Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 24(22): 6417-27, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26358771

RESUMO

Arginase deficiency is caused by deficiency of arginase 1 (ARG1), a urea cycle enzyme that converts arginine to ornithine. Clinical features of arginase deficiency include elevated plasma arginine levels, spastic diplegia, intellectual disability, seizures and growth deficiency. Unlike other urea cycle disorders, recurrent hyperammonemia is typically less severe in this disorder. Normalization of plasma arginine levels is the consensus treatment goal, because elevations of arginine and its metabolites are suspected to contribute to the neurologic features. Using data from patients enrolled in a natural history study conducted by the Urea Cycle Disorders Consortium, we found that 97% of plasma arginine levels in subjects with arginase deficiency were above the normal range despite conventional treatment. Recently, arginine-degrading enzymes have been used to deplete arginine as a therapeutic strategy in cancer. We tested whether one of these enzymes, a pegylated human recombinant arginase 1 (AEB1102), reduces plasma arginine in murine models of arginase deficiency. In neonatal and adult mice with arginase deficiency, AEB1102 reduced the plasma arginine after single and repeated doses. However, survival did not improve likely, because this pegylated enzyme does not enter hepatocytes and does not improve hyperammonemia that accounts for lethality. Although murine models required dosing every 48 h, studies in cynomolgus monkeys indicate that less frequent dosing may be possible in patients. Given that elevated plasma arginine rather than hyperammonemia is the major treatment challenge, we propose that AEB1102 may have therapeutic potential as an arginine-reducing agent in patients with arginase deficiency.


Assuntos
Arginase/uso terapêutico , Arginina/sangue , Hiperargininemia/tratamento farmacológico , Animais , Arginase/sangue , Arginase/genética , Arginina/metabolismo , Encéfalo/metabolismo , Criança , Pré-Escolar , Estudos de Coortes , Modelos Animais de Doenças , Feminino , Humanos , Hiperamonemia/sangue , Hiperamonemia/metabolismo , Hiperargininemia/sangue , Hiperargininemia/genética , Hiperargininemia/metabolismo , Estudos Longitudinais , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/uso terapêutico , Convulsões/sangue , Convulsões/metabolismo
2.
J Biol Chem ; 290(20): 12793-803, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25825493

RESUMO

Cyclooxygenase enzymes (COX-1 and COX-2) catalyze the conversion of arachidonic acid to prostaglandin G2. The inhibitory activity of rapid, reversible COX inhibitors (ibuprofen, naproxen, mefenamic acid, and lumiracoxib) demonstrated a significant increase in potency and time dependence of inhibition against double tryptophan murine COX-2 mutants at the 89/90 and 89/119 positions. In contrast, the slow, time-dependent COX inhibitors (diclofenac, indomethacin, and flurbiprofen) were unaffected by those mutations. Further mutagenesis studies suggested that mutation at position 89 was principally responsible for the changes in inhibitory potency of rapid, reversible inhibitors, whereas mutation at position 90 may exert some effect on the potency of COX-2-selective diarylheterocycle inhibitors; no effect was observed with mutation at position 119. Several crystal structures with or without NSAIDs indicated that placement of a bulky residue at position 89 caused a closure of a gap at the lobby, and alteration of histidine to tryptophan at position 90 changed the electrostatic profile of the side pocket of COX-2. Thus, these two residues, especially Val-89 at the lobby region, are crucial for the entrance and exit of some NSAIDs from the COX active site.


Assuntos
Inibidores de Ciclo-Oxigenase 2/química , Ciclo-Oxigenase 2/química , Mutação de Sentido Incorreto , Animais , Domínio Catalítico , Cristalografia por Raios X , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Camundongos , Ligação Proteica , Eletricidade Estática
3.
Int J Infect Dis ; 102: 566-570, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33160064

RESUMO

The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a source of significant morbidity and death worldwide, and effective treatments are urgently needed. Clinical trials have focused largely on direct antiviral therapies or on immunomodulation in patients with severe manifestations of COVID-19. One therapeutic approach that remains to be clinically investigated is disruption of the host-virus relationship through amino acid restriction, a strategy used successfully in the setting of cancer treatment. Arginine is an amino acid that has been shown in nonclinical studies to be essential in the life cycle of many viruses. Therefore, arginine depletion may be an effective therapeutic approach against SARS-CoV-2. Several arginine-metabolizing enzymes in clinical development may be a viable approach to induce a low arginine environment to treat COVID-19 and other viral diseases. Herein, we explore the rationale for arginine depletion as a therapeutic approach for COVID-19.


Assuntos
Arginina/deficiência , COVID-19/metabolismo , COVID-19/terapia , SARS-CoV-2/metabolismo , Animais , COVID-19/virologia , Humanos , SARS-CoV-2/genética
4.
Cancer Immunol Res ; 9(4): 415-429, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33500272

RESUMO

Metabolic dysregulation is a hallmark of cancer. Many tumors exhibit auxotrophy for various amino acids, such as arginine, because they are unable to meet the demand for these amino acids through endogenous production. This vulnerability can be exploited by employing therapeutic strategies that deplete systemic arginine in order to limit the growth and survival of arginine auxotrophic tumors. Pegzilarginase, a human arginase-1 enzyme engineered to have superior stability and enzymatic activity relative to the native human arginase-1 enzyme, depletes systemic arginine by converting it to ornithine and urea. Therapeutic administration of pegzilarginase in the setting of arginine auxotrophic tumors exerts direct antitumor activity by starving the tumor of exogenous arginine. We hypothesized that in addition to this direct effect, pegzilarginase treatment indirectly augments antitumor immunity through increased antigen presentation, thus making pegzilarginase a prime candidate for combination therapy with immuno-oncology (I-O) agents. Tumor-bearing mice (CT26, MC38, and MCA-205) receiving pegzilarginase in combination with anti-PD-L1 or agonist anti-OX40 experienced significantly increased survival relative to animals receiving I-O monotherapy. Combination pegzilarginase/immunotherapy induced robust antitumor immunity characterized by increased intratumoral effector CD8+ T cells and M1 polarization of tumor-associated macrophages. Our data suggest potential mechanisms of synergy between pegzilarginase and I-O agents that include increased intratumoral MHC expression on both antigen-presenting cells and tumor cells, and increased presence of M1-like antitumor macrophages. These data support the clinical evaluation of I-O agents in conjunction with pegzilarginase for the treatment of patients with cancer.


Assuntos
Antineoplásicos/farmacologia , Arginase/farmacologia , Linfócitos T CD8-Positivos/imunologia , Inibidores de Checkpoint Imunológico/farmacologia , Receptores OX40/antagonistas & inibidores , Transferência Adotiva , Animais , Arginase/análise , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/metabolismo , Receptores OX40/metabolismo
5.
Transl Res ; 217: 11-22, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31954097

RESUMO

Metabolic remodeling contributes to the development and progression of some cancers and exposes them to vulnerabilities, including specific nutrient dependencies that can be targeted therapeutically. Arginine is a semiessential amino acid, and several cancers are unable to endogenously synthesize sufficient levels of arginine for survival and proliferation, most commonly due to reduced expression of argininosuccinate synthase (ASS1). Such cancers are dependent on arginine and they can be targeted via enzyme-mediated depletion of systemic arginine. We report the preclinical safety, antitumor efficacy, and immune-potentiating effects of pegzilarginase, a highly potent human arginine-degrading enzyme. Toxicology studies showed that pegzilarginase-mediated arginine depletion is well tolerated at therapeutic levels that elicit an antitumor growth effect. To determine which tumor types are best suited for clinical development, we profiled clinical tumor samples for ASS1 expression, which correlated with pegzilarginase sensitivity in vivo in patient-derived xenograft (PDx) models. Among the histologies tested, malignant melanoma, small cell lung cancer and Merkel cell carcinoma had the highest prevalence of low ASS1 expression, the highest proportion of PDx models responding to pegzilarginase, and the strongest correlation between low or no ASS1 expression and sensitivity to pegzilarginase. In an immune-competent syngeneic mouse model, pegzilarginase slowed tumor growth and promoted the recruitment of CD8+ tumor infiltrating lymphocytes. This is consistent with the known autophagy-inducing effects of arginine depletion, and the link between autophagy and major histocompatibility complex antigen presentation to T cells. Our work supports the ongoing clinical investigations of pegzilarginase in solid tumors and clinical combination of pegzilarginase with immune checkpoint inhibitors.


Assuntos
Antineoplásicos/farmacologia , Arginase/farmacologia , Animais , Arginase/análise , Arginase/toxicidade , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos ICR , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/enzimologia , Proteínas Recombinantes/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cell Rep ; 18(11): 2547-2556, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28297659

RESUMO

Cancer cells reprogram their metabolism, altering both uptake and utilization of extracellular nutrients. We individually depleted amino acid nutrients from isogenic cells expressing commonly activated oncogenes to identify correspondences between nutrient supply and viability. In HME (human mammary epithelial) cells, deprivation of cystine led to increased cell death in cells expressing an activated epidermal growth factor receptor (EGFR) mutant. Cell death occurred via synchronous ferroptosis, with generation of reactive oxygen species (ROS). Hydrogen peroxide promoted cell death, as both catalase and inhibition of NADPH oxidase 4 (NOX4) blocked ferroptosis. Blockade of EGFR or mitogen-activated protein kinase (MAPK) signaling similarly protected cells from ferroptosis, whereas treatment of xenografts derived from EGFR mutant non-small-cell lung cancer (NSCLC) with a cystine-depleting enzyme inhibited tumor growth in mice. Collectively, our results identify a potentially exploitable sensitization of some EGFR/MAPK-driven tumors to ferroptosis following cystine depletion.


Assuntos
Aminoácidos/metabolismo , Cistina/farmacologia , Oncogenes , Animais , Mama/citologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisteína/metabolismo , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Receptores ErbB/genética , Feminino , Glutationa/farmacologia , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/metabolismo , Ferro/metabolismo , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos SCID , Mutação/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase
7.
Nat Cell Biol ; 10(6): 740-7, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18488018

RESUMO

The growth and metabolic actions of growth hormone (GH) are believed to be mediated through the GH receptor (GHR) by JAK2 activation. The GHR exists as a constitutive homodimer, with signal transduction by ligand-induced realignment of receptor subunits. Based on the crystal structures, we identify a conformational change in the F'G' loop of the lower cytokine module, which results from binding of hGH but not G120R hGH antagonist. Mutations disabling this conformational change cause impairment of ERK but not JAK2 and STAT5 activation by the GHR in FDC-P1 cells. This results from the use of two associated tyrosine kinases by the GHR, with JAK2 activating STAT5, and Lyn activating ERK1/2. We provide evidence that Lyn signals through phospholipase C gamma, leading to activation of Ras. Accordingly, mice with mutations in the JAK2 association motif respond to GH with activation of hepatic Src and ERK1/2, but not JAK2/STAT5. We suggest that F'G' loop movement alters the signalling choice between JAK2 and a Src family kinase by regulating TMD realignment. Our findings could explain debilitated ERK but not STAT5 signalling in some GH-resistant dwarfs and suggest pathway-specific cytokine agonists.


Assuntos
Receptores da Somatotropina/agonistas , Receptores da Somatotropina/química , Animais , Dimerização , Humanos , Janus Quinase 2/metabolismo , Camundongos , Modelos Biológicos , Conformação Molecular , Fosfolipase C gama/metabolismo , Conformação Proteica , Estrutura Terciária de Proteína , Coelhos , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Proteínas ras/metabolismo
8.
Bioorg Med Chem Lett ; 12(9): 1315-8, 2002 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-11965379

RESUMO

Cyclooxygenase inhibition studies with novel indomethacin alkanolamides demonstrate the potential for dramatic differences in inhibitor properties conferred by subtle structural modifications. The transformation of non-selective alpha-(S)-substituted indomethacin ethanolamides to potent, COX-2 selective inhibitors by simple stereocenter inversion highlights this property.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Isoenzimas/efeitos dos fármacos , Prostaglandina-Endoperóxido Sintases/efeitos dos fármacos , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/metabolismo , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Estereoisomerismo
10.
Arch Biochem Biophys ; 409(1): 127-33, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12464251

RESUMO

Selective inhibition of cyclooxygenase-2 (COX-2) leads to relief of pain and inflammation with reduced gastrointestinal side effects relative to nonsteroidal anti-inflammatory drugs. 2-Acetoxyphenylhept-2-ynyl sulfide (APHS) is a selective COX-2 inhibitor that covalently modifies the protein by acetylating Ser-530. We utilized site-directed mutants in the COX-2 active site to probe the molecular determinants of APHS acetylation of COX-2. Incorporation of acetyl groups into Ser-530 was monitored by HPLC and mass spectrometry. Mutations that introduce steric bulk into a channel at the top of the active site (e.g., G533A, G533V) lead to a significant reduction in APHS acetylation. Reduction in acetylation is also observed by mutation of the active-site tyrosine (Tyr-385) to phenylalanine. Mutations in the side-pocket region, into which diarylheterocycle inhibitors insert, do not affect the ability of APHS to acetylate COX-2. Surprisingly, mutation of Arg-120, which is located on the floor of the active site, strongly reduces acetylation. Based on these results, we propose that the heptynyl side chain of APHS inserts into the top channel and acetylates Ser-530 with the assistance of hydrogen bonding from Tyr-385. Arg-120 is proposed to fix the conformation of the active site to one that favors acetylation.


Assuntos
Inibidores Enzimáticos/farmacologia , Heptanos/farmacologia , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Sulfetos/farmacologia , Alcinos , Sequência de Aminoácidos , Animais , Arginina/química , Sítios de Ligação , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Brometo de Cianogênio/farmacologia , Ciclo-Oxigenase 2 , Hidrogênio/metabolismo , Ligação de Hidrogênio , Insetos , Ligantes , Espectrometria de Massas , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Conformação Proteica , Serina/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Tirosina/química , Tirosina/metabolismo
11.
Biochemistry ; 42(30): 9041-9, 2003 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12885237

RESUMO

The endocannabinoid arachidonylethanolamide (AEA, anandamide) is an endogenous ligand for the cannabinoid receptors and has been shown to be oxygenated by cyclooxygenase-2 (COX-2). We examined the structural requirements for COX-mediated, AEA oxygenation using a number of substrate analogues and site-directed mutants of COX-2. Fourteen AEA analogues were synthesized and tested as COX substrates. These studies identified the hydroxyl moiety of AEA as a critical determinant in the ability of COX enzymes to effect robust endocannabinoid oxygenation. In addition, these studies suggest that subtle structural modifications of AEA analogues near the ethanolamide moiety can result in pronounced changes in their ability to serve as COX-2 substrates. Site-directed mutagenesis studies have permitted the development of a model of AEA binding within the COX-2 active site. As with arachidonic acid, the omega-terminus of AEA binds in a hydrophobic alcove near the top of the COX-2 active site. The polar ethanolamide moiety of AEA, like the carboxylate of arachidonate, interacts with Arg-120 at the bottom of the COX-2 active site. Mutation of Tyr-385 prevents AEA oxygenation, suggesting that, as in the case of other COX substrates, AEA metabolism is initiated by Tyr-385-mediated hydrogen abstraction. Thus, AEA binds within the COX-2 active site in a conformation roughly similar to that of arachidonic acid. However, important differences have been identified that account for the isoform selectivity of AEA oxygenation. Importantly, the COX-2 side pocket and Arg-513 in particular are critical determinants of the ability of COX-2 to efficiently generate prostaglandin H(2) ethanolamide. The reduced efficiency of COX-1-mediated, AEA oxygenation can thus be explained by the absence of an arginine residue at position 513 in this isoform. Mutational analysis of Leu-531, an amino acid located directly across from the COX-2 side pocket, suggests that AEA is shifted away from this hydrophobic residue and toward Arg-513 relative to arachidonic acid. Coupled with earlier observations with the endocannabinoid 2-arachidonylglycerol, these results indicate that one possible function of the highly conserved COX-2 active site side pocket is to promote endocannabinoid oxygenation.


Assuntos
Aminoácidos/química , Ácidos Araquidônicos/metabolismo , Canabinoides/metabolismo , Ácidos Graxos Insaturados/metabolismo , Isoenzimas/química , Oxigênio/metabolismo , Prostaglandina-Endoperóxido Sintases/química , Acetilação , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Ácidos Araquidônicos/química , Sítios de Ligação/genética , Moduladores de Receptores de Canabinoides , Catálise , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Endocanabinoides , Ácidos Graxos Insaturados/química , Interações Hidrofóbicas e Hidrofílicas , Ácidos Hidroxieicosatetraenoicos/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Proteínas de Membrana , Metionina/genética , Camundongos , Mutagênese Sítio-Dirigida , Alcamidas Poli-Insaturadas , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandina-Endoperóxido Sintases/metabolismo , Serina/genética , Ovinos , Especificidade por Substrato/genética , Tirosina/genética , Tirosina/metabolismo
12.
J Biol Chem ; 277(1): 478-85, 2002 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-11677234

RESUMO

Prostaglandin synthesis by cyclooxygenases-1 and -2 (COX-1 and COX-2) involves an initial oxygenation of arachidonic acid at C-11, followed by endoperoxide and cyclopentane ring formation, and then a second reaction with molecular oxygen in the S configuration at C-15. The resulting 15S-hydroxyl group of prostaglandins is crucial for their bioactivity. Using human COX-1 and human and murine COX-2, we have identified two amino acids located in the oxygenase active site that control the stereochemistry at C-15. The most crucial determinant is Ser-530, the residue that is acetylated by aspirin. In COX-2, site-directed mutagenesis of Ser-530 to methionine, threonine, or valine produced highly active enzymes that formed 82-95% 15R-configuration prostaglandins; these have the opposite stereochemistry at C-15 to the natural products. In COX-1, the corresponding Ser-530 mutations inactivated the enzyme. The second residue, Val-349, exerts a more subtle influence. When Val-349 was replaced by isoleucine, the mutant COX-1 and COX-2 enzymes formed 41 and 65% 15R-prostaglandins, respectively. This change was highly specific for isoleucine, as mutations of Val-349 to alanine, leucine, asparagine, or threonine did not alter or only slightly altered (< or =13%) the S-configuration at C-15. These results establish a previously unrecognized role for Ser-530 and Val-349 in maintaining the correct S stereochemistry of the carbon-15 hydroxyl group during prostaglandin synthesis. The findings may also explain the absolute conservation of Ser-530, the target of aspirin, throughout the families of cyclooxygenase enzymes.


Assuntos
Isoenzimas/fisiologia , Prostaglandina-Endoperóxido Sintases/fisiologia , Prostaglandinas/química , Animais , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Humanos , Ácidos Hidroxieicosatetraenoicos/análise , Proteínas de Membrana , Camundongos , Conformação Molecular , Mutagênese Sítio-Dirigida , Serina
13.
Clin Endocrinol (Oxf) ; 56(4): 475-85, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11966740

RESUMO

OBJECTIVE: Because there is discordance between different immunoassay values for serum hGH, and because clinical state may not correlate with immunoreactive hGH, we have developed an assay to accurately measure serum hGH somatogenic bioactivity. The results of this assay were compared with the Elegance two-site ELISA assay across 135 patient samples in a variety of clinical states. DESIGN: The somatogenic assay was based on stable expression of hGH receptor in the murine BaF line, allowing these cells to proliferate in response to hGH. To eliminate interference by other growth factors in serum, we created a specific antagonist of the hGH receptor (similar to Trovert or Pegvisomant) which allowed us to obtain a true measure of hGH somatogenic activity by subtraction of the activity in the presence of the antagonist. The assay was carried out in microtiter plates over 24 h, with oxidation of a chromogenic tetrazolium salt (MTT) as the endpoint. PATIENTS: These encompassed a number of different clinical conditions related to short stature, including idiopathic short stature, neurosecretory dysfunction and renal failure, as well as obese patients on dietary restriction and normal volunteers. MEASUREMENTS: In addition to the colourimetric (MTT) response to hGH, we measured free hGH by stripping out GHBP-bound hGH using beads coupled to a monoclonal antibody to the GHBP (GH binding protein). All samples were measured in both bioassay and ELISA assay. RESULTS: This bioassay was sensitive (5 mU/l or 2 microg/l) and precise, and not subject to interference by the GHBP. There was a good correlation (r = 0.95) between bioactivity and immunoactivity across clinical states. There was, however, an increased bioactivity during secretory peaks (over 25 mU/l), which has been reported previously for the Nb2 bioassay. Free hGH did not correlate with clinical state. CONCLUSIONS: Because the results of the Elegance ELISA and the bioassay correlate well, even though there is greater bioactivity at higher hormone concentrations, it is evident that an appropriate immunoassay is able to act as a reliable indicator for clinical assessment. In those rare cases where bio-inactive GH exists, our bioassay should provide an appropriate means to demonstrate this.


Assuntos
Bioensaio/métodos , Transtornos do Crescimento/sangue , Hormônio do Crescimento Humano/sangue , Adulto , Animais , Técnicas de Cultura de Células , Linhagem Celular , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Hormônio do Crescimento Humano/antagonistas & inibidores , Humanos , Camundongos , Receptores da Somatotropina/antagonistas & inibidores , Receptores da Somatotropina/metabolismo , Reprodutibilidade dos Testes
14.
J Biol Chem ; 278(46): 45763-9, 2003 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-12925531

RESUMO

A variety of drugs inhibit the conversion of arachidonic acid to prostaglandin G2 by the cyclooxygenase (COX) activity of prostaglandin endoperoxide synthases. Several modes of inhibitor binding in the COX active site have been described including ion pairing of carboxylic acid containing inhibitors with Arg-120 of COX-1 and COX-2 and insertion of arylsulfonamides and sulfones into the COX-2 side pocket. Recent crystallographic evidence suggests that Tyr-385 and Ser-530 chelate polar or negatively charged groups in arachidonic acid and aspirin. We tested the generality of this binding mode by analyzing the action of a series of COX inhibitors against site-directed mutants of COX-2 bearing changes in Arg-120, Tyr-355, Tyr-348, and Ser-530. Interestingly, diclofenac inhibition was unaffected by the mutation of Arg-120 to alanine but was dramatically attenuated by the S530A mutation. Determination of the crystal structure of a complex of diclofenac with murine COX-2 demonstrates that diclofenac binds to COX-2 in an inverted conformation with its carboxylate group hydrogen-bonded to Tyr-385 and Ser-530. This finding represents the first experimental demonstration that the carboxylate group of an acidic non-steroidal anti-inflammatory drug can bind to a COX enzyme in an orientation that precludes the formation of a salt bridge with Arg-120. Mutagenesis experiments suggest Ser-530 is also important in time-dependent inhibition by nimesulide and piroxicam.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Inibidores Enzimáticos/farmacologia , Isoenzimas/antagonistas & inibidores , Serina/química , Tirosina/química , Animais , Ácido Araquidônico/química , Arginina/química , Ligação Competitiva , Linhagem Celular , Cristalografia por Raios X , Ciclo-Oxigenase 2 , Diclofenaco/antagonistas & inibidores , Diclofenaco/química , Relação Dose-Resposta a Droga , Insetos , Camundongos , Modelos Químicos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Piroxicam/química , Prostaglandina-Endoperóxido Sintases , Ligação Proteica , Sulfonamidas/química , Fatores de Tempo
15.
Protein Expr Purif ; 31(2): 305-10, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14550652

RESUMO

Two protein families that are critical for vesicle transport are the Syntaxin and Munc18/Sec1 families of proteins. These two molecules form a high affinity complex and play an essential role in vesicle docking and fusion. Munc18c was expressed as an N-terminally His-tagged fusion protein from recombinant baculovirus in Sf9 insect cells. His-tagged Munc18c was purified to homogeneity using both cobalt-chelating affinity chromatography and gel filtration chromatography. With this simple two-step protocol, 3.5 mg of purified Munc18c was obtained from a 1L culture. Further, the N-terminal His-tag could be removed by thrombin cleavage while the tagged protein was bound to metal affinity resin. Recombinant Munc18c produced in this way is functional, in that it forms a stable complex with the SNARE interacting partner, syntaxin4. Thus we have developed a method for producing and purifying large amounts of functional Munc18c--both tagged and detagged--from a baculovirus expression system. We have also developed a method to purify the Munc18c:syntaxin4 complex. These methods will be employed for future functional and structural studies.


Assuntos
Baculoviridae/genética , Proteínas de Membrana/metabolismo , Animais , Expressão Gênica , Histidina/genética , Histidina/metabolismo , Proteínas de Membrana/isolamento & purificação , Camundongos , Engenharia de Proteínas , Proteínas Qa-SNARE , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo
16.
Blood ; 102(9): 3206-9, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12855566

RESUMO

In a search for novel growth factors, we discovered that human interleukin-20 (IL-20) enhanced colony formation by CD34+ multipotential progenitors. IL-20 had no effect on erythroid, granulocyte-macrophage, or megakaryocyte progenitors. IL-20 transgenic mice increased the numbers and cell cycling of multipotential but not other progenitors. IL-20 administration to normal mice significantly increased only multipotential progenitor cells, demonstrating that IL-20 significantly influences hematopoiesis, with specificity toward multipotential progenitors. This is the first cytokine with such specificity identified.


Assuntos
Células-Tronco Hematopoéticas/citologia , Interleucinas/farmacologia , Células-Tronco Multipotentes/efeitos dos fármacos , Animais , Antígenos CD34 , Células da Medula Óssea/citologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Sangue Fetal/citologia , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Interleucinas/administração & dosagem , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA