Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
SLAS Discov ; 26(9): 1177-1188, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34112017

RESUMO

Regulators of G protein signaling (RGS) proteins serve as critical regulatory nodes to limit the lifetime and extent of signaling via G protein-coupled receptors (GPCRs). Previously, approaches to pharmacologically inhibit RGS activity have mostly focused on the inhibition of GTPase activity by interrupting the interaction of RGS proteins with the G proteins they regulate. However, several RGS proteins are also regulated by association with binding partners. A notable example is the mammalian RGS7 protein, which has prominent roles in metabolic control, vision, reward, and actions of opioid analgesics. In vivo, RGS7 exists in complex with the binding partners type 5 G protein ß subunit (Gß5) and R7 binding protein (R7BP), which control its stability and activity, respectively. Targeting the whole RGS7/Gß5/R7BP protein complex affords the opportunity to allosterically tune opioid receptor signaling following opioid engagement while potentially bypassing undesirable side effects. Hence, we implemented a novel strategy to pharmacologically target the interaction between RGS7/Gß5 and R7BP. To do so, we searched for protein complex inhibitors using a time-resolved fluorescence resonance energy transfer (FRET)-based high-throughput screening (HTS) assay that measures compound-mediated alterations in the FRET signal between RGS7/Gß5 and R7BP. We performed two HTS campaigns, each screening ~100,000 compounds from the Scripps Drug Discovery Library (SDDL). Each screen yielded more than 100 inhibitors, which will be described herein.


Assuntos
Descoberta de Drogas , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas RGS/metabolismo , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Humanos , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/antagonistas & inibidores , Ligação Proteica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas
2.
J Clin Invest ; 130(3): 1168-1184, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32039920

RESUMO

Dopamine receptor D1 modulates glutamatergic transmission in cortico-basal ganglia circuits and represents a major target of L-DOPA therapy in Parkinson's disease. Here we show that D1 and metabotropic glutamate type 5 (mGlu5) receptors can form previously unknown heteromeric entities with distinctive functional properties. Interacting with Gq proteins, cell-surface D1-mGlu5 heteromers exacerbated PLC signaling and intracellular calcium release in response to either glutamate or dopamine. In rodent models of Parkinson's disease, D1-mGlu5 nanocomplexes were strongly upregulated in the dopamine-denervated striatum, resulting in a synergistic activation of PLC signaling by D1 and mGlu5 receptor agonists. In turn, D1-mGlu5-dependent PLC signaling was causally linked with excessive activation of extracellular signal-regulated kinases in striatal neurons, leading to dyskinesia in animals treated with L-DOPA or D1 receptor agonists. The discovery of D1-mGlu5 functional heteromers mediating maladaptive molecular and motor responses in the dopamine-denervated striatum may prompt the development of new therapeutic principles for Parkinson's disease.


Assuntos
Corpo Estriado/metabolismo , Sistema de Sinalização das MAP Quinases , Neurônios/metabolismo , Doença de Parkinson Secundária/metabolismo , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de Dopamina D1/metabolismo , Animais , Corpo Estriado/patologia , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Neurônios/patologia , Doença de Parkinson Secundária/tratamento farmacológico , Doença de Parkinson Secundária/genética , Doença de Parkinson Secundária/patologia , Ratos , Receptor de Glutamato Metabotrópico 5/genética , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/genética
3.
Sci Rep ; 9(1): 16890, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31729429

RESUMO

Multiple voltage-gated Na+ (Nav) channelopathies can be ascribed to subtle changes in the Nav macromolecular complex. Fibroblast growth factor 14 (FGF14) is a functionally relevant component of the Nav1.6 channel complex, a causative link to spinocerebellar ataxia 27 (SCA27) and an emerging risk factor for neuropsychiatric disorders. Yet, how this protein:channel complex is regulated in the cell is still poorly understood. To search for key cellular pathways upstream of the FGF14:Nav1.6 complex, we have developed, miniaturized and optimized an in-cell assay in 384-well plates by stably reconstituting the FGF14:Nav1.6 complex using the split-luciferase complementation assay. We then conducted a high-throughput screening (HTS) of 267 FDA-approved compounds targeting known mediators of cellular signaling. Of the 65 hits initially detected, 24 were excluded based on counter-screening and cellular toxicity. Based on target analysis, potency and dose-response relationships, 5 compounds were subsequently repurchased for validation and confirmed as hits. Among those, the tyrosine kinase inhibitor lestaurtinib was highest ranked, exhibiting submicromolar inhibition of FGF14:Nav1.6 assembly. While providing evidence for a robust in-cell HTS platform that can be adapted to search for any channelopathy-associated regulatory proteins, these results lay the potential groundwork for repurposing cancer drugs for neuropsychopharmacology.


Assuntos
Antineoplásicos , Ensaios de Triagem em Larga Escala/métodos , Mapas de Interação de Proteínas/fisiologia , Agonistas do Canal de Sódio Disparado por Voltagem/isolamento & purificação , Bloqueadores do Canal de Sódio Disparado por Voltagem/isolamento & purificação , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Fatores de Crescimento de Fibroblastos/agonistas , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/química , Células HEK293 , Humanos , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/química , Canal de Sódio Disparado por Voltagem NAV1.6/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Ligação Proteica , Agonistas do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/metabolismo
4.
Essays Biochem ; 61(5): 543-560, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-29118099

RESUMO

Structure-based drug design plays a central role in therapeutic development. Until recently, protein crystallography and NMR have dominated experimental approaches to obtain structural information of biological molecules. However, in recent years rapid technical developments in single particle cryo-electron microscopy (cryo-EM) have enabled the determination to near-atomic resolution of macromolecules ranging from large multi-subunit molecular machines to proteins as small as 64 kDa. These advances have revolutionized structural biology by hugely expanding both the range of macromolecules whose structures can be determined, and by providing a description of macromolecular dynamics. Cryo-EM is now poised to similarly transform the discipline of structure-based drug discovery. This article reviews the potential of cryo-EM for drug discovery with reference to protein ligand complex structures determined using this technique.


Assuntos
Microscopia Crioeletrônica/métodos , Desenho de Fármacos , Complexos Multiproteicos/química , Proteínas/química , Bibliotecas de Moléculas Pequenas/química , Animais , Microscopia Crioeletrônica/instrumentação , Cristalografia por Raios X , Descoberta de Drogas , Escherichia coli/química , Humanos , Ligantes , Modelos Moleculares , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/antagonistas & inibidores , Proteínas/agonistas , Proteínas/antagonistas & inibidores , Saccharomyces cerevisiae/química , Bibliotecas de Moléculas Pequenas/síntese química , Relação Estrutura-Atividade
5.
Nature ; 541(7636): 228-232, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28024296

RESUMO

Although long non-coding RNAs (lncRNAs) are non-protein-coding transcripts by definition, recent studies have shown that a fraction of putative small open reading frames within lncRNAs are translated. However, the biological significance of these hidden polypeptides is still unclear. Here we identify and functionally characterize a novel polypeptide encoded by the lncRNA LINC00961. This polypeptide is conserved between human and mouse, is localized to the late endosome/lysosome and interacts with the lysosomal v-ATPase to negatively regulate mTORC1 activation. This regulation of mTORC1 is specific to activation of mTORC1 by amino acid stimulation, rather than by growth factors. Hence, we termed this polypeptide 'small regulatory polypeptide of amino acid response' (SPAR). We show that the SPAR-encoding lncRNA is highly expressed in a subset of tissues and use CRISPR/Cas9 engineering to develop a SPAR-polypeptide-specific knockout mouse while maintaining expression of the host lncRNA. We find that the SPAR-encoding lncRNA is downregulated in skeletal muscle upon acute injury, and using this in vivo model we establish that SPAR downregulation enables efficient activation of mTORC1 and promotes muscle regeneration. Our data provide a mechanism by which mTORC1 activation may be finely regulated in a tissue-specific manner in response to injury, and a paradigm by which lncRNAs encoding small polypeptides can modulate general biological pathways and processes to facilitate tissue-specific requirements, consistent with their restricted and highly regulated expression profile.


Assuntos
Complexos Multiproteicos/metabolismo , Músculos/fisiologia , Peptídeos/metabolismo , RNA Longo não Codificante/genética , Regeneração/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Adenosina Trifosfatases/metabolismo , Aminoácidos/metabolismo , Aminoácidos/farmacologia , Animais , Sistemas CRISPR-Cas/genética , Endossomos/metabolismo , Edição de Genes , Células HEK293 , Humanos , Lisossomos/enzimologia , Lisossomos/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/agonistas , Músculos/lesões , Especificidade de Órgãos , Peptídeos/deficiência , Peptídeos/genética , Transdução de Sinais/efeitos dos fármacos
6.
Cell Rep ; 17(1): 29-36, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27681418

RESUMO

FGF21 improves the metabolic profile of obese animals through its actions on adipocytes. To elucidate the signaling network responsible for mediating these effects, we quantified dynamic changes in the adipocyte phosphoproteome following acute exposure to FGF21. FGF21 regulated a network of 821 phosphosites on 542 proteins. A major FGF21-regulated signaling node was mTORC1/S6K. In contrast to insulin, FGF21 activated mTORC1 via MAPK rather than through the canonical PI3K/AKT pathway. Activation of mTORC1/S6K by FGF21 was surprising because this is thought to contribute to deleterious metabolic effects such as obesity and insulin resistance. Rather, mTORC1 mediated many of the beneficial actions of FGF21 in vitro, including UCP1 and FGF21 induction, increased adiponectin secretion, and enhanced glucose uptake without any adverse effects on insulin action. This study provides a global view of FGF21 signaling and suggests that mTORC1 may act to facilitate FGF21-mediated health benefits in vivo.


Assuntos
Adipócitos/efeitos dos fármacos , Adiponectina/genética , Fatores de Crescimento de Fibroblastos/farmacologia , Complexos Multiproteicos/genética , Fosfoproteínas/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Serina-Treonina Quinases TOR/genética , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Adiponectina/agonistas , Adiponectina/metabolismo , Animais , Diferenciação Celular , Desoxiglucose/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica , Redes Reguladoras de Genes/efeitos dos fármacos , Injeções Intraperitoneais , Marcação por Isótopo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/metabolismo , Fosfoproteínas/metabolismo , Proteoma/genética , Proteoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Gordura Subcutânea Abdominal/citologia , Gordura Subcutânea Abdominal/efeitos dos fármacos , Gordura Subcutânea Abdominal/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína Desacopladora 1/agonistas , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
7.
Am J Physiol Cell Physiol ; 310(11): C874-84, 2016 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-27053525

RESUMO

Protein synthesis is stimulated by resistance exercise and intake of amino acids, in particular leucine. Moreover, activation of mammalian target of rapamycin complex 1 (mTORC1) signaling by leucine is potentiated by the presence of other essential amino acids (EAA). However, the contribution of the branched-chain amino acids (BCAA) to this effect is yet unknown. Here we compare the stimulatory role of leucine, BCAA, and EAA ingestion on anabolic signaling following exercise. Accordingly, eight trained volunteers completed four sessions of resistance exercise during which they ingested either placebo, leucine, BCAA, or EAA (including the BCAA) in random order. Muscle biopsies were taken at rest, immediately after exercise, and following 90 and 180 min of recovery. Following 90 min of recovery the activity of S6 kinase 1 (S6K1) was greater than at rest in all four trials (Placebo

Assuntos
Aminoácidos de Cadeia Ramificada/administração & dosagem , Aminoácidos Essenciais/administração & dosagem , Leucina/administração & dosagem , Complexos Multiproteicos/agonistas , Músculo Esquelético/efeitos dos fármacos , Treinamento Resistido , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Biópsia , Glicemia/metabolismo , Proteínas de Ciclo Celular , Metabolismo Energético/efeitos dos fármacos , Fator de Iniciação 4E em Eucariotos/metabolismo , Voluntários Saudáveis , Humanos , Insulina/sangue , Ácido Láctico/sangue , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/metabolismo , Músculo Esquelético/enzimologia , Fosfoproteínas/metabolismo , Fosforilação , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo , Resultado do Tratamento
8.
J Mol Cell Cardiol ; 91: 6-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26739211

RESUMO

BACKGROUND: Activity of mTOR complex 1 (mTORC1) has been shown to be up-regulated in animal models of heart failure. Here, we investigated the change and role of mTORC1 in human nonischemic dilated cardiomyopathy (NICM). METHODS: Endomyocardial biopsy specimens were obtained from patients with NICM (n=52) and from Brugada syndrome patients with normal LVEF as controls (n=10). The specimens were stained for phospho-ribosomal protein S6 (p-Rps6) and phospho-p70S6K (p-p70S6K), and the area with p-Rps6 signal was used as an index of mTORC1 activity. Using median mTORC1 activity, patients were divided into a high mTORC1 activity (H-mTOR) group and a low mTORC1 activity (L-mTOR) group. RESULTS: The ratio of p-Rps6-positive area in biopsy samples was 10-fold larger in patients with NICM than in controls (2.0±2.2% vs. 0.2±0.2%, p<0.01). p-p70S6K signal level was higher in the H-mTOR group than in the L-mTOR group. The proportion of patients with a family history of cardiomyopathy was higher and the proportion of patients on ACE inhibitors or angiotensin receptor blockers was lower in the H-mTOR group than in the L-mTOR group. The p-Rps6-positive area was correlated with extent of myocardial fibrosis (r=0.46, p<0.01). The cardiac event-free survival rate during a 5-year follow-up period tended to be lower in the H-mTOR group than in the L-mTOR group (52.9% vs. 81.6%, P=0.10). CONCLUSION: Aberrant activation of mTORC1 in cardiomyocytes was associated with myocardial fibrosis and a trend for worse prognosis in patients with NICM, indicating that persistently activated mTORC1 contributes to progression of human heart failure.


Assuntos
Síndrome de Brugada/genética , Cardiomiopatia Dilatada/genética , Insuficiência Cardíaca/genética , Complexos Multiproteicos/metabolismo , Miocárdio/enzimologia , Serina-Treonina Quinases TOR/metabolismo , Adulto , Antagonistas de Receptores de Angiotensina/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Biópsia , Síndrome de Brugada/tratamento farmacológico , Síndrome de Brugada/mortalidade , Síndrome de Brugada/patologia , Cardiomiopatia Dilatada/tratamento farmacológico , Cardiomiopatia Dilatada/mortalidade , Cardiomiopatia Dilatada/patologia , Progressão da Doença , Endocárdio/efeitos dos fármacos , Endocárdio/enzimologia , Endocárdio/patologia , Ativação Enzimática , Feminino , Fibrose , Expressão Gênica , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/mortalidade , Insuficiência Cardíaca/patologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/enzimologia , Ventrículos do Coração/patologia , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Pessoa de Meia-Idade , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/genética , Miocárdio/patologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Estudos Retrospectivos , Proteínas Quinases S6 Ribossômicas/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Análise de Sobrevida , Serina-Treonina Quinases TOR/genética
10.
FEBS J ; 282(24): 4672-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26432171

RESUMO

p62/SQSTM1 is a stress-inducible cellular protein that is conserved among metazoans but not in plants and fungi. p62/SQSTM1 has multiple domains that mediate its interactions with various binding partners and it serves as a signaling hub for diverse cellular events such as amino acid sensing and the oxidative stress response. In addition, p62/SQSTM1 functions as a selective autophagy receptor for degradation of ubiqutinated substrates. In the present review, we describe the current knowledge about p62 with regard to mammalian target of rapamycin complex 1 activation, the Keap1-Nrf2 pathway and selective autophagy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia , Modelos Biológicos , Modelos Moleculares , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/química , Animais , Ativação Enzimática , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/agonistas , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/metabolismo , Fator 2 Relacionado a NF-E2/agonistas , Fator 2 Relacionado a NF-E2/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteína Sequestossoma-1 , Serina-Treonina Quinases TOR/metabolismo
11.
Cell Rep ; 11(9): 1446-57, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26004183

RESUMO

AMPK is a master regulator of cellular metabolism that exerts either oncogenic or tumor suppressor activity depending on context. Here, we report that the specific AMPK agonist GSK621 selectively kills acute myeloid leukemia (AML) cells but spares normal hematopoietic progenitors. This differential sensitivity results from a unique synthetic lethal interaction involving concurrent activation of AMPK and mTORC1. Strikingly, the lethality of GSK621 in primary AML cells and AML cell lines is abrogated by chemical or genetic ablation of mTORC1 signaling. The same synthetic lethality between AMPK and mTORC1 activation is established in CD34-positive hematopoietic progenitors by constitutive activation of AKT or enhanced in AML cells by deletion of TSC2. Finally, cytotoxicity in AML cells from GSK621 involves the eIF2α/ATF4 signaling pathway that specifically results from mTORC1 activation. AMPK activation may represent a therapeutic opportunity in mTORC1-overactivated cancers.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antineoplásicos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Imidazóis/farmacologia , Leucemia Mieloide Aguda/metabolismo , Complexos Multiproteicos/agonistas , Pirimidinonas/farmacologia , Animais , Imunofluorescência , Xenoenxertos , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Nus , Microscopia Eletrônica de Transmissão , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR
12.
Mol Ther ; 23(3): 445-55, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25502903

RESUMO

Recent evidence has shown that Ras homolog enriched in brain (Rheb) is dysregulated in Alzheimer's disease (AD) brains. However, it is still unclear whether Rheb activation contributes to the survival and protection of hippocampal neurons in the adult brain. To assess the effects of active Rheb in hippocampal neurons in vivo, we transfected neurons in the cornu ammonis 1 (CA1) region in normal adult rats with an adeno-associated virus containing the constitutively active human Rheb (hRheb(S16H)) and evaluated the effects on thrombin-induced neurotoxicity. Transduction with hRheb(S16H) significantly induced neurotrophic effects in hippocampal neurons through activation of mammalian target of rapamycin complex 1 (mTORC1) without side effects such as long-term potentiation impairment and seizures from the alteration of cytoarchitecture, and the expression of hRheb(S16H) prevented thrombin-induced neurodegeneration in vivo, an effect that was diminished by treatment with specific neutralizing antibodies against brain-derived neurotrophic factor (BDNF). In addition, our results showed that the basal mTORC1 activity might be insufficient to mediate the level of BDNF expression, but hRheb(S16H)-activated mTORC1 stimulated BDNF production in hippocampal neurons. These results suggest that viral vector transduction with hRheb(S16H) may have therapeutic value in the treatment of neurodegenerative diseases such as AD.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Região CA1 Hipocampal/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , Neurônios/metabolismo , Neuropeptídeos/genética , Transdução Genética/métodos , Animais , Anticorpos Neutralizantes/farmacologia , Fator Neurotrófico Derivado do Encéfalo/agonistas , Fator Neurotrófico Derivado do Encéfalo/antagonistas & inibidores , Fator Neurotrófico Derivado do Encéfalo/genética , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/efeitos dos fármacos , Dependovirus/genética , Dependovirus/metabolismo , Expressão Gênica , Vetores Genéticos/administração & dosagem , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neuropeptídeos/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Ratos , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Trombina/antagonistas & inibidores , Trombina/toxicidade
13.
Am J Physiol Endocrinol Metab ; 307(8): E686-94, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25159322

RESUMO

Skeletal muscle loading/overload stimulates the Ca²âº-activated, serine/threonine kinase Ca²âº/calmodulin-dependent protein kinase kinase-α (CaMKKα); yet to date, no studies have examined whether CaMKKα regulates muscle growth. The purpose of this study was to determine if constitutive activation of CaMKKα signaling could stimulate muscle growth and if so whether CaMKKα is essential for this process. CaMKKα signaling was selectively activated in mouse muscle via expression of a constitutively active form of CaMKKα using in vivo electroporation. After 2 wk, constitutively active CaMKKα expression increased muscle weight (~10%) and protein content (~10%), demonstrating that activation of CaMKKα signaling can stimulate muscle growth. To determine if active CaMKKα expression stimulated muscle growth via increased mammalian target of rapamycin complex 1 (mTORC1) signaling and protein synthesis, [³H]phenylalanine incorporation into proteins was assessed with or without the mTORC1 inhibitor rapamycin. Constitutively active CaMKKα increased protein synthesis ~60%, and this increase was prevented by rapamycin, demonstrating a critical role for mTORC1 in this process. To determine if CaMKKα is essential for growth, muscles from CaMKKα knockout mice were stimulated to hypertrophy via unilateral ablation of synergist muscles (overload). Surprisingly, compared with wild-type mice, muscles from CaMKKα knockout mice exhibited greater growth (~15%) and phosphorylation of the mTORC1 substrate 70-kDa ribosomal protein S6 kinase (Thr³89; ~50%), demonstrating that CaMKKα is not essential for overload-induced mTORC1 activation or muscle growth. Collectively, these results demonstrate that activation of CaMKKα signaling is sufficient but not necessary for activation of mTORC1 signaling and growth in mouse skeletal muscle.


Assuntos
Sinalização do Cálcio , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Complexos Multiproteicos/agonistas , Desenvolvimento Muscular , Proteínas Musculares/biossíntese , Músculo Esquelético/metabolismo , Regulação para Cima , Técnicas de Ablação/efeitos adversos , Animais , Sinalização do Cálcio/efeitos dos fármacos , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/química , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/genética , Cruzamentos Genéticos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hipertrofia , Técnicas In Vitro , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/metabolismo , Desenvolvimento Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/patologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima/efeitos dos fármacos
14.
Nature ; 512(7512): 49-53, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25043012

RESUMO

In the 1950s, the drug thalidomide, administered as a sedative to pregnant women, led to the birth of thousands of children with multiple defects. Despite the teratogenicity of thalidomide and its derivatives lenalidomide and pomalidomide, these immunomodulatory drugs (IMiDs) recently emerged as effective treatments for multiple myeloma and 5q-deletion-associated dysplasia. IMiDs target the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4(CRBN)) and promote the ubiquitination of the IKAROS family transcription factors IKZF1 and IKZF3 by CRL4(CRBN). Here we present crystal structures of the DDB1-CRBN complex bound to thalidomide, lenalidomide and pomalidomide. The structure establishes that CRBN is a substrate receptor within CRL4(CRBN) and enantioselectively binds IMiDs. Using an unbiased screen, we identified the homeobox transcription factor MEIS2 as an endogenous substrate of CRL4(CRBN). Our studies suggest that IMiDs block endogenous substrates (MEIS2) from binding to CRL4(CRBN) while the ligase complex is recruiting IKZF1 or IKZF3 for degradation. This dual activity implies that small molecules can modulate an E3 ubiquitin ligase and thereby upregulate or downregulate the ubiquitination of proteins.


Assuntos
Peptídeo Hidrolases/química , Talidomida/química , Ubiquitina-Proteína Ligases/química , Proteínas Adaptadoras de Transdução de Sinal , Cristalografia por Raios X , Proteínas de Ligação a DNA/agonistas , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Lenalidomida , Modelos Moleculares , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Peptídeo Hidrolases/metabolismo , Ligação Proteica , Relação Estrutura-Atividade , Especificidade por Substrato , Talidomida/análogos & derivados , Talidomida/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/metabolismo
15.
PLoS One ; 9(4): e95393, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24740400

RESUMO

Ursolic acid (UA), a pentacyclic triterpenoid widely found in medicinal herbs and fruits, has been reported to possess a wide range of beneficial properties including anti-hyperglycemia, anti-obesity, and anti-cancer. However, the molecular mechanisms underlying the action of UA remain largely unknown. Here we show that UA inhibits leucine-induced activation of the mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway in C2C12 myotubes. The UA-mediated inhibition of mTORC1 is independent of Akt, tuberous sclerosis complex 1/2 (TSC1/2), and Ras homolog enriched in brain (Rheb), suggesting that UA negatively regulates mTORC1 signaling by targeting at a site downstream of these mTOR regulators. UA treatment had no effect on the interaction between mTOR and its activator Raptor or inhibitor Deptor, but suppressed the binding of RagB to Raptor and inhibited leucine-induced mTOR lysosomal localization. Taken together, our study identifies UA as a direct negative regulator of the mTORC1 signaling pathway and suggests a novel mechanism by which UA exerts its beneficial function.


Assuntos
Leucina/farmacologia , Complexos Multiproteicos/genética , Fibras Musculares Esqueléticas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Triterpenos/farmacologia , Animais , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leucina/antagonistas & inibidores , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Aves Predatórias/genética , Aves Predatórias/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ácido Ursólico
16.
PLoS One ; 8(7): e69420, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936011

RESUMO

Glycogen phosphorylase (GP) catalyzes the breakdown of glycogen and largely contributes to hepatic glucose production making GP inhibition an attractive target to modulate glucose levels in diabetes. Hereby we present the metabolic effects of a novel, potent, glucose-based GP inhibitor (KB228) tested in vitro and in vivo under normoglycemic and diabetic conditions. KB228 administration enhanced glucose sensitivity in chow-fed and obese, diabetic mice that was a result of higher hepatic glucose uptake. Besides improved glucose sensitivity, we have observed further unexpected metabolic rearrangements. KB228 administration increased oxygen consumption that was probably due to the overexpression of uncoupling protein-2 (UCP2) that was observed in animal and cellular models. Furthermore, KB228 treatment induced mammalian target of rapamycin complex 2 (mTORC2) in mice. Our data demonstrate that glucose based GP inhibitors are capable of reducing glucose levels in mice under normo and hyperglycemic conditions. Moreover, these GP inhibitors induce accommodation in addition to GP inhibition--such as enhanced mitochondrial oxidation and mTORC2 signaling--to cope with the glucose influx and increased glycogen deposition in the cells, however the molecular mechanism of accommodation is unexplored.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Glucose/farmacologia , Glicogênio Fosforilase/antagonistas & inibidores , Glicogênio Hepático/metabolismo , Ureia/farmacologia , Animais , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/fisiopatologia , Inibidores Enzimáticos/síntese química , Expressão Gênica/efeitos dos fármacos , Glucose/análogos & derivados , Glucose/síntese química , Teste de Tolerância a Glucose , Glicogênio Fosforilase/metabolismo , Canais Iônicos/agonistas , Canais Iônicos/genética , Canais Iônicos/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Glicogênio Hepático/antagonistas & inibidores , Masculino , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Obesos , Proteínas Mitocondriais/agonistas , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Proteína Desacopladora 2 , Ureia/análogos & derivados , Ureia/síntese química
17.
Mol Biol Cell ; 24(13): 2146-55, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23657816

RESUMO

Global stimulation of Dictyostelium with different chemoattractants elicits multiple transient signaling responses, including synthesis of cAMP and cGMP, actin polymerization, activation of kinases ERK2, TORC2, and phosphatidylinositide 3-kinase, and Ras-GTP accumulation. Mechanisms that down-regulate these responses are poorly understood. Here we examine transient activation of TORC2 in response to chemically distinct chemoattractants, cAMP and folate, and suggest that TORC2 is regulated by adaptive, desensitizing responses to stimulatory ligands that are independent of downstream, feedback, or feedforward circuits. Cells with acquired insensitivity to either folate or cAMP remain fully responsive to TORC2 activation if stimulated with the other ligand. Thus TORC2 responses to cAMP or folate are not cross-inhibitory. Using a series of signaling mutants, we show that folate and cAMP activate TORC2 through an identical GEF/Ras pathway but separate receptors and G protein couplings. Because the common GEF/Ras pathway also remains fully responsive to one chemoattractant after desensitization to the other, GEF/Ras must act downstream and independent of adaptation to persistent ligand stimulation. When initial chemoattractant concentrations are immediately diluted, cells rapidly regain full responsiveness. We suggest that ligand adaptation functions in upstream inhibitory pathways that involve chemoattractant-specific receptor/G protein complexes and regulate multiple response pathways.


Assuntos
Dictyostelium/efeitos dos fármacos , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas de Protozoários/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Quimiotaxia , AMP Cíclico/farmacologia , Dictyostelium/genética , Dictyostelium/metabolismo , Ácido Fólico/farmacologia , Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/genética , Alvo Mecanístico do Complexo 2 de Rapamicina , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/genética , Mutação , Proteínas de Protozoários/genética , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Proteínas ras/genética , Proteínas ras/metabolismo
18.
J Neurochem ; 114(4): 972-80, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20477947

RESUMO

Pharmacological characterization of adenosine A(1) and A(2A) receptors in human brain caudate nucleus membranes led to non-cooperative binding of radiolabelled ligands. In human caudate nucleus but not in cortex, the agonist binding to A(1) receptors was modulated by the agonist binding to A(2A) receptors indicating a functional negative cross-talk. Accordingly, the A(1) receptor-activation-mediated G(i)-dependent guanosine 5'-o-(3-[(35)S]thio-triphosphate) binding was modulated by agonist binding to A(2A) receptors. A(2A) receptors occupation led to a decrease in the potency of A(1) receptor agonists. These results indicate that A(1) but not A(2A) receptors activation, likely occurring at low adenosine concentrations, engages a G(i)-mediated signaling; however, when both receptors are occupied by adenosine, there is an A(2A) receptor-mediated impairment of G(i)-operated transducing units. These findings are relevant to get insight into the complex relationships derived from co-expression of multiple neurotransmitter/neuromodulator receptors subtypes that individually are coupled to different G proteins. A further finding was the demonstration that the A(2A) receptor agonist, CGS 21680, at high concentrations able to significantly bind to the A(1) receptor, behaved as a partial agonist of the later receptor. This fact might be taken into account when characterizing CGS 21680 actions in human cells expressing A(1) receptors when the compound is used at micromolar concentrations.


Assuntos
Núcleo Caudado/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Receptor Cross-Talk/fisiologia , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Agonistas do Receptor A1 de Adenosina , Agonistas do Receptor A2 de Adenosina , Ligação Competitiva/fisiologia , Núcleo Caudado/efeitos dos fármacos , Membrana Celular/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/agonistas , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Humanos , Complexos Multiproteicos/agonistas , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA