Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Genes Dev ; 38(11-12): 528-535, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38960718

RESUMO

As part of the efforts to understand nuclear IκB function in NF-κB-dependent gene expression, we report an X-ray crystal structure of the IκBζ ankyrin repeat domain in complex with the dimerization domain of the NF-κB p50 homodimer. IκBζ possesses an N-terminal α helix that conveys domain folding stability. Affinity and specificity of the complex depend on a small portion of p50 at the nuclear localization signal. The model suggests that only one p50 subunit supports binding with IκBζ, and biochemical experiments confirm that IκBζ associates with DNA-bound NF-κB p50:RelA heterodimers. Comparisons of IκBζ:p50 and p50:κB DNA complex crystallographic models indicate that structural rearrangement is necessary for ternary complex formation of IκBζ and p50 with DNA.


Assuntos
Modelos Moleculares , Subunidade p50 de NF-kappa B , Ligação Proteica , Multimerização Proteica , Humanos , Sequência de Aminoácidos , Núcleo Celular/metabolismo , Cristalografia por Raios X , DNA/metabolismo , DNA/química , Proteínas I-kappa B/metabolismo , Proteínas I-kappa B/química , Proteínas I-kappa B/genética , Subunidade p50 de NF-kappa B/metabolismo , Subunidade p50 de NF-kappa B/química , Subunidade p50 de NF-kappa B/genética , Fator de Transcrição RelA/metabolismo , Fator de Transcrição RelA/química , Fator de Transcrição RelA/genética
2.
Nat Commun ; 15(1): 3785, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38710674

RESUMO

Mutations in human isocitrate dehydrogenase 1 (IDH1) drive tumor formation in a variety of cancers by replacing its conventional activity with a neomorphic activity that generates an oncometabolite. Little is understood of the mechanistic differences among tumor-driving IDH1 mutants. We previously reported that the R132Q mutant unusually preserves conventional activity while catalyzing robust oncometabolite production, allowing an opportunity to compare these reaction mechanisms within a single active site. Here, we employ static and dynamic structural methods and observe that, compared to R132H, the R132Q active site adopts a conformation primed for catalysis with optimized substrate binding and hydride transfer to drive improved conventional and neomorphic activity over R132H. This active site remodeling reveals a possible mechanism of resistance to selective mutant IDH1 therapeutic inhibitors. This work enhances our understanding of fundamental IDH1 mechanisms while pinpointing regions for improving inhibitor selectivity.


Assuntos
Domínio Catalítico , Isocitrato Desidrogenase , Mutação , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Humanos , Cinética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores Enzimáticos/farmacologia
3.
Res Sq ; 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38464189

RESUMO

Mutations in human isocitrate dehydrogenase 1 (IDH1) drive tumor formation in a variety of cancers by replacing its conventional activity with a neomorphic activity that generates an oncometabolite. Little is understood of the mechanistic differences among tumor-driving IDH1 mutants. We previously reported that the R132Q mutant uniquely preserves conventional activity while catalyzing robust oncometabolite production, allowing an opportunity to compare these reaction mechanisms within a single active site. Here, we employed static and dynamic structural methods and found that, compared to R132H, the R132Q active site adopted a conformation primed for catalysis with optimized substrate binding and hydride transfer to drive improved conventional and neomorphic activity over R132H. This active site remodeling revealed a possible mechanism of resistance to selective mutant IDH1 therapeutic inhibitors. This work enhances our understanding of fundamental IDH1 mechanisms while pinpointing regions for improving inhibitor selectivity.

4.
bioRxiv ; 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38260668

RESUMO

Mutations in human isocitrate dehydrogenase 1 (IDH1) drive tumor formation in a variety of cancers by replacing its conventional activity with a neomorphic activity that generates an oncometabolite. Little is understood of the mechanistic differences among tumor-driving IDH1 mutants. We previously reported that the R132Q mutant uniquely preserves conventional activity while catalyzing robust oncometabolite production, allowing an opportunity to compare these reaction mechanisms within a single active site. Here, we employed static and dynamic structural methods and found that, compared to R132H, the R132Q active site adopted a conformation primed for catalysis with optimized substrate binding and hydride transfer to drive improved conventional and neomorphic activity over R132H. This active site remodeling revealed a possible mechanism of resistance to selective mutant IDH1 therapeutic inhibitors. This work enhances our understanding of fundamental IDH1 mechanisms while pinpointing regions for improving inhibitor selectivity.

5.
Biomolecules ; 13(9)2023 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-37759710

RESUMO

Though originally characterized as an inactive or transcriptionally repressive factor, the NF-κB p50 homodimer has become appreciated as a physiologically relevant driver of specific target gene expression. By virtue of its low affinity for cytoplasmic IκB protein inhibitors, p50 accumulates in the nucleus of resting cells, where it is a binding target for the transcriptional co-activator IκBζ. In this study, we employed X-ray crystallography to analyze the structure of the p50 homodimer on κB DNA from the promoters of human interleukin-6 (IL-6) and neutrophil-gelatinase-associated lipocalin (NGAL) genes, both of which respond to IκBζ. The NF-κB p50 homodimer binds 11-bp on IL-6 κB DNA, while, on NGAL κB DNA, the spacing is 12-bp. This begs the question: what DNA binding mode is preferred by NF-κB p50 homodimer? To address this, we engineered a "Test" κB-like DNA containing the core sequence 5'-GGGGAATTCCCC-3' and determined its X-ray crystal structure in complex with p50. This revealed that, when presented with multiple options, NF-κB p50 homodimer prefers to bind 11-bp, which necessarily imposes asymmetry on the complex despite the symmetry inherent in both the protein and its target DNA, and that the p50 dimerization domain can contact DNA via distinct modes.


Assuntos
Interleucina-6 , Subunidade p50 de NF-kappa B , NF-kappa B , Humanos , Cristalografia por Raios X , DNA , Lipocalina-2 , Raios X , Subunidade p50 de NF-kappa B/química , Subunidade p50 de NF-kappa B/fisiologia
6.
Mol Cancer Res ; 21(2): 170-186, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36214671

RESUMO

Disease recurrence in high-grade serous ovarian cancer may be due to cancer stem-like cells (CSC) that are resistant to chemotherapy and capable of reestablishing heterogeneous tumors. The alternative NF-κB signaling pathway is implicated in this process; however, the mechanism is unknown. Here we show that TNF-like weak inducer of apoptosis (TWEAK) and its receptor, Fn14, are strong inducers of alternative NF-κB signaling and are enriched in ovarian tumors following chemotherapy treatment. We further show that TWEAK enhances spheroid formation ability, asymmetric division capacity, and expression of SOX2 and epithelial-to-mesenchymal transition genes VIM and ZEB1 in ovarian cancer cells, phenotypes that are enhanced when TWEAK is combined with carboplatin. Moreover, TWEAK in combination with chemotherapy induces expression of the CSC marker CD117 in CD117- cells. Blocking the TWEAK-Fn14-RelB signaling cascade with a small-molecule inhibitor of Fn14 prolongs survival following carboplatin chemotherapy in a mouse model of ovarian cancer. These data provide new insights into ovarian cancer CSC biology and highlight a signaling axis that should be explored for therapeutic development. IMPLICATIONS: This study identifies a unique mechanism for the induction of ovarian cancer stem cells that may serve as a novel therapeutic target for preventing relapse.


Assuntos
NF-kappa B , Neoplasias Ovarianas , Humanos , Animais , Feminino , Camundongos , NF-kappa B/metabolismo , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/metabolismo , Carboplatina/farmacologia , Receptores do Fator de Necrose Tumoral/genética , Receptor de TWEAK/genética , Linhagem Celular Tumoral , Recidiva Local de Neoplasia/tratamento farmacológico , Citocina TWEAK , Transdução de Sinais/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Células-Tronco/metabolismo , Fator de Transcrição RelB/metabolismo
7.
Bioorg Med Chem Lett ; 65: 128713, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35367592

RESUMO

The IKK-NFκB complex is a key signaling node that facilitates activation of gene expression in response to extracellular signals. The kinase IKKß and the transcription factor RELA have been targeted by covalent modifiers that bind to surface exposed cysteine residues. A common feature in well characterized covalent modifiers of RELA and IKKß is the Michael acceptor containing α-methylene-γ-butyrolactone functionality. Through synthesis and evaluation of a focused set of α-methylene-γ-butyrolactone containing spirocyclic dimers (SpiDs) we identified SpiD3 as an anticancer agent with low nanomolar potency. Using cell-free and cell-based studies we show that SpiD3 is a covalent modifier that generates stable RELA containing high molecular weight complexes. SpiD3 inhibits TNFα-induced IκBα phosphorylation resulting in the blockade of RELA nuclear translocation. SpiD3 induces apoptosis, inhibits colony formation and migration of cancer cells. The NCI-60 cell line screen revealed that SpiD3 potently inhibits growth of leukemia cell lines, making it a suitable pre-therapeutic lead for hematological malignancies.


Assuntos
Antineoplásicos , Isatina , 4-Butirolactona/análogos & derivados , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Quinase I-kappa B/metabolismo , Isatina/farmacologia , NF-kappa B/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
8.
RSC Chem Biol ; 3(1): 32-36, 2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-35128406

RESUMO

Tumor necrosis factor (TNF) α-induced nuclear translocation of the NF-κB subunit RELA has been implicated in several pathological conditions. Here we report the discovery of a spirocyclic dimer (SpiD7) that covalently modifies RELA to inhibit TNFα-induced nuclear translocation. This is a previously unexplored strategy to inhibit TNFα-induced NF-κB activation.

9.
Chem Commun (Camb) ; 57(38): 4678-4681, 2021 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-33977973

RESUMO

Inhibitor of nuclear factor kappa-B kinase subunit beta (IKKß) is a key regulator of the cannonical NF-κB pathway. IKKß has been validated as a drug target for pathological conditions, which include chronic inflammatory diseases and cancer. Pharmacological studies revealed that chronic administration of ATP-competitive IKKß inhibitors resulted in unexpected toxicity. We previously reported the discovery of 13-197 as a non-toxic IKKß inhibitor that reduced tumor growth. Here, we show that 13-197 inhibits IKKß in a ATP non-competitive manner and an allosteric pocket at the interface of the kinase and ubiquitin like domains was identified as the potential binding site.


Assuntos
Trifosfato de Adenosina/metabolismo , Quinase I-kappa B/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Sítios de Ligação/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Quinase I-kappa B/metabolismo , Estrutura Molecular , Inibidores de Proteínas Quinases/química , Bibliotecas de Moléculas Pequenas/química
10.
Biochemistry ; 57(20): 2943-2957, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29708732

RESUMO

Transcription activator proteins typically contain two functional domains: a DNA binding domain (DBD) that binds to DNA with sequence specificity and an activation domain (AD) whose established function is to recruit RNA polymerase. In this report, we show that purified recombinant nuclear factor κB (NF-κB) RelA dimers bind specific κB DNA sites with an affinity significantly lower than that of the same dimers from nuclear extracts of activated cells, suggesting that additional nuclear cofactors might facilitate DNA binding by the RelA dimers. Additionally, recombinant RelA binds DNA with relatively low affinity at a physiological salt concentration in vitro. The addition of p53 or RPS3 (ribosomal protein S3) increases RelA:DNA binding affinity 2- to >50-fold depending on the protein and ionic conditions. These cofactor proteins do not form stable ternary complexes, suggesting that they stabilize the RelA:DNA complex through dynamic interactions. Surprisingly, the RelA-DBD alone fails to bind DNA under the same solution conditions even in the presence of cofactors, suggesting an important role of the RelA-AD in DNA binding. Reduced RelA:DNA binding at a physiological ionic strength suggests that multiple cofactors might be acting simultaneously to mitigate the electrolyte effect and stabilize the RelA:DNA complex in vivo. Overall, our observations suggest that the RelA-AD and multiple cofactor proteins function cooperatively to prime the RelA-DBD and stabilize the RelA:DNA complex in cells. Our study provides a mechanism for nuclear cofactor proteins in NF-κB-dependent gene regulation.


Assuntos
Coenzimas/química , Proteínas de Ligação a DNA/química , NF-kappa B/química , Fator de Transcrição RelA/química , Linhagem Celular , Núcleo Celular/química , Núcleo Celular/genética , Coenzimas/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Humanos , NF-kappa B/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Domínios Proteicos , Proteínas Ribossômicas/química , Fator de Transcrição RelA/genética , Proteína Supressora de Tumor p53/química
11.
J Lipid Res ; 58(7): 1386-1398, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28507038

RESUMO

Cysteinyl leukotrienes (CysLTs) are a small family of biological signaling lipids produced by active leukocytes that contribute to diverse inflammatory disease states as a consequence of their engagement with dedicated G protein-coupled receptors. Immunization of mice with a CysLT-modified hapten carrier protein yielded novel monoclonal antibodies that display variable binding affinity to CysLTs. Solution binding assays indicated differing specificities among the antibodies tested, with antibody 10G4 displaying a preference for leukotriene C4 (LTC4). X-ray crystallography of a humanized 10G4 Fab fragment in complex with LTC4 revealed that binding induces a hook-like conformation within the hydrocarbon tail of the lipid arachidonic acid moiety. Specific hydrogen bonding to the LTC4 carboxylate groups further stabilized the complex, while a water molecule mediated a hydrogen bond network that connected the N-terminal arm of l-glutathione to both the arachidonyl carboxylate of LTC4 and the antibody heavy chain. Prophylactic administration of two anti-CysLT antibodies in mice followed by challenge with LTC4 demonstrated their in vivo efficacy against acute inflammation in a vascular permeability model. 10G4 ameliorated the effects of acute dextran sulfate sodium-induced colitis, suggesting that anti-CysLT antibodies could provide a therapeutic benefit in the treatment of inflammatory diseases.


Assuntos
Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Especificidade de Anticorpos , Colite/imunologia , Colite/terapia , Cisteína/imunologia , Leucotrienos/imunologia , Doença Aguda , Animais , Anticorpos Monoclonais Humanizados/química , Vasos Sanguíneos/metabolismo , Colite/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imunização , Camundongos , Modelos Moleculares , Permeabilidade , Conformação Proteica
12.
J Am Chem Soc ; 135(39): 14504-7, 2013 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-24028339

RESUMO

Boron in the ocean is generally considered a nonbiological element due to its relatively high concentration (0.4 mM) and depth independent concentration profile. Here we report an unexpected role for boron in the iron transport system of the marine bacterium Marinobacter algicola. Proteome analysis under varying boron concentrations revealed that the periplasmic ferric binding protein (Mb-FbpA) was among the proteins whose expression was most affected, strongly implicating the involvement of boron in iron utilization. Here we show that boron facilitates Fe(3+) sequestration by Mb-FbpA at pH 8 (oceanic pH) by acting as a synergistic anion (B(OH)4(1-)). Fe(3+) sequestration does not occur at pH 6.5 where boric acid (B(OH)3; pK(a) = 8.55) is the predominant species. Borate anion is also shown to bind to apo-Mb-FbpA with mM affinity at pH 8, consistent with the biological relevance implied from boron's oceanic concentration (0.4 mM). Borate is among those synergistic anions tested which support the strongest Fe(3+) binding to Mb-FbpA, where the range of anion dependent affinity constants is log K'(eff) = 21-22. Since the pKa of boric acid (8.55) lies near the pH of ocean water, changes in oceanic pH, as a consequence of fluctuations in atmospheric CO2, may perturb iron uptake in many marine heterotrophic bacteria due to a decrease in oceanic borate anion concentration.


Assuntos
Proteínas de Bactérias/metabolismo , Boratos/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Marinobacter/metabolismo , Ânions/metabolismo , Boro/metabolismo , Ferro/metabolismo , Modelos Moleculares
13.
Methods ; 56(1): 25-32, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22178692

RESUMO

Biophysical and structural studies on muscle myosin rely upon milligram quantities of extremely pure material. However, many biologically interesting myosin isoforms are expressed at levels that are too low for direct purification from primary tissues. Efforts aimed at recombinant expression of functional striated muscle myosin isoforms in bacterial or insect cell culture have largely met with failure, although high level expression in muscle cell culture has recently been achieved at significant expense. We report a novel method for the use of strains of the fruit fly Drosophila melanogaster genetically engineered to produce histidine-tagged recombinant muscle myosin isoforms. This method takes advantage of the single muscle myosin heavy chain gene within the Drosophila genome, the high level of expression of accessible myosin in the thoracic indirect flight muscles, the ability to knock out endogenous expression of myosin in this tissue and the relatively low cost of fruit fly colony production and maintenance. We illustrate this method by expressing and purifying a recombinant histidine-tagged variant of embryonic body wall skeletal muscle myosin II from an engineered fly strain. The recombinant protein shows the expected ATPase activity and is of sufficient purity and homogeneity for crystallization. This system may prove useful for the expression and isolation of mutant myosins associated with skeletal muscle diseases and cardiomyopathies for their biochemical and structural characterization.


Assuntos
Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Voo Animal , Expressão Gênica , Miosinas/genética , Miosinas/isolamento & purificação , Transgenes/genética , Animais , Cromatografia de Afinidade/métodos , Técnicas de Inativação de Genes , Miosinas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/metabolismo
14.
Structure ; 19(3): 397-408, 2011 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-21397190

RESUMO

UCS proteins, such as UNC-45, influence muscle contraction and other myosin-dependent motile processes. We report the first X-ray crystal structure of a UCS domain-containing protein, the UNC-45 myosin chaperone from Drosophila melanogaster (DmUNC-45). The structure reveals that the central and UCS domains form a contiguous arrangement of 17 consecutive helical layers that arrange themselves into five discrete armadillo repeat subdomains. Small-angle X-ray scattering data suggest that free DmUNC-45 adopts an elongated conformation and exhibits flexibility in solution. Protease sensitivity maps to a conserved loop that contacts the most carboxy-terminal UNC-45 armadillo repeat subdomain. Amino acid conservation across diverse UCS proteins maps to one face of this carboxy-terminal subdomain, and the majority of mutations that affect myosin-dependent cellular activities lie within or around this region. Our crystallographic, biophysical, and biochemical analyses suggest that DmUNC-45 function is afforded by its flexibility and by structural integrity of its UCS domain.


Assuntos
Proteínas do Domínio Armadillo/química , Proteínas de Drosophila/química , Chaperonas Moleculares/química , Proteínas Recombinantes/química , Sequência de Aminoácidos , Animais , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/metabolismo , Caenorhabditis elegans , Cristalografia por Raios X , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/química , Escherichia coli , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Dados de Sequência Molecular , Mutação , Miosinas/metabolismo , Maleabilidade , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espalhamento a Baixo Ângulo , Alinhamento de Sequência
15.
Proc Natl Acad Sci U S A ; 106(42): 17717-22, 2009 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-19815502

RESUMO

The pleiotropic signaling lipid sphingosine-1-phosphate (S1P) plays significant roles in angiogenesis, heart disease, and cancer. LT1009 (also known as sonepcizumab) is a humanized monoclonal antibody that binds S1P with high affinity and specificity. Because the antibody is currently in clinical trials, it is important to confirm by structural and biochemical analyses that it binds its target in a predictable manner. Therefore, we determined the structure of a complex between the LT1009 antibody Fab fragment and S1P refined to 1.90 A resolution. The antibody employs unique and diverse strategies to recognize its antigen. Two metal ions bridge complementarity determining regions from the antibody light chain and S1P. The coordination geometry, inductively coupled plasma spectroscopy, surface plasmon resonance spectroscopy, and biochemical assays suggest that these are Ca(2+). The amino alcohol head group of the sphingosine backbone is recognized through hydrogen bonding interactions from 1 aa side chain and polypeptide backbone atoms of the antibody light and heavy chains. The S1P hydrophobic tail is almost completely enclosed within a hydrophobic channel formed primarily by the heavy chain. Both treatment of the complex with metal chelators and mutation of amino acids in the light chain that coordinate the metal atoms or directly contact the polar head group abrogate binding, while mutations within the hydrophobic cavity also decrease S1P binding affinity. The structure suggests mechanistic details for recognition of a signaling lipid by a therapeutic antibody candidate. Moreover, this study provides direct structural evidence that antibodies are capable of using metals to bridge antigen:antibody complexes.


Assuntos
Anticorpos Monoclonais/química , Complexo Antígeno-Anticorpo/química , Fragmentos Fab das Imunoglobulinas/química , Lisofosfolipídeos/química , Lisofosfolipídeos/imunologia , Esfingosina/análogos & derivados , Animais , Anticorpos Monoclonais/genética , Afinidade de Anticorpos , Especificidade de Anticorpos , Sítios de Ligação de Anticorpos/genética , Cálcio/química , Cristalografia por Raios X , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Técnicas In Vitro , Lisofosfolipídeos/antagonistas & inibidores , Camundongos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Conformação Proteica , Esfingosina/antagonistas & inibidores , Esfingosina/química , Esfingosina/imunologia , Ressonância de Plasmônio de Superfície
16.
Cold Spring Harb Perspect Biol ; 1(3): a000075, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20066103

RESUMO

The prosurvival transcription factor NF-kappaB specifically binds promoter DNA to activate target gene expression. NF-kappaB is regulated through interactions with IkappaB inhibitor proteins. Active proteolysis of these IkappaB proteins is, in turn, under the control of the IkappaB kinase complex (IKK). Together, these three molecules form the NF-kappaB signaling module. Studies aimed at characterizing the molecular mechanisms of NF-kappaB, IkappaB, and IKK in terms of their three-dimensional structures have lead to a greater understanding of this vital transcription factor system.


Assuntos
Proteínas I-kappa B/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Animais , DNA/química , Dimerização , Humanos , Modelos Biológicos , Conformação Molecular , Complexo de Endopeptidases do Proteassoma/química , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Fator de Transcrição RelA/química
17.
J Mol Biol ; 379(1): 122-35, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18436238

RESUMO

Although they share sequence homology to classical cytoplasmic I kappaB inhibitors of transcription factor NF-kappaB, the proteins I kappaB zeta, Bcl-3, and I kappa BNS function in the nucleus as factors that influence NF-kappaB-dependent gene expression profiles. Through the use of purified recombinant proteins and by comparison with the classical I kappaB protein I kappaB alpha, we have discovered mechanistic details of the interaction between I kappaB zeta and functional NF-kappaB dimers. Whereas I kappaB alpha and other classical I kappaB proteins bind tightly to NF-kappaB dimers that possess the p65 subunit, I kappaB zeta binds preferentially to NF-kappaB p50 homodimers. This altered specificity is particularly interesting in light of the fact that both NF-kappaB subunits exhibit high sequence and structural homology, while the I kappaB alpha and I kappaB zeta proteins are also conserved in primary amino acid sequence. We further show that I kappaB zeta is capable of forming a stable ternary complex with the NF-kappaB p50 homodimer and kappaB DNA. Again, this is a stark contrast from I kappaB alpha, which inhibits NF-kappaB p65 homodimer binding to NF-kappaB target DNA sequences. Removal of the DNA sequences flanking the NF-kappaB binding site does not directly affect the interaction of p50 and I kappaB zeta. Rather, we have discovered that the carboxy-terminal glycine-rich region of the NF-kappaB p50 homodimer is involved in mediating high-affinity binding of I kappaB zeta and NF-kappaB p50. We conclude that the NF-kappaB p50 homodimer functions as a legitimate activator of gene expression through formation of a ternary complex between itself, I kappaB zeta, and DNA. The requirement for formation of this complex could explain why the nuclear I kappaB protein I kappaB zeta is absolutely required for expression of the pluripotent pro-inflammatory cytokine interleukin-6 in peritoneal macrophages.


Assuntos
DNA/química , Subunidade p50 de NF-kappa B/química , Proteínas Nucleares/química , Proteínas Adaptadoras de Transdução de Sinal , Animais , Sequência de Bases , DNA/metabolismo , Dimerização , Ensaio de Desvio de Mobilidade Eletroforética , Glicina/química , Humanos , Proteínas I-kappa B , Interleucina-6/genética , Camundongos , Dados de Sequência Molecular , Subunidade p50 de NF-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
18.
J Mol Neurosci ; 30(1-2): 103-4, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17192648

RESUMO

Many peptidic toxins from animal venoms target neuronal or peripheral synaptic receptors with high affinities and specificities. Hence, these toxins are not only potent natural weapons but also precise molecular tools for pharmacological studies of their receptors. Although they belong to various structural and/or functional subfamilies, they often share similar molecular features, such as a highly reticulated scaffold presenting specific binding determinants.


Assuntos
Acetilcolina/metabolismo , Venenos Elapídicos/química , Proteínas do Tecido Nervoso/química , Peptídeos/química , Sinapses/fisiologia , Animais , Inibidores da Colinesterase
19.
J Mol Biol ; 360(2): 421-34, 2006 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-16756995

RESUMO

IkappaBalpha is an ankyrin repeat protein that inhibits NF-kappaB transcriptional activity by sequestering NF-kappaB outside of the nucleus in resting cells. We have characterized the binding thermodynamics and kinetics of the IkappaBalpha ankyrin repeat domain to NF-kappaB(p50/p65) using surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC). SPR data showed that the IkappaBalpha and NF-kappaB associate rapidly but dissociate very slowly, leading to an extremely stable complex with a K(D,obs) of approximately 40 pM at 37 degrees C. As reported previously, the amino-terminal DNA-binding domain of p65 contributes little to the overall binding affinity. Conversely, helix four of p65, which forms part of the nuclear localization sequence, was essential for high-affinity binding. This was surprising, given the small size of the binding interface formed by this part of p65. The NF-kappaB(p50/p65) heterodimer and p65 homodimer bound IkappaBalpha with almost indistinguishable thermodynamics, except that the NF-kappaB p65 homodimer was characterized by a more favorable DeltaH(obs) relative to the NF-kappaB(p50/p65) heterodimer. Both interactions were characterized by a large negative heat capacity change (DeltaC(P,obs)), approximately half of which was contributed by the p65 helix four that was necessary for tight binding. This could not be accounted for readily by the small loss of buried non-polar surface area and we hypothesize that the observed effect is due to additional folding of some regions of the complex.


Assuntos
Proteínas I-kappa B/metabolismo , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/metabolismo , Fator de Transcrição RelA/química , Fator de Transcrição RelA/metabolismo , Animais , DNA/metabolismo , Dimerização , Humanos , Cinética , Camundongos , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Inibidor de NF-kappaB alfa , Subunidade p50 de NF-kappa B/química , Subunidade p50 de NF-kappa B/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Ressonância de Plasmônio de Superfície , Temperatura , Termodinâmica
20.
Protein Sci ; 13(7): 1767-77, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15215520

RESUMO

The crystal structure of IkappaBalpha in complex with the transcription factor, nuclear factor kappa-B (NF-kappaB) shows six ankyrin repeats, which are all ordered. Electron density was not observed for most of the residues within the PEST sequence, although it is required for high-affinity binding. To characterize the folded state of IkappaBalpha (67-317) when it is not in complex with NF-kappaB, we have carried out circular dichroism (CD) spectroscopy, 8-anilino-1-napthalenesulphonic acid (ANS) binding, differential scanning calorimetry, and amide hydrogen/deuterium exchange experiments. The CD spectrum shows the presence of helical structure, consistent with other ankyrin repeat proteins. The large amount of ANS-binding and amide exchange suggest that the protein may have molten globule character. The amide exchange experiments show that the third ankyrin repeat is the most compact, the second and fourth repeats are somewhat less compact, and the first and sixth repeats are solvent exposed. The PEST extension is also highly solvent accessible. Ikappa Balpha unfolds with a T(m) of 42 degrees C, and forms a soluble aggregate that sequesters helical and variable loop parts of the first, fourth, and sixth repeats and the PEST extension. The second and third repeats, which conform most closely to a consensus for stable ankyrin repeats, appear to remain outside of the aggregate. The ramifications of these observations for the biological function of IkappaBalpha are discussed.


Assuntos
Repetição de Anquirina , Deutério/química , Hidrogênio/química , Proteínas I-kappa B/química , Amidas/química , Motivos de Aminoácidos , Animais , Varredura Diferencial de Calorimetria , Dicroísmo Circular , Humanos , Inibidor de NF-kappaB alfa , NF-kappa B/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Fator de Transcrição RelA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA