Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 20(20)2019 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-31635305

RESUMO

The AMP-activated protein kinase (AMPK) is principally known as a major regulator of cellular energy status, but it has been recently shown to play a key structural role in cell-cell junctions. The aim of this study was to evaluate the impact of AMPK activation on the reassembly of tight junctions in intestinal epithelial Caco-2 cells. We generated Caco-2 cells invalidated for AMPK α1/α2 (AMPK dKO) by CRISPR/Cas9 technology and evaluated the effect of the direct AMPK activator 991 on the reassembly of tight junctions following a calcium switch assay. We analyzed the integrity of the epithelial barrier by measuring the trans-epithelial electrical resistance (TEER), the paracellular permeability, and quantification of zonula occludens 1 (ZO-1) deposit at plasma membrane by immunofluorescence. Here, we demonstrated that AMPK deletion induced a delay in tight junction reassembly and relocalization at the plasma membrane during calcium switch, leading to impairments in the establishment of TEER and paracellular permeability. We also showed that 991-induced AMPK activation accelerated the reassembly and reorganization of tight junctions, improved the development of TEER and paracellular permeability after calcium switch. Thus, our results show that AMPK activation ensures a better recovery of epithelial barrier function following injury.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Junções Íntimas/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Células CACO-2 , Cálcio/metabolismo , Permeabilidade da Membrana Celular , Análise Mutacional de DNA , Humanos , Sistema de Sinalização das MAP Quinases , Mutação , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo
2.
FASEB J ; 28(7): 3211-24, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24652947

RESUMO

AMP-activated protein kinase (AMPK) is a sensor of cellular energy status that plays a central role in skeletal muscle metabolism. We used skeletal muscle-specific AMPKα1α2 double-knockout (mdKO) mice to provide direct genetic evidence of the physiological importance of AMPK in regulating muscle exercise capacity, mitochondrial function, and contraction-stimulated glucose uptake. Exercise performance was significantly reduced in the mdKO mice, with a reduction in maximal force production and fatigue resistance. An increase in the proportion of myofibers with centralized nuclei was noted, as well as an elevated expression of interleukin 6 (IL-6) mRNA, possibly consistent with mild skeletal muscle injury. Notably, we found that AMPKα1 and AMPKα2 isoforms are dispensable for contraction-induced skeletal muscle glucose transport, except for male soleus muscle. However, the lack of skeletal muscle AMPK diminished maximal ADP-stimulated mitochondrial respiration, showing an impairment at complex I. This effect was not accompanied by changes in mitochondrial number, indicating that AMPK regulates muscle metabolic adaptation through the regulation of muscle mitochondrial oxidative capacity and mitochondrial substrate utilization but not baseline mitochondrial muscle content. Together, these results demonstrate that skeletal muscle AMPK has an unexpected role in the regulation of mitochondrial oxidative phosphorylation that contributes to the energy demands of the exercising muscle.-Lantier, L., Fentz, J., Mounier, R., Leclerc, J., Treebak, J. T., Pehmøller, C., Sanz, N., Sakakibara, I., Saint-Amand, E., Rimbaud, S., Maire, P., Marette, A., Ventura-Clapier, R., Ferry, A., Wojtaszewski, J. F. P., Foretz, M., Viollet, B. AMPK controls exercise endurance, mitochondrial oxidative capacity, and skeletal muscle integrity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Resistência Física/fisiologia , Animais , Glucose/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular/fisiologia , Fadiga Muscular/fisiologia , Oxirredução , Fosforilação/fisiologia , Condicionamento Físico Animal
3.
J Hepatol ; 60(1): 152-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24012615

RESUMO

BACKGROUND: AMP-activated protein kinase (AMPK) is an evolutionarily conserved sensor of cellular energy status that contributes to restoration of energy homeostasis by slowing down ATP-consuming pathways and activating ATP-producing pathways. Unexpectedly, in different systems, AMPK is also required for proper cell division. In the current study, we evaluated the potential effect of the AMPK catalytic subunit, AMPKα1, on hepatocyte proliferation. METHODS: Hepatocyte proliferation was determined in AMPKα1 knockout and wild-type mice in vivo after two thirds partial hepatectomy, and in vitro in primary hepatocyte cultures. The activities of metabolic and cell cycle-related signaling pathways were measured. RESULTS: After partial hepatectomy, hepatocytes proliferated rapidly, correlating with increased AMPK phosphorylation. Deletion of AMPKα1 delayed liver regeneration by impacting on G1/S transition phase. The proliferative defect of AMPKα1-deficient hepatocytes was cell autonomous, and independent of energy balance. The priming phase, lipid droplet accumulation, protein anabolic responses and growth factor activation after partial hepatectomy occurred normally in the absence of AMPKα1 activity. By contrast, mRNA and protein expression of cyclin A2, a key driver of S phase progression, were compromised in the absence of AMPK activity. Importantly, AMPKα1 controlled cyclin A2 transcription mainly through the ATF/CREB element. CONCLUSIONS: Our study highlights a novel role for AMPKα1 as a positive regulator of hepatocyte division occurring independently of energy balance.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Proliferação de Células , Ciclina A2/fisiologia , Hepatócitos/fisiologia , Animais , Ciclina A2/genética , Metabolismo Energético , Regeneração Hepática , Camundongos , Camundongos Endogâmicos C57BL , Fase S
4.
Crit Rev Biochem Mol Biol ; 45(4): 276-95, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20522000

RESUMO

All living organisms depend on dynamic mechanisms that repeatedly reassess the status of amassed energy, in order to adapt energy supply to demand. The AMP-activated protein kinase (AMPK) alphabetagamma heterotrimer has emerged as an important integrator of signals managing energy balance. Control of AMPK activity involves allosteric AMP and ATP regulation, auto-inhibitory features and phosphorylation of its catalytic (alpha) and regulatory (beta and gamma) subunits. AMPK has a prominent role not only as a peripheral sensor but also in the central nervous system as a multifunctional metabolic regulator. AMPK represents an ideal second messenger for reporting cellular energy state. For this reason, activated AMPK acts as a protective response to energy stress in numerous systems. However, AMPK inhibition also actively participates in the control of whole body energy homeostasis. In this review, we discuss recent findings that support the role and function of AMPK inhibition under physiological and pathological states.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Metabolismo Energético , Proteínas Quinases Ativadas por AMP/química , Animais , Regulação para Baixo , Ativação Enzimática , Humanos
5.
Clin Sci (Lond) ; 122(6): 253-70, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22117616

RESUMO

Considerable efforts have been made since the 1950s to better understand the cellular and molecular mechanisms of action of metformin, a potent antihyperglycaemic agent now recommended as the first-line oral therapy for T2D (Type 2 diabetes). The main effect of this drug from the biguanide family is to acutely decrease hepatic glucose production, mostly through a mild and transient inhibition of the mitochondrial respiratory chain complex I. In addition, the resulting decrease in hepatic energy status activates AMPK (AMP-activated protein kinase), a cellular metabolic sensor, providing a generally accepted mechanism for the action of metformin on hepatic gluconeogenesis. The demonstration that respiratory chain complex I, but not AMPK, is the primary target of metformin was recently strengthened by showing that the metabolic effect of the drug is preserved in liver-specific AMPK-deficient mice. Beyond its effect on glucose metabolism, metformin has been reported to restore ovarian function in PCOS (polycystic ovary syndrome), reduce fatty liver, and to lower microvascular and macrovascular complications associated with T2D. Its use has also recently been suggested as an adjuvant treatment for cancer or gestational diabetes and for the prevention in pre-diabetic populations. These emerging new therapeutic areas for metformin will be reviewed together with recent findings from pharmacogenetic studies linking genetic variations to drug response, a promising new step towards personalized medicine in the treatment of T2D.


Assuntos
Hipoglicemiantes/farmacologia , Metformina/farmacologia , Animais , Sistema Cardiovascular/efeitos dos fármacos , Relógios Circadianos/efeitos dos fármacos , Nefropatias Diabéticas/tratamento farmacológico , Feminino , Humanos , Metformina/uso terapêutico , Neoplasias/tratamento farmacológico , Síndrome do Ovário Policístico/tratamento farmacológico
6.
FASEB J ; 25(1): 337-47, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20881209

RESUMO

AMP-activated protein kinase (AMPK) is an αßγ heterotrimer conserved throughout evolution and important for energy sensing in all eukaryote cells. AMPK controls metabolism and various cellular events in response to both hormones and changes in cellular energy status. The γ subunit senses intracellular energy status through the competitive binding of AMP and ATP. We show here that targeted disruption of the mouse AMPKγ1 gene (Prkag1) causes regenerative hemolytic anemia by increasing the sequestration of abnormal erythrocytes. Prkag1(-/-) mice displayed splenomegaly and iron accumulation due to compensatory splenic erythropoiesis and erythrophagocytosis. Moreover, AMPKγ1-deficient erythrocytes were highly resistant to osmotic hemolysis and poorly deformable in response to increasing shear stress, consistent with greater membrane rigidity. No change in cytoskeletal protein composition was observed; however, the phosphorylation level of adducin, a protein promoting the binding of spectrin to actin, was higher in AMPKγ1-deficient erythrocytes. Together, these results demonstrate that AMPKγ1 subunit is required for the maintenance of erythrocyte membrane elasticity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Anemia/enzimologia , Membrana Eritrocítica/metabolismo , Esplenomegalia/enzimologia , Proteínas Quinases Ativadas por AMP/genética , Anemia/sangue , Anemia/genética , Anemia Hemolítica/enzimologia , Anemia Hemolítica/genética , Animais , Western Blotting , Elasticidade , Eritroblastos/metabolismo , Eritroblastos/patologia , Contagem de Eritrócitos , Deformação Eritrocítica , Feminino , Hiperplasia , Ferro/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Baço/metabolismo , Baço/patologia , Esplenomegalia/sangue , Esplenomegalia/genética
7.
J Behav Med ; 35(2): 167-78, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21487723

RESUMO

The cognitive-behavioural model of health anxiety hypothesizes that the degree of health threat experienced by an individual is a function of the perceived: (1) likelihood of illness; (2) awfulness of illness; (3) difficulty coping with illness; and (4) inadequacy of medical services. While research has examined cognitions in health anxiety, it is not known whether these cognitions predict health anxiety in individuals who do or do not report medical conditions and whether these cognitions are uniquely related to health anxiety. After developing the Health Cognitions Questionnaire to assess these specific cognitions, we examined the extent to which the cognitions predicted health anxiety and poor response to reassurance in a healthy community sample (n = 273) and a sample who self-reported various medical conditions (n = 208). Supporting the cognitive-behavioural model, these cognitions predicted health anxiety and poor response to reassurance in both samples, with some differences observed between those who did or did not report medical conditions. The cognitions were uniquely related to health anxiety even after controlling for depression and general anxiety. Clinical and theoretical implications are discussed. Overall, the Health Cognitions Questionnaire has potential to facilitate further research on the development, maintenance, and treatment of health anxiety.


Assuntos
Ansiedade/psicologia , Atitude Frente a Saúde , Medo/psicologia , Adolescente , Adulto , Depressão/psicologia , Feminino , Humanos , Masculino , Autorrelato , Inquéritos e Questionários
8.
FASEB J ; 24(9): 3555-61, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20460585

RESUMO

Skeletal muscle mass is regulated by signaling pathways that govern protein synthesis and cell proliferation, and the mammalian target of rapamycin (mTOR) plays a key role in these processes. Recent studies suggested the crucial role of AMP-activated protein kinase (AMPK) in the inhibition of protein synthesis and cell growth. Here, we address the role of AMPK in the regulation of muscle cell size in vitro and in vivo. The size of AMPK-deficient myotubes was 1.5-fold higher than for controls. A marked increase in p70S6K Thr(389) and rpS6 Ser-235/236 phosphorylation was observed concomitantly with an up-regulation of protein synthesis rate. Treatment with rapamycin prevented p70S6K phosphorylation and rescued cell size control in AMPK-deficient cells. Importantly, myotubes lacking AMPK were resistant to further cell size increase beyond AMPK deletion alone, as MyrAkt-induced hypertrophy was absent in these cells. Moreover, in skeletal muscle-specific deficient AMPKalpha1/alpha2 KO mice, soleus muscle showed a higher mass with myofibers of larger size and was associated with increased p70S6K and rpS6 phosphorylation. Our results uncover the role of AMPK in the maintenance of muscle cell size control and highlight the crosstalk between AMPK and mTOR/p70S6K signaling pathways coordinating a metabolic checkpoint on cell growth.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Tamanho Celular , Proteínas Quinases Ativadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Células Cultivadas , Immunoblotting , Técnicas In Vitro , Metformina/farmacologia , Camundongos , Camundongos Knockout , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Fosforilação/efeitos dos fármacos , Ribonucleotídeos/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo
9.
Mol Metab ; 47: 101183, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33548500

RESUMO

OBJECTIVE: The intestinal epithelial barrier (IEB) restricts the passage of microbes and potentially harmful substances from the lumen through the paracellular space, and rupture of its integrity is associated with a variety of gastrointestinal disorders and extra-digestive diseases. Increased IEB permeability has been linked to disruption of metabolic homeostasis leading to obesity and type 2 diabetes. Interestingly, recent studies have uncovered compelling evidence that the AMP-activated protein kinase (AMPK) signaling pathway plays an important role in maintaining epithelial cell barrier function. However, our understanding of the function of intestinal AMPK in regulating IEB and glucose homeostasis remains sparse. METHODS: We generated mice lacking the two α1 and α2 AMPK catalytic subunits specifically in intestinal epithelial cells (IEC AMPK KO) and determined the physiological consequences of intestinal-specific deletion of AMPK in response to high-fat diet (HFD)-induced obesity. We combined histological, functional, and integrative analyses to ascertain the effects of gut AMPK loss on intestinal permeability in vivo and ex vivo and on the development of obesity and metabolic dysfunction. We also determined the impact of intestinal AMPK deletion in an inducible mouse model (i-IEC AMPK KO) by measuring IEB function, glucose homeostasis, and the composition of gut microbiota via fecal 16S rRNA sequencing. RESULTS: While there were no differences in in vivo intestinal permeability in WT and IEC AMPK KO mice, ex vivo transcellular and paracellular permeability measured in Ussing chambers was significantly increased in the distal colon of IEC AMPK KO mice. This was associated with a reduction in pSer425 GIV phosphorylation, a marker of leaky gut barrier. However, the expression of tight junction proteins in intestinal epithelial cells and pro-inflammatory cytokines in the lamina propria were not different between genotypes. Although the HFD-fed AMPK KO mice displayed suppression of the stress polarity signaling pathway and a concomitant increase in colon permeability, loss of intestinal AMPK did not exacerbate body weight gain or adiposity. Deletion of AMPK was also not sufficient to alter glucose homeostasis or the acute glucose-lowering action of metformin in control diet (CD)- or HFD-fed mice. CD-fed i-IEC AMPK KO mice also presented higher permeability in the distal colon under homeostatic conditions but, surprisingly, this was not detected upon HFD feeding. Alteration in epithelial barrier function in the i-IEC AMPK KO mice was associated with a shift in the gut microbiota composition with higher levels of Clostridiales and Desulfovibrionales. CONCLUSIONS: Altogether, our results revealed a significant role of intestinal AMPK in maintaining IEB integrity in the distal colon but not in regulating glucose homeostasis. Our data also highlight the complex interaction between gut microbiota and host AMPK.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Colo/metabolismo , Glucose/metabolismo , Homeostase , Animais , Bactérias/classificação , Bactérias/genética , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Fezes/microbiologia , Microbioma Gastrointestinal , Mucosa Intestinal/metabolismo , Masculino , Metformina/farmacologia , Camundongos , Camundongos Knockout , Obesidade/metabolismo , Permeabilidade/efeitos dos fármacos , RNA Ribossômico 16S
10.
FASEB J ; 23(7): 2264-73, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19237506

RESUMO

Activation of AMP-activated protein kinase (AMPK) inhibits protein synthesis through the suppression of the mammalian target of rapamycin complex 1 (mTORC1), a critical regulator of muscle growth. The purpose of this investigation was to determine the role of the AMPKalpha1 catalytic subunit on muscle cell size control and adaptation to muscle hypertrophy. We found that AMPKalpha1(-/-) primary cultured myotubes and myofibers exhibit larger cell size compared with control cells in response to chronic Akt activation. We next subjected the plantaris muscle of AMPKalpha1(-/-) and control mice to mechanical overloading to induce muscle hypertrophy. We observed significant elevations of AMPKalpha1 activity in the control muscle at days 7 and 21 after the overload. Overloading-induced muscle hypertrophy was significantly accelerated in AMPKalpha1(-/-) mice than in control mice [+32 vs. +53% at day 7 and +57 vs. +76% at day 21 in control vs. AMPKalpha1(-/-) mice, respectively]. This enhanced growth of AMPKalpha1-deficient muscle was accompanied by increased phosphorylation of mTOR signaling downstream targets and decreased phosphorylation of eukaryotic elongation factor 2. These results demonstrate that AMPKalpha1 plays an important role in limiting skeletal muscle overgrowth during hypertrophy through inhibition of the mTOR-signaling pathway.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Proteínas de Transporte/metabolismo , Hipertrofia , Músculo Esquelético/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Quinases Ativadas por AMP/deficiência , Animais , Crescimento Celular , Camundongos , Camundongos Knockout , Fator 2 de Elongação de Peptídeos/metabolismo , Fosforilação , Substâncias Protetoras , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Estresse Mecânico , Serina-Treonina Quinases TOR
11.
Cell Rep ; 22(8): 1994-2005, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29466728

RESUMO

Liver kinase B1 (LKB1) is involved in several biological processes and is a key regulator of hepatic metabolism and polarity. Here, we demonstrate that the master kinase LKB1 plays a dual role in liver regeneration, independently of its major target, AMP-activated protein kinase (AMPK). We found that the loss of hepatic Lkb1 expression promoted hepatocyte proliferation acceleration independently of metabolic/energetic balance. LKB1 regulates G0/G1 progression, specifically by controlling epidermal growth factor receptor (EGFR) signaling. Furthermore, later in regeneration, LKB1 controls mitotic fidelity. The deletion of Lkb1 results in major alterations to mitotic spindle formation along the polarity axis. Thus, LKB1 deficiency alters ploidy profile at late stages of regeneration. Our findings highlight the dual role of LKB1 in liver regeneration, as a guardian of hepatocyte proliferation and genomic integrity.


Assuntos
Genoma , Hepatócitos/citologia , Hepatócitos/metabolismo , Regeneração Hepática/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP , Animais , Proliferação de Células , Ativação Enzimática , Receptores ErbB/metabolismo , Deleção de Genes , Inativação Gênica , Fígado/citologia , Camundongos , Mitose , Ploidias , Proteínas Serina-Treonina Quinases/deficiência , Transdução de Sinais
12.
Endocrinology ; 148(1): 422-32, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17038560

RESUMO

We previously have described molecular mechanisms converging at the Nur response element-signal transducer and activator of transcription (STAT) composite site responsible for synergistic activation of the proopiomelanocortin (POMC) gene promoter by leukemia inhibitory factor (LIF) and CRH. In this study, we asked how glucocorticoids (GC), the physiological negative regulators of POMC gene expression, modulate this synergism. In the corticotroph cell line AtT-20, the response of the wild-type promoter to LIF+CRH was barely inhibited by GC, whereas a distal promoter subregion (-414/-293) encompassing the Nur response element-STAT site and devoid of the negative GC-responsive element located in the proximal domain, displayed a cooperative response to LIF+dexamethasone (DEX) and LIF+CRH+DEX treatments. LIF+CRH-stimulated ACTH secretion was also inefficiently inhibited by DEX in the same cell line. This study was focused thereafter on LIF+DEX cooperativity, which may be responsible, on the wild-type promoter, for lack of negative regulation by DEX of the LIF+CRH synergy. The STAT1-3 low-affinity site, in the context of the (-414/-293) subregion of the POMC promoter, was found necessary and sufficient for transcriptional synergism between activated GC receptor (GR) and STAT1-3. Moreover the activities of reporters specific for STAT1-3 or GR were reciprocally enhanced by DEX or LIF. Single and sequential chromatin immunoprecipitations revealed 1) a STAT-dependent corecruitment of coactivators after LIF and LIF+DEX stimulation and 2) a more lasting recruitment of both STAT3 and GR in the same enhanceosome on the endogenous POMC promoter after LIF+DEX joint stimulation than after the single one. Such events may be responsible for a lack of repressive property of GR unmasked on the whole POMC promoter during LIF+CRH stimulation and may contribute to the tonicity of the hypothalamic-pituitary-adrenal axis during inflammatory-infectious diseases.


Assuntos
Dexametasona/farmacologia , Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Fator Inibidor de Leucemia/farmacologia , Pró-Opiomelanocortina/genética , Hormônio Adrenocorticotrópico/metabolismo , Animais , Linhagem Celular , Sinergismo Farmacológico , Expressão Gênica/fisiologia , Adeno-Hipófise/citologia , Adeno-Hipófise/fisiologia , Regiões Promotoras Genéticas/fisiologia , Ratos , Receptores de Glucocorticoides/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
13.
Mol Endocrinol ; 18(12): 2997-3010, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15319449

RESUMO

Leukemia inhibitory factor (LIF) cooperates with CRH at the pituitary level to induce POMC gene transcription, resulting in activation of the pituitary-adrenal axis. However, the underlying molecular mechanisms remain elusive. Here, we show that the NurRE-signal transducers and activators of transcription (STAT) composite element of the POMC promoter was the predominant target of the LIF-CRH synergy. Whereas NurRE or STAT sites alone conferred synergy, the maximal response was found with the NurRE-STAT reporter, suggesting that direct DNA binding of both transcription factors is required for an optimal synergy. During LIF-CRH stimulation, Nur77 and activated STAT1-3 were bound to the composite element, and the binding of each factor was abolished by appropriate mutations. CREB was also detected in this complex in a stimulation-dependent and DNA binding-independent manner. Nur77 and STAT1-3 bound to the NurRE-STAT site were each sufficient for CREB recruitment. Recombinant CREB directly interacted with recombinant Nur77 or STAT1-3. Moreover, CREB-Nur77 interaction was increased by CREB phosphorylation at Ser-133 and the dominant-negative mutant CREB-M1 efficiently inhibited the synergistic LIF-CRH response. This synergism was also inhibited after transfection of CREB-small interfering RNA. We conclude that both CREB phosphorylation at Ser-133 and level of CREB expression are crucial in LIF-CRH synergism where CREB, without direct DNA binding, could improve the stability of Nur77 and STAT1-3 binding to POMC promoter and facilitate the recruitment of coactivators. This novel intrapituitary signaling mechanism may have more general implications in cross talks between cAMP-protein kinase A and Janus kinase-STAT pathways.


Assuntos
Hormônio Liberador da Corticotropina/fisiologia , Proteínas de Ligação a DNA/metabolismo , Interleucina-6/fisiologia , Pró-Opiomelanocortina/genética , Elementos de Resposta/genética , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Hormônio Liberador da Corticotropina/genética , AMP Cíclico/metabolismo , AMP Cíclico/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Interleucina-6/genética , Fator Inibidor de Leucemia , Camundongos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Fosforilação , Hipófise/metabolismo , Hipófise/fisiologia , Regiões Promotoras Genéticas , Conformação Proteica , RNA Interferente Pequeno/genética , Receptores Citoplasmáticos e Nucleares , Receptores de Esteroides , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Fator de Transcrição STAT3 , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Transativadores/fisiologia , Fatores de Transcrição/genética , Regulação para Cima/genética
14.
Endocrinology ; 143(10): 3916-24, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12239103

RESUMO

To better understand how leukemia inhibitory factor (LIF) activates proopiomelanocortin (POMC) gene transcription in pituitary corticotrophs, time-course studies of the induction of POMC promoter activity and specific tyrosine phosphorylation of signal transducer and activator of transcription 1 (STAT1) and STAT3 were performed. It was found that both phosphorylation of STAT1 and -3 and activation of the promoter activity rapidly and transiently take place within minutes and 2-6 h, respectively, in favor of a direct effect of the LIF pathway on POMC promoter. Activated STAT1 and -3 form homo-/heterodimers able to bind the Sis-inducible element. The most abundant Sis-inducible element binding dimers are STAT3/3 and STAT1/3. Degenerated STAT1/3-binding sites from the POMC promoter were tested for their ability to bind activated STAT1 and 3; only the -390/-379 site, partially overlapping the Nur response element, binds with low affinity activated STAT1 and -3. Analysis of the three domains and subregions of the POMC promoter showed that two subregions are specifically responsive to LIF. The response of the distal subregion requires the intact STAT1 and -3 DNA-binding site -390/-379, whereas the responsiveness of the proximal subregion takes place despite the absence of direct STAT1 and -3 DNA binding and may imply interaction of activated STAT with basal transcription factors.


Assuntos
Inibidores do Crescimento/fisiologia , Interleucina-6 , Linfocinas/fisiologia , Pró-Opiomelanocortina/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , Animais , Sequência de Bases/genética , Sítios de Ligação/genética , Linhagem Celular , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Dimerização , Fator Inibidor de Leucemia , Fosforilação , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Fatores de Tempo , Transativadores/metabolismo , Transativadores/fisiologia
15.
Autophagy ; 10(9): 1522-34, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24988326

RESUMO

AMP-activated protein kinase α1 knockout (prkaa1(-/-)) mice manifest splenomegaly and anemia. The underlying molecular mechanisms, however, remain to be established. In this study, we tested the hypothesis that defective autophagy-dependent mitochondrial clearance in prkaa1(-/-) mice exacerbates oxidative stress, thereby enhancing erythrocyte destruction. The levels of ULK1 phosphorylation, autophagical flux, mitochondrial contents, and reactive oxygen species (ROS) were examined in human erythroleukemia cell line, K562 cells, as well as prkaa1(-/-) mouse embryonic fibroblasts and erythrocytes. Deletion of Prkaa1 resulted in the inhibition of ULK1 phosphorylation at Ser555, prevented the formation of ULK1 and BECN1- PtdIns3K complexes, and reduced autophagy capacity. The suppression of autophagy was associated with enhanced damaged mitochondrial accumulation and ROS production. Compared with wild-type (WT) mice, prkaa1(-/-) mice exhibited a shortened erythrocyte life span, hemolytic destruction of erythrocytes, splenomegaly, and anemia, all of which were alleviated by the administration of either rapamycin to activate autophagy or Mito-tempol, a mitochondria-targeted antioxidant, to scavenge mitochondrial ROS. Furthermore, transplantation of WT bone marrow into prkaa1(-/-) mice restored mitochondrial removal, reduced intracellular ROS levels, and normalized hematologic parameters and spleen size. Conversely, transplantation of prkaa1 (-/-) bone marrow into WT mice recapitulated the prkaa1(-/-) mouse phenotypes. We conclude that PRKAA1-dependent autophagy-mediated clearance of damaged mitochondria is required for erythrocyte maturation and homeostasis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/genética , Eritrócitos/citologia , Eritrócitos/metabolismo , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Antioxidantes/farmacologia , Autofagia/efeitos dos fármacos , Linhagem Celular , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo
16.
Endocrinology ; 153(7): 3468-81, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22581459

RESUMO

AMP-activated protein kinase (AMPK), a key regulator of cellular energy homeostasis, is present in metabolic tissues (muscle and liver) and has been identified as a modulator of the female reproductive functions. However, its function in the testis has not yet been clearly defined. We have investigated the potential role of AMPK in male reproduction by using transgenic mice lacking the activity of AMPK catalytic subunit α1 gene [α1AMPK knockout (KO)]. In the testis, the α1AMPK subunit is expressed in germ cells and also in somatic cells (Sertoli and Leydig cells). α1AMPK KO male mice show a decrease in fertility, despite no clear alteration in the testis morphology or sperm production. However, in α1AMPK(-/-) mice, we demonstrate that spermatozoa have structural abnormalities and are less motile than in control mice. These spermatozoa alterations are associated with a 50% decrease in mitochondrial activity, a 60% decrease in basal oxygen consumption, and morphological defects. The α1AMPK KO male mice had high androgen levels associated with a 5- and 3-fold increase in intratesticular cholesterol and testosterone concentrations, respectively. High concentrations of proteins involved in steroid production (3ß-hydroxysteroid dehydrogenase, cytochrome steroid 17 alpha-hydroxylase/17,20 lysate, and steroidogenic acute regulatory protein) were also detected in α1AMPK(-/-) testes. In the pituitary, the LH and FSH concentrations tended to be lower in α1AMPK(-/-) male mice, probably due to the negative feedback of the high testosterone levels. These results suggest that total α1AMPK deficiency in male mice affects androgen production and quality of spermatozoa, leading to a decrease in fertility.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Astenozoospermia/metabolismo , Espermatozoides/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Apoptose , Metabolismo Energético , Feminino , Fertilidade , Receptores X do Fígado , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão/métodos , Modelos Biológicos , Receptores Nucleares Órfãos/metabolismo , Ovário/fisiologia , Consumo de Oxigênio , Espermatozoides/fisiologia , Testículo/metabolismo , beta-Galactosidase/metabolismo
17.
Cell Cycle ; 10(16): 2640-6, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21799304

RESUMO

Nutrition and physical activity have profound effects on skeletal muscle metabolism and growth. Regulation of muscle mass depends on a thin balance between growth-promoting and growth-suppressing factors. Over the past decade, the mammalian target of rapamycin (mTOR) kinase has emerged as an essential factor for muscle growth by mediating the anabolic response to nutrients, insulin, insulin-like growth factors and resistance exercise. As opposed to the mTOR signaling pathway, the AMP-activated protein kinase (AMPK) is switched on during starvation and endurance exercise to upregulate energy-conserving processes. Recent evidence indicates that mTORC1 (mTOR Complex 1) and AMPK represent two antagonistic forces governing muscle adaption to nutrition, starvation and growth stimulation. Animal knockout models with impaired mTORC1 signaling showed decreased muscle mass correlated with increased AMPK activation. Interestingly, AMPK inhibition in p70S6K-deficient muscle cells restores cell growth and sensitivity to nutrients. Conversely, muscle cells lacking AMPK have increased mTORC1 activation with increased cell size and protein synthesis rate. We also demonstrated that the hypertrophic action of MyrAkt is enhanced in AMPK-deficient muscle, indicating that AMPK acts as a negative feedback control to restrain muscle hypertrophy. Our recent results extend this notion by showing that AMPKα1, but not AMPKα2, regulates muscle cell size through the control of mTORC1 signaling. These results reveal the diverse functions of the two catalytic isoforms of AMPK, with AMPKα1 playing a predominant role in the control of muscle cell size and AMPKα2 mediating muscle metabolic adaptation. Thus, the crosstalk between AMPK and mTORC1 signaling is a highly regulated way to control changes in muscle growth and metabolic rate imposed by external cues.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Músculos/citologia , Músculos/metabolismo , Proteínas/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Tamanho Celular/efeitos dos fármacos , Alimentos , Técnicas de Inativação de Genes , Humanos , Hipertrofia/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Atividade Motora , Complexos Multiproteicos , Desenvolvimento Muscular , Músculos/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Proteínas/antagonistas & inibidores , Proteínas/genética , Ribonucleotídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sirolimo/farmacologia , Inanição , Serina-Treonina Quinases TOR
18.
J Clin Invest ; 120(7): 2355-69, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20577053

RESUMO

Metformin is widely used to treat hyperglycemia in individuals with type 2 diabetes. Recently the LKB1/AMP-activated protein kinase (LKB1/AMPK) pathway was proposed to mediate the action of metformin on hepatic gluconeogenesis. However, the molecular mechanism by which this pathway operates had remained elusive. Surprisingly, here we have found that in mice lacking AMPK in the liver, blood glucose levels were comparable to those in wild-type mice, and the hypoglycemic effect of metformin was maintained. Hepatocytes lacking AMPK displayed normal glucose production and gluconeogenic gene expression compared with wild-type hepatocytes. In contrast, gluconeogenesis was upregulated in LKB1-deficient hepatocytes. Metformin decreased expression of the gene encoding the catalytic subunit of glucose-6-phosphatase (G6Pase), while cytosolic phosphoenolpyruvate carboxykinase (Pepck) gene expression was unaffected in wild-type, AMPK-deficient, and LKB1-deficient hepatocytes. Surprisingly, metformin-induced inhibition of glucose production was amplified in both AMPK- and LKB1-deficient compared with wild-type hepatocytes. This inhibition correlated in a dose-dependent manner with a reduction in intracellular ATP content, which is crucial for glucose production. Moreover, metformin-induced inhibition of glucose production was preserved under forced expression of gluconeogenic genes through PPARgamma coactivator 1alpha (PGC-1alpha) overexpression, indicating that metformin suppresses gluconeogenesis via a transcription-independent process. In conclusion, we demonstrate that metformin inhibits hepatic gluconeogenesis in an LKB1- and AMPK-independent manner via a decrease in hepatic energy state.


Assuntos
Gluconeogênese/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Fígado/metabolismo , Metformina/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP , Animais , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Gluconeogênese/genética , Glucose/genética , Glucose/metabolismo , Glucose/farmacologia , Glucose-6-Fosfatase/biossíntese , Glucose-6-Fosfatase/genética , Glucose-6-Fosfatase/metabolismo , Hepatócitos/metabolismo , Hiperglicemia/genética , Hiperglicemia/metabolismo , Hipoglicemiantes/metabolismo , Fígado/efeitos dos fármacos , Metformina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoenolpiruvato Carboxiquinase (ATP)/genética , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/farmacologia , Regulação para Cima/efeitos dos fármacos
19.
FEBS Lett ; 584(16): 3667-71, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20670625

RESUMO

AMP-activated protein kinase (AMPK) plays a pivotal role in regulating cellular energy metabolism. We previously showed that AMPKalpha1-/- mice develop moderate anemia associated with splenomegaly and high reticulocytosis. Here, we report that splenectomy of AMPKalpha1-/- mice worsened anemia supporting evidence that AMPKalpha1-/- mice developed a compensatory response through extramedullary erythropoiesis in the spleen. Transplantation of bone marrow from AMPKalpha1-/- mice into wild-type recipients recapitulated the hematologic phenotype. Further, AMPKalpha1-/- red blood cells (RBC) showed less deformability in response to shear stress limiting their membrane flexibility. Thus, our results highlight the crucial role of AMPK to preserve RBC integrity.


Assuntos
Proteínas Quinases Ativadas por AMP/sangue , Deformação Eritrocítica/fisiologia , Proteínas Quinases Ativadas por AMP/deficiência , Proteínas Quinases Ativadas por AMP/genética , Anemia/sangue , Anemia/enzimologia , Anemia/genética , Animais , Transplante de Medula Óssea , Deformação Eritrocítica/genética , Eritropoese , Hematopoese Extramedular , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragilidade Osmótica , Esplenectomia , Esplenomegalia/sangue , Esplenomegalia/enzimologia , Esplenomegalia/genética
20.
Front Biosci (Landmark Ed) ; 14(9): 3380-400, 2009 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-19273282

RESUMO

Type 2 diabetes is one of the fastest growing public health problems worldwide, resulting from both genetic factors and inadequate adaptation to environmental changes. It is characterized by abnormal glucose and lipid metabolism due in part to resistance to the actions of insulin in skeletal muscle, liver and fat. AMP-activated protein kinase (AMPK), a phylogenetically conserved serine/threonine protein kinase, acts as an integrator of regulatory signals monitoring systemic and cellular energy status. The growing realization that AMPK regulates the coordination of anabolic and catabolic metabolic processes represents an attractive concept for type 2 diabetes therapy. Recent findings showing that pharmacological activation of AMPK improves blood glucose homeostasis, lipid profile and blood pressure in insulin-resistant rodents suggest that this kinase could be a novel therapeutic target in the treatment of type 2 diabetes. Consistent with these results, physical exercise and major classes of antidiabetic drugs have recently been reported to activate AMPK. In the present review, we update these topics and discuss the concept of targeting the AMPK pathway for the treatment of type 2 diabetes.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Diabetes Mellitus Tipo 2/terapia , Proteínas Quinases Ativadas por AMP/química , Restrição Calórica , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/terapia , Diabetes Mellitus Tipo 2/enzimologia , Ativação Enzimática , Exercício Físico , Glucose/metabolismo , Homeostase , Humanos , Ilhotas Pancreáticas/fisiopatologia , Metabolismo dos Lipídeos , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA