Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Comput Math Methods Med ; 2022: 7187940, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309839

RESUMEN

Objective: To explore the functional role of Calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) in the progression of ovarian carcinoma (OC). Methods: RT-qPCR analysis and western blot were conducted to detect the mRNA and protein expression of CaMKK2, PI3K, PDK1 and Akt in OC tissues and cells, respectively. CCK-8 assay, transwell migration assay and flow cytometry were used to measure cell proliferation, migration and apoptosis, respectively. Results: CaMKK2, PI3K, PDK1 and Akt were highly expressed in OC tissues compared with the corresponding controls. CaMKK2 knockdown significantly suppressed the mRNA and protein expression of PI3K, PDK1 and Akt in HO8910 and OV90 cells. Moreover, CaMKK2 knockdown could dramatically repress cell proliferation, migration, and markedly elevate cell apoptosis in HO8910 and OV90 cells. Conclusions: CaMKK2 played a promotion role in OC progression via activating the PI3K/PDK1/Akt axis.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Neoplasias Ováricas/enzimología , Apoptosis/fisiología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Biología Computacional , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
2.
Osteoarthritis Cartilage ; 30(1): 124-136, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34506942

RESUMEN

OBJECTIVE: To investigate the role of Ca2+/calmodulin-dependent protein kinase 2 (CaMKK2) in post-traumatic osteoarthritis (PTOA). METHODS: Destabilization of the medial meniscus (DMM) or sham surgeries were performed on 10-week-old male wild-type (WT) and Camkk2-/- mice. Half of the DMM-WT mice and all other cohorts (n = 6/group) received tri-weekly intraperitoneal (i.p.) injections of saline whereas the remaining DMM-WT mice (n = 6/group) received i.p. injections of the CaMKK2 inhibitor STO-609 (0.033 mg/kg body weight) thrice a week. Study was terminated at 8- or 12-weeks post-surgery, and knee joints processed for microcomputed tomography imaging followed by histology and immunohistochemistry. Primary articular chondrocytes were isolated from knee joints of 4-6-day-old WT and Camkk2-/- mice, and treated with 10 ng/ml interleukin-1ß (IL)-1ß for 24 or 48 h to investigate gene and protein expression. RESULTS: CaMKK2 levels and activity became elevated in articular chondrocytes following IL-1ß treatment or DMM surgery. Inhibition or absence of CaMKK2 protected against DMM-associated destruction of the cartilage, subchondral bone alterations and synovial inflammation. When challenged with IL-1ß, chondrocytes lacking CaMKK2 displayed attenuated inflammation, cartilage catabolism, and resistance to suppression of matrix synthesis. IL-1ß-treated CaMKK2-null chondrocytes displayed decreased IL-6 production, activation of signal transducer and activator of transcription 3 (Stat3) and matrix metalloproteinase 13 (MMP13), indicating a potential mechanism for the regulation of inflammatory responses in chondrocytes by CaMKK2. CONCLUSIONS: Our findings reveal a novel function for CaMKK2 in chondrocytes and highlight the potential for its inhibition as an innovative therapeutic strategy in the prevention of PTOA.


Asunto(s)
Bencimidazoles/uso terapéutico , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Cartílago Articular/lesiones , Naftalimidas/uso terapéutico , Osteoartritis/etiología , Osteoartritis/prevención & control , Animales , Masculino , Ratones , Heridas y Lesiones/complicaciones
3.
Mol Carcinog ; 60(11): 769-783, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34437731

RESUMEN

Although CAMKK2 is overexpressed in several cancers, its role and relevant downstream signaling pathways in gastric cancer (GC) are poorly understood. Treatment of AGS GC cells with a CAMKK2 inhibitor, STO-609, resulted in decreased cell proliferation, cell migration, invasion, colony-forming ability, and G1/S-phase arrest. Quantitative phosphoproteomics in AGS cells with the CAMKK2 inhibitor led to the identification of 9603 unique phosphosites mapping to 3120 proteins. We observed decreased phosphorylation of 1101 phosphopeptides (1.5-fold) corresponding to 752 proteins upon CAMKK2 inhibition. Bioinformatics analysis of hypo-phosphorylated proteins revealed enrichment of MAPK1/MAPK3 signaling. Kinase enrichment analysis of hypo-phosphorylated proteins using the X2K Web tool identified ERK1, cyclin-dependant kinase 1 (CDK1), and CDK2 as downstream substrates of CAMKK2. Moreover, inhibition of CAMKK2 and MEK1 resulted in decreased phosphorylation of ERK1, CDK1, MCM2, and MCM3. Immunofluorescence results were in concordance with our mass spectroscopy data and Western blot analysis results. Taken together, our data reveal the essential role of CAMKK2 in the pathobiology of GC through the activation of the MEK/ERK1 signaling cascade.


Asunto(s)
Bencimidazoles/farmacología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Naftalimidas/farmacología , Proteómica/métodos , Neoplasias Gástricas/metabolismo , Proteína Quinasa CDC2/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cromatografía Liquida , Quinasa 2 Dependiente de la Ciclina/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Espectrometría de Masas en Tándem
4.
J Med Chem ; 64(15): 10849-10877, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34264658

RESUMEN

CAMKK2 is a serine/threonine kinase and an activator of AMPK whose dysregulation is linked with multiple diseases. Unfortunately, STO-609, the tool inhibitor commonly used to probe CAMKK2 signaling, has limitations. To identify promising scaffolds as starting points for the development of high-quality CAMKK2 chemical probes, we utilized a hinge-binding scaffold hopping strategy to design new CAMKK2 inhibitors. Starting from the potent but promiscuous disubstituted 7-azaindole GSK650934, a total of 32 compounds, composed of single-ring, 5,6-, and 6,6-fused heteroaromatic cores, were synthesized. The compound set was specifically designed to probe interactions with the kinase hinge-binding residues. Compared to GSK650394 and STO-609, 13 compounds displayed similar or better CAMKK2 inhibitory potency in vitro, while compounds 13g and 45 had improved selectivity for CAMKK2 across the kinome. Our systematic survey of hinge-binding chemotypes identified several potent and selective inhibitors of CAMKK2 to serve as starting points for medicinal chemistry programs.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Calcio/farmacología , Calmodulina/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Calcio/química , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Calmodulina/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
5.
Oncol Rep ; 44(5): 2288-2296, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33000264

RESUMEN

Triptolide, a triterpene extracted from the Chinese herb Tripterygium wilfordii, has been reported to exert multiple bioactivities, including immunosuppressive, anti­inflammatory and anticancer effects. Although the anticancer effect of triptolide has attracted significant attention, the specific anticancer mechanism in non­small­cell lung cancer (NSCLC) remains unclear. The present study aimed to investigate the anticancer effect of triptolide in the H1395 NSCLC cell line and to determine its mechanism of action. The results revealed that triptolide significantly inhibited the cell viability of NSCLC cells in a dose­dependent manner, which was suggested to be through inducing apoptosis. In addition, triptolide was revealed to activate the calcium (Ca2+)/calmodulin­dependent protein kinase kinase ß (CaMKKß)/AMP­activated protein kinase (AMPK) signaling pathway by regulating the intracellular Ca2+ concentration levels, which increased the phosphorylation levels of AMPK and reduced the phosphorylation levels of AKT, ultimately leading to apoptosis. The CaMKKß blocker STO­609 and the AMPK blocker Compound C significantly inhibited the apoptosis­promoting effect of triptolide. In conclusion, the results of the present study suggested that triptolide may induce apoptosis through the CaMKKß­AMPK signaling pathway and may be a promising drug for the treatment of NSCLC.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Diterpenos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Fenantrenos/farmacología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Bencimidazoles/farmacología , Calcio/análisis , Calcio/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Diterpenos/uso terapéutico , Compuestos Epoxi/farmacología , Compuestos Epoxi/uso terapéutico , Humanos , Neoplasias Pulmonares/patología , Naftalimidas/farmacología , Fenantrenos/uso terapéutico , Fosforilación/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos
6.
PLoS One ; 15(10): e0240230, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33119597

RESUMEN

OBJECTIVE: Our study is to identify DEGs (Differentially Expressed Genes), comprehensively investigate hub genes, annotate enrichment functions and key pathways of Non-functional pituitary adenomas (NFPAs), and also to verify STO-609 therapeutic effect. METHODS: The gene expression level of NFPA and normal tissues were compared to identify the DEGs (Differential expressed genes) based on gene expression profiles (GSE2175, GSE26966 and GSE51618). Enrichment functions, pathways and key genes were identified by carrying out GO (Gene Ontology), KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis and PPI (Protein-Protein Interation) network analysis. Moreover, experiments in vitro were conducted to verify the anti-NFPAs effects of STO-609. RESULTS: 169 over-expression genes and 182 low expression genes were identified among 3 datasets. Dopaminergic synapse and vibrio cholerae infection pathways have distinctly changed in NFPA tissues. The Ca2+/CaM pathway played important roles in NFPA. Four hub proteins encoded by genes CALM1, PRDM10, RIPK4 and MAD2L1 were recognized as hub proteins. In vitro, assays showed that STO-609 induced apoptosis of NFPA cells to inhibit the hypophysoma cellular viability, diffusion and migration. CONCLUSION: Four hub proteins, encoded by gene CALM1, PRDM10, RIPK4 and MAD2L1, played important roles in NFPA development. The Ca2+/CaM signaling pathway had significant alternations during NFPA forming process, the STO-609, a selective CaM-KK inhibitor, inhibited NFPA cellular viability, proliferation and migration. Meanwhile, NFPA was closely related to parkinson's disease (PD) in many aspects.


Asunto(s)
Antineoplásicos/uso terapéutico , Bencimidazoles/uso terapéutico , Naftalimidas/uso terapéutico , Neoplasias Hipofisarias/tratamiento farmacológico , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Línea Celular Tumoral , Biología Computacional , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Ratones , Análisis por Micromatrices , Neoplasias Hipofisarias/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal
7.
Biochemistry ; 59(17): 1701-1710, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32298102

RESUMEN

Ca2+/calmodulin-dependent protein kinase kinase (CaMKK) activates particular multifunctional kinases, including CaMKI, CaMKIV, and 5'AMP-activated protein kinase (AMPK), resulting in the regulation of various Ca2+-dependent cellular processes, including neuronal, metabolic, and pathophysiological pathways. We developed and characterized a novel pan-CaMKK inhibitor, TIM-063 (2-hydroxy-3-nitro-7H-benzo[de]benzo[4,5]imidazo[2,1-a]isoquinolin-7-one) derived from STO-609 (7H-benzimidazo[2,1-a]benz[de]isoquinoline-7-one-3-carboxylic acid), and an inactive analogue (TIM-062) as molecular probes for the analysis of CaMKK-mediated cellular responses. Unlike STO-609, TIM-063 had an inhibitory activity against CaMKK isoforms (CaMKKα and CaMKKß) with a similar potency (Ki = 0.35 µM for CaMKKα, and Ki = 0.2 µM for CaMKKß) in vitro. Two TIM-063 analogues lacking a nitro group (TIM-062) or a hydroxy group (TIM-064) completely impaired CaMKK inhibitory activities, indicating that both substituents are necessary for the CaMKK inhibitory activity of TIM-063. Enzymatic analysis revealed that TIM-063 is an ATP-competitive inhibitor that directly targets the catalytic domain of CaMKK, similar to STO-609. TIM-063 suppressed the ionomycin-induced phosphorylation of exogenously expressed CaMKI, CaMKIV, and endogenous AMPKα in HeLa cells with an IC50 of ∼0.3 µM, and it suppressed CaMKK isoform-mediated CaMKIV phosphorylation in transfected COS-7 cells. Thus, TIM-063, but not the inactive analogue (TIM-062), displayed cell permeability and the ability to inhibit CaMKK activity in cells. Taken together, these results indicate that TIM-063 could be a useful tool for the precise analysis of CaMKK-mediated signaling pathways and may be a promising lead compound for the development of therapeutic agents for the treatment of CaMKK-related diseases.


Asunto(s)
Bencimidazoles/química , Bencimidazoles/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Naftalimidas/química , Naftalimidas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Células COS , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Chlorocebus aethiops , Células HeLa , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología
8.
Molecules ; 25(2)2020 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-31941153

RESUMEN

The calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) activates CAMK1, CAMK4, AMPK, and AKT, leading to numerous physiological responses. The deregulation of CAMKK2 is linked to several diseases, suggesting the utility of CAMKK2 inhibitors for oncological, metabolic and inflammatory indications. In this work, we demonstrate that STO-609, frequently described as a selective inhibitor for CAMKK2, potently inhibits a significant number of other kinases. Through an analysis of literature and public databases, we have identified other potent CAMKK2 inhibitors and verified their activities in differential scanning fluorimetry and enzyme inhibition assays. These inhibitors are potential starting points for the development of selective CAMKK2 inhibitors and will lead to tools that delineate the roles of this kinase in disease biology.


Asunto(s)
Bencimidazoles/química , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina , Naftalimidas/química , Inhibidores de Proteínas Quinasas/química , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/química , Humanos
9.
Sci Rep ; 9(1): 16452, 2019 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-31712618

RESUMEN

Calcium/Calmodulin-dependent Protein Kinase Kinase 2 (CAMKK2) acts as a signaling hub, receiving signals from various regulatory pathways and decoding them via phosphorylation of downstream protein kinases - such as AMPK (AMP-activated protein kinase) and CAMK types I and IV. CAMKK2 relevance is highlighted by its constitutive activity being implicated in several human pathologies. However, at present, there are no selective small-molecule inhibitors available for this protein kinase. Moreover, CAMKK2 and its closest human homolog, CAMKK1, are thought to have overlapping biological roles. Here we present six new co-structures of potent ligands bound to CAMKK2 identified from a library of commercially-available kinase inhibitors. Enzyme assays confirmed that most of these compounds are equipotent inhibitors of both human CAMKKs and isothermal titration calorimetry (ITC) revealed that binding to some of these molecules to CAMKK2 is enthalpy driven. We expect our results to advance current efforts to discover small molecule kinase inhibitors selective to each human CAMKK.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/química , Inhibidores de Proteínas Quinasas/química , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Descubrimiento de Drogas , Humanos , Ligandos , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Recombinantes , Relación Estructura-Actividad
10.
Int J Mol Sci ; 20(11)2019 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-31146414

RESUMEN

AMP-activated protein kinase (AMPK) has been implicated in contractility changes in bladders with partial bladder outlet obstruction (PBOO), but the role of AMPK in the contractile response of normal bladder remains unclear. We investigated the phosphorylation of AMPKα and expression of the involved upstream AMPK kinases (AMPKKs) in a model of bladders with PBOO and sought to determine whether the pharmacological inhibition of these two factors affected detrusor contractility in normal bladders, using female Sprague-Dawley rats. Cystometry and Western blot analysis were performed in rats that were subjected to PBOO induction or a sham operation. Cystometry was performed in normal rats that received selective inhibitors of AMPKα and Ca2+/calmodulin-dependent protein kinase kinase (CaMKKß) (compound C and STO-609, respectively) at doses determined in the experiments. In the PBOO bladders, bladder weight and micturition pressure (MP) were higher and AMPKα phosphorylation (T172) and CaMKKß expression was significantly reduced. Compound C and STO-609 increased MP. The increased contractile response in bladders with PBOO-induced hypertrophy was related to decreased CaMKKß/AMPK signaling activity, and the pharmacological inhibition of this pathway in normal bladders increased detrusor contractility, implying a role of CaMKKß/AMPK signaling in the bladder in the regulation of detrusor contractility.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Contracción Muscular , Proteínas Quinasas/metabolismo , Obstrucción del Cuello de la Vejiga Urinaria/metabolismo , Vejiga Urinaria/metabolismo , Micción , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Femenino , Naftalimidas/farmacología , Naftalimidas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/fisiología , Vejiga Urinaria/fisiopatología , Obstrucción del Cuello de la Vejiga Urinaria/tratamiento farmacológico
11.
Nat Commun ; 10(1): 2450, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31164648

RESUMEN

Tumor-associated myeloid cells regulate tumor growth and metastasis, and their accumulation is a negative prognostic factor for breast cancer. Here we find calcium/calmodulin-dependent kinase kinase (CaMKK2) to be highly expressed within intratumoral myeloid cells in mouse models of breast cancer, and demonstrate that its inhibition within myeloid cells suppresses tumor growth by increasing intratumoral accumulation of effector CD8+ T cells and immune-stimulatory myeloid subsets. Tumor-associated macrophages (TAMs) isolated from Camkk2-/- mice expressed higher levels of chemokines involved in the recruitment of effector T cells compared to WT. Similarly, in vitro generated Camkk2-/- macrophages recruit more T cells, and have a reduced capability to suppress T cell proliferation, compared to WT. Treatment with CaMKK2 inhibitors blocks tumor growth in a CD8+ T cell-dependent manner, and facilitates a favorable reprogramming of the immune cell microenvironment. These data, credential CaMKK2 as a myeloid-selective checkpoint, the inhibition of which may have utility in the immunotherapy of breast cancer.


Asunto(s)
Neoplasias de la Mama/inmunología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/inmunología , Carcinoma/inmunología , Neoplasias Mamarias Animales/inmunología , Células Mieloides/inmunología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Linfocitos T CD8-positivos/inmunología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Proliferación Celular , Quimiocinas/inmunología , Femenino , Humanos , Inmunohistoquímica , Técnicas In Vitro , Macrófagos/inmunología , Macrófagos/metabolismo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Escape del Tumor/genética
12.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1627-1641, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30946956

RESUMEN

Lipopolysaccharide (LPS), an essential component of outer membrane of the Gram-negative bacteria, plays a pivotal role in myocardial anomalies in sepsis. Recent evidence depicted an essential role for mitochondrial aldehyde dehydrogenase (ALDH2) in cardiac homeostasis. This study examined the effect of ALDH2 on endotoxemia-induced cardiac anomalies. Echocardiographic, cardiac contractile and intracellular Ca2+ properties were examined. Our results indicated that LPS impaired cardiac contractile function (reduced fractional shortening, LV end systolic diameter, peak shortening, maximal velocity of shortening/relengthening, prolonged relengthening duration, oxidation of SERCA, and intracellular Ca2+ mishandling), associated with ER stress, inflammation, O2- production, increased autophagy, CAMKKß, phosphorylated AMPK and suppressed phosphorylation of mTOR, the effects of which were significantly attenuated or negated by ALDH2. LPS promoted early endosomal formation (as evidenced by RAB4 and RAB5a), apoptosis and necrosis (MTT and LDH) while decreasing late endosomal formation (RAB7 and RAB 9), the effects were reversed by ALDH2. In vitro study revealed that LPS-induced SERCA oxidation, autophagy and cardiac dysfunction were abrogated by ALDH2 activator Alda-1, the ER chaperone TUDCA, the autophagy inhibitor 3-MA, or the AMPK inhibitor Compound C. The beneficial effect of Alda-1 against LPS was nullified by AMPK activator AICAR or rapamycin. CAMKKß inhibition failed to rescue LPS-induced ER stress. Tunicamycin-induced cardiomyocyte dysfunction was ameliorated by Alda-1 and autophagy inhibition, the effect of which was abolished by rapamycin. These data suggested that ALDH2 protected against LPS-induced cardiac anomalies via suppression of ER stress, autophagy in a CAMKKß/AMPK/mTOR-dependent manner.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/genética , Calcio/metabolismo , Cardiomiopatías/genética , Lipopolisacáridos/farmacología , Mitocondrias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Aldehído Deshidrogenasa Mitocondrial/antagonistas & inhibidores , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Benzamidas/farmacología , Benzodioxoles/farmacología , Señalización del Calcio , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Cardiomiopatías/inducido químicamente , Cardiomiopatías/enzimología , Cardiomiopatías/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/enzimología , Mitocondrias/patología , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/genética , Miocardio/enzimología , Miocardio/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Cultivo Primario de Células , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Ácido Tauroquenodesoxicólico/farmacología , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab4/genética , Proteínas de Unión al GTP rab4/metabolismo , Proteínas de Unión a GTP rab7
13.
Int J Mol Sci ; 20(9)2019 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-31027360

RESUMEN

Neonatal hypoxia-ischemia (HI) is a major cause of death and disability in neonates. HI leads to a dramatic rise in intracellular calcium levels, which was originally thought to be detrimental to the brain. However, it has been increasingly recognized that this calcium signaling may also play an important protective role after injury by triggering endogenous neuroprotective pathways. Calcium/calmodulin-dependent protein kinase kinase ß (CaMKK ß) is a major kinase activated by elevated levels of intracellular calcium. Here we evaluated the functional role of CaMKK ß in neonatal mice after HI in both acute and chronic survival experiments. Postnatal day ten wild-type (WT) and CaMKK ß knockout (KO) mouse male pups were subjected to unilateral carotid artery ligation, followed by 40 min of hypoxia (10% O2 in N2). STO-609, a CaMKK inhibitor, was administered intraperitoneally to WT mice at 5 minutes after HI. TTC (2,3,5-triphenyltetrazolium chloride monohydrate) staining was used to assess infarct volume 24 h after HI. CaMKK ß KO mice had larger infarct volume than WT mice and STO-609 increased the infarct volume in WT mice after HI. In chronic survival experiments, WT mice treated with STO-609 showed increased tissue loss in the ipsilateral hemisphere three weeks after HI. Furthermore, when compared with vehicle-treated mice, they showed poorer functional recovery during the three week survival period, as measured by the wire hang test and corner test. Loss of blood-brain barrier proteins, a reduction in survival protein (Bcl-2), and an increase in pro-apoptotic protein Bax were also seen after HI with CaMKK ß inhibition. In conclusion, inhibition of CaMKK ß exacerbated neonatal hypoxia-ischemia injury in mice. Our data suggests that enhancing CaMKK signaling could be a potential target for the treatment of hypoxic-ischemic brain injury.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Hipoxia-Isquemia Encefálica/enzimología , Hipoxia-Isquemia Encefálica/patología , Animales , Animales Recién Nacidos , Bencimidazoles/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Western Blotting , Muerte Celular/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Naftalimidas/farmacología
14.
PLoS One ; 14(1): e0211420, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30695053

RESUMEN

Capsaicin is a natural compound present in chili and red peppers and the responsible of their spicy flavor. It has recently provoked interest because of its antitumoral effects in many cell types although its action mechanism is not clearly understood. As metabolic dysregulation is one of the hallmarks of cancer cells and the key metabolic sensor in the AMP-activated kinase (AMPK), in this study we explored the ability of capsaicin to modulate AMPK activity. We found that capsaicin activated AMPK in HepG2 cells by increasing AMPK phosphorylation and its downstream target ACC. Mechanistically, we determined that capsaicin activated AMPK through the calcium/calmodulin-dependent protein kinase kinase ß, CaMKKß as either the CaMKK inhibitor STO-609 or CaMKK knock down with siRNA abrogated the activation of AMPK. Moreover, capsaicin decreased cell viability, inhibited Akt/mTOR pathway and increased reactive oxygen species (ROS) in HepG2 cells. AMPK activation was involved in the underpinning mechanism of capsaicin-induced cell death.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Calcio/metabolismo , Capsaicina/farmacología , Capsicum/química , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Fármacos del Sistema Sensorial/farmacología , Bencimidazoles/farmacología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Supervivencia Celular , Activación Enzimática , Células Hep G2 , Humanos , Naftalimidas/farmacología , Fosforilación , Transducción de Señal
15.
Eur J Neurosci ; 49(1): 27-39, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30422362

RESUMEN

Brain microvascular endothelial cells play an essential role in maintaining blood-brain barrier (BBB) integrity, and disruption of the BBB aggravates the ischemic injury. CaMKK (α and ß) is a major kinase activated by elevated intracellular calcium. Previously, we demonstrated that inhibition of CaMKK exacerbated outcomes, conversely, overexpression reduced brain injury after stroke in mice. Interestingly, CaMKK has been shown to activate a key endothelial protector, sirtuin 1 (SIRT1). We hypothesized that CaMKK protects brain endothelial cells via SIRT1 activation after stroke. In this study, Oxygen-Glucose Deprivation (OGD) was performed in human brain microvascular endothelial cells. Stroke was induced by middle cerebral artery occlusion (MCAO) in male mice. Knockdown of CaMKK ß using siRNA increased cell death following OGD. Inhibition of CaMKK ß by STO-609 significantly and selectively down-regulated levels of phosphorylated SIRT1 after OGD. Changes in the downstream targets of SIRT1 were observed following STO-609 treatment. The effect of STO-609 on cell viability after OGD was absent, when SIRT1 was concurrently inhibited. We also demonstrated that STO-609 increased endothelial expression of the pro-inflammatory proteins ICAM-1 and VCAM-1 and inhibition of CaMKK exacerbated OGD-induced leukocyte-endothelial adhesion. Finally, intracerebroventricular injection of STO-609 exacerbated endothelial apoptosis and reduced BBB integrity after 24-hr reperfusion following MCAO in vivo. Collectively, these results demonstrated that CaMKK inhibition reduced endothelial cell viability, exacerbated inflammatory responses and aggravated BBB impairment after ischemia. CaMKK activation may attenuate ischemic brain injury via protection of the microvascular system and a reduction in the infiltration of pro-inflammatory factors.


Asunto(s)
Barrera Hematoencefálica/enzimología , Isquemia Encefálica/enzimología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Células Endoteliales/enzimología , Accidente Cerebrovascular/enzimología , Animales , Bencimidazoles/farmacología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Muerte Celular , Células Cultivadas , Humanos , Masculino , Ratones Endogámicos C57BL , Naftalimidas/farmacología , Sirtuina 1/metabolismo
16.
Sci Rep ; 8(1): 14800, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30287839

RESUMEN

The calcium/calmodulin-dependent protein kinases (CAMKKs) are upstream activators of CAMK1 and CAMK4 signalling and have important functions in neural development, maintenance and signalling, as well as in other aspects of biology such as Ca2+ signalling in the cardiovascular system. To support the development of specific inhibitors of CAMKKs we have determined the crystal structure of CAMKK1 with two ATP-competitive inhibitors. The structures reveal small but exploitable differences between CAMKK1 and CAMKK2, despite the high sequence identity, which could be used in the generation of specific inhibitors. Screening of a kinase inhibitor library revealed molecules that bind potently to CAMKK1. Isothermal titration calorimetry revealed that the most potent inhibitors had binding energies largely dependent on favourable enthalpy. Together, the data provide a foundation for future inhibitor development activities.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/química , Diseño de Fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Calorimetría , Dominio Catalítico , Evaluación Preclínica de Medicamentos , Hesperidina/química , Hesperidina/farmacología , Humanos , Estructura Secundaria de Proteína
17.
Virol J ; 15(1): 120, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-30081955

RESUMEN

BACKGROUND: The results of our previous study showed that impaired cellular energy metabolism contributes to duck enteritis virus-induced autophagy via the 5`-adenosine monophosphate-activated protein kinase (AMPK)/tuberous sclerosis complex 2/mammalian target of rapamycin pathway in duck embryo fibroblast (DEF) cells. However, it remains unknown whether any other underlying mechanisms of AMPK activation are involved in autophagy induction. METHODS: The activity of CaMKKß and AMPK in DEF cells infected with DEV were evaluated.The Effect of inhibitory activity of CaMKKß on DEV-induced autophagy was investigated. In addtion to, the cytosolic calcium level in DEF cells infected with DEV were evaluated.The Effect of inhibitory cytosolic calcium level on DEV-induced autophagy was investigated. RESULTS: In this study, duck enteritis virus (DEV) infection activated CaMKKß and its substrate molecule AMPK at 36, 48, and 60 h post-infection (hpi). STO-609, a CaMKKß inhibitor, or CaMKKß siRNA significantly inhibited the activation of DEV to AMPK, LC3I to LC3II transformation, and GFP-LC3 puncta distribution. In addition, inhibition of CaMKKß activity also significantly reduced progeny DEV titer and gB protein expression. Besides, cytosolic calcium (Ca2+) was higher in DEV-infected cells than mock controls at 36, 48, and 60 hpi, respectively. Treatment of DEV-infected cells with 1,2-Bis (2-aminophenoxy) ethane-N, N, N', N-tetraacetic acid (BAPTA-AM) significantly reduced intracellular Ca2+ ion concentrations, as well as CaMKKß and AMPK activities, and subsequent autophagy, in addition to viral protein synthesis and viral titer. CONCLUSIONS: These results showed that elevated [Ca2+]cyto-mediated activation of CaMKKß managed the activation of AMPK, which then positively regulated autophagy, thereby providing further insight into DEV-host interactions.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Calcio/metabolismo , Mardivirus/fisiología , Transducción de Señal , Animales , Antígenos Virales/genética , Autofagia/efectos de los fármacos , Autofagia/fisiología , Bencimidazoles/farmacología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Células Cultivadas , Citosol/metabolismo , Patos , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Fibroblastos/patología , Fibroblastos/virología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno , Naftalimidas/farmacología , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas del Envoltorio Viral/genética , Proteínas Virales/genética
18.
Biochim Biophys Acta Gen Subj ; 1862(10): 2304-2313, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30053538

RESUMEN

BACKGROUND: Calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2), a member of the Ca⁠2+/calmodulin-dependent kinase (CaMK) family, functions as an upstream activator of CaMKI, CaMKIV and AMP-activated protein kinase. Thus, CaMKK2 is involved in the regulation of several key physiological and pathophysiological processes. Previous studies have suggested that Ca2+/CaM binding may cause unique conformational changes in the CaMKKs compared with other CaMKs. However, the underlying mechanistic details remain unclear. METHODS: In this study, hydrogen-deuterium exchange coupled to mass spectrometry, time-resolved fluorescence spectroscopy, small-angle x-ray scattering and chemical cross-linking were used to characterize Ca2+/CaM binding-induced structural changes in CaMKK2. RESULTS: Our data suggest that: (i) the CaMKK2 kinase domain interacts with the autoinhibitory region (AID) through the N-terminal lobe of the kinase domain including the RP insert, a segment important for targeting downstream substrate kinases; (ii) Ca2+/CaM binding affects the structure of several regions surrounding the ATP-binding pocket, including the activation segment; (iii) although the CaMKK2:Ca2+/CaM complex shows high conformational flexibility, most of its molecules are rather compact; and (iv) AID-bound Ca2+/CaM transiently interacts with the CaMKK2 kinase domain. CONCLUSIONS: Interactions between the CaMKK2 kinase domain and the AID differ from those of other CaMKs. In the absence of Ca2+/CaM binding the autoinhibitory region inhibits CaMKK2 by both blocking access to the RP insert and by affecting the structure of the ATP-binding pocket. GENERAL SIGNIFICANCE: Our results corroborate the hypothesis that Ca2+/CaM binding causes unique conformational changes in the CaMKKs relative to other CaMKs.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/química , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Calcio/metabolismo , Calmodulina/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Humanos , Modelos Moleculares , Fosforilación , Unión Proteica , Conformación Proteica , Dominios Proteicos
19.
Molecules ; 23(5)2018 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-29783765

RESUMEN

We demonstrate for the first time that 4H-1,2,6-thiadiazin-4-one (TDZ) can function as a chemotype for the design of ATP-competitive kinase inhibitors. Using insights from a co-crystal structure of a 3,5-bis(arylamino)-4H-1,2,6-thiadiazin-4-one bound to calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2), several analogues were identified with micromolar activity through targeted displacement of bound water molecules in the active site. Since the TDZ analogues showed reduced promiscuity compared to their 2,4-dianilinopyrimidine counter parts, they represent starting points for development of highly selective kinase inhibitors.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Tiadiazoles/síntesis química , Tiadiazoles/farmacología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/química , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Tiadiazoles/química , Agua/química
20.
Bioorg Med Chem Lett ; 28(10): 1958-1963, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29653895

RESUMEN

Hypothalamic CAMKK2 represents a potential mechanism for chemically affecting satiety and promoting weight loss in clinically obese patients. Single-digit nanomolar inhibitors of CAMKK2 were identified in three related ATP-competitive series. Limited optimization of kinase selectivity, solubility, and pharmacokinetic properties were undertaken on all three series, as SAR was often transferrable. Ultimately, a 2,4-diaryl 7-azaindole was optimized to afford a tool molecule that potently inhibits AMPK phosphorylation in a hypothalamus-derived cell line, is orally bioavailable, and crosses the blood-brain barrier. When dosed orally in rodents, compound 4 t limited ghrelin-induced food intake.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Administración Oral , Animales , Encéfalo/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ghrelina/farmacología , Enlace de Hidrógeno , Indoles/química , Indoles/metabolismo , Concentración 50 Inhibidora , Ratones , Mutagénesis , Inhibidores de Proteínas Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA