Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 53(1): 140-7, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24374310

RESUMO

Eukaryotic protein kinases are generally classified as being either tyrosine or serine-threonine specific. Though not evident from inspection of their primary sequences, many serine-threonine kinases display a significant preference for serine or threonine as the phosphoacceptor residue. Here we show that a residue located in the kinase activation segment, which we term the "DFG+1" residue, acts as a major determinant for serine-threonine phosphorylation site specificity. Mutation of this residue was sufficient to switch the phosphorylation site preference for multiple kinases, including the serine-specific kinase PAK4 and the threonine-specific kinase MST4. Kinetic analysis of peptide substrate phosphorylation and crystal structures of PAK4-peptide complexes suggested that phosphoacceptor residue preference is not mediated by stronger binding of the favored substrate. Rather, favored kinase-phosphoacceptor combinations likely promote a conformation optimal for catalysis. Understanding the rules governing kinase phosphoacceptor preference allows kinases to be classified as serine or threonine specific based on their sequence.


Assuntos
Peptídeos/química , Proteínas Serina-Treonina Quinases/química , Quinases Ativadas por p21/química , Sítios de Ligação , Cristalografia por Raios X , Células HEK293 , Humanos , Cinética , Peptídeos/genética , Peptídeos/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Especificidade por Substrato/fisiologia , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
2.
J Cell Sci ; 131(17)2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30154209

RESUMO

T cell-mediated adaptive immunity requires naïve, unstimulated T cells to transition from a quiescent metabolic state into a highly proliferative state upon T cell receptor engagement. This complex process depends on transcriptional changes mediated by Ca2+-dependent NFAT signaling, mTOR-mediated signaling and increased activity of the guanine nucleotide biosynthetic inosine-5'-monophosphate (IMP) dehydrogenase 1 and 2 enzymes (IMPDH1 and IMPDH2, hereafter IMPDH). Inhibitors of these pathways serve as potent immunosuppressants. Unexpectedly, we discovered that all three pathways converge to promote the assembly of IMPDH protein into micron-scale macromolecular filamentous structures in response to T cell activation. Assembly is post-transcriptionally controlled by mTOR and the Ca2+ influx regulator STIM1. Furthermore, IMPDH assembly and catalytic activity were negatively regulated by guanine nucleotide levels, suggesting a negative feedback loop that limits biosynthesis of guanine nucleotides. Filamentous IMPDH may be more resistant to this inhibition, facilitating accumulation of the higher GTP levels required for T cell proliferation.


Assuntos
IMP Desidrogenase/metabolismo , Linfócitos T/enzimologia , Animais , Células Cultivadas , Nucleotídeos de Guanina/metabolismo , IMP Desidrogenase/genética , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Baço/enzimologia , Baço/imunologia , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Linfócitos T/imunologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
3.
Mol Cell ; 40(3): 493-500, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-21070974

RESUMO

Phospholipid-enriched membranes such as the plasma membrane can serve as direct regulators of kinase signaling. Pak1 is involved in growth factor signaling at the plasma membrane, and its dysregulation is implicated in cancer. Pak1 adopts an autoinhibited conformation that is relieved upon binding to membrane-bound Rho GTPases Rac1 or Cdc42, but whether lipids also regulate Pak1 in vivo is unknown. We show here that phosphoinositides, particularly PIP(2), potentiate Rho-GTPase-mediated Pak1 activity. A positively charged region of Pak1 binds to phosphoinositide-containing membranes, and this interaction is essential for membrane recruitment and activation of Pak1 in response to extracellular signals. Our results highlight an active role for lipids as allosteric regulators of Pak1 and suggest that Pak1 is a "coincidence detector" whose activation depends on GTPases present in phosphoinositide-rich membranes. These findings expand the role of phosphoinositides in kinase signaling and suggest how altered phosphoinositide metabolism may upregulate Pak1 activity in cancer cells.


Assuntos
Ativadores de Enzimas/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Quinases Ativadas por p21/metabolismo , Sequência de Aminoácidos , Animais , Extratos Celulares , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Xenopus , Quinases Ativadas por p21/química , Proteínas rac1 de Ligação ao GTP/metabolismo
4.
EMBO Rep ; 15(11): 1184-91, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25223282

RESUMO

The enzyme CTP synthase (CTPS) dynamically assembles into macromolecular filaments in bacteria, yeast, Drosophila, and mammalian cells, but the role of this morphological reorganization in regulating CTPS activity is controversial. During Drosophila oogenesis, CTPS filaments are transiently apparent in ovarian germline cells during a period of intense genomic endoreplication and stockpiling of ribosomal RNA. Here, we demonstrate that CTPS filaments are catalytically active and that their assembly is regulated by the non-receptor tyrosine kinase DAck, the Drosophila homologue of mammalian Ack1 (activated cdc42-associated kinase 1), which we find also localizes to CTPS filaments. Egg chambers from flies deficient in DAck or lacking DAck catalytic activity exhibit disrupted CTPS filament architecture and morphological defects that correlate with reduced fertility. Furthermore, ovaries from these flies exhibit reduced levels of total RNA, suggesting that DAck may regulate CTP synthase activity. These findings highlight an unexpected function for DAck and provide insight into a novel pathway for the developmental control of an essential metabolic pathway governing nucleotide biosynthesis.


Assuntos
Carbono-Nitrogênio Ligases/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Oogênese , Proteínas Tirosina Quinases/metabolismo , Animais , Carbono-Nitrogênio Ligases/genética , Drosophila/genética , Drosophila/fisiologia , Proteínas de Drosophila/genética , Feminino , Ovário/metabolismo , Transporte Proteico , Proteínas Tirosina Quinases/genética , RNA/metabolismo
5.
Proc Natl Acad Sci U S A ; 109(11): 4116-21, 2012 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-22371566

RESUMO

Synaptic transmission is mediated by a complex set of molecular events that must be coordinated in time and space. While many proteins that function at the synapse have been identified, the signaling pathways regulating these molecules are poorly understood. Pak5 (p21-activated kinase 5) is a brain-specific isoform of the group II Pak kinases whose substrates and roles within the central nervous system are largely unknown. To gain insight into the physiological roles of Pak5, we engineered a Pak5 mutant to selectively radiolabel its substrates in murine brain extract. Using this approach, we identified two novel Pak5 substrates, Pacsin1 and Synaptojanin1, proteins that directly interact with one another to regulate synaptic vesicle endocytosis and recycling. Pacsin1 and Synaptojanin1 were phosphorylated by Pak5 and the other group II Paks in vitro, and Pak5 phosphorylation promoted Pacsin1-Synaptojanin1 binding both in vitro and in vivo. These results implicate Pak5 in Pacsin1- and Synaptojanin1-mediated synaptic vesicle trafficking and may partially account for the cognitive and behavioral deficits observed in group II Pak-deficient mice.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Neurônios/enzimologia , Neuropeptídeos/metabolismo , Fosfoproteínas/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Vesículas Sinápticas/enzimologia , Quinases Ativadas por p21/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Transporte Biológico , Encéfalo/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Modelos Biológicos , Fosforilação , Ligação Proteica , Especificidade por Substrato
6.
J Biol Chem ; 288(13): 8887-97, 2013 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-23393142

RESUMO

Autoinhibited p21-activated kinase 1 (Pak1) can be activated in vitro by the plasma membrane-bound Rho GTPases Rac1 and Cdc42 as well as by the lipid phosphatidylinositol (4,5)-bisphosphate (PIP2). Activator binding is mediated by a GTPase-binding motif and an adjacent phosphoinositide-binding motif. Whether these two classes of activators play alternative, additive, or synergistic roles in Pak1 activation is unknown, as is their contributions to Pak1 activation in vivo. To address these questions, we developed a system to mimic the membrane anchoring of Rho GTPases by creating liposomes containing both PIP2 and a Ni(2+)-NTA modified lipid capable of binding hexahistidine-tagged Cdc42. We find that among all biologically relevant phosphoinositides, only PIP2 is able to synergistically activate Pak1 in concert with Cdc42. Membrane binding of the kinase was highly sensitive to the spatial density of PIP2 and Pak1 demonstrated dramatically enhanced affinity for Cdc42 anchored in a PIP2 environment. To validate these findings in vivo, we utilized an inducible recruitment system to drive the ectopic synthesis of PIP2 on Golgi membranes, which normally have active Cdc42 but lack significant concentrations of PIP2. Pak1 was recruited to PIP2-containing membranes in a manner dependent on the ability of Pak1 to bind to both PIP2 and Cdc42. These findings provide a mechanistic explanation for the essential role of both phosphoinositides and GTPases in Pak1 recruitment and activation. In contrast, Ack, another Cdc42 effector kinase that lacks an analogous phosphoinositide-binding motif, fails to show the same enhancement of membrane binding and activation by PIP2, thus indicating that regulation by PIP2 and Cdc42 could provide a combinatorial code for activation of different GTPase effectors in different subcellular locations.


Assuntos
Regulação Enzimológica da Expressão Gênica , Fosfatidilinositol 4,5-Difosfato/química , Quinases Ativadas por p21/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Complexo de Golgi/metabolismo , Células HEK293 , Humanos , Lipossomos/química , Níquel/química , Fosfatidilinositóis/química , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
7.
J Biol Chem ; 288(12): 8762-8771, 2013 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-23386615

RESUMO

The serine/threonine protein kinases Mst1 and Mst2 can be activated by cellular stressors including hydrogen peroxide. Using two independent protein interaction screens, we show that these kinases associate, in an oxidation-dependent manner, with Prdx1, an enzyme that regulates the cellular redox state by reducing hydrogen peroxide to water and oxygen. Mst1 inactivates Prdx1 by phosphorylating it at Thr-90 and Thr-183, leading to accumulation of hydrogen peroxide in cells. These results suggest that hydrogen peroxide-stimulated Mst1 activates a positive feedback loop to sustain an oxidizing cellular state.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Peroxirredoxinas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Dano ao DNA , Células HEK293 , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos , Dados de Sequência Molecular , Oxirredução , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Serina-Treonina Quinase 3 , Técnicas do Sistema de Duplo-Híbrido
8.
J Biol Chem ; 288(39): 28068-77, 2013 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-23935097

RESUMO

Dual inhibitors of the closely related receptor tyrosine kinases insulin-like growth factor 1 receptor (IGF-1R) and insulin receptor (IR) are promising therapeutic agents in cancer. Here, we report an unusually selective class of dual inhibitors of IGF-1R and IR identified in a parallel screen of known kinase inhibitors against a panel of 300 human protein kinases. Biochemical and structural studies indicate that this class achieves its high selectivity by binding to the ATP-binding pocket of inactive, unphosphorylated IGF-1R/IR and stabilizing the activation loop in a native-like inactive conformation. One member of this compound family was originally reported as an inhibitor of the serine/threonine kinase ERK, a kinase that is distinct in the structure of its unphosphorylated/inactive form from IR/IGF-1R. Remarkably, this compound binds to the ATP-binding pocket of ERK in an entirely different conformation to that of IGF-1R/IR, explaining the potency against these two structurally distinct kinase families. These findings suggest a novel approach to polypharmacology in which two or more unrelated kinases are inhibited by a single compound that targets different conformations of each target kinase.


Assuntos
Regulação da Expressão Gênica , Inibidores de Proteínas Quinases/química , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Trifosfato de Adenosina/química , Animais , Células CHO , Cricetulus , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Desenho de Fármacos , Humanos , Sistema de Sinalização das MAP Quinases , Mutação , Fosforilação , Conformação Proteica , Inibidores de Proteínas Quinases/classificação , Pirazóis/química , Piridazinas/química
9.
J Neurosci ; 32(14): 4878-86, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22492044

RESUMO

Hypertension, a powerful risk factor for stroke and dementia, has damaging effects on the brain and its vessels. In particular, hypertension alters vital cerebrovascular control mechanisms linking neural activity to cerebral perfusion. In experimental models of slow-developing hypertension, free radical signaling in the subfornical organ (SFO), one of the forebrain circumventricular organs, is critical for the hormonal release and sympathetic activation driving the elevation in arterial pressure. However, the contribution of this central mechanism to the cerebrovascular alterations induced by hypertension remains uncertain. We tested the hypothesis that free radical production in the SFO is involved in the alterations in cerebrovascular regulation produced by hypertension. In a mouse model of gradual hypertension induced by chronic administration of subpressor doses of angiotensin II (AngII), suppression of free radicals in the SFO by overexpression of CuZn-superoxide dismutase (CuZnSOD) prevented the alteration in neurovascular coupling and endothelium-dependent responses in somatosensory cortex induced by hypertension. The SFO mediates the dysfunction via two signaling pathways. One involves SFO-dependent activation of the paraventricular hypothalamic nucleus, elevations in plasma vasopressin, upregulation of endothelin-1 in cerebral resistance arterioles and activation of endothelin type A receptors. The other pathway depends on activation of cerebrovascular AngII type 1 (AT1) receptors by AngII. Both pathways mediate vasomotor dysfunction by inducing vascular oxidative stress. The findings implicate for the first time the SFO and its efferent hypothalamic pathways in the cerebrovascular alterations induced by AngII, and identify vasopressin and endothelin-1 as potential therapeutic targets to counteract the devastating effects of hypertension on the brain.


Assuntos
Angiotensina II/fisiologia , Angiotensina II/toxicidade , Circulação Cerebrovascular/fisiologia , Hipertensão/fisiopatologia , Órgão Subfornical/fisiopatologia , Animais , Circulação Cerebrovascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Técnicas de Transferência de Genes , Hipertensão/induzido quimicamente , Hipertensão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Receptor Tipo 1 de Angiotensina/agonistas , Receptor Tipo 1 de Angiotensina/fisiologia , Órgão Subfornical/efeitos dos fármacos
10.
Am J Physiol Heart Circ Physiol ; 305(10): H1451-61, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24014678

RESUMO

Regulation of blood pressure by angiotensin II (ANG II) is a process that involves the reactive oxygen species (ROS) and calcium. We have shown that ANG-II type 1 receptor (AT1R) and prostaglandin E2 (PGE2) type 1 receptors (EP1R) are required in the subfornical organ (SFO) for ROS-mediated hypertension induced by slow-pressor ANG-II infusion. However, the signaling pathway associated with this process remains unclear. We sought to determine mechanisms underlying the ANG II-induced ROS and calcium influx in mouse SFO cells. Ultrastructural studies showed that cyclooxygenase 1 (COX-1) codistributes with AT1R in the SFO, indicating spatial proximity. Functional studies using SFO cells revealed that ANG II potentiated PGE2 release, an effect dependent on AT1R, phospholipase A2 (PLA2) and COX-1. Furthermore, both ANG II and PGE2 increased ROS formation. While the increase in ROS initiated by ANG II, but not PGE2, required the activation of the AT1R/PLA2/COX-1 pathway, both ANG II and PGE2 were dependent on EP1R and Nox2 as downstream effectors. Finally, ANG II potentiated voltage-gated L-type Ca(2+) currents in SFO neurons via the same signaling pathway required for PGE2 production. Blockade of EP1R and Nox2-derived ROS inhibited ANG II and PGE2-mediated Ca(2+) currents. We propose a mechanism whereby ANG II increases COX-1-derived PGE2 through the AT1R/PLA2 pathway, which promotes ROS production by EP1R/Nox2 signaling in the SFO. ANG II-induced ROS are coupled with Ca(2+) influx in SFO neurons, which may influence SFO-mediated sympathoexcitation. Our findings provide the first evidence of a spatial and functional framework that underlies ANG-II signaling in the SFO and reveal novel targets for antihypertensive therapies.


Assuntos
Angiotensina II/metabolismo , Sinalização do Cálcio , Ciclo-Oxigenase 1/metabolismo , Dinoprostona/metabolismo , Hipertensão/enzimologia , Proteínas de Membrana/metabolismo , Neurônios/enzimologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Órgão Subfornical/enzimologia , Potenciais de Ação , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Pressão Sanguínea , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Ciclo-Oxigenase 1/deficiência , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Hipertensão/patologia , Hipertensão/fisiopatologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Fosfolipases A2/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores de Prostaglandina E Subtipo EP1/deficiência , Receptores de Prostaglandina E Subtipo EP1/genética , Órgão Subfornical/efeitos dos fármacos , Órgão Subfornical/fisiopatologia , Órgão Subfornical/ultraestrutura
11.
Cell Death Discov ; 8(1): 253, 2022 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-35523770

RESUMO

Ferroptosis is a caspase-independent form of regulated cell death strongly linked to the accumulation of reactive lipid hydroperoxides. Lipid hydroperoxides are neutralized in cells by glutathione peroxidase 4 (GPX4) and inhibitors of GPX4 are potent ferroptosis inducers with therapeutic potential in cancer. Here we report that siRNA-mediated silencing of the AMPK-related kinase NUAK2 suppresses cell death by small-molecule inducers of ferroptosis but not apoptosis. Mechanistically we find that NUAK2 suppresses the expression of GPX4 at the RNA level and enhances ferroptosis triggered by GPX4 inhibitors in a manner independent of its kinase activity. NUAK2 is amplified along with MDM4 in a subset of breast cancers, particularly the claudin-low subset, suggesting that this may predict vulnerability to GPX4 inhibitors. These findings identify a novel pathway regulating GPX4 expression as well as ferroptotic sensitivity with potential as a biomarker of breast cancer patients that might respond to GPX4 inhibition as a therapeutic strategy.

12.
Nat Struct Mol Biol ; 29(1): 47-58, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35013599

RESUMO

Inosine-5'-monophosphate dehydrogenase (IMPDH), a key regulatory enzyme in purine nucleotide biosynthesis, dynamically assembles filaments in response to changes in metabolic demand. Humans have two isoforms: IMPDH2 filaments reduce sensitivity to feedback inhibition, while IMPDH1 assembly remains uncharacterized. IMPDH1 plays a unique role in retinal metabolism, and point mutants cause blindness. Here, in a series of cryogenic-electron microscopy structures we show that human IMPDH1 assembles polymorphic filaments with different assembly interfaces in extended and compressed states. Retina-specific splice variants introduce structural elements that reduce sensitivity to GTP inhibition, including stabilization of the extended filament form. Finally, we show that IMPDH1 disease mutations fall into two classes: one disrupts GTP regulation and the other has no effect on GTP regulation or filament assembly. These findings provide a foundation for understanding the role of IMPDH1 in retinal function and disease and demonstrate the diverse mechanisms by which metabolic enzyme filaments are allosterically regulated.


Assuntos
IMP Desidrogenase/genética , Retina/enzimologia , Trifosfato de Adenosina/metabolismo , Regulação Alostérica , Sítios de Ligação , Domínio Catalítico , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , IMP Desidrogenase/química , IMP Desidrogenase/ultraestrutura , Modelos Moleculares , NAD/metabolismo , Doenças Retinianas/genética
13.
Blood ; 114(3): 580-8, 2009 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-19451552

RESUMO

CD4(+) T cells use the chemokine receptor CCR7 to home to and migrate within lymphoid tissue, where T-cell activation takes place. Using primary T-cell receptor (TCR)-transgenic (tg) CD4(+) T cells, we explored the effect of CCR7 ligands, in particular CCL21, on T-cell activation. We found that the presence of CCL21 during early time points strongly increased in vitro T-cell proliferation after TCR stimulation, correlating with increased expression of early activation markers. CCL21 costimulation resulted in increased Ras- and Rac-GTP formation and enhanced phosphorylation of Akt, MEK, and ERK but not p38 or JNK. Kinase-dead PI3Kdelta(D910A/D910A) or PI3Kgamma-deficient TCR-tg CD4(+) T cells showed similar responsiveness to CCL21 costimulation as control CD4(+) T cells. Conversely, deficiency in the Rac guanine exchange factor DOCK2 significantly impaired CCL21-mediated costimulation in TCR-tg CD4(+) T cells, concomitant with impaired Rac- but not Ras-GTP formation. Using lymph node slices for live monitoring of T-cell behavior and activation, we found that G protein-coupled receptor signaling was required for early CD69 expression but not for Ca(2+) signaling. Our data suggest that the presence of CCL21 during early TCR signaling lowers the activation threshold through Ras- and Rac-dependent pathways leading to increased ERK phosphorylation.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Quimiocina CCL21/fisiologia , Proteínas Ativadoras de GTPase/metabolismo , Ativação Linfocitária , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Troca do Nucleotídeo Guanina , Linfonodos , Camundongos , Camundongos Knockout , Fosforilação , Transdução de Sinais , Proteínas ras/metabolismo
14.
Nat Commun ; 12(1): 2244, 2021 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-33854057

RESUMO

Ferroptosis is associated with lipid hydroperoxides generated by the oxidation of polyunsaturated acyl chains. Lipid hydroperoxides are reduced by glutathione peroxidase 4 (GPX4) and GPX4 inhibitors induce ferroptosis. However, the therapeutic potential of triggering ferroptosis in cancer cells with polyunsaturated fatty acids is unknown. Here, we identify conjugated linoleates including α-eleostearic acid (αESA) as ferroptosis inducers. αESA does not alter GPX4 activity but is incorporated into cellular lipids and promotes lipid peroxidation and cell death in diverse cancer cell types. αESA-triggered death is mediated by acyl-CoA synthetase long-chain isoform 1, which promotes αESA incorporation into neutral lipids including triacylglycerols. Interfering with triacylglycerol biosynthesis suppresses ferroptosis triggered by αESA but not by GPX4 inhibition. Oral administration of tung oil, naturally rich in αESA, to mice limits tumor growth and metastasis with transcriptional changes consistent with ferroptosis. Overall, these findings illuminate a potential approach to ferroptosis, complementary to GPX4 inhibition.


Assuntos
Coenzima A Ligases/metabolismo , Ferroptose , Ácidos Linolênicos/metabolismo , Neoplasias de Mama Triplo Negativas/enzimologia , Neoplasias de Mama Triplo Negativas/fisiopatologia , Animais , Morte Celular , Coenzima A Ligases/genética , Humanos , Camundongos , Camundongos Endogâmicos NOD , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo
15.
Mol Biol Cell ; 31(12): 1201-1205, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32463766

RESUMO

Many different enzymes in intermediate metabolism dynamically assemble filamentous polymers in cells, often in response to changes in physiological conditions. Most of the enzyme filaments known to date have only been observed in cells, but in a handful of cases structural and biochemical studies have revealed the mechanisms and consequences of assembly. In general, enzyme polymerization functions as a mechanism to allosterically tune enzyme kinetics, and it may play a physiological role in integrating metabolic signaling. Here, we highlight some principles of metabolic filaments by focusing on two well-studied examples in nucleotide biosynthesis pathways-inosine-5'-monophosphate (IMP) dehydrogenase and cytosine triphosphate (CTP) synthase.


Assuntos
Carbono-Nitrogênio Ligases/metabolismo , IMP Desidrogenase/metabolismo , Carbono-Nitrogênio Ligases/fisiologia , Citoplasma/metabolismo , Citoesqueleto/metabolismo , Células HeLa , Humanos , IMP Desidrogenase/fisiologia , Polimerização , Multimerização Proteica/fisiologia
16.
Biol Open ; 9(7)2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32580972

RESUMO

Polymerization of metabolic enzymes into micron-scale assemblies is an emerging mechanism for regulating their activity. CTP synthase (CTPS) is an essential enzyme in the biosynthesis of the nucleotide CTP and undergoes regulated and reversible assembly into large filamentous structures in organisms from bacteria to humans. The purpose of these assemblies is unclear. A major challenge to addressing this question has been the inability to abolish assembly without eliminating CTPS protein. Here we demonstrate that a recently reported point mutant in CTPS, Histidine 355A (H355A), prevents CTPS filament assembly in vivo and dominantly inhibits the assembly of endogenous wild-type CTPS in the Drosophila ovary. Expressing this mutant in ovarian germline cells, we show that disruption of CTPS assembly in early stage egg chambers reduces egg production. This effect is exacerbated in flies fed the glutamine antagonist 6-diazo-5-oxo-L-norleucine, which inhibits de novo CTP synthesis. These findings introduce a general approach to blocking the assembly of polymerizing enzymes without eliminating their catalytic activity and demonstrate a role for CTPS assembly in supporting egg production, particularly under conditions of limited glutamine metabolism.This article has an associated First Person interview with the first author of the paper.


Assuntos
Carbono-Nitrogênio Ligases/metabolismo , Drosophila/fisiologia , Células Germinativas/metabolismo , Multimerização Proteica , Reprodução , Animais , Carbono-Nitrogênio Ligases/química , Carbono-Nitrogênio Ligases/genética , Metabolismo Energético , Imunofluorescência , Expressão Gênica , Glutamina/metabolismo , Mutação
17.
Biochemistry ; 48(40): 9327-9, 2009 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-19764708

RESUMO

Formins are potent actin assembly factors. Diaphanous formins, including mDia1, mDia2, and mDia3 in mammals, are implicated in mitosis and cytokinesis, but no chemical interactors have been reported. We developed an in vitro screen for inhibitors of actin assembly by mDia1 and identified an inhibitor of mDia1 and mDia2 that does not inhibit mDia3 at the concentrations tested. These results establish the druggability of mDia formins and introduce a first-generation inhibitor.


Assuntos
Actinas/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas de Transporte/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , NADPH Desidrogenase/antagonistas & inibidores , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Proteínas Fetais/antagonistas & inibidores , Forminas , Humanos , Camundongos , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/fisiologia , NADPH Desidrogenase/química , NADPH Desidrogenase/fisiologia , Proteínas Nucleares/antagonistas & inibidores , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiologia , Estrutura Terciária de Proteína/fisiologia , Pirenos/química , Homologia de Sequência de Aminoácidos
18.
Chem Biol ; 15(4): 322-31, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18420139

RESUMO

Autoregulatory domains found within kinases may provide more unique targets for chemical inhibitors than the conserved ATP-binding pocket targeted by most inhibitors. The kinase Pak1 contains an autoinhibitory domain that suppresses the catalytic activity of its kinase domain. Pak1 activators relieve this autoinhibition and initiate conformational rearrangements and autophosphorylation events leading to kinase activation. We developed a screen for allosteric inhibitors targeting Pak1 activation and identified the inhibitor IPA-3. Remarkably, preactivated Pak1 is resistant to IPA-3. IPA-3 also inhibits activation of related Pak isoforms regulated by autoinhibition, but not more distantly related Paks, nor >200 other kinases tested. Pak1 inhibition by IPA-3 in live cells supports a critical role for Pak in PDGF-stimulated Erk activation. These studies illustrate an alternative strategy for kinase inhibition and introduce a highly selective, cell-permeable chemical inhibitor of Pak.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Homeostase/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Quinases Ativadas por p21/antagonistas & inibidores , Quinases Ativadas por p21/metabolismo , Animais , Dissulfetos/química , Dissulfetos/metabolismo , Dissulfetos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Camundongos , Naftóis/química , Naftóis/metabolismo , Naftóis/farmacologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Conformação Proteica/efeitos dos fármacos , Inibidores de Proteínas Quinases/metabolismo , Bibliotecas de Moléculas Pequenas/metabolismo , Especificidade por Substrato
19.
Nat Struct Mol Biol ; 11(8): 747-55, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15235593

RESUMO

Current drug discovery efforts focus primarily on proteins with defined enzymatic or small molecule binding sites. Autoregulatory domains represent attractive alternative targets for small molecule inhibitors because they also occur in noncatalytic proteins and because allosteric inhibitors may avoid specificity problems inherent in active site-directed inhibitors. We report here the identification of wiskostatin, a chemical inhibitor of the neural Wiskott-Aldrich syndrome protein (N-WASP). Wiskostatin interacts with a cleft in the regulatory GTPase-binding domain (GBD) of WASP in the solution structure of the complex. Wiskostatin induces folding of the isolated, unstructured GBD into its autoinhibited conformation, suggesting that wiskostatin functions by stabilizing N-WASP in its autoinhibited state. The use of small molecules to bias conformational equilibria represents a potentially general strategy for chemical inhibition of autoinhibited proteins, even in cases where such sites have not been naturally evolved in a target.


Assuntos
Carbazóis/farmacologia , Proteínas do Tecido Nervoso/fisiologia , Propanolaminas/farmacologia , Sítio Alostérico , Animais , Sítios de Ligação , Carbazóis/química , Catálise , Citoplasma/metabolismo , Relação Dose-Resposta a Droga , Desenho de Fármacos , Espectroscopia de Ressonância Magnética , Modelos Químicos , Modelos Moleculares , Proteínas do Tecido Nervoso/química , Propanolaminas/química , Conformação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Transdução de Sinais , Relação Estrutura-Atividade , Fatores de Tempo , Proteína Neuronal da Síndrome de Wiskott-Aldrich , Xenopus laevis/metabolismo
20.
Structure ; 15(2): 201-13, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17292838

RESUMO

p21-activated kinases have been classified into two groups based on their domain architecture. Group II PAKs (PAK4-6) regulate a wide variety of cellular functions, and PAK deregulation has been linked to tumor development. Structural comparison of five high-resolution structures comprising all active, monophosphorylated group II catalytic domains revealed a surprising degree of domain plasticity, including a number of catalytically productive and nonproductive conformers. Rearrangements of helix alphaC, a key regulatory element of kinase function, resulted in an additional helical turn at the alphaC N terminus and a distortion of its C terminus, a movement hitherto unseen in protein kinases. The observed structural changes led to the formation of interactions between conserved residues that structurally link the glycine-rich loop, alphaC, and the activation segment and firmly anchor alphaC in an active conformation. Inhibitor screening identified six potent PAK inhibitors from which a tri-substituted purine inhibitor was cocrystallized with PAK4 and PAK5.


Assuntos
Domínio Catalítico , Proteínas Serina-Treonina Quinases/química , Sequência de Aminoácidos , Animais , Domínio Catalítico/efeitos dos fármacos , Domínio Catalítico/genética , Cristalografia , Dados de Sequência Molecular , Conformação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Purinas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA