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










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-22684055

RESUMO

The role of ADAM-8 in cancer and inflammatory diseases such as allergy, arthritis and asthma makes it an attractive target for drug development. Therefore, the catalytic domain of human ADAM-8 was expressed, purified and crystallized in complex with a hydroxamic acid inhibitor, batimastat. The crystal structure of the enzyme-inhibitor complex was refined to 2.1 Å resolution. ADAM-8 has an overall fold similar to those of other ADAM members, including a central five-stranded ß-sheet and a catalytic Zn(2+) ion. However, unique differences within the S1' binding loop of ADAM-8 are observed which might be exploited to confer specificity and selectivity to ADAM-8 competitive inhibitors for the treatment of diseases involving this enzyme.


Assuntos
Proteínas ADAM/química , Domínio Catalítico , Proteínas de Membrana/química , Fenilalanina/análogos & derivados , Inibidores de Proteases/química , Tiofenos/química , Proteínas ADAM/metabolismo , Humanos , Ligantes , Proteínas de Membrana/metabolismo , Modelos Moleculares , Fenilalanina/química , Fenilalanina/metabolismo , Inibidores de Proteases/metabolismo , Ligação Proteica , Desdobramento de Proteína , Tiofenos/metabolismo
2.
Protein Pept Lett ; 19(5): 485-91, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22486643

RESUMO

The Janus kinase (JAK) family consists of four members: JAK-1, -2, -3 and tyrosine kinase 2 (TYK-2). Recent work suggests that cytokine signaling through TYK-2 may play a critical role in a number of inflammatory processes. We recently described the purification and characterization of phosphorylated isoforms of the TYK-2 kinase domain (TYK-2 KD) and its high resolution 3D structure in the presence of inhibitors. We now report the expression and a two-step purification procedure for the doubly tagged full-length construct, H6-FL-TYK-2-FLAG, and examine its properties compared to those of TYK-2 KD. In the presence of ATP and a peptide substrate, H6-FL-TYK-2-FLAG showed a marked lag in phosphopeptide product formation, while TYK-2 KD showed no such lag. This lag could be eliminated by ATP pretreatment, suggesting that the H6-FL-TYK-2-FLAG enzyme was activated by phosphorylation. The potencies of several nanomolar inhibitors were similar for TYK-2 KD and H6-FL-TYK-2-FLAG. However, these same inhibitors were about 1000 times less potent inhibiting the autophosphorylation of H6-FL-TYK-2-FLAG than they were inhibiting the phosphorylation of a peptide substrate modeled after the activation loop sequence of TYK-2. This intriguing result suggests that autophosphorylation and, thus, activation of H6-FL-TYK-2-FLAG is relatively insensitive to inhibition and that present inhibitors act to inhibit TYK-2 subsequent to activation. Inhibition of TYK-2 autophosphorylation may represent a new area of investigation for the JAK family.


Assuntos
TYK2 Quinase/antagonistas & inibidores , TYK2 Quinase/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Domínio Catalítico , Eletroforese em Gel de Poliacrilamida , Histidina/química , Humanos , Cinética , Oligopeptídeos/química , Fosforilação , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , TYK2 Quinase/química , TYK2 Quinase/isolamento & purificação
3.
Protein Sci ; 20(4): 735-44, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21370305

RESUMO

A ((1S,2R)-2-hydroxy-2,3-dihydro-1H-inden-1-yl) succinamide derivative (here referred to as Compound 12) shows significant activity toward many matrix metalloproteinases (MMPs), including MMP-2, MMP-8, MMP-9, and MMP-13. Modeling studies had predicted that this compound would not bind to ADAMTS-5 (a disintegrin and metalloproteinase with thrombospondin motifs-5) due to its shallow S1' pocket. However, inhibition analysis revealed it to be a nanomolar inhibitor of both ADAMTS-4 and -5. The observed inconsistency was explained by analysis of crystallographic structures, which showed that Compound 12 in complex with the catalytic domain of ADAMTS-5 (cataTS5) exhibits an unusual conformation in the S1' pocket of the protein. This first demonstration that cataTS5 can undergo an induced conformational change in its active site pocket by a molecule like Compound 12 should enable the design of new aggrecanase inhibitors with better potency and selectivity profiles.


Assuntos
Proteínas ADAM/química , Amidas/química , Conformação Proteica , Proteína ADAMTS5 , Animais , Domínio Catalítico , Bovinos , Desenho de Fármacos , Humanos , Inibidores de Metaloproteinases de Matriz , Metaloproteinases da Matriz/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Succinatos
4.
Biochim Biophys Acta ; 1804(9): 1817-31, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20580860

RESUMO

Because of their central role in programmed cell death, the caspases are attractive targets for developing new therapeutics against cancer and autoimmunity, myocardial infarction and ischemic damage, and neurodegenerative diseases. We chose to target caspase-3, an executioner caspase, and caspase-8, an initiator caspase, based on the vast amount of information linking their functions to diseases. Through a structure-based drug design approach, a number of novel beta-strand peptidomimetic compounds were synthesized. Kinetic studies of caspase-3 and caspase-8 inhibition were carried out with these urazole ring-containing irreversible peptidomimetics and a known irreversible caspase inhibitor, Z-VAD-fmk. Using a stopped-flow fluorescence assay, we were able to determine individual kinetic parameters of caspase-3 and caspase-8 inhibition by these inhibitors. Z-VAD-fmk and the peptidomimetic inhibitors inhibit caspase-3 and caspase-8 via a three-step kinetic mechanism. Inhibition of both caspase-3 and caspase-8 by Z-VAD-fmk and of caspase-3 by the peptidomimetic inhibitors proceeds via two rapid equilibrium steps followed by a relatively fast inactivation step. However, caspase-8 inhibition by the peptidomimetics goes through a rapid equilibrium step, a slow-binding reversible step, and an extremely slow inactivation step. The crystal structures of inhibitor complexes of caspases-3 and -8 validate the design of the inhibitors by illustrating in detail how they mimic peptide substrates. One of the caspase-8 structures also shows binding at a secondary, allosteric site, providing a possible route to the development of noncovalent small molecule modulators of caspase activity.


Assuntos
Caspase 3/química , Caspase 8/química , Inibidores de Cisteína Proteinase/farmacologia , Inibidores de Caspase , Cristalização , Cristalografia por Raios X , Inibidores de Cisteína Proteinase/síntese química , Humanos , Cinética , Estrutura Molecular , Conformação Proteica
5.
Biochem Biophys Res Commun ; 396(2): 543-8, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20435017

RESUMO

The Janus kinase family consists of four members: JAK-1, -2, -3 and TYK-2. While JAK-2 and JAK-3 have been well characterized biochemically, there is little data on TYK-2. Recent work suggests that TYK-2 may play a critical role in the development of a number of inflammatory processes. We have carried out a series of biochemical studies to better understand TYK-2 enzymology and its inhibition profile, in particular how the TYK-2 phosphorylated forms differ from each other and from the other JAK family members. We have expressed and purified milligram quantities of the TYK-2 kinase domain (KD) to high purity and developed a method to separate the non-, mono- (pY(1054)) and di-phosphorylated forms of the enzyme. Kinetic studies (k(cat(app))/K(m(app))) indicated that phosphorylation of the TYK-2-KD (pY(1054)) increased the catalytic efficiency 4.4-fold compared to its non-phosphorylated form, while further phosphorylation to generate the di-phosphorylated enzyme imparted no further increase in activity. These results are in contrast to those obtained with the JAK-2-KD and JAK-3-KD, where little or no increase in activity occurred upon mono-phosphorylation, while di-phosphorylation resulted in a 5.1-fold increase in activity for the JAK-2-KD. Moreover, ATP-competitive inhibitors demonstrated 10-30-fold shifts in potency (K(i(app))) as a result of the TYK-2-KD phosphorylation state, while the shifts for JAK-3-KD were only 2-3-fold and showed little or no change for JAK-2-KD. Thus, the phosphorlyation state imparted differential effects on both activity and inhibition within the JAK family of kinases.


Assuntos
Janus Quinase 2/biossíntese , Janus Quinase 2/isolamento & purificação , Janus Quinase 3/biossíntese , Janus Quinase 3/isolamento & purificação , TYK2 Quinase/biossíntese , TYK2 Quinase/isolamento & purificação , Animais , Catálise , Humanos , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 3/antagonistas & inibidores , Camundongos , Fosforilação , Estrutura Terciária de Proteína , TYK2 Quinase/antagonistas & inibidores
6.
J Mol Biol ; 400(3): 413-33, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20478313

RESUMO

Janus kinases (JAKs) are critical regulators of cytokine pathways and attractive targets of therapeutic value in both inflammatory and myeloproliferative diseases. Although the crystal structures of active JAK1 and JAK2 kinase domains have been reported recently with the clinical compound CP-690550, the structures of both TYK2 and JAK3 with CP-690550 have remained outstanding. Here, we report the crystal structures of TYK2, a first in class structure, and JAK3 in complex with PAN-JAK inhibitors CP-690550 ((3R,4R)-3-[4-methyl-3-[N-methyl-N-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]piperidin-1-yl]-3-oxopropionitrile) and CMP-6 (tetracyclic pyridone 2-t-butyl-9-fluoro-3,6-dihydro-7H-benz[h]-imidaz[4,5-f]isoquinoline-7-one), both of which bind in the ATP-binding cavities of both JAK isozymes in orientations similar to that observed in crystal structures of JAK1 and JAK2. Additionally, a complete thermodynamic characterization of JAK/CP-690550 complex formation was completed by isothermal titration calorimetry, indicating the critical role of the nitrile group from the CP-690550 compound. Finally, computational analysis using WaterMap further highlights the critical positioning of the CP-690550 nitrile group in the displacement of an unfavorable water molecule beneath the glycine-rich loop. Taken together, the data emphasize the outstanding properties of the kinome-selective JAK inhibitor CP-690550, as well as the challenges in obtaining JAK isozyme-selective inhibitors due to the overall structural and sequence similarities between the TYK2, JAK1, JAK2 and JAK3 isozymes. Nevertheless, subtle amino acid variations of residues lining the ligand-binding cavity of the JAK enzymes, as well as the global positioning of the glycine-rich loop, might provide the initial clues to obtaining JAK-isozyme selective inhibitors.


Assuntos
Benzimidazóis/metabolismo , Inibidores Enzimáticos/metabolismo , Janus Quinase 3/química , Piridonas/metabolismo , Pirimidinas/metabolismo , Pirróis/metabolismo , TYK2 Quinase/química , Sítios de Ligação , Calorimetria , Humanos , Janus Quinase 3/metabolismo , Cinética , Modelos Moleculares , Piperidinas , Ligação Proteica , Estrutura Terciária de Proteína , TYK2 Quinase/metabolismo
7.
Protein Expr Purif ; 69(1): 54-63, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19781647

RESUMO

Janus-associated kinases (JAKs) play critical roles in cytokine signaling, and have emerged as viable therapeutic targets in inflammation and oncology related diseases. To date, targeting JAK proteins with highly selective inhibitor compounds have remained elusive. We have expressed the active kinase domains for both JAK2 and JAK3 and devised purification protocols to resolve the non-, mono- (Y1007) and diphosphorylated (Y1007 and Y1008) states of JAK2 and non- and monophosphorylated states of JAK3 (Y980). An optimal purified protein yield of 20, 29 and 69mg per 20L cell culture was obtained for the three JAK2 forms, respectively, and 12.2 and 2.3mg per 10L fermentation for the two JAK3 forms allowing detailed biochemical and biophysical studies. To monitor the purification process we developed a novel HPLC activity assay where a sequential order of phosphorylation was observed whereby the first tyrosine residue was completely phosphorylated prior to phosphorylation of the tandem tyrosine residue. A Caliper-based microfluidics assay was used to determine the kinetic parameters (K(m) and k(cat)) for each phosphorylated state, showing that monophosphorylated (Y1007) JAK2 enzyme activity increased 9-fold over that of the nonphosphorylated species, and increased an additional 6-fold for the diphosphorylated (Y1007/Y1008) species, while phosphorylation of JAK3 resulted in a negligible increase in activity. Moreover, crystal structures have been generated for each isolated state of JAK2 and JAK3 with resolutions better than 2.4A. The generation of these reagents has enabled kinetic and structural characterization to inform the design of potent and selective inhibitors of the JAK family.


Assuntos
Janus Quinase 2/química , Janus Quinase 2/isolamento & purificação , Janus Quinase 3/química , Janus Quinase 3/isolamento & purificação , Sequência de Aminoácidos , Biocatálise , Cromatografia Líquida de Alta Pressão , Cristalização , Eletroforese em Gel de Poliacrilamida , Fermentação , Humanos , Cinética , Dados de Sequência Molecular , Fosforilação , Estrutura Terciária de Proteína
8.
Arch Biochem Biophys ; 491(1-2): 106-11, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19766586

RESUMO

A disintegrin and metalloprotease-8 (ADAM8) is thought to play a role in cancer and inflammatory diseases such as allergy, arthritis, and asthma. Despite the implication of ADAM8 in these diseases, the functional role of ADAM8 catalytic activity remains unclear. In this report, we demonstrate that an early critical autolytic event, we have termed pre-processing, is accelerated at acidic pH (pH 5.5) while autolytic activation is abrogated under the same conditions. Likewise, we found that pre-processing is hindered and autolytic activation is facilitated in neutral pH conditions, and thus demonstrates a pH-dependent shift in substrate selectivity. This finding is further supported by two peptide substrates corresponding to the pre-processing and C-terminal scissile bonds that were preferentially cleaved at acidic and neutral pH, respectively. Lastly, we found fibronectin cleavage to be attenuated at pH 5.5, while two novel substrates, brevican, and vitronectin, were readily cleaved in neutral or acidic conditions.


Assuntos
Proteínas ADAM/metabolismo , Proteínas de Membrana/metabolismo , Proteoglicanas/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Proteínas ADAM/isolamento & purificação , Biocatálise , Ativação Enzimática , Proteínas da Matriz Extracelular/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Cinética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Peptídeos/metabolismo , Estrutura Terciária de Proteína , Especificidade por Substrato
9.
J Biol Chem ; 284(36): 24185-91, 2009 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-19586907

RESUMO

Several inhibitors of a series of cis-1(S)2(R)-amino-2-indanol-based compounds were reported to be selective for the aggrecanases, ADAMTS-4 and -5 over other metalloproteases. To understand the nature of this selectivity for aggrecanases, the inhibitors, along with the broad spectrum metalloprotease inhibitor marimastat, were independently bound to the catalytic domain of ADAMTS-5, and the corresponding crystal structures were determined. By comparing the structures, it was determined that the specificity of the relative inhibitors for ADAMTS-5 was not driven by a specific interaction, such as zinc chelation, hydrogen bonding, or charge interactions, but rather by subtle and indirect factors, such as water bridging, ring rigidity, pocket size, and shape, as well as protein conformation flexibility.


Assuntos
Endopeptidases/química , Inibidores Enzimáticos/química , Proteínas ADAM/química , Proteína ADAMTS4 , Proteína ADAMTS5 , Animais , Bovinos , Humanos , Ligação de Hidrogênio , Pró-Colágeno N-Endopeptidase/química , Estrutura Terciária de Proteína , Homologia Estrutural de Proteína , Zinco/química
10.
Protein Expr Purif ; 65(2): 133-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19189860

RESUMO

Soluble guanylate cyclase (sGC) has been purified from 100 L cell culture infected by baculovirus using the newer and highly effective titerless infected-cells preservation and scale-up (TIPS) method. Successive passage of the enzyme through DEAE, Ni(2+)-NTA, and POROS Q columns obtained approximately 100mg of protein. The sGC obtained by this procedure was already about 90% pure and suitable for various studies which include high throughput screening (HTS) and hit follow-up. However, in order to obtain enzyme of greater homogeneity and purity for crystallographic and high precision spectroscopic and kinetic studies of sGC with select stimulators, the sGC solution after the POROS Q step was further purified by GTP-agarose affinity chromatography. This additional step led to the generation of 26 mg of enzyme that was about 99% pure. This highly pure and active enzyme exhibited a M(r)=144,933 by static light scattering supportive of a dimeric structure. It migrated as a two-band protein, each of equal intensity, on SDS-PAGE corresponding to the alpha (M(r) approximately 77,000) and beta (M(r) approximately 70,000) sGC subunits. It showed an A(430)/A(280)=1.01, indicating one heme per heterodimer, and a maximum of the Soret band at 430 nm indicative of a penta-coordinated ferrous heme with a histidine as the axial ligand. The Soret band shifted to 398 nm in the presence of an NO donor as expected for the formation of a penta-coordinated nitrosyl-heme complex. Non-stimulated sGC had k(cat)/K(m)=1.7 x 10(-3)s(-1)microM(-1) that increased to 5.8 x 10(-1)s(-1)microM(-1) upon stimulation with an NO donor which represents a 340-fold increase due to stimulation. The novel combination of using the TIPS method for co-expression of a heterodimeric heme-containing enzyme, along with the application of a reproducible ligand affinity purification method, has enabled us to obtain recombinant human sGC of both the quality and quantity needed to study structure-function relationships.


Assuntos
Baculoviridae/genética , Guanilato Ciclase/isolamento & purificação , Guanilato Ciclase/metabolismo , Insetos/citologia , Insetos/virologia , Receptores Citoplasmáticos e Nucleares/isolamento & purificação , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Animais , Baculoviridae/fisiologia , Técnicas de Cultura de Células , Cromatografia de Afinidade , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Guanilato Ciclase/química , Humanos , Cinética , Fosfoenolpiruvato Carboxiquinase (GTP)/química , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Proteínas Recombinantes/química , Sefarose/química , Guanilil Ciclase Solúvel
11.
Protein Expr Purif ; 65(2): 122-32, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19174191

RESUMO

Compounds capable of stimulating soluble guanylate cyclase (sGC) activity might become important new tools to treat hypertension. While rational design of these drugs would be aided by elucidation of the sGC three-dimensional structure and molecular mechanism of activation, such efforts also require quantities of high quality enzyme that are challenging to produce. We implemented the titerless infected-cells preservation and scale-up (TIPS) methodology to express the heterodimeric sGC. In the TIPS method, small-scale insect cell cultures were first incubated with a recombinant baculovirus which replicated in the cells. The baculovirus-infected insect cells (BIIC) were harvested and frozen prior to cell lysis and the subsequent escape of the newly replicated virus into the culture supernatant. Thawed BIIC stocks were ultimately used for subsequent scale up. As little as 1 mL of BIIC was needed to infect a 100-L insect cell culture, in contrast to the usual 1L of high-titer, virus stock supernatants. The TIPS method eliminates the need and protracted time for titering virus supernatants, and provides stable, concentrated storage of recombinant baculovirus in the form of infected cells. The latter is particularly advantageous for virus stocks which are unstable, such as those for sGC, and provides a highly efficient alternative for baculovirus storage and expression. The TIPS process enabled efficient scale up to 100-L batches, each producing about 200mg of active sGC. Careful adjustment of expression culture conditions over the course of several 100-L runs provided uniform starting titers, specific activity, and composition of contaminating proteins that facilitated development of a process that reproducibly yielded highly active, purified sGC.


Assuntos
Baculoviridae/genética , Guanilato Ciclase/biossíntese , Receptores Citoplasmáticos e Nucleares/biossíntese , Spodoptera/citologia , Spodoptera/metabolismo , Animais , Baculoviridae/fisiologia , Western Blotting , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Guanilato Ciclase/química , Guanilato Ciclase/metabolismo , Humanos , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Guanilil Ciclase Solúvel , Spodoptera/virologia , Fatores de Tempo
12.
Biosci Rep ; 29(4): 217-28, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18811590

RESUMO

Members of the ADAM (a disintegrin and metalloproteinase) family of proteins possess a multidomain architecture which permits functionalities as adhesion molecules, signalling intermediates and proteolytic enzymes. ADAM8 is found on immune cells and is induced by multiple pro-inflammatory stimuli suggesting a role in inflammation. Here we describe an activation mechanism for recombinant human ADAM8 that is independent from classical PC (pro-protein convertase)-mediated activation. N-terminal sequencing revealed that, unlike other ADAMs, ADAM8 undergoes pre-processing at Glu(158), which fractures the Pro (pro-segment)-domain before terminal activation takes place to remove the putative cysteine switch (Cys(167)). ADAM8 lacking the DIS (disintegrin) and/or CR (cysteine-rich) and EGF (epidermal growth factor) domains displayed impaired ability to complete this event. Thus pre-processing of the Pro-domain is co-ordinated by DIS and CR/EGF domains. Furthermore, by placing an EK (enterokinase) recognition motif between the Pro- and catalytic domains of multiple constructs, we were able to artificially remove the pro-segment prior to pre-processing. In the absence of pre-processing of the Pro-domain a marked decrease in specific activity was observed with the autoactivated enzyme, suggesting that the Pro-domain continued to associate and inhibit active enzyme. Thus, pre-processing of the Pro-domain of human ADAM8 is important for enzyme maturation by preventing re-association of the pro-segment with the catalytic domain. Given the observed necessity of DIS and CR/EGF for pre-processing, we conclude that these domains are crucial for the proper activation and maturation of human ADAM8.


Assuntos
Proteínas ADAM/metabolismo , Proteínas de Membrana/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Proteínas ADAM/isolamento & purificação , Catálise , Domínio Catalítico , Ativação Enzimática , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Análise de Sequência de Proteína
13.
Protein Expr Purif ; 60(1): 58-63, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18430586

RESUMO

Soluble guanylate cyclase (sGC), the main target of nitric oxide (NO), is a cytosolic, heme-containing, heterodimeric enzyme that catalyzes the conversion of guanosine 5'-triphosphate (GTP) to 3,5'-cyclic guanosine monophosphate (cGMP) and pyrophosphate (PPi) in the presence of Mg2+. Cyclic GMP is then involved in transmitting the NO activating signals to a variety of downstream effectors such as cyclic-nucleotide-gated channels, protein kinases, and phosphodiesterases. In this work, sGC has been purified from bovine lung. The lungs were subjected to grinding and extraction with buffer at physiological pH followed by centrifugation. The resulting solution was subjected to successive column chromatography on DEAE- and Q-Sepharose, Ceramic Hydroxyapatite, Resource Q, and GTP-agarose. The purified enzyme migrated as a two-band protein on SDS-PAGE corresponding to sGC subunits alpha (M(r)=77,532) and beta (M(r)=70,500) and had an A(280 nm)/A(430 nm) of approximately 1 indicating one heme per heterodimer. The yield of enzyme was 8-10mg from 4 to 5 kg bovine lungs. V(max) and K(m) of non-stimulated sGC were 22 nmol/mg/min and 180 microM, respectively. Upon stimulation with the NO donor 3-ethyl-3-(ethylaminoethyl)-1-hydroxy-2-oxo-1-triazene, the V(max) increased to 1330 nmol/mg/min while the K(m) dropped to 43 microM. The quality and quantity of enzyme make it suitable for studies to probe the structure and catalytic mechanism of this enzyme and for research related to drug discovery.


Assuntos
GMP Cíclico/metabolismo , Guanilato Ciclase/isolamento & purificação , Pulmão/enzimologia , Receptores Citoplasmáticos e Nucleares/isolamento & purificação , Animais , Bovinos , Cromatografia de Afinidade/métodos , Guanilato Ciclase/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Guanilil Ciclase Solúvel
15.
J Chromatogr A ; 1148(1): 46-54, 2007 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-17391681

RESUMO

A rapid HPLC assay was developed for monitoring the activity of the two proprotein convertases, PACE-4 and furin. Six novel peptide substrates were synthesized containing the minimal PC recognition sequence (Arg-X-X-Arg), as well as tryptophan residue(s) for easy detection. Four of the peptides were cleaved by both PCs and their kinetic parameters determined. Two peptides were not cleaved but were shown to be good negative controls although not inhibitors of either PC. In addition, inhibition curves were plotted and IC(50) values calculated for PACE-4 and furin in the presence of two polyarginine peptides, hexa and deca-D-arginine.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Pró-Proteína Convertases/metabolismo , Cinética , Pró-Proteína Convertases/análise , Pró-Proteína Convertases/antagonistas & inibidores , Reprodutibilidade dos Testes , Especificidade por Substrato , Fatores de Tempo
19.
Protein Expr Purif ; 46(2): 299-308, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16256367

RESUMO

A truncated form of human procaspase-9 missing the first 111 amino acids, and a variety of mutants derived therefrom, have been expressed in Escherichia coli inclusion bodies. Upon refolding to active enzymes, Delta(1-111) procaspase-9 and mutants were recovered at purity greater than 95% and with a final yield of 20-35 mg/L cell culture. Our active procaspase-9 retains its pro-segment, while undergoing major auto processing at Asp315 and a minor (20%) cleavage at Glu306. This unusual cleavage at a Glu-X bond also took place in the D315E mutant, and we describe herein the inhibitor Z-VAE-fmk that shows enhanced inactivation of procaspase-9 over caspases-3. The bond at Asp330, not processed by procaspase-9, is cleaved by caspase-3 and the resulting procaspase-9 variant, missing the 316-330 bridge, is six times as active as the non-mutated Delta(1-111) proenzyme. A deletion mutant lacking residues 316-330 underwent auto activation by cleavage at Asp315-Ala331 bond. Moreover, substitution of Glu306 by an Asp residue in this mutant led to rapid removal of the peptide spanning Ser307 to Asp330, and resulted in an enzyme that was 7.6 times as active as the non-mutated Delta(1-111) procaspase-9. Finally, replacing both Asp315 and Glu306 with Ala generated a procaspase-9 mutant incapable of auto processing. This single chain procaspase-9 was fully as active as the non-mutated Delta(1-111) enzyme processed at Asp315 or Glu306. Our demonstration that unprocessed procaspase-9 mutants are active as proteases with caspase-type specificity suggests that the role of procaspase-9 in cascade activation of executioner caspases might, in some circumstances, be carried out alone and without association of the apoptosome.


Assuntos
Caspases/biossíntese , Caspases/química , Expressão Gênica , Dobramento de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Erros Inatos do Metabolismo dos Aminoácidos , Caspase 9 , Caspases/genética , Ativação Enzimática/genética , Escherichia coli , Humanos , Corpos de Inclusão/química , Corpos de Inclusão/genética , Proteínas Recombinantes/genética , Deleção de Sequência
20.
J Neurochem ; 84(5): 1006-17, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12603825

RESUMO

The involvement of beta-secretase (BACE1; beta-site APP-cleaving enzyme) in producing the beta-amyloid component of plaques found in the brains of Alzheimer's patients, has fueled a major research effort to characterize this protease. Here, we describe work toward understanding the substrate specificity of BACE1 that began by considering the natural APP substrate and its Swedish mutant, APPSw, and proceeded on to include oxidized insulin B chain and ubiquitin substrates. From these findings, and the study of additional synthetic peptides, we determined that a decapeptide derived from APP in which the P3-P2' sequence, ...VKM--DA..., was replaced by ...ISY--EV... (-- = beta site of cleavage), yielded a substrate that was cleaved by BACE1 seven times faster than the corresponding APPSw peptide, SEVNL--DAEFR. The expanded peptide, GLTNIKTEEISEISY--EVEFRWKK, was cleaved an additional seven times faster than its decapeptide counterpart (boldface), and provides a substrate allowing assay of BACE1 at picomolar concentrations. Several APP mutants reflecting these beta-site amino acid changes were prepared as the basis for cellular assays. The APPISYEV mutant proved to be a cellular substrate that was superior to APPSw. The assay based on APPISYEV is highly specific for measuring BACE1 activity in cells; its homolog, BACE2, barely cleaved APPISYEV at the beta-site. Insertion of the optimized ISY--EV motif at either the beta-site (Asp1) or beta'-site (Glu11) directs the rate of cellular processing of APP at these two accessible sites. Thus, we have identified optimal BACE1 substrates that will be useful to elucidate the cellular enzymatic actions of BACE1, and for design of inhibitors that might be of therapeutic benefit in Alzheimer's disease.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases/química , Ácido Aspártico Endopeptidases/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/química , Animais , Ácido Aspártico Endopeptidases/genética , Sítios de Ligação/fisiologia , Células CHO , Cricetinae , Endopeptidases , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Processamento de Proteína Pós-Traducional , Relação Estrutura-Atividade , Especificidade por Substrato/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...