Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Biol ; 436(4): 168432, 2024 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-38161000

RESUMO

Helicobacter pylori colonizes the stomach in about half of the human population, leading to an increased risk of peptic ulcer disease and gastric cancer. H. pylori secretes an 88 kDa VacA toxin that contributes to pathogenesis. VacA assembles into oligomeric complexes in solution and forms anion-selective channels in cell membranes. Cryo-electron microscopy (cryo-EM) analyses of VacA oligomers in solution provided insights into VacA oligomerization but failed to reveal the structure of the hydrophobic N-terminal region predicted to be a pore-forming domain. In this study, we incubated VacA with liposomes and used single particle cryo-EM to analyze detergent-extracted VacA oligomers. A 3D structure of detergent-solubilized VacA hexamers revealed the presence of six α-helices extending from the center of the oligomers, a feature not observed in previous studies of water-soluble VacA oligomers. Cryo-electron tomography analysis and 2D averages of VacA associated with liposomes confirmed that central regions of the membrane-associated VacA oligomers can insert into the lipid bilayer. However, insertion is heterogenous, with some membrane-associated oligomers appearing only partially inserted and others sitting on top of the bilayer. These studies indicate that VacA undergoes a conformational change when contacting the membrane and reveal an α-helical region positioned to extend into the membrane. Although the reported VacA 3D structure does not represent a selective anion channel, our combined single particle 3D analysis, cryo-electron tomography, and modeling allow us to propose a model for the structural organization of the VacA N-terminus in the context of a hexamer as it inserts into the membrane.


Assuntos
Proteínas de Bactérias , Helicobacter pylori , Toxinas Biológicas , Canais de Ânion Dependentes de Voltagem , Humanos , Proteínas de Bactérias/química , Microscopia Crioeletrônica/métodos , Detergentes , Helicobacter pylori/química , Lipossomos/química , Toxinas Biológicas/química , Canais de Ânion Dependentes de Voltagem/química , Multimerização Proteica
2.
Biochemistry ; 62(11): 1725-1734, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37130292

RESUMO

Dicer is an RNase III enzyme that is responsible for the maturation of small RNAs such as microRNAs. As Dicer's cleavage products play key roles in promoting cellular homeostasis through the fine-tuning of gene expression, dysregulation of Dicer activity can lead to several human diseases, including cancers. Mutations in Dicer have been found to induce tumorigenesis and lead to the development of a rare pleiotropic tumor predisposition syndrome found in children and young adults called DICER1 syndrome. These patients harbor germline and somatic mutations in Dicer that lead to defective microRNA processing and activity. While most mutations occur within Dicer's catalytic RNase III domains, alterations within the Platform-PAZ (Piwi-Argonaute-Zwille) domain also cause loss of microRNA production. Using a combination of in vitro biochemical and cellular studies, we characterized the effect of disease-relevant Platform-PAZ-associated mutations on the processing of a well-studied oncogenic microRNA, pre-microRNA-21. We then compared these results to those of a representative from another Dicer substrate class, the small nucleolar RNA, snord37. From this analysis, we provide evidence that mutations within the Platform-PAZ domain result in differential impacts on RNA binding and processing, adding new insights into the complexities of Dicer processing of small RNA substrates.


Assuntos
MicroRNAs , RNA Nucleolar Pequeno , Criança , Humanos , RNA Nucleolar Pequeno/genética , Ribonuclease III/química , MicroRNAs/química , Mutação , RNA Helicases DEAD-box/genética
3.
J Biol Chem ; 299(4): 104574, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36870682

RESUMO

Caveolin-1 (CAV1) is a membrane-sculpting protein that oligomerizes to generate flask-shaped invaginations of the plasma membrane known as caveolae. Mutations in CAV1 have been linked to multiple diseases in humans. Such mutations often interfere with oligomerization and the intracellular trafficking processes required for successful caveolae assembly, but the molecular mechanisms underlying these defects have not been structurally explained. Here, we investigate how a disease-associated mutation in one of the most highly conserved residues in CAV1, P132L, affects CAV1 structure and oligomerization. We show that P132 is positioned at a major site of protomer-protomer interactions within the CAV1 complex, providing a structural explanation for why the mutant protein fails to homo-oligomerize correctly. Using a combination of computational, structural, biochemical, and cell biological approaches, we find that despite its homo-oligomerization defects P132L is capable of forming mixed hetero-oligomeric complexes with WT CAV1 and that these complexes can be incorporated into caveolae. These findings provide insights into the fundamental mechanisms that control the formation of homo- and hetero-oligomers of caveolins that are essential for caveolae biogenesis, as well as how these processes are disrupted in human disease.


Assuntos
Caveolina 1 , Caveolinas , Doença , Humanos , Cavéolas/metabolismo , Caveolina 1/genética , Caveolina 1/metabolismo , Caveolinas/metabolismo , Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Mutação , Subunidades Proteicas/metabolismo , Doença/genética
4.
Elife ; 102021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34519271

RESUMO

Legionella pneumophila is an opportunistic pathogen that causes the potentially fatal pneumonia known as Legionnaires' disease. The pathology associated with infection depends on bacterial delivery of effector proteins into the host via the membrane spanning Dot/Icm type IV secretion system (T4SS). We have determined sub-3.0 Å resolution maps of the Dot/Icm T4SS core complex by single particle cryo-EM. The high-resolution structural analysis has allowed us to identify proteins encoded outside the Dot/Icm genetic locus that contribute to the core T4SS structure. We can also now define two distinct areas of symmetry mismatch, one that connects the C18 periplasmic ring (PR) and the C13 outer membrane cap (OMC) and one that connects the C13 OMC with a 16-fold symmetric dome. Unexpectedly, the connection between the PR and OMC is DotH, with five copies sandwiched between the OMC and PR to accommodate the symmetry mismatch. Finally, we observe multiple conformations in the reconstructions that indicate flexibility within the structure.


Assuntos
Proteínas de Bactérias/isolamento & purificação , Microscopia Crioeletrônica/métodos , Legionella pneumophila/química , Proteínas de Bactérias/química , Conformação Proteica , Especificidade da Espécie , Sistemas de Secreção Tipo IV/química
5.
J Mol Biol ; 433(16): 166909, 2021 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-33676924

RESUMO

Structural studies of membrane proteins, especially small membrane proteins, are associated with well-known experimental challenges. Complexation with monoclonal antibody fragments is a common strategy to augment such proteins; however, generating antibody fragments that specifically bind a target protein is not trivial. Here we identify a helical epitope, from the membrane-proximal external region (MPER) of the gp41-transmembrane subunit of the HIV envelope protein, that is recognized by several well-characterized antibodies and that can be fused as a contiguous extension of the N-terminal transmembrane helix of a broad range of membrane proteins. To analyze whether this MPER-epitope tag might aid structural studies of small membrane proteins, we determined an X-ray crystal structure of a membrane protein target that does not crystallize without the aid of crystallization chaperones, the Fluc fluoride channel, fused to the MPER epitope and in complex with antibody. We also demonstrate the utility of this approach for single particle electron microscopy with Fluc and two additional small membrane proteins that represent different membrane protein folds, AdiC and GlpF. These studies show that the MPER epitope provides a structurally defined, rigid docking site for antibody fragments that is transferable among diverse membrane proteins and can be engineered without prior structural information. Antibodies that bind to the MPER epitope serve as effective crystallization chaperones and electron microscopy fiducial markers, enabling structural studies of challenging small membrane proteins.


Assuntos
Epitopos/química , Proteínas de Membrana/química , Modelos Moleculares , Domínios e Motivos de Interação entre Proteínas , Cristalografia por Raios X , Epitopos/imunologia , Humanos , Proteínas de Membrana/imunologia , Microscopia Eletrônica , Conformação Proteica , Relação Estrutura-Atividade
6.
J Control Release ; 330: 529-539, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33358977

RESUMO

The current health crisis of corona virus disease 2019 (COVID-19) highlights the urgent need for vaccine systems that can generate potent and protective immune responses. Protein vaccines are safe, but conventional approaches for protein-based vaccines often fail to elicit potent and long-lasting immune responses. Nanoparticle vaccines designed to co-deliver protein antigens and adjuvants can promote their delivery to antigen-presenting cells and improve immunogenicity. However, it remains challenging to develop vaccine nanoparticles that can preserve and present conformational epitopes of protein antigens for induction of neutralizing antibody responses. Here, we have designed a new lipid-based nanoparticle vaccine platform (NVP) that presents viral proteins (HIV-1 and SARS-CoV-2 antigens) in a conformational manner for induction of antigen-specific antibody responses. We show that NVP was readily taken up by dendritic cells (DCs) and promoted DC maturation and antigen presentation. NVP loaded with BG505.SOSIP.664 (SOSIP) or SARS-CoV-2 receptor-binding domain (RBD) was readily recognized by neutralizing antibodies, indicating the conformational display of antigens on the surfaces of NVP. Rabbits immunized with SOSIP-NVP elicited strong neutralizing antibody responses against HIV-1. Furthermore, mice immunized with RBD-NVP induced robust and long-lasting antibody responses against RBD from SARS-CoV-2. These results suggest that NVP is a promising platform technology for vaccination against infectious pathogens.


Assuntos
Vacinas contra a AIDS/química , Vacinas contra COVID-19/química , Imunidade Humoral/efeitos dos fármacos , Lipídeos/química , Nanopartículas , Vacinas Virais/química , Vacinas contra a AIDS/administração & dosagem , Adjuvantes Imunológicos , Animais , Apresentação de Antígeno , Reações Antígeno-Anticorpo , Vacinas contra COVID-19/administração & dosagem , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , HIV-1 , Humanos , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , SARS-CoV-2 , Vacinas Virais/administração & dosagem
7.
Br J Pharmacol ; 176(11): 1649-1663, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30710458

RESUMO

BACKGROUND AND PURPOSE: The δ-opioid receptor is an emerging target for the management of chronic pain and depression. Biased signalling, the preferential activation of one signalling pathway over another downstream of δ-receptors, may generate better therapeutic profiles. BMS 986187 is a positive allosteric modulator of δ-receptors. Here, we ask if BMS 986187 can directly activate the receptor from an allosteric site, without an orthosteric ligand, and if a signalling bias is generated. EXPERIMENTAL APPROACH: We used several clonal cell lines expressing δ-receptors, to assess effects of BMS 986187 on events downstream of δ-receptors by measuring G-protein activation, ß-arrestin 2 recruitment, receptor phosphorylation, loss of surface receptor expression, ERK1/ERK2 phosphorylation, and receptor desensitization. KEY RESULTS: BMS 986187 is a G protein biased allosteric agonist, relative to ß-arrestin 2 recruitment. Despite showing direct and potent G protein activation, BMS 986187 has a low potency to recruit ß-arrestin 2. This appears to reflect the inability of BMS 986187 to elicit any significant receptor phosphorylation, consistent with low receptor internalization and a slower onset of desensitization, compared with the full agonist SNC80. CONCLUSIONS AND IMPLICATIONS: This is the first evidence of biased agonism mediated through direct binding to an allosteric site on an opioid receptor, without a ligand at the orthosteric site. Our data suggest that agonists targeting δ-receptors, or indeed any GPCR, through allosteric sites may be a novel way to promote signalling bias and thereby potentially produce a more specific pharmacology than can be observed by activation via the orthosteric site.


Assuntos
Proteínas de Ligação ao GTP/agonistas , Receptores Opioides delta/metabolismo , Xantonas/farmacologia , Sítio Alostérico , Animais , Células CHO , Cricetulus , Proteínas de Ligação ao GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos
8.
Elife ; 72018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30479275

RESUMO

Lecithin:cholesterol acyltransferase (LCAT) and LCAT-activating compounds are being investigated as treatments for coronary heart disease (CHD) and familial LCAT deficiency (FLD). Herein we report the crystal structure of human LCAT in complex with a potent piperidinylpyrazolopyridine activator and an acyl intermediate-like inhibitor, revealing LCAT in an active conformation. Unlike other LCAT activators, the piperidinylpyrazolopyridine activator binds exclusively to the membrane-binding domain (MBD). Functional studies indicate that the compound does not modulate the affinity of LCAT for HDL, but instead stabilizes residues in the MBD and facilitates channeling of substrates into the active site. By demonstrating that these activators increase the activity of an FLD variant, we show that compounds targeting the MBD have therapeutic potential. Our data better define the substrate binding site of LCAT and pave the way for rational design of LCAT agonists and improved biotherapeutics for augmenting or restoring reverse cholesterol transport in CHD and FLD patients.


Assuntos
HDL-Colesterol/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferase/metabolismo , Domínio Catalítico , Ativadores de Enzimas/química , Ativadores de Enzimas/farmacologia , Estabilidade Enzimática/efeitos dos fármacos , Células HEK293 , Humanos , Lipídeos de Membrana/metabolismo , Mutação/genética , Fosfatidilcolina-Esterol O-Aciltransferase/química , Conformação Proteica , Eletricidade Estática , Relação Estrutura-Atividade
9.
Bioorg Med Chem ; 26(20): 5443-5461, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30270002

RESUMO

The non-canonical IκB kinases TANK-binding kinase 1 (TBK1) and inhibitor of nuclear factor kappa-B kinase ε (IKKε) play a key role in insulin-independent pathways that promote energy storage and block adaptive energy expenditure during obesity. Utilizing docking calculations and the x-ray structure of TBK1 bound to amlexanox, an inhibitor of these kinases with modest potency, a series of analogues was synthesized to develop a structure activity relationship (SAR) around the A- and C-rings of the core scaffold. A strategy was developed wherein R7 and R8 A-ring substituents were incorporated late in the synthetic sequence by utilizing palladium-catalyzed cross-coupling reactions on appropriate bromo precursors. Analogues display IC50 values as low as 210 nM and reveal A-ring substituents that enhance selectivity toward either kinase. In cell assays, selected analogues display enhanced phosphorylation of p38 or TBK1 and elicited IL-6 secretion in 3T3-L1 adipocytes better than amlexanox. An analogue bearing a R7 cyclohexyl modification demonstrated robust IL-6 production in 3T3-L1 cells as well as a phosphorylation marker of efficacy and was tested in obese mice where it promoted serum IL-6 response, weight loss, and insulin sensitizing effects comparable to amlexanox. These studies provide impetus to expand the SAR around the amlexanox core toward uncovering analogues with development potential.


Assuntos
Quinase I-kappa B/antagonistas & inibidores , Obesidade/tratamento farmacológico , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Piridinas/química , Piridinas/farmacologia , Células 3T3-L1 , Aminação , Animais , Fármacos Antiobesidade/síntese química , Fármacos Antiobesidade/química , Fármacos Antiobesidade/farmacologia , Fármacos Antiobesidade/uso terapêutico , Cromanos/síntese química , Cromanos/química , Cromanos/farmacologia , Cromanos/uso terapêutico , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Quinase I-kappa B/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Obesidade/metabolismo , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/metabolismo , Piridinas/síntese química , Piridinas/uso terapêutico
10.
Mol Pharmacol ; 94(4): 1210-1219, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30082428

RESUMO

Chronic low-grade inflammation is a hallmark of obesity, which is a risk factor for the development of type 2 diabetes. The drug amlexanox inhibits IκB kinase ε (IKKε) and TANK binding kinase 1 (TBK1) to promote energy expenditure and improve insulin sensitivity. Clinical studies have demonstrated efficacy in a subset of diabetic patients with underlying adipose tissue inflammation, albeit with moderate potency, necessitating the need for improved analogs. Herein we report crystal structures of TBK1 in complex with amlexanox and a series of analogs that modify its carboxylic acid moiety. Removal of the carboxylic acid or mutation of the adjacent Thr156 residue significantly reduces potency toward TBK1, whereas conversion to a short amide or ester nearly abolishes the inhibitory effects. IKKε is less affected by these modifications, possibly due to variation in its hinge that allows for increased conformational plasticity. Installation of a tetrazole carboxylic acid bioisostere improved potency to 200 and 400 nM toward IKKε and TBK1, respectively. Despite improvements in the in vitro potency, no analog produced a greater response in adipocytes than amlexanox, perhaps because of altered absorption and distribution. The structure-activity relationships and cocrystal structures described herein will aid in future structure-guided inhibitor development using the amlexanox pharmacophore for the treatment of obesity and type 2 diabetes.


Assuntos
Aminopiridinas/farmacologia , Ácidos Carboxílicos/farmacologia , Quinase I-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Linhagem Celular , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Relação Estrutura-Atividade
11.
J Biol Chem ; 292(49): 20313-20327, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29030428

RESUMO

Lecithin:cholesterol acyltransferase (LCAT) plays a key role in reverse cholesterol transport by transferring an acyl group from phosphatidylcholine to cholesterol, promoting the maturation of high-density lipoproteins (HDL) from discoidal to spherical particles. LCAT is activated through an unknown mechanism by apolipoprotein A-I (apoA-I) and other mimetic peptides that form a belt around HDL. Here, we report the crystal structure of LCAT with an extended lid that blocks access to the active site, consistent with an inactive conformation. Residues Thr-123 and Phe-382 in the catalytic domain form a latch-like interaction with hydrophobic residues in the lid. Because these residues are mutated in genetic disease, lid displacement was hypothesized to be an important feature of apoA-I activation. Functional studies of site-directed mutants revealed that loss of latch interactions or the entire lid enhanced activity against soluble ester substrates, and hydrogen-deuterium exchange (HDX) mass spectrometry revealed that the LCAT lid is extremely dynamic in solution. Upon addition of a covalent inhibitor that mimics one of the reaction intermediates, there is an overall decrease in HDX in the lid and adjacent regions of the protein, consistent with ordering. These data suggest a model wherein the active site of LCAT is shielded from soluble substrates by a dynamic lid until it interacts with HDL to allow transesterification to proceed.


Assuntos
Apolipoproteína A-I/fisiologia , Fosfatidilcolina-Esterol O-Aciltransferase/química , Domínio Catalítico , Cristalografia por Raios X , Medição da Troca de Deutério , Ativação Enzimática , Humanos , Lipoproteínas HDL/metabolismo , Mutagênese Sítio-Dirigida , Fosfatidilcolina-Esterol O-Aciltransferase/metabolismo , Conformação Proteica
12.
Sci Signal ; 10(471)2017 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-28325821

RESUMO

Insulin stimulates glucose uptake through the translocation of the glucose transporter GLUT4 to the plasma membrane. The exocyst complex tethers GLUT4-containing vesicles to the plasma membrane, a process that requires the binding of the G protein (heterotrimeric guanine nucleotide-binding protein) RalA to the exocyst complex. We report that upon activation of RalA, the protein kinase TBK1 phosphorylated the exocyst subunit Exo84. Knockdown of TBK1 blocked insulin-stimulated glucose uptake and GLUT4 translocation; knockout of TBK1 in adipocytes blocked insulin-stimulated glucose uptake; and ectopic overexpression of a kinase-inactive mutant of TBK1 reduced insulin-stimulated glucose uptake in 3T3-L1 adipocytes. The phosphorylation of Exo84 by TBK1 reduced its affinity for RalA and enabled its release from the exocyst. Overexpression of a kinase-inactive mutant of TBK1 blocked the dissociation of the TBK1/RalA/exocyst complex, and treatment of 3T3-L1 adipocytes with specific inhibitors of TBK1 reduced the rate of complex dissociation. Introduction of phosphorylation-mimicking or nonphosphorylatable mutant forms of Exo84 blocked insulin-stimulated GLUT4 translocation. Thus, these data indicate that TBK1 controls GLUT4 vesicle engagement and disengagement from the exocyst, suggesting that exocyst components not only constitute a tethering complex for the GLUT4 vesicle but also act as "gatekeepers" controlling vesicle fusion at the plasma membrane.


Assuntos
Adipócitos/efeitos dos fármacos , Transportador de Glucose Tipo 4/metabolismo , Insulina/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Transportador de Glucose Tipo 4/genética , Hipoglicemiantes/farmacologia , Immunoblotting , Camundongos , Mutação , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico/efeitos dos fármacos , Interferência de RNA , Proteínas de Transporte Vesicular/genética , Proteínas ral de Ligação ao GTP/genética , Proteínas ral de Ligação ao GTP/metabolismo
13.
Nat Commun ; 6: 6047, 2015 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-25581158

RESUMO

The search for effective treatments for obesity and its comorbidities is of prime importance. We previously identified IKK-ε and TBK1 as promising therapeutic targets for the treatment of obesity and associated insulin resistance. Here we show that acute inhibition of IKK-ε and TBK1 with amlexanox treatment increases cAMP levels in subcutaneous adipose depots of obese mice, promoting the synthesis and secretion of the cytokine IL-6 from adipocytes and preadipocytes, but not from macrophages. IL-6, in turn, stimulates the phosphorylation of hepatic Stat3 to suppress expression of genes involved in gluconeogenesis, in the process improving glucose handling in obese mice. Preliminary data in a small cohort of obese patients show a similar association. These data support an important role for a subcutaneous adipose tissue-liver axis in mediating the acute metabolic benefits of amlexanox on glucose metabolism, and point to a new therapeutic pathway for type 2 diabetes.


Assuntos
Gluconeogênese , Fígado/metabolismo , Transdução de Sinais , Gordura Subcutânea/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adulto , Idoso , Aminopiridinas/farmacologia , Animais , AMP Cíclico/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Gluconeogênese/efeitos dos fármacos , Glucose-6-Fosfatase/metabolismo , Humanos , Inflamação/patologia , Resistência à Insulina , Interleucina-6/metabolismo , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Receptores Adrenérgicos beta/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Gordura Subcutânea/efeitos dos fármacos , Adulto Jovem , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Diabetes ; 63(9): 2935-48, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24722244

RESUMO

Glycogen and lipids are major storage forms of energy that are tightly regulated by hormones and metabolic signals. We demonstrate that feeding mice a high-fat diet (HFD) increases hepatic glycogen due to increased expression of the glycogenic scaffolding protein PTG/R5. PTG promoter activity was increased and glycogen levels were augmented in mice and cells after activation of the mechanistic target of rapamycin complex 1 (mTORC1) and its downstream target SREBP1. Deletion of the PTG gene in mice prevented HFD-induced hepatic glycogen accumulation. Of note, PTG deletion also blocked hepatic steatosis in HFD-fed mice and reduced the expression of numerous lipogenic genes. Additionally, PTG deletion reduced fasting glucose and insulin levels in obese mice while improving insulin sensitivity, a result of reduced hepatic glucose output. This metabolic crosstalk was due to decreased mTORC1 and SREBP activity in PTG knockout mice or knockdown cells, suggesting a positive feedback loop in which once accumulated, glycogen stimulates the mTORC1/SREBP1 pathway to shift energy storage to lipogenesis. Together, these data reveal a previously unappreciated broad role for glycogen in the control of energy homeostasis.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Metabolismo dos Lipídeos , Glicogênio Hepático/metabolismo , Obesidade/metabolismo , Animais , Dieta Hiperlipídica , Metabolismo Energético/fisiologia , Retroalimentação , Glicogênio/metabolismo , Células HEK293 , Humanos , Resistência à Insulina , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Lipogênese , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Serina-Treonina Quinases TOR/metabolismo
15.
Elife ; 2: e01119, 2013 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-24368730

RESUMO

Obesity produces a chronic inflammatory state involving the NFκB pathway, resulting in persistent elevation of the noncanonical IκB kinases IKKε and TBK1. In this study, we report that these kinases attenuate ß-adrenergic signaling in white adipose tissue. Treatment of 3T3-L1 adipocytes with specific inhibitors of these kinases restored ß-adrenergic signaling and lipolysis attenuated by TNFα and Poly (I:C). Conversely, overexpression of the kinases reduced induction of Ucp1, lipolysis, cAMP levels, and phosphorylation of hormone sensitive lipase in response to isoproterenol or forskolin. Noncanonical IKKs reduce catecholamine sensitivity by phosphorylating and activating the major adipocyte phosphodiesterase PDE3B. In vivo inhibition of these kinases by treatment of obese mice with the drug amlexanox reversed obesity-induced catecholamine resistance, and restored PKA signaling in response to injection of a ß-3 adrenergic agonist. These studies suggest that by reducing production of cAMP in adipocytes, IKKε and TBK1 may contribute to the repression of energy expenditure during obesity. DOI: http://dx.doi.org/10.7554/eLife.01119.001.


Assuntos
Adipócitos/enzimologia , Tecido Adiposo Branco/enzimologia , Catecolaminas/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Quinase I-kappa B/metabolismo , Inflamação/enzimologia , Obesidade/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Tecido Adiposo Branco/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos beta 3/farmacologia , Aminopiridinas/farmacologia , Animais , Células COS , Chlorocebus aethiops , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/genética , Dioxóis/farmacologia , Modelos Animais de Doenças , Metabolismo Energético , Ativação Enzimática , Células HEK293 , Humanos , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/genética , Inflamação/genética , Canais Iônicos/metabolismo , Lipólise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/metabolismo , Obesidade/genética , Fosforilação , Poli I-C/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Esterol Esterase/metabolismo , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Proteína Desacopladora 1
16.
Nat Med ; 19(3): 313-21, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23396211

RESUMO

Emerging evidence suggests that inflammation provides a link between obesity and insulin resistance. The noncanonical IκB kinases IKK-ɛ and TANK-binding kinase 1 (TBK1) are induced in liver and fat by NF-κB activation upon high-fat diet feeding and in turn initiate a program of counterinflammation that preserves energy storage. Here we report that amlexanox, an approved small-molecule therapeutic presently used in the clinic to treat aphthous ulcers and asthma, is an inhibitor of these kinases. Treatment of obese mice with amlexanox elevates energy expenditure through increased thermogenesis, producing weight loss, improved insulin sensitivity and decreased steatosis. Because of its record of safety in patients, amlexanox may be an interesting candidate for clinical evaluation in the treatment of obesity and related disorders.


Assuntos
Aminopiridinas/farmacologia , Fármacos Antiobesidade/farmacologia , Metabolismo Energético/efeitos dos fármacos , Quinase I-kappa B/antagonistas & inibidores , Resistência à Insulina , Obesidade/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Antialérgicos/farmacologia , Linhagem Celular , Dieta Hiperlipídica , Ativação Enzimática , Fígado Gorduroso/tratamento farmacológico , Transtornos do Metabolismo de Glucose/tratamento farmacológico , Quinase I-kappa B/metabolismo , Resistência à Insulina/imunologia , Gordura Intra-Abdominal/efeitos dos fármacos , Gordura Intra-Abdominal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , NF-kappa B/metabolismo , Obesidade/tratamento farmacológico , Obesidade/imunologia , Consumo de Oxigênio/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Redução de Peso/efeitos dos fármacos
17.
PLoS One ; 7(8): e42451, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22900022

RESUMO

BACKGROUND: TC10 is a small GTPase found in lipid raft microdomains of adipocytes. The protein undergoes activation in response to insulin, and plays a key role in the regulation of glucose uptake by the hormone. METHODOLOGY/PRINCIPAL FINDINGS: TC10 requires high concentrations of magnesium in order to stabilize guanine nucleotide binding. Kinetic analysis of this process revealed that magnesium acutely decreased the nucleotide release and exchange rates of TC10, suggesting that the G protein may behave as a rapidly exchanging, and therefore active protein in vivo. However, in adipocytes, the activity of TC10 is not constitutive, indicating that mechanisms must exist to maintain the G protein in a low activity state in untreated cells. Thus, we searched for proteins that might bind to and stabilize TC10 in the inactive state. We found that Caveolin interacts with TC10 only when GDP-bound and stabilizes GDP binding. Moreover, knockdown of Caveolin 1 in 3T3-L1 adipocytes increased the basal activity state of TC10. CONCLUSIONS/SIGNIFICANCE: Together these data suggest that TC10 is intrinsically active in vivo, but is maintained in the inactive state by binding to Caveolin 1 in 3T3-L1 adipocytes under basal conditions, permitting its activation by insulin.


Assuntos
Adipócitos/metabolismo , Caveolinas/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Células 3T3-L1 , Animais , Sítios de Ligação , Caveolina 1/metabolismo , Caveolinas/genética , Estabilidade Enzimática , Guanosina Difosfato/metabolismo , Magnésio/química , Magnésio/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Nucleotídeos/metabolismo , Ligação Proteica , Interferência de RNA , Proteína cdc42 de Ligação ao GTP/química , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/química
18.
J Biol Chem ; 282(22): 16553-66, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17412693

RESUMO

In the process of insulin-stimulated GLUT4 vesicle exocytosis, Munc18c has been proposed to control SNARE complex formation by inactivating syntaxin 4 in a self-associated conformation. Using in vivo fluorescence resonance energy transfer in 3T3L1 adipocytes, co-immunoprecipitation, and in vitro binding assays, we provide data to indicate that Munc18c also associates with nearly equal affinity to a mutant of syntaxin 4 in a constitutively open (unfolded) state (L173A/E174A; LE). To bind to the open conformation of syntaxin 4, we found that Munc18c requires an interaction with the N terminus of syntaxin 4, which resembles Sly1 interaction with the N terminus of ER/Golgi syntaxins. However, both N and C termini of syntaxin 4 are required for Munc18c binding, since a mutation in the syntaxin 4 SNARE domain (I241A) reduces the interaction, irrespective of syntaxin 4 conformation. Using an optical reporter for syntaxin 4-SNARE pairings in vivo, we demonstrate that Munc18c blocks recruitment of SNAP23 to wild type syntaxin 4 yet associates with syntaxin 4LE-SNAP23 Q-SNARE complexes. Fluorescent imaging of GLUT4 vesicles in 3T3L1 adipocytes revealed that syntaxin 4LE expressed with Munc18c bypasses the requirement of insulin for GLUT4 vesicle plasma membrane docking. This effect was attenuated by reducing the Munc18c-syntaxin 4LE interaction with the I241A mutation, indicating that Munc18c facilitates vesicle docking. Therefore, in contradiction to previous models, our data indicates that the conformational "opening" of syntaxin 4 rather than the dissociation of Munc18c is the critical event required for GLUT4 vesicle docking.


Assuntos
Adipócitos/metabolismo , Vesículas Citoplasmáticas/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Munc18/metabolismo , Proteínas Q-SNARE/metabolismo , Proteínas Qa-SNARE/metabolismo , Células 3T3 , Adipócitos/citologia , Animais , Transporte Biológico Ativo/fisiologia , Vesículas Citoplasmáticas/genética , Transportador de Glucose Tipo 4/genética , Camundongos , Modelos Biológicos , Complexos Multiproteicos/genética , Proteínas Munc18/genética , Mutação , Dobramento de Proteína , Proteínas Q-SNARE/genética , Proteínas Qa-SNARE/genética , Proteínas Qb-SNARE , Proteínas Qc-SNARE
19.
Endocrinology ; 148(1): 27-33, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17008399

RESUMO

Previous studies have suggested that activation of the Rho family member GTPase TC10 is necessary but not sufficient for the stimulation of glucose transport by insulin. We show here that endogenous TC10alpha is rapidly activated in response to insulin in 3T3L1 adipocytes in a phosphatidylinositol 3-kinase-independent manner, whereas platelet-derived growth factor was without effect. Knockdown of TC10alpha but not TC10beta by RNA interference inhibited insulin-stimulated glucose uptake as well as the translocation of the insulin-sensitive glucose transporter GLUT4 from intracellular sites to the plasma membrane. In contrast, loss of TC10alpha had no effect on the stimulation of Akt by insulin. Additionally, knockdown of TC10alpha inhibited insulin-stimulated translocation of its effector CIP4. These data indicate that TC10alpha is specifically required for insulin-stimulated glucose uptake in adipocytes.


Assuntos
Adipócitos/metabolismo , Glucose/farmacocinética , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Proteínas rho de Ligação ao GTP/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Animais , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Antígenos de Histocompatibilidade Menor , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas rho de Ligação ao GTP/genética
20.
Cell Metab ; 5(1): 59-72, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17189207

RESUMO

Insulin stimulates glucose uptake by promoting translocation of the Glut4 glucose transporter from intracellular storage compartments to the plasma membrane. In the absence of insulin, Glut4 is retained intracellularly; the mechanism underlying this process remains uncertain. Using the TC10-interacting protein CIP4 as bait in a yeast two-hybrid screen, we cloned a RasGAP and VPS9 domain-containing protein, Gapex-5/RME-6. The VPS9 domain is a guanine nucleotide exchange factor for Rab31, a Rab5 subfamily GTPase implicated in trans-Golgi network (TGN)-to-endosome trafficking. Overexpression of Rab31 blocks insulin-stimulated Glut4 translocation, whereas knockdown of Rab31 potentiates insulin-stimulated Glut4 translocation and glucose uptake. Gapex-5 is predominantly cytosolic in untreated cells; its overexpression promotes intracellular retention of Glut4 in adipocytes. Insulin recruits the CIP4/Gapex-5 complex to the plasma membrane, thus reducing Rab31 activity and permitting Glut4 vesicles to translocate to the cell surface, where Glut4 docks and fuses to transport glucose into the cell.


Assuntos
Adipócitos/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Membrana Celular/metabolismo , Fibroblastos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/química , Insulina/metabolismo , Camundongos , Antígenos de Histocompatibilidade Menor , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Transporte Proteico , Alinhamento de Sequência , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...