Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
Front Immunol ; 12: 754083, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712241

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a hetero geneous group of cells, which can suppress the immune response, promote tumor progression and impair the efficacy of immunotherapies. Consequently, the pharmacological targeting of MDSC is emerging as a new immunotherapeutic strategy to stimulate the natural anti-tumor immune response and potentiate the efficacy of immunotherapies. Herein, we leveraged genetically modified models and a small molecule inhibitor to validate Calcium-Calmodulin Kinase Kinase 2 (CaMKK2) as a druggable target to control MDSC accumulation in tumor-bearing mice. The results indicated that deletion of CaMKK2 in the host attenuated the growth of engrafted tumor cells, and this phenomenon was associated with increased antitumor T cell response and decreased accumulation of MDSC. The adoptive transfer of MDSC was sufficient to restore the ability of the tumor to grow in Camkk2-/- mice, confirming the key role of MDSC in the mechanism of tumor rejection. In vitro studies indicated that blocking of CaMKK2 is sufficient to impair the yield of MDSC. Surprisingly, MDSC generated from Camkk2-/- bone marrow cells also showed a higher ability to terminally differentiate toward more immunogenic cell types (e.g inflammatory macrophages and dendritic cells) compared to wild type (WT). Higher intracellular levels of reactive oxygen species (ROS) accumulated in Camkk2-/- MDSC, increasing their susceptibility to apoptosis and promoting their terminal differentiation toward more mature myeloid cells. Mechanistic studies indicated that AMP-activated protein kinase (AMPK), which is a known CaMKK2 proximal target controlling the oxidative stress response, fine-tunes ROS accumulation in MDSC. Accordingly, failure to activate the CaMKK2-AMPK axis can account for the elevated ROS levels in Camkk2-/- MDSC. These results highlight CaMKK2 as an important regulator of the MDSC lifecycle, identifying this kinase as a new druggable target to restrain MDSC expansion and enhance the efficacy of anti-tumor immunotherapy.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Células Supresoras de Origen Mieloide/enzimología , Proteínas de Neoplasias/fisiología , Proteínas Quinasas Activadas por AMP/fisiología , Traslado Adoptivo , Animales , Apoptosis , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/deficiencia , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Femenino , Depleción Linfocítica , Linfoma/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Células Supresoras de Origen Mieloide/trasplante , Mielopoyesis , Especies Reactivas de Oxígeno , Microambiente Tumoral
3.
Cell Commun Signal ; 18(1): 80, 2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32460794

RESUMEN

BACKGROUND: Circulatory iron is a hazardous biometal. Therefore, iron is transported in a redox-safe state by a serum glycoprotein - transferrin (TF). Different organs acquire iron from the systemic circulation through a tightly regulated mechanism at the blood-tissue interface which involves receptor-mediated internalization of TF. Thus, abnormal TF trafficking may lead to iron dyshomeostasis associated with several diseases including neurodegeneration. Iron -induced toxicity can cause neuronal damage to iron-sensitive brain regions. Recently, it was discovered that CAMKK2, a calcium (Ca2+)/calmodulin-activated kinase, controls receptor-mediated TF trafficking in mouse tissues, specifically in the brain. The biological function of CAMKK2 is mediated through multiple downstream effectors. Both CAMKK2 and one of its downstream kinase, CAMK4, exhibit overlapping expression in mouse brain. The role of CAMK4 in vesicular transport has been reported and loss of CAMKK2 or CAMK4 leads to cognitive defects in mouse. Therefore, it was hypothesized that CAMKK2-CAMK4 signaling regulates receptor-mediated TF trafficking and iron homeostasis which may be responsible for the neuronal malfunction observed in CAMKK2- or CAMK4-deficient mice. METHODS: CAMK4-/- mouse was used to study tissue-specific turnover of TF, TF-receptor (TFRC) and iron. CRISPR/Cas9-based CAMKK2 and/or CAMK4 deleted human embryonic kidney-derived HEK293 cell clones were used to study the molecular defects in receptor-mediated TF trafficking. Further, a "zero functional G protein" condition in HEK293 cell was exploited to study CAMKK2-CAMK4 signaling-mediated regulation of intracellular Ca2+ homeostasis which was linked to calcium signaling during TF trafficking. RESULTS: Loss of CAMK4 leads to abnormal post-translational modifications (PTMs) and turnover of TF in mouse cerebellum and liver which was associated with iron dyshomeostasis in these tissues. The HEK293 cell-based study revealed that the absence of CAMKK2-CAMK4 signaling altered intracellular Ca2+ homeostasis and lead to abnormal calcium signaling during TF trafficking. Also, CAMKK2-CAMK4 signaling deficiency affected the molecular interaction of TF and TF-receptor-associated protein complexes which indicated a potential failure in the recruitment of interacting proteins due to differential PTMs in TF. CONCLUSION: Overall, this study established a novel mechanistic link between intracellular Ca2+ level, receptor-mediated TF trafficking, and iron homeostasis, all regulated by CAMKK2-CAMK4 signaling. Video Abstract.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/fisiología , Hierro/metabolismo , Transferrina/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Toxicol Lett ; 323: 1-9, 2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-31982503

RESUMEN

Zearalenone (ZEA) is a prevalent non-steroidal estrogenic mycotoxin produced mainly by Fusarium contamination. Our previous study showed that ZEA induces the autophagy of Sertoli cells (SCs). However, the underlying mechanisms are still unknown. Several studies have indicated that the increasing level of cytoplasmic Ca2+ could induce autophagy through CaMKKß and AMPK pathways. Thus in order to investigate the potential mechanism underlying ZEA-induced autophagy, the activity of calmodulin-dependent kinase kinase ß(CaMKKß)and AMP-activated protein kinase (AMPK) signaling pathway in ZEA-infected TM4 cells was studied. In the present study, ZEA activated the CaMKKß and AMPK signaling pathways. The AMPK inhibitor and activator significantly inhibited and stimulated the effect of ZEA on AMPK, the transformation from LC3I to LC3II, and the distribution of LC3 dots. In addition, cytosolic calcium (Ca2+) was increased gradually with the concentration of ZEA. After treatment of ZEA-infected cells with 1, 2-bis (2-aminophenoxy) ethane-N, N, N', N'- tetraacetic acid- tetraac etoxymethyl ester (BAPTA-AM) and 2-aminoethyl diphenylborinate (2-APB), the intracellular concentration of Ca2+ reduced significantly. Also, the activities of CaMKKß and AMPK and subsequent autophagy decreased. Moreover, the antioxidant NAC significantly decreased activities of AMPK and autophagy -related protein. Therefore, it can be speculated that ROS- mediated ER-stress induced by ZEA activates AMPK via Ca2+-CaMKKß leading to autophagy in TM4 cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Autofagia/efectos de los fármacos , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Calcio/fisiología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Zearalenona/toxicidad , Animales , Células Cultivadas , Ratones , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/fisiología , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/fisiología
5.
Mol Nutr Food Res ; 63(22): e1801377, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31454158

RESUMEN

SCOPE: Nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide, defined by hepatic over-accumulation of lipids without significant ethanol consumption. Pharmacological or bioactive food ingredients that suppress hepatic lipid accumulation through AMP-activated protein kinase (AMPK) signaling, which plays a critical role in the regulation of lipid metabolism, are searched. METHODS AND RESULTS: It is found that tomatidine, the aglycone of α-tomatine abundant in green tomatoes, significantly inhibits palmitate-provoked lipid accumulation and stimulates phosphorylation of AMPK and acetyl-CoA carboxylase 1 (ACC1) in human HepG2 hepatocytes. The results also indicate that tomatidine can enhance triglyceride turnover and decline in lipogenesis by upregulating adipose triglyceride lipase (ATGL) and downregulating fatty acid synthase (FAS) via the AMPK signaling-dependent regulation of transcription factors, element-binding protein-1c (SREBP-1c) and forkhead box protein O1 (FoxO1). Furthermore, mechanistic studies demonstrate that tomatidine-stimulated AMPK phosphorylation is due to CaMKKß activation in response to an increase in intracellular Ca2+ concentration. Finally, it is discovered that tomatidine functions as an agonist for vitamin D receptor to elicit AMPK-dependent suppression of lipid accumulation. CONCLUSION: The in vitro study suggests the potential efficacy of tomatidine as a preventive and therapeutic treatment in obesity-related fatty liver diseases.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Hepatocitos/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Palmitatos/farmacología , Receptores de Calcitriol/fisiología , Tomatina/análogos & derivados , Calcio/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Activación Enzimática/efectos de los fármacos , Proteína Forkhead Box O1/genética , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Transducción de Señal/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Tomatina/farmacología
6.
Biochem Pharmacol ; 169: 113613, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31445019

RESUMEN

Previously, we reported that hepatic muscarinic receptors modulate both acute and chronic liver injury, however, the role of muscarinic receptors in fatty liver disease is unclear. We observed in patients who underwent weight loss surgery, a decrease in hepatic expression of M3 muscarinic receptors (M3R). We also observed that fat loading of hepatocytes, increased M3R expression. Based on these observations, we tested the hypothesis that M3R regulate hepatocyte lipid accumulation. Incubation of AML12 hepatocytes with 1 mM oleic acid resulted in lipid accumulation that was significantly reduced by co-treatment with a muscarinic agonist (pilocarpine or carbachol), an effect blocked by atropine (a muscarinic antagonist). Similar treatment of Hepa 1-6 cells, a mouse hepatoblastoma cell line, showed comparable results. In both, control and fat-loaded AML12 cells, pilocarpine induced time-dependent AMPKα phosphorylation and significantly up-regulated lipolytic genes (ACOX1, CPT1, and PPARα). Compound C, a selective and reversible AMPK inhibitor, significantly blunted pilocarpine-mediated reduction of lipid accumulation and pilocarpine-mediated up-regulation of lipolytic genes. BAPTA-AM, a calcium chelator, and STO-609, a calcium/calmodulin-dependent protein kinase kinase inhibitor, attenuated agonist-induced AMPKα phosphorylation. Finally, M3R siRNA attenuated agonist-induced AMPKα phosphorylation as well as agonist-mediated reduction of hepatocyte steatosis. In conclusion, this proof-of-concept study demonstrates that M3R has protective effects against hepatocyte lipid accumulation by activating AMPK pathway and is a potential therapeutic target for non-alcoholic fatty liver disease.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Receptor Muscarínico M3/fisiología , Animales , Células Cultivadas , Humanos , Ratones , PPAR alfa/fisiología , Fosforilación , Receptor Muscarínico M1/fisiología , Transducción de Señal/fisiología
7.
Aging (Albany NY) ; 10(9): 2394-2406, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30227388

RESUMEN

Rolipram is a selective phosphodiesterase 4 (PDE4) inhibitor that exerts a variety of effects, including anti-inflammatory, immunosuppressive, and anti-tumor effects. The aim of this study was to investigate the effect of rolipram on metabolic disorder and its underlying mechanisms. Metabolic disorder was induced in 8-week-old wild type BABL/c mice by administration of D-galactose for 4 weeks. Simultaneously the mice were administered vehicle or rolipram. Alternatively, beginning at 3 or 21 months, the mice were administered db-cAMP for 3 months, with or without a high-fat-diet (HFD) to induce metabolic disorder. In both models, better metabolic function was observed in rolipram-treated mice. Rolipram reduced adipose deposition and inflammation and reserved metabolic disorder. Treatment with rolipram increased the AMPK phosphorylation and SIRT6 levels in the liver and kidney while reducing NF-κB acetylation. In vitro, these effects were blocked by suppression of SIRT6 expression using specific siRNA. Increased cAMP levels reduced excessive adipose deposition, and improved adipose distribution in presenile mice. These findings provide a promising strategy for the treatment of aging-related metabolic dysfunctions and suggest that selective PDE4 inhibitors may be useful agents for the treatment of aging-related metabolic diseases.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Tejido Adiposo/metabolismo , Envejecimiento/metabolismo , Enfermedades Metabólicas/tratamiento farmacológico , Inhibidores de Fosfodiesterasa 4/farmacología , Sirtuinas/fisiología , Células 3T3-L1 , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , AMP Cíclico/análisis , Masculino , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Rolipram/farmacología , Transducción de Señal/efectos de los fármacos , Sirtuinas/análisis
8.
Int J Cancer ; 136(6): 1434-44, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25080865

RESUMEN

The multikinase inhibitor sorafenib is under clinical investigation for the treatment of many solid tumors, but in most cases, the molecular target responsible for the clinical effect is unknown. Furthermore, enhancing the effectiveness of sorafenib using combination strategies is a major clinical challenge. Here, we identify sorafenib as an activator of AMP-activated protein kinase (AMPK), in a manner that involves either upstream LKB1 or CAMKK2. We further show in a phase II clinical trial in KRAS mutant advanced non-small cell lung cancer (NSCLC) with single agent sorafenib an improved disease control rate in patients using the antidiabetic drug metformin. Consistent with this, sorafenib and metformin act synergistically in inhibiting cellular proliferation in NSCLC in vitro and in vivo. A synergistic effect of both drugs is also seen on phosphorylation of the AMPKα activation site. Our results provide a rationale for the synergistic antiproliferative effects, given that AMPK inhibits downstream mTOR signaling. These data suggest that the combination of sorafenib with AMPK activators could have beneficial effects on tumor regression by AMPK pathway activation. The combination of metformin or other AMPK activators and sorafenib could be tested in prospective clinical trials.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/congénito , Neoplasias Pulmonares/tratamiento farmacológico , Metformina/farmacología , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Transducción de Señal , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Mutación , Niacinamida/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Sorafenib , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/genética
9.
J Neurochem ; 126(5): 565-78, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23692284

RESUMEN

We have investigated the mechanisms underlying the facilitatory modulation mediated by kainate receptor (KAR) activation in the cortex, using isolated nerve terminals (synaptosomes) and slice preparations. In cortical nerve terminals, kainate (KA, 100 µM) produced an increase in 4-aminopyridine (4-AP)-evoked glutamate release. In thalamocortical slices, KA (1 µM) produced an increase in the amplitude of evoked excitatory post-synaptic currents (eEPSCs) at synapses established between thalamic axon terminals from the ventrobasal nucleus onto stellate neurons of L4 of the somatosensory cortex. In both, synaptosomes and slices, the effect of KA was antagonized by 6-cyano-7-nitroquinoxaline-2,3-dione, and persisted after pre-treatment with a cocktail of antagonists of other receptors whose activation could potentially have produced facilitation of release indirectly. Mechanistically, the observed effects of KA appear to be congruent in synaptosomal and slice preparations. Thus, the facilitation by KA of synaptosomal glutamate release and thalamocortical synaptic transmission were suppressed by the inhibition of protein kinase A and occluded by the stimulation of adenylyl cyclase. Dissecting this G-protein-independent regulation further in thalamocortical slices, the KAR-mediated facilitation of synaptic transmission was found to be sensitive to the block of Ca(2+) permeant KARs by philanthotoxin. Intriguingly, the synaptic facilitation was abrogated by depletion of intracellular Ca(2+) stores by thapsigargin, or inhibition of Ca(2+) -induced Ca(2+) -release by ryanodine. Thus, the KA-mediated modulation was contingent on both Ca(2+) entry through Ca(2+) -permeable KARs and liberation of intracellular Ca(2+) stores. Finally, sensitivity to W-7 indicated that the increased cytosolic [Ca(2+) ] underpinning KAR-mediated regulation of synaptic transmission at thalamocortical synapses, requires downstream activation of calmodulin. We conclude that neocortical pre-synaptic KARs mediate the facilitation of glutamate release and synaptic transmission by a Ca(2+) -calmodulin dependent activation of an adenylyl cyclase/cAMP/protein kinase A signalling cascade, independent of G-protein involvement.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Corteza Cerebral/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Glutamatos/metabolismo , Receptores de Ácido Kaínico/fisiología , Receptores Presinapticos/fisiología , Sinapsis/fisiología , Tálamo/fisiología , Algoritmos , Animales , Corteza Cerebral/efectos de los fármacos , AMP Cíclico/metabolismo , Interpretación Estadística de Datos , Fenómenos Electrofisiológicos , Agonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/fisiología , Técnicas In Vitro , Ácido Kaínico/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Técnicas de Placa-Clamp , Receptores de Ácido Kaínico/efectos de los fármacos , Receptores Presinapticos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sinaptosomas/metabolismo , Tálamo/efectos de los fármacos
10.
Toxicol Appl Pharmacol ; 267(2): 174-83, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23319015

RESUMEN

AMP-activated protein kinase (AMPK) plays a central role in controlling hepatic lipid metabolism through modulating the downstream acetyl CoA carboxylase (ACC) and sterol regulatory element-binding protein-1c (SREBP-1c) pathway. Saponins, particularly platycodin D, from the roots of Platycodon grandiflorum (Changkil saponins, CKS) have a variety of pharmacological properties, including antioxidant and hepatoprotective properties. The aim of this study was to investigate the effects of CKS on hepatic lipogenesis and on the expression of genes involved in lipogenesis, and the mechanisms involved. CKS attenuated fat accumulation and the induction of the lipogenic genes encoding SREBP-1c and fatty acid synthase in the livers of HFD-fed rats and in steatotic HepG2 cells. Blood biochemical analyses and histopathological examinations showed that CKS prevented liver injury. CKS and platycodin D each increased the phosphorylation of AMPK and acetyl-CoA carboxylase in HFD-fed rats and HepG2 cells. The use of specific inhibitors showed that platycodin D activated AMPK via SIRT1/CaMKKß in HepG2 cells. This study demonstrates that CKS or platycodin D alone can regulate hepatic lipogenesis via an AMPK-dependent signalling pathway.


Asunto(s)
Dieta Alta en Grasa , Glucosa/farmacología , Lipogénesis/efectos de los fármacos , Hígado/efectos de los fármacos , Platycodon/química , Saponinas/farmacología , Triterpenos/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Ácido Graso Sintasas/genética , Células Hep G2 , Humanos , Hígado/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Sirtuina 1/antagonistas & inhibidores , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
11.
Proc Natl Acad Sci U S A ; 109(50): E3405-13, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23184977

RESUMEN

Autophagy is a cellular degradation process involving an intracellular membrane trafficking pathway that recycles cellular components or eliminates intracellular microbes in lysosomes. Many pathogens subvert autophagy to enhance their replication, but the mechanisms these pathogens use to initiate the autophagy process have not been elucidated. This study identifies rotavirus as a pathogen that encodes a viroporin, nonstructural protein 4, which releases endoplasmic reticulum calcium into the cytoplasm, thereby activating a calcium/calmodulin-dependent kinase kinase-ß and 5' adenosine monophosphate-activated protein kinase-dependent signaling pathway to initiate autophagy. Rotavirus hijacks this membrane trafficking pathway to transport viral proteins from the endoplasmic reticulum to sites of viral replication to produce infectious virus. This process requires PI3K activity and autophagy-initiation proteins Atg3 and Atg5, and it is abrogated by chelating cytoplasmic calcium or inhibiting calcium/calmodulin-dependent kinase kinase-ß. Although the early stages of autophagy are initiated, rotavirus infection also blocks autophagy maturation. These studies identify a unique mechanism of virus-mediated, calcium-activated signaling that initiates autophagy and hijacks this membrane trafficking pathway to transport viral proteins to sites of viral assembly.


Asunto(s)
Autofagia/fisiología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Rotavirus/fisiología , Replicación Viral/fisiología , Animales , Proteína 5 Relacionada con la Autofagia , Proteínas Relacionadas con la Autofagia , Señalización del Calcio , Línea Celular , Células Cultivadas , Activación Enzimática , Glicoproteínas/fisiología , Macaca mulatta , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/fisiología , Transporte de Proteínas , Rotavirus/patogenicidad , Transducción de Señal , Toxinas Biológicas/fisiología , Enzimas Ubiquitina-Conjugadoras/deficiencia , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/fisiología , Respuesta de Proteína Desplegada , Proteínas no Estructurales Virales/fisiología , Ensamble de Virus/fisiología
12.
Adv Exp Med Biol ; 740: 703-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22453966

RESUMEN

Calcium/calmodulin-stimulated protein kinases can be classified as one of two types - restricted or multifunctional. This family of kinases contains several structural similarities: all possess a calmodulin binding motif and an autoinhibitory region. In addition, all of the calcium/calmodulin-stimulated protein kinases examined in this chapter are regulated by phosphorylation, which either activates or inhibits their kinase activity. However, as the multifunctional calcium/calmodulin-stimulated protein kinases are ubiquitously expressed, yet regulate a broad range of cellular functions, additional levels of regulation that control these cell-specific functions must exist. These additional layers of control include gene expression, signaling pathways, and expression of binding proteins and molecular targeting. All of the multifunctional calcium/calmodulin-stimulated protein kinases examined in this chapter appear to be regulated by these additional layers of control, however, this does not appear to be the case for the restricted kinases.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/química , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/química , Quinasa de la Caseína I/síntesis química , Quinasa de la Caseína I/fisiología , Humanos , Quinasa de Cadena Ligera de Miosina/química , Quinasa de Cadena Ligera de Miosina/fisiología , Fosforilasa Quinasa/química , Fosforilasa Quinasa/fisiología , Fosforilación
14.
Neurobiol Aging ; 33(5): 1006.e11-23, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22048125

RESUMEN

Pathological autophagic vacuoles (AVs) accumulate in the brains of Alzheimer's disease (AD) patients, but the mechanisms by which they are induced are unknown. In this study, we found that the formation of AVs was mediated by activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) in the brains of APP/PS1 double transgenic mice, amyloid-beta peptide (Aß) pathology-bearing model mouse. Injection of sunitinib malate, AMPK inhibitor, to the mice lowered AV formation in their brains. Consistent with our in vivo observations, treatment of SH-SY5Y cells with Aß enhanced the induction of autophagosomes, which was mediated by Ca(2+)/calmodulin-dependent protein kinase kinase-beta (CaMKKß)-AMPK signaling, as shown using various inhibitors and small interfering RNA (siRNA). CaMKKß is a calcium-activated kinase, and the depletion of intracellular calcium by BAPTA-AM, a Ca(2+) chelator, also curtailed Aß-induced autophagy. Finally, the inhibition of receptor for advanced glycation end products (RAGE) attenuated autophagsome formation and AMPK signaling. Conversely, RAGE overexpression amplified the induction of autophagy. These results implicate the regulation of the Aß-induced formation of AVs by the RAGE-calcium-CaMKKß-AMPK pathway and suggest that modulation of autophagosome formation and the interaction between Aß and RAGE are beneficial in the treatment and prevention of Alzheimer's disease.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/fisiología , Autofagia/fisiología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Fragmentos de Péptidos/fisiología , Receptores Inmunológicos/fisiología , Transducción de Señal/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Transgénicos , Neuroblastoma/metabolismo , Neuroblastoma/patología , Fragmentos de Péptidos/genética , Presenilina-1/genética , Receptor para Productos Finales de Glicación Avanzada , Vacuolas/metabolismo
15.
Toxicol Appl Pharmacol ; 257(1): 48-58, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21884717

RESUMEN

The cardioprotective properties of puerarin, a natural product, have been attributed to the endothelial nitric oxide synthase (eNOS)-mediated production of nitric oxide (NO) in EA.hy926 endothelial cells. However, the mechanism by which puerarin activates eNOS remains unclear. In this study, we sought to identify the intracellular pathways underlying eNOS activation by puerarin. Puerarin induced the activating phosphorylation of eNOS on Ser1177 and the production of NO in EA.hy926 cells. Puerarin-induced eNOS phosphorylation required estrogen receptor (ER)-mediated phosphatidylinositol 3-kinase (PI3K)/Akt signaling and was reversed by AMP-activated protein kinase (AMPK) and calcium/calmodulin-dependent kinase II (CaMKII) inhibition. Importantly, puerarin inhibited the adhesion of tumor necrosis factor (TNF)-α-stimulated monocytes to endothelial cells and suppressed the TNF-α induced expression of intercellular cell adhesion molecule-1. Puerarin also inhibited the TNF-α-induced nuclear factor-κB activation, which was attenuated by pretreatment with N(G)-nitro-L-arginine methyl ester, a NOS inhibitor. These results indicate that puerarin stimulates eNOS phosphorylation and NO production via activation of an estrogen receptor-mediated PI3K/Akt- and CaMKII/AMPK-dependent pathway. Puerarin may be useful for the treatment or prevention of endothelial dysfunction associated with diabetes and cardiovascular disease.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Isoflavonas/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Quinasas/fisiología , Receptores de Estrógenos/fisiología , Quinasas de la Proteína-Quinasa Activada por el AMP , Western Blotting , Calcio/análisis , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Línea Celular , Células Endoteliales/química , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
16.
Endocrinology ; 152(10): 3668-79, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21862616

RESUMEN

When fed a standard chow diet, CaMKK2 null mice have increased adiposity and larger adipocytes than do wild-type mice, whereas energy balance is unchanged. Here, we show that Ca(2+)/calmodulin-dependent protein kinase kinase 2 (CaMKK2) is expressed in preadipocytes, where it functions as an AMP-activated protein kinase (AMPK)α kinase. Acute inhibition or deletion of CaMKK2 in preadipocytes enhances their differentiation into mature adipocytes, which can be reversed by 5-aminoimidazole-4-carboxamide ribonucleotide-mediated activation of AMPK. During adipogenesis, CaMKK2 expression is markedly decreased and temporally accompanied by increases in mRNA encoding the early adipogenic genes CCAAT/enhancer binding protein (C/EBP) ß and C/EBP δ. Preadipocyte factor 1 has been reported to inhibit adipogenesis by up-regulating sex determining region Y-box 9 (Sox9) expression in preadipocytes and Sox9 suppresses C/EBPß and C/EBPδ transcription. We show that inhibition of the CaMKK2/AMPK signaling cascade in preadipocytes reduces preadipocyte factor 1 and Sox9 mRNA resulting in accelerated adipogenesis. We conclude that CaMKK2 and AMPK function in a signaling pathway that participates in the regulation of adiposity.


Asunto(s)
Adipocitos/citología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Diferenciación Celular , Células Madre/citología , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/metabolismo , Adipogénesis , Animales , Proteínas de Unión al Calcio , Células Cultivadas , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ratones , Fosforilación , Transducción de Señal
17.
J Leukoc Biol ; 90(5): 897-909, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21816924

RESUMEN

Granulocytes serve a critical function in host organisms by recognizing and destroying invading microbes, as well as propagating and maintaining inflammation at sites of infection. However, the molecular pathways underpinning the development of granulocytes are poorly understood. Here, we identify a role for CaMKK2 in the restriction of granulocytic fate commitment and differentiation of myeloid progenitor cells. Following BMT, engraftment by Camkk2(-/-) donor cells resulted in the increased production of mature granulocytes in the BM and peripheral blood. Similarly, Camkk2(-/-) mice possessed elevated numbers of CMP cells and exhibited an accelerated granulopoietic phenotype in the BM. Camkk2(-/-) myeloid progenitors expressed increased levels of C/EBPα and PU.1 and preferentially differentiated into Gr1(+)Mac1(+) granulocytes and CFU-G in vitro. During normal granulopoiesis in vivo or G-CSF-induced differentiation of 32D myeloblast cells in vitro, CaMKK2 mRNA and protein were decreased as a function of time and were undetectable in mature granulocytes. Expression of ectopic CaMKK2 in Camkk2(-/-) CMPs was sufficient to rescue aberrant granulocyte differentiation and when overexpressed in 32D cells, was also sufficient to impede granulocyte differentiation in a kinase activity-dependent manner. Collectively, our results reveal a novel role for CaMKK2 as an inhibitor of granulocytic fate commitment and differentiation in early myeloid progenitors.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Linaje de la Célula , Granulocitos , Células Progenitoras Mieloides , Animales , Trasplante de Médula Ósea , Diferenciación Celular , Línea Celular , Microambiente Celular , Técnicas de Cocultivo , Granulocitos/citología , Granulocitos/inmunología , Granulocitos/metabolismo , Antígenos Comunes de Leucocito , Ratones , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/metabolismo , Células del Estroma , Irradiación Corporal Total
18.
J Pharmacol Exp Ther ; 339(1): 257-66, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21765041

RESUMEN

Flufenamic acid (FFA) is a nonsteroidal anti-inflammatory drug (NSAID). It has anti-inflammatory and antipyretic properties. In addition, it modulates multiple channel activities. The mechanisms underlying the pharmacological actions of FFA are presently unclear. Given that AMP-activated protein kinase (AMPK) has both anti-inflammatory and channel-regulating functions, we examined whether FFA induces AMPK activation. 1) Exposure of several different types of cells to FFA resulted in an elevation of AMPKα phosphorylation at Thr172. This effect of FFA was reproduced by functionally and structurally similar mefenamic acid, tolfenamic acid, niflumic acid, and meclofenamic acid. 2) FFA-induced activation of AMPK was largely abolished by the treatment of cells with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl ester) (an intracellular Ca(2+) chelator) or depletion of extracellular Ca(2+), whereas it was mimicked by stimulation of cells with the Ca(2+) ionophore 5-(methylamino)-2-({(2R,3R,6S,8S,9R,11R)-3,9,11-trimethyl-8-[(1S)-1-methyl-2-oxo-2-(1H-pyrrol-2-yl)ethyl]-1,7-dioxaspiro[5.5]undec-2-yl}methyl)-1,3-benzoxazole-4-carboxylic acid (A23187) or ionomycin. 3) FFA triggered a rise in intracellular Ca(2+), which was abolished by cyclosporine, a blocker of mitochondrial permeability transition pore. Cyclosporine also abolished FFA-induced activation of AMPK. 4) Inhibition of Ca(2+)/calmodulin-dependent kinase kinase ß (CaMKKß) with 7-oxo-7H-benzimidazo[2,1-a]benz[de]isoquinoline-3-carboxylic acid acetate (STO-609) or down-regulation of CaMKKß with short interfering RNA largely abrogated FFA-induced activation of AMPK. 5) FFA significantly suppressed nuclear factor-κB activity and inducible nitric-oxide synthase expression triggered by interleukin-1ß and tumor necrosis factor α. This suppression was also largely abrogated by STO-609. Taken together, we conclude that FFA induces AMPK activation through the Ca(2+)-CaMKKß pathway. Activation of AMPK is a presently unrecognized important mechanism underlying the pharmacological effects of FFA.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Antiinflamatorios no Esteroideos/farmacología , Ácido Flufenámico/farmacología , Fosfatasa Alcalina/metabolismo , Animales , Western Blotting , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Especificidad de la Especie , Porcinos , Transfección
19.
Biochem J ; 434(3): 503-12, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21204788

RESUMEN

Hyperphosphorylation of tau is a hallmark of Alzheimer's disease and other tauopathies. Although the mechanisms underlying hyperphosphorylation are not fully understood, cellular stresses such as impaired energy metabolism are thought to influence the signalling cascade. The AMPK (AMP-activated protein kinase)-related kinases MARK (microtubule-associated protein-regulating kinase/microtubule affinity-regulating kinase) and BRSK (brain-specific kinase) have been implicated in tau phosphorylation, but are insensitive to activation by cellular stress. In the present study, we show that AMPK itself phosphorylates tau on a number of sites, including Ser²6² and Ser³96, altering microtubule binding of tau. In primary mouse cortical neurons, CaMKKß (Ca²+/calmodulin-dependent protein kinase kinase ß) activation of AMPK in response to Aß (amyloid-ß peptide)-(1-42) leads to increased phosphorylation of tau at Ser²6²/Ser³56 and Ser3³96. Activation of AMPK by Aß-(1-42) is inhibited by memantine, a partial antagonist of the NMDA (N-methyl-D-aspartate) receptor and currently licensed for the treatment of Alzheimer's disease. These findings identify a pathway in which Aß-(1-42) activates CaMKKß and AMPK via the NMDA receptor, suggesting the possibility that AMPK plays a role in the pathophysiological phosphorylation of tau.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Péptidos beta-Amiloides/fisiología , Fragmentos de Péptidos/fisiología , Proteínas tau/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Dominio Catalítico , Células Cultivadas , Corteza Cerebral/citología , Activación Enzimática , Memantina/farmacología , Ratones , Microtúbulos/metabolismo , Neuronas/metabolismo , Fragmentos de Péptidos/farmacología , Fosforilación , Unión Proteica , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
20.
J Surg Res ; 169(2): 179-87, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20189584

RESUMEN

BACKGROUND: Adenosine monophosphate-activated protein kinase (AMPK) orchestrates the regulation of energy-generating and -consuming pathways, and protects the heart against ischemic injury and apoptosis. Recent progress shed light on various factors, including adiponectin, MIF, H11K, and metformin in the activation of AMPK. It is uncertain whether the activation of AMPK is contributed to cardioprotection of opioids. Here we show that morphine, an exogenous non-peptide opioid receptor agonist, can modulate the activation of the cardioprotective AMPK pathway during ischemia and exert anti-apoptotic effects through AMPK. METHODS: Isolated rat hearts were perfused on a constant pressure Langendorff system and subjected to 30 min of global ischemia followed by 60 min of reperfusion. The hearts received vehicles, morphine, a combination of morphine and compound C, a combination of morphine and STO609, a combination of morphine and BAPTA-AM at the onset of ischemia. Hemodynamics parameters, infarct size, release of intracellular creatine kinase, expression of AMPK, and terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling staining were analyzed. RESULTS: Morphine significantly increased phosphorylation level of Thr172 site on AMPK, left ventricular function, and reduced infarct size as a percentage of the area at risk (IS/AAR from 63% ± 7% to 40% ± 5%), release of intracellular creatine kinase (from 319 ± 46 to 156 ± 42IU/60 min/gdw), apoptosis ratio (from 16% ± 2% to 5% ± 1.4%) during reperfusion in comparison with the control group. A inhibitor of AMPK, compound C abrogated phosphorylation of AMPK induced by morphine, the improvement in myocardial function, and the reduction of IS/AAR (58% ± 6%), release of intracellular creatine kinase (270 ± 40IU/60 min/gdw), apoptosis ratio (13% ± 1.5%). A Ca(2+)/calmodulin-dependent protein kinase kinase inhibitor STO609 and a chelator of intracellular Ca(2+) stores BAPTA-AM also abolished the cardioprotection of morphine. CONCLUSIONS: Morphine can ameliorate myocardial contractile dysfunction and limit infarct size following ischemia and reperfusion by a mechanism involving activation of AMPK, and activate AMPK by Ca(2+)-CaMKKß-dependent phosphorylation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/fisiología , Analgésicos Opioides/farmacología , Precondicionamiento Isquémico Miocárdico/métodos , Morfina/farmacología , Daño por Reperfusión Miocárdica/prevención & control , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Calcio/fisiología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Masculino , Modelos Animales , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/fisiopatología , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...