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











Base de dados
Intervalo de ano de publicação
1.
Osteoarthritis Cartilage ; 30(1): 124-136, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34506942

RESUMO

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.


Assuntos
Benzimidazóis/uso terapêutico , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Cartilagem Articular/lesões , Naftalimidas/uso terapêutico , Osteoartrite/etiologia , Osteoartrite/prevenção & controle , Animais , Masculino , Camundongos , Ferimentos e Lesões/complicações
2.
Front Immunol ; 12: 754083, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34712241

RESUMO

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.


Assuntos
Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Células Supressoras Mieloides/enzimologia , Proteínas de Neoplasias/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Transferência Adotiva , Animais , Apoptose , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/deficiência , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/genética , Feminino , Depleção Linfocítica , Linfoma/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/metabolismo , Células Supressoras Mieloides/fisiologia , Células Supressoras Mieloides/transplante , Mielopoese , Espécies Reativas de Oxigênio , Microambiente Tumoral
3.
Mol Nutr Food Res ; 63(22): e1801377, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31454158

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Hepatócitos/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Palmitatos/farmacologia , Receptores de Calcitriol/fisiologia , Tomatina/análogos & derivados , Cálcio/metabolismo , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Ativação Enzimática/efeitos dos fármacos , Proteína Forkhead Box O1/genética , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Transdução de Sinais/fisiologia , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Tomatina/farmacologia
4.
Biochem Pharmacol ; 169: 113613, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31445019

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Hepatócitos/metabolismo , Metabolismo dos Lipídeos , Receptor Muscarínico M3/fisiologia , Animais , Células Cultivadas , Humanos , Camundongos , PPAR alfa/fisiologia , Fosforilação , Receptor Muscarínico M1/fisiologia , Transdução de Sinais/fisiologia
5.
Aging (Albany NY) ; 10(9): 2394-2406, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30227388

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Tecido Adiposo/metabolismo , Envelhecimento/metabolismo , Doenças Metabólicas/tratamento farmacológico , Inibidores da Fosfodiesterase 4/farmacologia , Sirtuínas/fisiologia , Células 3T3-L1 , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , AMP Cíclico/análise , Masculino , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Inibidores da Fosfodiesterase 4/uso terapêutico , Rolipram/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sirtuínas/análise
6.
Int J Cancer ; 136(6): 1434-44, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25080865

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/congênito , Neoplasias Pulmonares/tratamento farmacológico , Metformina/farmacologia , Niacinamida/análogos & derivados , Compostos de Fenilureia/farmacologia , Transdução de Sinais , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Niacinamida/farmacologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Sorafenibe , Serina-Treonina Quinases TOR/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/genética
7.
J Neurochem ; 126(5): 565-78, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23692284

RESUMO

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.


Assuntos
Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Córtex Cerebral/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Glutamatos/metabolismo , Receptores de Ácido Caínico/fisiologia , Receptores Pré-Sinápticos/fisiologia , Sinapses/fisiologia , Tálamo/fisiologia , Algoritmos , Animais , Córtex Cerebral/efeitos dos fármacos , AMP Cíclico/metabolismo , Interpretação Estatística de Dados , Fenômenos Eletrofisiológicos , Agonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas In Vitro , Ácido Caínico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Técnicas de Patch-Clamp , Receptores de Ácido Caínico/efeitos dos fármacos , Receptores Pré-Sinápticos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sinaptossomos/metabolismo , Tálamo/efeitos dos fármacos
8.
Toxicol Appl Pharmacol ; 267(2): 174-83, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23319015

RESUMO

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.


Assuntos
Dieta Hiperlipídica , Glucose/farmacologia , Lipogênese/efeitos dos fármacos , Fígado/efeitos dos fármacos , Platycodon/química , Saponinas/farmacologia , Triterpenos/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Ácido Graxo Sintases/genética , Células Hep G2 , Humanos , Fígado/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Sirtuína 1/antagonistas & inibidores , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
9.
Proc Natl Acad Sci U S A ; 109(50): E3405-13, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23184977

RESUMO

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.


Assuntos
Autofagia/fisiologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Rotavirus/fisiologia , Replicação Viral/fisiologia , Animais , Proteína 5 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Sinalização do Cálcio , Linhagem Celular , Células Cultivadas , Ativação Enzimática , Glicoproteínas/fisiologia , Macaca mulatta , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/fisiologia , Transporte Proteico , Rotavirus/patogenicidade , Transdução de Sinais , Toxinas Biológicas/fisiologia , Enzimas de Conjugação de Ubiquitina/deficiência , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/fisiologia , Resposta a Proteínas não Dobradas , Proteínas não Estruturais Virais/fisiologia , Montagem de Vírus/fisiologia
10.
Neurobiol Aging ; 33(5): 1006.e11-23, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22048125

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/fisiologia , Autofagia/fisiologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Fragmentos de Peptídeos/fisiologia , Receptores Imunológicos/fisiologia , Transdução de Sinais/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Fragmentos de Peptídeos/genética , Presenilina-1/genética , Receptor para Produtos Finais de Glicação Avançada , Vacúolos/metabolismo
11.
Toxicol Appl Pharmacol ; 257(1): 48-58, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21884717

RESUMO

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.


Assuntos
Células Endoteliais/efeitos dos fármacos , Isoflavonas/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Quinases/fisiologia , Receptores de Estrogênio/fisiologia , Quinases Proteína-Quinases Ativadas por AMP , Western Blotting , Cálcio/análise , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Linhagem Celular , Células Endoteliais/química , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
12.
J Leukoc Biol ; 90(5): 897-909, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21816924

RESUMO

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.


Assuntos
Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Linhagem da Célula , Granulócitos , Células Progenitoras Mieloides , Animais , Transplante de Medula Óssea , Diferenciação Celular , Linhagem Celular , Microambiente Celular , Técnicas de Cocultura , Granulócitos/citologia , Granulócitos/imunologia , Granulócitos/metabolismo , Antígenos Comuns de Leucócito , Camundongos , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/metabolismo , Células Estromais , Irradiação Corporal Total
13.
J Pharmacol Exp Ther ; 339(1): 257-66, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21765041

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Anti-Inflamatórios não Esteroides/farmacologia , Ácido Flufenâmico/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Western Blotting , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Especificidade da Espécie , Suínos , Transfecção
14.
Metabolism ; 60(4): 579-85, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20598720

RESUMO

The GLUT2 glucose transporter plays an important role in glucose-induced insulin secretion in pancreatic ß-cells by catalyzing the uptake of glucose into the cell. In this study, we investigated whether exendin-4, a long-acting agonist of glucagon-like peptide-1, mediates stimulatory effects on GLUT2 gene expression through the Ca²+/calmodulin (CaM)-dependent protein kinase IV (CaMKIV) cascade. GLUT2 expression was examined by real-time polymerase chain reaction, Western blot analysis, and a reporter gene assay in rat insulin-secreting INS-1 cells incubated with exendin-4. An increased expression level of GLUT2 protein was noted in response to increasing concentrations of exendin-4, with maximal induction at 10 nmol/L. Real-time polymerase chain reaction analysis similarly revealed a significant increase in the amount of GLUT2 messenger RNA by 10 nmol/L exendin-4. Exendin-4 also stimulated GLUT2 promoter activity in response to increasing exendin-4 concentrations, but failed to do so in the presence of STO-609, a CaMKK inhibitor. We also investigated the effect of the constitutively active form of CaMKK (CaMKKc) on GLUT2 promoter activity. The result is consistent with the observations that CaMKKc/CaMKIV enhanced or up-regulated GLUT2 promoter activity in INS-1 cells. Furthermore, exendin-4 induction of GLUT2 protein expression was significantly suppressed in the cells knocking down the CaMKIV. In summary, activation of the CaMKK/CaMKIV cascade might be required for exendin-4-induced GLUT2 gene transcription, indicating that exendin-4 plays an important role in insulin secretion in pancreatic ß-cells.


Assuntos
Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/fisiologia , Transportador de Glucose Tipo 2/biossíntese , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/metabolismo , Peptídeos/farmacologia , Peçonhas/farmacologia , Animais , Western Blotting , Sinalização do Cálcio/genética , Sinalização do Cálcio/fisiologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/genética , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Exenatida , Genes Reporter , Transportador de Glucose Tipo 2/genética , Células Secretoras de Insulina/enzimologia , Luciferases/genética , RNA Interferente Pequeno/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Transcrição Gênica/genética , Transfecção
15.
J Surg Res ; 169(2): 179-87, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20189584

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/fisiologia , Analgésicos Opioides/farmacologia , Precondicionamento Isquêmico Miocárdico/métodos , Morfina/farmacologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Cálcio/fisiologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Masculino , Modelos Animais , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Fosforilação/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
16.
Protein Expr Purif ; 73(2): 189-97, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20451617

RESUMO

AMP-activated protein kinase (AMPK) is an energy-sensing serine/threonine protein kinase that plays a central role in whole-body energy homeostasis. AMPK is a heterotrimeric enzyme with a catalytic (alpha) subunit and two regulatory (beta and gamma) subunits. The muscle-specific AMPK heterotrimeric complex (alpha2beta2gamma3) is involved in glucose and fat metabolism in skeletal muscle and therefore has emerged as an attractive target for drug development for diabetes and metabolic syndrome. To date, expression of recombinant full-length human AMPK alpha2beta2gamma3 has not been reported. Here we describe the expression, purification and biochemical characterization of functional full-length AMPK alpha2beta2gamma3 heterotrimeric complex using an Escherichia coli expression system. All three subunits of AMPK alpha2beta2gamma3 were transcribed as a single tricistronic transcript driven by the T7 RNA polymerase promoter, allowing spontaneous formation of the heterotrimeric complex in the bacterial cytosol. The self-assembled trimeric complex was purified from the cell lysate by nickel-ion chromatography using the hexahistidine tag fused exclusively at the N-terminus of the alpha 2 domain. The un-assembled beta 2 and gamma 3 domains were removed by extensive washing of the column. Further purification of the heterotrimer was performed using size exclusion chromatography. The final yield of the recombinant AMPK alpha2beta2gamma3 complex was 1.1mg/L culture in shaker flasks. The E. coli expressed enzyme was catalytically inactive after purification, but was activated in vitro by upstream kinases such as CaMKKbeta and LKB1. The kinase activity of activated AMPK alpha2beta2gamma3 complex was significantly enhanced by AMP (an allosteric activator) but not by thienopyridone A-769662, a known small molecule activator of AMPK. Mass spectrometric characterization of recombinant AMPK alpha2beta2gamma3 showed significant heterogeneity before and after activation that could potentially hamper crystallographic studies of this complex.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Escherichia coli/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Monofosfato de Adenosina/metabolismo , Compostos de Bifenilo , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/genética , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Domínio Catalítico , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Escherichia coli/genética , Homeostase , Humanos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Pironas/farmacologia , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Tiofenos/farmacologia
17.
Cell Metab ; 7(5): 377-88, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18460329

RESUMO

Detailed knowledge of the pathways by which ghrelin and leptin signal to AMPK in hypothalamic neurons and lead to regulation of appetite and glucose homeostasis is central to the development of effective means to combat obesity. Here we identify CaMKK2 as a component of one of these pathways, show that it regulates hypothalamic production of the orexigenic hormone NPY, provide evidence that it functions as an AMPKalpha kinase in the hypothalamus, and demonstrate that it forms a unique signaling complex with AMPKalpha and beta. Acute pharmacologic inhibition of CaMKK2 in wild-type mice, but not CaMKK2 null mice, inhibits appetite and promotes weight loss consistent with decreased NPY and AgRP mRNAs. Moreover, the loss of CaMKK2 protects mice from high-fat diet-induced obesity, insulin resistance, and glucose intolerance. These data underscore the potential of targeting CaMKK2 as a therapeutic intervention.


Assuntos
Regulação do Apetite/fisiologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Metabolismo Energético/fisiologia , Hipotálamo/enzimologia , Resistência à Insulina/fisiologia , Quinases Proteína-Quinases Ativadas por AMP , Acetil-CoA Carboxilase/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Benzimidazóis/farmacologia , Células Cultivadas , Dieta Aterogênica , Feminino , Intolerância à Glucose/etiologia , Teste de Tolerância a Glucose , Hipotálamo/patologia , Immunoblotting , Técnicas Imunoenzimáticas , Imunoprecipitação , Hibridização In Situ , Insulina/metabolismo , Integrases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Naftalimidas/farmacologia , Neuropeptídeo Y/metabolismo , Proteínas Quinases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transfecção , Redução de Peso
18.
Apoptosis ; 13(6): 803-10, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18465250

RESUMO

Sodium arsenite induces apoptosis in PC12 cells by activating the stress-activated p38 MAP kinase and the pro-apoptotic Bcl-2 family protein Bim(EL). However, the relationship between p38 and Bim(EL) in this apoptosis has not been fully defined. Here, we report that sodium arsenite stimulates the protein expression and promoter activity of Bim(EL) in a p38-dependent manner. Sodium arsenite also caused nuclear translocation of FOXO3a, indicative of FOXO3a activation. Addition of a p38 inhibitor prevented FOXO3a nuclear translocation. RNAi knock down of FOXO3a inhibited Bim promoter activity, Bim(EL) protein expression, and arsenite-induced apoptosis. Our data identify p38 activation of FOXO3a and subsequent induction of Bim(EL) expression as a novel apoptotic mechanism. Together with our previous finding that Bim(EL) is phosphorylated and activated by p38, these results demonstrate that p38 induces apoptosis by regulating Bim(EL) at both the transcriptional and post-translational levels.


Assuntos
Apoptose/efeitos dos fármacos , Arsenitos/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/biossíntese , Compostos de Sódio/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Ativação Enzimática , Proteína Forkhead Box O3 , Imidazóis/farmacologia , Modelos Biológicos , Células PC12 , Transporte Proteico/efeitos dos fármacos , Piridinas/farmacologia , Interferência de RNA , Ratos , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
19.
PLoS One ; 3(2): e1606, 2008 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-18270593

RESUMO

Macrophages are capable of assuming numerous phenotypes in order to adapt to endogenous and exogenous challenges but many of the factors that regulate this process are still unknown. We report that Ca(2+)/calmodulin-dependent kinase kinase alpha (CaMKKalpha) is expressed in human monocytic cells and demonstrate that its inhibition blocks type-II monocytic cell activation and promotes classical activation. Affinity chromatography with paramagnetic beads isolated an approximately 50 kDa protein from nuclear lysates of U937 human monocytic cells activated with phorbol-12-myristate-13-acetate (PMA). This protein was identified as CaMKKalpha by mass spectrometry and Western analysis. The function of CaMKKalpha in monocyte activation was examined using the CaMKKalpha inhibitors (STO-609 and forskolin) and siRNA knockdown. Inhibition of CaMKKalpha, enhanced PMA-dependent CD86 expression and reduced CD11b expression. In addition, inhibition was associated with decreased translocation of CaMKKalpha to the nucleus. Finally, to further examine monocyte activation profiles, TNFalpha and IL-10 secretion were studied. CaMKKalpha inhibition attenuated PMA-dependent IL-10 production and enhanced TNFalpha production indicating a shift from type-II to classical monocyte activation. Taken together, these findings indicate an important new role for CaMKKalpha in the differentiation of monocytic cells.


Assuntos
Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Monócitos/química , Transporte Ativo do Núcleo Celular , Antígenos CD/análise , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/análise , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Diferenciação Celular , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Humanos , Interleucina-10/biossíntese , Monócitos Matadores Ativados , Acetato de Tetradecanoilforbol/farmacologia , Fator de Necrose Tumoral alfa/biossíntese
20.
Subcell Biochem ; 45: 169-214, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18193638

RESUMO

Numerous hormones, growth factors and physiological processes cause a rise in cytosolic Ca2+, which is translated into meaningful cellular responses by interacting with a large number of Ca2(+)-binding proteins. The Ca2(+)-binding protein that is most pervasive in mediating these responses is calmodulin (CaM), which acts as a primary receptor for Ca2+ in all eukaryotic cells. In turn, Ca2+/CaM functions as an allosteric activator of a host of enzymatic proteins including a considerable number of protein kinases. The topic of this review is to discuss the physiological roles of a sub-set of these protein kinases which can function in cells as a Ca2+/CaM-dependent kinase signaling cascade. The cascade was originally believed to consist of a CaM kinase kinase that phosphorylates and activates one of two CaM kinases, CaMKI or CaMKIV. The unusual aspect of this cascade is that both the kinase kinase and the kinase require the binding of Ca2+/CaM for activation. More recently, one of the CaM kinase kinases has been found to activate another important enzyme, the AMP-dependent protein kinase so the concept of the CaM kinase cascade must be expanded. A CaM kinase cascade is important for many normal physiological processes that when misregulated can lead to a variety of disease states. These processes include: cell proliferation and apoptosis that may conspire in the genesis of cancer; neuronal growth and function related to brain development, synaptic plasticity as well as memory formation and maintenance; proper function of the immune system including the inflammatory response, activation of T lymphocytes and hematopoietic stem cell maintenance; and the central control of energy balance that, when altered, can lead to obesity and diabetes. Although the study of the CaM-dependent kinase cascades is still in its infancy continued analysis of the pathways regulated by these Ca2(+)-initiated signaling cascades holds considerable promise for the future of disease-related research.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Memória/fisiologia , Animais , Caenorhabditis elegans/fisiologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Cerebelo/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Sistema Imunitário/fisiologia , Potenciação de Longa Duração/fisiologia , Camundongos , Neoplasias/fisiopatologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Fosfotransferases (Aceptor do Grupo Fosfato)/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA