSA-defined subtypes of myositis and IBM suggest that different pathological mechanisms underly muscle damage in each of these diseases.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Musculares/genética , Miositis por Cuerpos de Inclusión/genética , Miositis/genética , Adulto , Animales , Apolipoproteínas A/metabolismo , Biopsia , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Moléculas de Adhesión Celular/metabolismo , Técnicas de Cultivo de Célula , Dermatomiositis/genética , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Humanos , Hidroximetilglutaril-CoA Reductasas/metabolismo , Interleucina-8/metabolismo , Aprendizaje Automático , Masculino , Ratones , Mucoproteínas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miositis/patología , Polimiositis/genética , Transcriptoma
20.
Curr Biol ; 29(22): 3887-3898.e4, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31679932

RESUMEN

Critical to evolutionary fitness, animals regulate social behaviors by integrating signals from both their external environments and internal states. Here, we find that population density modulates the courtship behavior of male Drosophila melanogaster in an age-dependent manner. In a competitive mating assay, males reared in a social environment have a marked advantage in courting females when pitted against males reared in isolation. Group housing promotes courtship in mature (7-day) but not immature (2-day) males; this behavioral plasticity requires the Or47b pheromone receptor. Using single-sensillum recordings, we find that group housing increases the response of Or47b olfactory receptor neurons (ORNs) only in mature males. The effect of group housing on olfactory response and behavior can be mimicked by chronically exposing single-housed males to an Or47b ligand. At the molecular level, group housing elevates Ca2+ levels in Or47b ORNs, likely leading to CaMKI-mediated activation of the histone-acetyl transferase CBP. This signaling event in turn enhances the efficacy of juvenile hormone, an age-related regulator of reproductive maturation in flies. Furthermore, the male-specific Fruitless isoform (FruM) is required for the sensory plasticity, suggesting that FruM functions as a downstream genomic coincidence detector in Or47b ORNs-integrating reproductive maturity, signaled by juvenile hormone, and population density, signaled by CBP. In all, we identify a neural substrate and activity-dependent mechanism by which social context can directly influence pheromone sensitivity, thereby modulating social behavior according to animals' life-history stage.


Asunto(s)
Feromonas/metabolismo , Conducta Sexual Animal/fisiología , Factores de Edad , Animales , Conducta Animal/fisiología , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Copulación/fisiología , Cortejo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Femenino , Histona Acetiltransferasas/metabolismo , Masculino , Proteínas del Tejido Nervioso/genética , Neuronas Receptoras Olfatorias/fisiología , Feromonas/fisiología , Densidad de Población , Isoformas de Proteínas , Olfato/fisiología , Conducta Social , Medio Social , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...