Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Commun Signal ; 21(1): 30, 2023 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-36737758

RESUMO

BACKGROUND: C3G is a guanine nucleotide exchange factor (GEF) that activates Rap1 to promote cell adhesion. Resting C3G is autoinhibited and the GEF activity is released by stimuli that signal through tyrosine kinases. C3G is activated by tyrosine phosphorylation and interaction with Crk adaptor proteins, whose expression is elevated in multiple human cancers. However, the molecular details of C3G activation and the interplay between phosphorylation and Crk interaction are poorly understood. METHODS: We combined biochemical, biophysical, and cell biology approaches to elucidate the mechanisms of C3G activation. Binding of Crk adaptor proteins to four proline-rich motifs (P1 to P4) in C3G was characterized in vitro using isothermal titration calorimetry and sedimentation velocity, and in Jurkat and HEK293T cells by affinity pull-down assays. The nucleotide exchange activity of C3G over Rap1 was measured using nucleotide-dissociation kinetic assays. Jurkat cells were also used to analyze C3G translocation to the plasma membrane and the C3G-dependent activation of Rap1 upon ligation of T cell receptors. RESULTS: CrkL interacts through its SH3N domain with sites P1 and P2 of inactive C3G in vitro and in Jurkat and HEK293T cells, and these sites are necessary to recruit C3G to the plasma membrane. However, direct stimulation of the GEF activity requires binding of Crk proteins to the P3 and P4 sites. P3 is occluded in resting C3G and is essential for activation, while P4 contributes secondarily towards complete stimulation. Tyrosine phosphorylation of C3G alone causes marginal activation. Instead, phosphorylation primes C3G lowering the concentration of Crk proteins required for activation and increasing the maximum activity. Unexpectedly, optimal activation also requires the interaction of CrkL-SH2 domain with phosphorylated C3G. CONCLUSION: Our study revealed that phosphorylation of C3G by Src and Crk-binding form a two-factor mechanism that ensures tight control of C3G activation. Additionally, the simultaneous SH2 and SH3N interaction of CrkL with C3G, required for the activation, reveals a novel adaptor-independent function of Crk proteins relevant to understanding their role in physiological signaling and their deregulation in diseases. Video abstract.


Assuntos
Fator 2 de Liberação do Nucleotídeo Guanina , Proteínas Nucleares , Humanos , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Células HEK293 , Proteínas Nucleares/metabolismo , Nucleotídeos/metabolismo , Proteínas Proto-Oncogênicas c-crk/metabolismo , Domínios de Homologia de src , Tirosina/metabolismo
2.
Cancer Res ; 81(21): 5438-5450, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34417202

RESUMO

Despite the approval of several multikinase inhibitors that target SRC and the overwhelming evidence of the role of SRC in the progression and resistance mechanisms of many solid malignancies, inhibition of its kinase activity has thus far failed to improve patient outcomes. Here we report the small molecule eCF506 locks SRC in its native inactive conformation, thereby inhibiting both enzymatic and scaffolding functions that prevent phosphorylation and complex formation with its partner FAK. This mechanism of action resulted in highly potent and selective pathway inhibition in culture and in vivo. Treatment with eCF506 resulted in increased antitumor efficacy and tolerability in syngeneic murine cancer models, demonstrating significant therapeutic advantages over existing SRC/ABL inhibitors. Therefore, this mode of inhibiting SRC could lead to improved treatment of SRC-associated disorders. SIGNIFICANCE: Small molecule-mediated inhibition of SRC impairing both catalytic and scaffolding functions confers increased anticancer properties and tolerability compared with other SRC/ABL inhibitors.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Quinase 1 de Adesão Focal/antagonistas & inibidores , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Pirazóis/farmacologia , Pirimidinas/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Quinases da Família src/antagonistas & inibidores , Animais , Apoptose , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Conformação Proteica , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases da Família src/química , Quinases da Família src/metabolismo
3.
Nucleic Acids Res ; 48(14): 8113-8127, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32658272

RESUMO

Quorum sensing allows bacterial cells to communicate through the release of soluble signaling molecules into the surrounding medium. It plays a pivotal role in controlling bacterial conjugation in Gram-positive cells, a process that has tremendous impact on health. Intracellular regulatory proteins of the RRNPP family are common targets of these signaling molecules. The RRNPP family of gene regulators bind signaling molecules at their C-terminal domain (CTD), but have highly divergent functionalities at their N-terminal effector domains (NTD). This divergence is also reflected in the functional states of the proteins, and is highly interesting from an evolutionary perspective. RappLS20 is an RRNPP encoded on the Bacillus subtilis plasmid pLS20. It relieves the gene repression effectuated by RcopLS20 in the absence of the mature pLS20 signaling peptide Phr*pLS20. We report here an in-depth structural study of apo and Phr*pLS20-bound states of RappLS20 at various levels of atomic detail. We show that apo-RappLS20 is dimeric and that Phr*pLS20-bound Rap forms NTD-mediated tetramers. In addition, we show that RappLS20 binds RcopLS20 directly in the absence of Phr*pLS20 and that addition of Phr*pLS20 releases RcopLS20 from RappLS20. This allows RcopLS20 to bind the promotor region of crucial conjugation genes blocking their expression.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Óperon , Multimerização Proteica , Transativadores/metabolismo , Bacillus subtilis , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Conjugação Genética/genética , Peptídeos/metabolismo , Regiões Promotoras Genéticas , Repetições de Tetratricopeptídeos , Transativadores/química , Transativadores/genética
4.
Structure ; 25(7): 1145-1152.e4, 2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28648606

RESUMO

The R2TP complex, comprising the Rvb1p-Rvb2p AAA-ATPases, Tah1p, and Pih1p in yeast, is a specialized Hsp90 co-chaperone required for the assembly and maturation of multi-subunit complexes. These include the small nucleolar ribonucleoproteins, RNA polymerase II, and complexes containing phosphatidylinositol-3-kinase-like kinases. The structure and stoichiometry of yeast R2TP and how it couples to Hsp90 are currently unknown. Here, we determine the 3D organization of yeast R2TP using sedimentation velocity analysis and cryo-electron microscopy. The 359-kDa complex comprises one Rvb1p/Rvb2p hetero-hexamer with domains II (DIIs) forming an open basket that accommodates a single copy of Tah1p-Pih1p. Tah1p-Pih1p binding to multiple DII domains regulates Rvb1p/Rvb2p ATPase activity. Using domain dissection and cross-linking mass spectrometry, we identified a unique region of Pih1p that is essential for interaction with Rvb1p/Rvb2p. These data provide a structural basis for understanding how R2TP couples an Hsp90 dimer to a diverse set of client proteins and complexes.


Assuntos
Adenosina Trifosfatases/química , Proteínas de Choque Térmico HSP90/química , Chaperonas Moleculares/química , Proteínas Nucleares/química , Proteínas de Saccharomyces cerevisiae/química , Fatores de Transcrição/química , Adenosina Trifosfatases/metabolismo , Sítios de Ligação , Proteínas de Choque Térmico HSP90/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo
5.
Mol Pharm ; 7(5): 1608-17, 2010 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-20715776

RESUMO

IB-01212, an antitumoral cyclodepsipeptide isolated from the mycelium of the marine fungus Clonostachys sp., showed leishmanicidal activity at a low micromolar range of concentrations on promastigote and amastigote forms of the parasite. Despite its cationic and amphipathic character, shared with other membrane active antibiotic peptides, IB-01212 did not cause plasma membrane lesions large enough to allow the entrance of the vital dye SYTOX green (MW = 600), even at concentrations causing full lethality of the parasite. Having ruled out massive disruption of the plasma membrane, we surmised the involvement of intracellular targets. Proof of concept for this assumption was provided by the mitochondrial dysfunction caused by IB-01212, which finally caused the death of the parasite through an apoptotic-like process. The size of the cycle, the preservation of the C2 symmetry, and the nature of the bonds linking the two tetrapeptide halves participate in the modulation of the leishmanicidal activity exerted by this compound. Here we discuss the potential of IB-01212 as a lead for new generations of surrogates to be used in chemotherapy treatments against Leishmania .


Assuntos
Antiprotozoários/farmacologia , Depsipeptídeos/farmacologia , Leishmania/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Antiprotozoários/administração & dosagem , Antiprotozoários/química , Apoptose/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Depsipeptídeos/administração & dosagem , Depsipeptídeos/química , Proteínas Fúngicas/administração & dosagem , Proteínas Fúngicas/química , Proteínas Fúngicas/farmacologia , Leishmania/citologia , Leishmania/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C
6.
Mol Pharm ; 6(3): 813-24, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19317431

RESUMO

Leishmaniasis is a human parasitic disease caused by infection by the protozoan Leishmania spp. Chemotherapy is currently the only treatment available, but its efficacy is increasingly challenged by the rising incidence of resistance and the frequent severe side effects associated with first-line drugs. Thus the development of leads with distinct mechanisms of action is urgently needed. A strategy often used for this purpose consists of assaying for leishmanicidal activity drugs formerly developed for other applications, such as amphotericin B (antifungal) or miltefosine (antitumor), among others, to profit from previous pharmacological and toxicological studies. Kahalalide F (KF) is a tumoricidal cyclic depsipeptide currently under phase II clinical trials for several types of cancer and psoriasis. Its mechanism of action has not been fully elucidated. Here we report the leishmanicidal activity of KF and its synthetic analogues at a micromolar range of concentrations. Its lethality is strongly linked to the alteration of the plasma membrane (PM) of the parasite based on (i) a rapid depolarization of the PM and uptake of the vital dye SYTOX Green upon its addition; (ii) evidence of severe morphological damage to the membrane of the parasite, as shown by transmission electron microscopy; and (iii) a rapid drop in the intracellular ATP levels, which correlates significantly with the leishmanicidal activity for active analogues, some of them with significant improvement of their therapeutic index with respect to the parental molecule. In addition to the basic knowledge obtained, this class of lethal mechanism is considerably less prone to the induction of resistance than classical drugs. All together, these observations foster further studies for the optimization of KF and its analogues as new anti-Leishmania leads with a new mode of action.


Assuntos
Antineoplásicos/farmacologia , Antiprotozoários/farmacologia , Antiprotozoários/uso terapêutico , Depsipeptídeos/farmacologia , Depsipeptídeos/uso terapêutico , Leishmania/efeitos dos fármacos , Leishmaniose/tratamento farmacológico , Animais , Antineoplásicos/química , Antiprotozoários/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/ultraestrutura , Células Cultivadas , Ensaios Clínicos como Assunto , Depsipeptídeos/química , Leishmania/ultraestrutura , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Estrutura Molecular
7.
Biochim Biophys Acta ; 1763(1): 110-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16377003

RESUMO

There is an increasing awareness of immune cell modulation by antimicrobial peptides. While this process often requires specific receptors for the peptides involved, several reports point out to a receptor-independent process. The cecropin A-melittin hybrid peptide CA(1-8)M(1-18) (KWKLFKKIGIGAVLKVLTTGLPALIS-amide) modifies gene expression in the macrophage line RAW 264.7 in the absence of any previous macrophage priming, suggesting a membrane permeation process. To further analyze the initial steps of this mechanism, we have studied the interaction of the peptide with these cells. Below 2 microM, CA(1-8)M(1-18) causes a concentration-dependent membrane depolarization partially reversible with time. At 2 microM, the accumulation of the SYTOX green vital dye is one half of that achieved with 0.05% Triton X-100. The binding level, as assessed by fluorescein-labeled CA(1-8)M(1-18), varies from 7.7+/-1.2 to 37.4+/-3.9 x 10(6) molecules/cell over a 0.5-4.0 microM concentration range. Electrophysiological experiments with 0.5 microM CA(1-8)M(1-18), a concentration that triggers maximal NOS2 expression and minimal toxicity, show a reversible current induction in the RAW 264.7 plasma membrane that is maintained as far as peptide is present. This activation of the macrophage involves the production of nitric oxide, a metabolite lethal for many pathogens that results from unspecific membrane permeation by antimicrobial peptides, and represents a new mode of action that may open new therapeutic possibilities for these compounds against intracellular pathogens.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Meliteno/farmacologia , Monócitos/enzimologia , Óxido Nítrico Sintase Tipo II/biossíntese , Peptídeos/farmacologia , Animais , Células Cultivadas , Eletrofisiologia , Indução Enzimática/efeitos dos fármacos , Corantes Fluorescentes , Camundongos , Monócitos/citologia , NF-kappa B/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA