Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
1.
Eur J Med Chem ; 275: 116604, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-38917665

RESUMO

The endogenous opioid system regulates pain through local release of neuropeptides and modulation of their action on opioid receptors. However, the effect of opioid peptides, the enkephalins, is short-lived due to their rapid hydrolysis by enkephalin-degrading enzymes. In turn, an innovative approach to the management of pain would be to increase the local concentration and prolong the stability of enkephalins by preventing their inactivation by neural enkephalinases such as puromycin-sensitive aminopeptidase (PSA). Our previous structure-activity relationship studies offered the S-diphenylmethyl cysteinyl derivative of puromycin (20) as a nanomolar inhibitor of PSA. This chemical class, however, suffered from undesirable metabolism to nephrotoxic puromycin aminonucleoside (PAN). To prevent such toxicity, we designed and synthesized 5'-chloro substituted derivatives. The compounds retained the PSA inhibitory potency of the corresponding 5'-hydroxy analogs and had improved selectivity toward PSA. In vivo treatment with the lead compound 19 caused significantly reduced pain response in antinociception assays, alone and in combination with Met-enkephalin. The analgesic effect was reversed by the opioid antagonist naloxone, suggesting the involvement of opioid receptors. Further, PSA inhibition by compound 19 in brain slices caused local increase in endogenous enkephalin levels, corroborating our rationale. Pharmacokinetic assessment of compound 19 showed desirable plasma stability and identified the cysteinyl sulfur as the principal site of metabolic liability. We gained additional insight into inhibitor-PSA interactions by molecular modeling, which underscored the importance of bulky aromatic amino acid in puromycin scaffold. The results of this study strongly support our rationale for the development of PSA inhibitors for effective pain management.


Assuntos
Transdução de Sinais , Animais , Relação Estrutura-Atividade , Transdução de Sinais/efeitos dos fármacos , Masculino , Camundongos , Estrutura Molecular , Relação Dose-Resposta a Droga , Humanos , Antígenos CD13/antagonistas & inibidores , Antígenos CD13/metabolismo , Encefalinas/química , Encefalinas/metabolismo , Encefalinas/farmacologia , Puromicina/farmacologia , Puromicina/metabolismo , Puromicina/química , Analgésicos/farmacologia , Analgésicos/química , Aminopeptidases/antagonistas & inibidores , Aminopeptidases/metabolismo , Ratos
2.
Sheng Wu Gong Cheng Xue Bao ; 39(10): 4098-4107, 2023 Oct 25.
Artigo em Chinês | MEDLINE | ID: mdl-37877393

RESUMO

Human induced pluripotent stem cells (hiPSCs) are promising in regenerative medicine. However, the pluripotent stem cells (PSCs) may form clumps of cancerous tissue, which is a major safety concern in PSCs therapies. Rapamycin is a safe and widely used immunosuppressive pharmaceutical that acts through heterodimerization of the FKBP12 and FRB fragment. Here, we aimed to insert a rapamycin inducible caspase 9 (riC9) gene in a safe harbor AAVS1 site to safeguard hiPSCs therapy by drug induced homodimerization. The donor vector containing an EF1α promoter, a FRB-FKBP-Caspase 9 (CARD domain) fusion protein and a puromycin resistant gene was constructed and co-transfected with sgRNA/Cas9 vector into hiPSCs. After one to two weeks screening with puromycin, single clones were collected for genotype and phenotype analysis. Finally, rapamycin was used to induce the homodimerization of caspase 9 to activate the apoptosis of the engineered cells. After transfection of hiPSCs followed by puromycin screening, five cell clones were collected. Genome amplification and sequencing showed that the donor DNA has been precisely knocked out at the endogenous AAVS1 site. The engineered hiPSCs showed normal pluripotency and proliferative capacity. Rapamycin induced caspase 9 activation, which led to the apoptosis of all engineered hiPSCs and its differentiated cells with different sensitivity to drugs. In conclusion, we generated a rapamycin-controllable hiPSCs survival by homodimerization of caspase 9 to turn on cell apoptosis. It provides a new strategy to guarantee the safety of the hiPSCs therapy.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Humanos , Sirolimo/farmacologia , Sirolimo/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , RNA Guia de Sistemas CRISPR-Cas , Células-Tronco Pluripotentes/metabolismo , Diferenciação Celular , Puromicina/metabolismo
3.
Appl Biochem Biotechnol ; 195(12): 7379-7396, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37000351

RESUMO

Minimal change disease (MCD) is the most common cause of idiopathic nephrotic syndrome in children. The current major therapy is hormones for most steroid-sensitive patients. However, many patients have recurrent relapses of the disease and require long-term immunosuppression, leading to significant morbidity due to the side effects of the drugs. Therefore, better drugs need to be urgently explored to treat nephrotic syndrome while avoiding the side effects of drugs. Minnelide, a water-soluble prodrug of triptolide, has been proved to be effective in treating cancers in many clinical trials. This study aimed to investigate the therapeutic effect of minnelide in mice with adriamycin (ADR) nephropathy, its underlying protection mechanisms, and its reproductive toxicity. Minnelide was administered intraperitoneally to 6-8-week female mice with adriamycin nephropathy for 2 weeks, and the urine, blood, and kidney tissues were taken to analyze the therapeutic effect. In addition, we evaluated reproductive toxicity by measuring the levels of gonadal hormones and observing the histological changes in ovaries and testes. Primary mouse podocytes were exposed to puromycin (PAN) to damage the cytoskeleton and induce apoptosis, and then, triptolide was used to evaluate the therapeutic effect and underlying protection mechanisms in vitro. It was observed that minnelide dramatically alleviated proteinuria and apoptosis in mice with adriamycin nephropathy. In vitro, triptolide ameliorated puromycin-induced cytoskeletal rearrangement and apoptosis via reactive oxygen species-mediated mitochondrial pathway. In addition, minnelide caused no reproductive toxicity to male and female mice. The results suggested that minnelide might be a promising drug for nephrotic syndrome.


Assuntos
Nefropatias , Síndrome Nefrótica , Podócitos , Humanos , Criança , Camundongos , Masculino , Feminino , Animais , Doxorrubicina/toxicidade , Síndrome Nefrótica/induzido quimicamente , Síndrome Nefrótica/tratamento farmacológico , Síndrome Nefrótica/metabolismo , Podócitos/metabolismo , Podócitos/patologia , Nefropatias/induzido quimicamente , Proteinúria/tratamento farmacológico , Proteinúria/metabolismo , Proteinúria/patologia , Puromicina/metabolismo , Puromicina/farmacologia , Puromicina/uso terapêutico
4.
Elife ; 92020 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-32844746

RESUMO

Puromycin is an amino-acyl transfer RNA analog widely employed in studies of protein synthesis. Since puromycin is covalently incorporated into nascent polypeptide chains, anti-puromycin immunofluorescence enables visualization of nascent protein synthesis. A common assumption in studies of local messenger RNA translation is that the anti-puromycin staining of puromycylated nascent polypeptides in fixed cells accurately reports on their original site of translation, particularly when ribosomes are stalled with elongation inhibitors prior to puromycin treatment. However, when we attempted to implement a proximity ligation assay to detect ribosome-puromycin complexes, we found no evidence to support this assumption. We further demonstrated, using biochemical assays and live cell imaging of nascent polypeptides in mammalian cells, that puromycylated nascent polypeptides rapidly dissociate from ribosomes even in the presence of elongation inhibitors. Our results suggest that attempts to define precise subcellular translation sites using anti-puromycin immunostaining may be confounded by release of puromycylated nascent polypeptide chains prior to fixation.


Assuntos
Elongação Traducional da Cadeia Peptídica/efeitos dos fármacos , Inibidores da Síntese de Proteínas , Puromicina , Ribossomos , Animais , Linhagem Celular Tumoral , Camundongos , Inibidores da Síntese de Proteínas/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Proteínas/química , Proteínas/metabolismo , Puromicina/metabolismo , Puromicina/farmacologia , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Aminoacil-RNA de Transferência/química , Aminoacil-RNA de Transferência/metabolismo , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo
5.
Elife ; 92020 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-32844748

RESUMO

Puromycin is a tyrosyl-tRNA mimic that blocks translation by labeling and releasing elongating polypeptide chains from translating ribosomes. Puromycin has been used in molecular biology research for decades as a translation inhibitor. The development of puromycin antibodies and derivatized puromycin analogs has enabled the quantification of active translation in bulk and single-cell assays. More recently, in vivo puromycylation assays have become popular tools for localizing translating ribosomes in cells. These assays often use elongation inhibitors to purportedly inhibit the release of puromycin-labeled nascent peptides from ribosomes. Using in vitro and in vivo experiments in various eukaryotic systems, we demonstrate that, even in the presence of elongation inhibitors, puromycylated peptides are released and diffuse away from ribosomes. Puromycylation assays reveal subcellular sites, such as nuclei, where puromycylated peptides accumulate post-release and which do not necessarily coincide with sites of active translation. Our findings urge caution when interpreting puromycylation assays in vivo.


Assuntos
Núcleo Celular , Biossíntese de Proteínas , Inibidores da Síntese de Proteínas , Puromicina , Animais , Caenorhabditis elegans , Núcleo Celular/química , Núcleo Celular/metabolismo , Emetina/metabolismo , Emetina/farmacologia , Peptídeos/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/fisiologia , Inibidores da Síntese de Proteínas/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Puromicina/metabolismo , Puromicina/farmacologia , RNA de Transferência/metabolismo , Coelhos , Ribossomos/metabolismo , Análise de Célula Única
6.
Nat Protoc ; 14(2): 441-460, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30610239

RESUMO

Although protein synthesis is a conserved and essential cellular function, it is often regulated in a cell-type-specific manner to influence cell fate, growth and homeostasis. Most methods used to measure protein synthesis depend on metabolically labeling large numbers of cells with radiolabeled amino acids or amino acid analogs. Because these methods typically depend on specialized growth conditions, they have been largely restricted to yeast, bacteria and cell lines. Application of these techniques to investigating protein synthesis within mammalian systems in vivo has been challenging. The synthesis of O-propargyl-puromycin (OP-Puro), an analog of puromycin that contains a terminal alkyne group, has facilitated the quantification of protein synthesis within individual cells in vivo. OP-Puro enters the acceptor site of ribosomes and incorporates into nascent polypeptide chains. Incorporated OP-Puro can be detected through a click-chemistry reaction that links it to a fluorescently tagged azide molecule. In this protocol, we describe how to administer OP-Puro to mice, obtain cells of interest (here, we use bone marrow cells) just 1 h later, and quantify the amount of protein synthesized per hour by flow cytometry on the basis of OP-Puro incorporation. We have used this approach to show that hematopoietic stem cells (HSCs) exhibit an unusually low rate of protein synthesis relative to other hematopoietic cells, and it can be easily adapted to quantify cell-type-specific rates of protein synthesis across diverse mammalian tissues in vivo. Measurement of protein synthesis within bone marrow cells in a cohort of six mice can be achieved in 8-10 h.


Assuntos
Química Click/métodos , Células-Tronco Hematopoéticas/metabolismo , Biossíntese de Proteínas , Puromicina/análogos & derivados , Análise de Célula Única/métodos , Coloração e Rotulagem/métodos , Animais , Azidas/química , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Citometria de Fluxo , Corantes Fluorescentes/química , Células-Tronco Hematopoéticas/citologia , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Puromicina/metabolismo , Rodaminas/química , Ribossomos/genética , Ribossomos/metabolismo , Ácidos Sulfônicos/química
7.
Am J Physiol Heart Circ Physiol ; 313(6): H1119-H1129, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28822967

RESUMO

Hypertension is one of the most important risk factors of heart failure. In response to high blood pressure, the left ventricle manifests hypertrophic growth to ameliorate wall stress, which may progress into decompensation and trigger pathological cardiac remodeling. Despite the clinical importance, the temporal dynamics of pathological cardiac growth remain elusive. Here, we took advantage of the puromycin labeling approach to measure the relative rates of protein synthesis as a way to delineate the temporal regulation of cardiac hypertrophic growth. We first identified the optimal treatment conditions for puromycin in neonatal rat ventricular myocyte culture. We went on to demonstrate that myocyte growth reached its peak rate after 8-10 h of growth stimulation. At the in vivo level, with the use of an acute surgical model of pressure-overload stress, we observed the maximal growth rate to occur at day 7 after surgery. Moreover, RNA sequencing analysis supports that the most profound transcriptomic changes occur during the early phase of hypertrophic growth. Our results therefore suggest that cardiac myocytes mount an immediate growth response in reply to pressure overload followed by a gradual return to basal levels of protein synthesis, highlighting the temporal dynamics of pathological cardiac hypertrophic growth.NEW & NOTEWORTHY We determined the optimal conditions of puromycin incorporation in cardiac myocyte culture. We took advantage of this approach to identify the growth dynamics of cardiac myocytes in vitro. We went further to discover the protein synthesis rate in vivo, which provides novel insights about cardiac temporal growth dynamics in response to pressure overload.


Assuntos
Aorta Torácica/fisiopatologia , Pressão Arterial , Cardiomegalia/patologia , Proliferação de Células , Miócitos Cardíacos/patologia , Animais , Animais Recém-Nascidos , Aorta Torácica/cirurgia , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Constrição , Modelos Animais de Doenças , Regulação da Expressão Gênica , Masculino , Camundongos Endogâmicos C57BL , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Biossíntese de Proteínas , Puromicina/metabolismo , Ratos Sprague-Dawley , Fatores de Tempo
8.
FEBS Lett ; 591(1): 186-195, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27926780

RESUMO

The ubiquitin-like modifier, FAT10, is involved in proteasomal degradation and antigen processing. As ubiquitin and the ubiquitin-like modifier, ISG15, cotranslationally modify proteins, we investigated whether FAT10 could also be conjugated to newly synthesized proteins. Indeed, we found that nascent proteins are modified with FAT10, but not with the same preference for newly synthesized proteins as observed for ISG15. Our data show that puromycin-labeled polypeptides are strongly modified by ISG15 and less intensely by ubiquitin and FAT10. Nevertheless, conjugates of all three modifiers copurify with ribosomes. Taken together, we show that unlike ISG15, ubiquitin and FAT10 are conjugated to a similar degree to newly translated and pre-existing proteins.


Assuntos
Citocinas/metabolismo , Biossíntese de Proteínas , Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Células HEK293 , Humanos , Puromicina/metabolismo , Ribossomos/metabolismo , Especificidade por Substrato
9.
Nat Commun ; 7: 11657, 2016 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-27216360

RESUMO

The ribosome stalls on translation of polyproline sequences due to inefficient peptide bond formation between consecutive prolines. The translation factor EF-P is able to alleviate this stalling by accelerating Pro-Pro formation. However, the mechanism by which EF-P recognizes the stalled complexes and accelerates peptide bond formation is not known. Here, we use genetic code reprogramming through a flexible in-vitro translation (FIT) system to investigate how mutations in tRNA(Pro) affect EF-P function. We show that the 9-nt D-loop closed by the stable D-stem sequence in tRNA(Pro) is a crucial recognition determinant for EF-P. Such D-arm structures are shared only among the tRNA(Pro) isoacceptors and tRNA(fMet) in Escherichia coli, and the D-arm of tRNA(fMet) is essential for EF-P-induced acceleration of fMet-puromycin formation. Thus, the activity of EF-P is controlled by recognition elements in the tRNA D-arm.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/fisiologia , Fatores de Alongamento de Peptídeos/metabolismo , Biossíntese de Proteínas , RNA de Transferência de Prolina/metabolismo , Sítios de Ligação/genética , Proteínas de Escherichia coli/genética , Mutação , Motivos de Nucleotídeos/genética , Fatores de Alongamento de Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica/genética , Puromicina/química , Puromicina/metabolismo , RNA de Transferência de Metionina/química , RNA de Transferência de Metionina/metabolismo , RNA de Transferência de Prolina/química , RNA de Transferência de Prolina/genética , Ribossomos/genética , Ribossomos/metabolismo
10.
Biochem Biophys Res Commun ; 474(2): 247-251, 2016 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-27125456

RESUMO

The amino-nucleoside antibiotic, puromycin, acts by covalently linking to elongating polypeptide chains on ribosomes to generate prematurely terminated immature polypeptides. The trafficking of puromycin-conjugated (puromycylated) immature polypeptides within cell has, however, remained elusive. In this study, using O-propargyl-puromycin (OP-Puro), the distribution of puromycylated polypeptides was assessed in HeLa cells by click chemistry. Under standard culture conditions, OP-Puro signals were detected in the cytoplasm and nucleus with the highest concentrations in the nucleolus. Intriguingly, when proteasome activities were aborted using MG132, OP-Puro signals began to accumulate at promyelocytic leukemia nuclear bodies (PML-NBs) in addition to the nucleolus. We also found promiscuous association of OP-Puro signals with SUMO-2/3 and ubiquitin at PML-NBs, but not at the nucleolus, during abortive proteasome activities. This study reveals a previously unknown distribution of OP-Puro that argues for a nuclear function in regulating immature protein homeostasis.


Assuntos
Núcleo Celular/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Puromicina/análogos & derivados , Proteína SUMO-1/metabolismo , Ubiquitina/metabolismo , Química Click/métodos , Proteína da Leucemia Promielocítica/metabolismo , Puromicina/análise , Puromicina/metabolismo , Proteína SUMO-1/química , Ubiquitina/química
11.
Glia ; 64(3): 440-56, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26539695

RESUMO

The spatial organization of vascular endothelial growth factor (VEGF) signaling is a key determinant of vascular patterning during development and tissue repair. How VEGF signaling becomes spatially restricted and the role of VEGF secreting astrocytes in this process remains poorly understood. Using a VEGF-GFP fusion protein and confocal time-lapse microscopy, we observed the intracellular routing, secretion and immobilization of VEGF in scratch-activated living astrocytes. We found VEGF to be directly transported to cell-extracellular matrix attachments where it is incorporated into fibronectin fibrils. VEGF accumulated at ß1 integrin containing fibrillar adhesions and was translocated along the cell surface prior to internalization and degradation. We also found that only the astrocyte-derived, matrix-bound, and not soluble VEGF decreases ß1 integrin turnover in fibrillar adhesions. We suggest that polarized VEGF release and ECM remodeling by VEGF secreting cells is key to control the local concentration and signaling of VEGF. Our findings highlight the importance of astrocytes in directing VEGF functions and identify these mechanisms as promising target for angiogenic approaches.


Assuntos
Astrócitos/metabolismo , Polaridade Celular/fisiologia , Matriz Extracelular/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Astrócitos/ultraestrutura , Polaridade Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hidrazonas/metabolismo , Antígeno Ki-67/metabolismo , Microscopia Confocal , Neurônios/metabolismo , Fotodegradação , Puromicina/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/genética , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo , Fatores de Tempo , Transfecção
12.
Biotechnol Bioeng ; 111(10): 2041-55, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24842774

RESUMO

A common problem with using embryonic stem (ES) cells as a source for analysis of gene expression, drug toxicity, or functional characterization studies is the heterogeneity that results from many differentiation protocols. The ability to generate large numbers of high purity differentiated cells from pluripotent stem cells could greatly enhance their utility for in vitro characterization studies and transplantation in pre-clinical injury models. Population heterogeneity is particularly troublesome for post-mitotic neurons, including motoneurons, because they do not proliferate and are quickly diluted in culture by proliferative phenotypes, such as glia. Studies of motoneuron biology and disease, in particular amyotrophic lateral sclerosis, can benefit from high purity motoneuron cultures. In this study, we engineered a transgenic-ES cell line where highly conserved enhancer elements for the motoneuron transcription factor Hb9 were used to drive puromycin N-acetyltransferase expression in ES cell-derived motoneurons. Antibiotic selection with puromycin was then used to obtain high purity motoneuron cultures following differentiation of mouse ES cells. Purity was maintained during maturation allowing the production of consistent, uniform populations of cholinergic ES cell-derived motoneurons. Appropriate functional properties of purified motoneurons were verified by acetylcholinesterase activity and electrophysiology. Antibiotic selection, therefore, can provide an inexpensive alternative to current methods for isolating ES cell-derived motoneurons at high purity that does not require specialized laboratory equipment and provides a unique platform for studies in motoneuron development and degeneration.


Assuntos
Células-Tronco Embrionárias/citologia , Proteínas de Homeodomínio/genética , Neurônios Motores/citologia , Neurogênese , Fatores de Transcrição/genética , Acetiltransferases/genética , Animais , Antimetabólitos Antineoplásicos/metabolismo , Técnicas de Cultura de Células/métodos , Engenharia Celular , Células-Tronco Embrionárias/metabolismo , Regulação Enzimológica da Expressão Gênica , Camundongos , Neurônios Motores/metabolismo , Inibidores da Síntese de Proteínas/metabolismo , Puromicina/metabolismo , Transgenes
13.
Nature ; 509(7498): 49-54, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24670665

RESUMO

Many aspects of cellular physiology remain unstudied in somatic stem cells, for example, there are almost no data on protein synthesis in any somatic stem cell. Here we set out to compare protein synthesis in haematopoietic stem cells (HSCs) and restricted haematopoietic progenitors. We found that the amount of protein synthesized per hour in HSCs in vivo was lower than in most other haematopoietic cells, even if we controlled for differences in cell cycle status or forced HSCs to undergo self-renewing divisions. Reduced ribosome function in Rpl24(Bst/+) mice further reduced protein synthesis in HSCs and impaired HSC function. Pten deletion increased protein synthesis in HSCs but also reduced HSC function. Rpl24(Bst/+) cell-autonomously rescued the effects of Pten deletion in HSCs; blocking the increase in protein synthesis, restoring HSC function, and delaying leukaemogenesis. Pten deficiency thus depletes HSCs and promotes leukaemia partly by increasing protein synthesis. Either increased or decreased protein synthesis impairs HSC function.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Biossíntese de Proteínas , Animais , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Feminino , Citometria de Fluxo , Teste de Complementação Genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/patologia , Homeostase/efeitos dos fármacos , Homeostase/genética , Cinética , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Masculino , Camundongos , Mutação/genética , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/genética , Puromicina/análogos & derivados , Puromicina/metabolismo , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Ribossomos/genética , Ribossomos/metabolismo , Fatores de Tempo
14.
J Biol Chem ; 289(18): 12693-704, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24652291

RESUMO

Expression of CGS1, which codes for an enzyme of methionine biosynthesis, is feedback-regulated by mRNA degradation in response to S-adenosyl-L-methionine (AdoMet). In vitro studies revealed that AdoMet induces translation arrest at Ser-94, upon which several ribosomes stack behind the arrested one, and mRNA degradation occurs at multiple sites that presumably correspond to individual ribosomes in a stacked array. Despite the significant contribution of stacked ribosomes to inducing mRNA degradation, little is known about the ribosomes in the stacked array. Here, we assigned the peptidyl-tRNA species of the stacked second and third ribosomes to their respective codons and showed that they are arranged at nine-codon intervals behind the Ser-94 codon, indicating tight stacking. Puromycin reacts with peptidyl-tRNA in the P-site, releasing the nascent peptide as peptidyl-puromycin. This reaction is used to monitor the activity of the peptidyltransferase center (PTC) in arrested ribosomes. Puromycin reaction of peptidyl-tRNA on the AdoMet-arrested ribosome, which is stalled at the pre-translocation step, was slow. This limited reactivity can be attributed to the peptidyl-tRNA occupying the A-site at this step rather than to suppression of PTC activity. In contrast, puromycin reactions of peptidyl-tRNA with the stacked second and third ribosomes were slow but were not as slow as pre-translocation step ribosomes. We propose that the anticodon end of peptidyl-tRNA resides in the A-site of the stacked ribosomes and that the stacked ribosomes are stalled at an early step of translocation, possibly at the P/E hybrid state.


Assuntos
Proteínas de Arabidopsis/metabolismo , Carbono-Oxigênio Liases/metabolismo , Elongação Traducional da Cadeia Peptídica , Ribossomos/metabolismo , S-Adenosilmetionina/metabolismo , Sequência de Aminoácidos , Proteínas de Arabidopsis/genética , Sequência de Bases , Sítios de Ligação/genética , Carbono-Oxigênio Liases/genética , Eletroforese em Gel de Poliacrilamida , Cinética , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Peptídeos/genética , Peptídeos/metabolismo , Puromicina/análogos & derivados , Puromicina/metabolismo , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA de Plantas/genética , RNA de Plantas/metabolismo , Aminoacil-RNA de Transferência/metabolismo , Ribossomos/genética , S-Adenosilmetionina/genética , Transcrição Gênica
15.
Nat Commun ; 4: 2735, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24193185

RESUMO

Eradication of cancer cells while minimizing damage to healthy cells is a primary goal of cancer therapy. Highly selective drugs are urgently needed. Here we demonstrate a new prodrug strategy for selective cancer therapy that utilizes increased histone deacetylase (HDAC) and tumour-associated protease activities produced in malignant cancer cells. By coupling an acetylated lysine group to puromycin, a masked cytotoxic agent is created, which is serially activated by HDAC and an endogenous protease cathepsin L (CTSL) that remove the acetyl group first and then the unacetylated lysine group liberating puromycin. The agent selectively kills human cancer cell lines with high HDAC and CTSL activities. In vivo studies confirm tumour growth inhibition in prodrug-treated mice bearing human cancer xenografts. This cancer-selective cleavage of the masking group is a promising strategy for the next generation of anticancer drug development that could be applied to many other cytotoxic agents.


Assuntos
Antineoplásicos/uso terapêutico , Catepsina L/metabolismo , Histona Desacetilases/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Pró-Fármacos , Puromicina/uso terapêutico , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Camundongos , Neoplasias Experimentais/enzimologia , Puromicina/química , Puromicina/metabolismo
16.
Biotechnol Prog ; 29(4): 1043-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23749410

RESUMO

Due to the high medical and commercial value of recombinant proteins for clinical and diagnostic purposes, the protein synthesis machinery of mammalian host cells is the subject of extensive research by the biopharmaceutical industry. RNA translation and protein synthesis are steps that may determine the extent of growth and productivity of host cells. To address the problems of utilization of current radioisotope methods with proprietary media, we have focused on the application of an alternative method of measuring protein synthesis in recombinant Chinese hamster ovary (CHO) cells. This method employs puromycin as a nonradioactive label which incorporates into nascent polypeptide chains and is detectable by western blotting. This method, which is referred to as SUnSET, successfully demonstrated the expected changes in protein synthesis in conditions that inhibit and restore translation activity and was reproducibly quantifiable. The study of the effects of feed and sodium butyrate addition on protein synthesis by SUnSET revealed an increase following 1 h feed supplementation while a high concentration of sodium butyrate was able to decrease translation during the same treatment period. Finally, SUnSET was used to compare protein synthesis activity during batch culture of the CHO cell line in relation to growth. The results indicate that as the cells approached the end of batch culture, the global rate of protein synthesis declined in parallel with the decreasing growth rate. In conclusion, this method can be used as a "snapshot" to directly monitor the effects of different culture conditions and treatments on translation in recombinant host cells.


Assuntos
Biossíntese de Proteínas , Proteínas/metabolismo , Puromicina/análise , Animais , Western Blotting , Células CHO , Técnicas de Cultura de Células , Células Cultivadas , Cricetulus , Puromicina/metabolismo
17.
J Cell Sci ; 123(Pt 13): 2266-72, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20554896

RESUMO

Continuous and regulated remodelling of the cytoskeleton is crucial for many basic cell functions. In contrast to actin filaments and microtubules, it is not understood how this is accomplished for the third major cytoskeletal filament system, which consists of intermediate-filament polypeptides. Using time-lapse fluorescence microscopy of living interphase cells, in combination with photobleaching, photoactivation and quantitative fluorescence measurements, we observed that epithelial keratin intermediate filaments constantly release non-filamentous subunits, which are reused in the cell periphery for filament assembly. This cycle is independent of protein biosynthesis. The different stages of the cycle occur in defined cellular subdomains: assembly takes place in the cell periphery and newly formed filaments are constantly transported toward the perinuclear region while disassembly occurs, giving rise to diffusible subunits for another round of peripheral assembly. Remaining juxtanuclear filaments stabilize and encage the nucleus. Our data suggest that the keratin-filament cycle of assembly and disassembly is a major mechanism of intermediate-filament network plasticity, allowing rapid adaptation to specific requirements, notably in migrating cells.


Assuntos
Citoesqueleto/metabolismo , Queratinas/metabolismo , Citoesqueleto de Actina/metabolismo , Linhagem Celular , Movimento Celular/fisiologia , Núcleo Celular/metabolismo , Cicloeximida/metabolismo , Citoesqueleto/ultraestrutura , Recuperação de Fluorescência Após Fotodegradação , Humanos , Queratinas/ultraestrutura , Microtúbulos/metabolismo , Subunidades Proteicas/metabolismo , Inibidores da Síntese de Proteínas/metabolismo , Pseudópodes/metabolismo , Puromicina/metabolismo
18.
J Mol Biol ; 396(4): 1043-52, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20045415

RESUMO

EF4, although structurally similar to the translocase EF-G, promotes back-translocation of tRNAs on the ribosome and is important for bacterial growth under certain conditions. Here, using a coordinated set of in vitro kinetic measures, including changes in the puromycin reactivity of peptidyl-tRNA and in the fluorescence of labeled tRNAs and mRNA, we elucidate the kinetic mechanism of EF4-catalyzed back-translocation and determine the effects of the translocation inhibitors spectinomycin and viomycin on the process. EF4-dependent back-translocation proceeds from a post-translocation (POST) complex to a pre-translocation (PRE) complex via a four-step kinetic scheme (i.e., POST-->I(1)-->I(2)-->I(3)-->PRE, which is not the simple reverse of translocation). During back-translocation, movements of the tRNA core regions and of mRNA are closely coupled to one another but are sometimes decoupled from movement of the 3'-end of peptidyl-tRNA. EF4 may be thought of as performing an interrupted catalysis of back-translocation, stopping at the formation of I(3) rather than catalyzing the complete process of back-translocation culminating in PRE complex formation. The delay in polypeptide elongation resulting from transient accumulation of I(3) is likely to be important for optimizing functional protein biosynthesis.


Assuntos
Proteínas de Escherichia coli/metabolismo , Fatores de Elongação da Transcrição/metabolismo , Sequência de Bases , Catálise , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Cinética , Modelos Biológicos , Fatores de Iniciação de Peptídeos , Puromicina/metabolismo , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Aminoacil-RNA de Transferência/genética , Aminoacil-RNA de Transferência/metabolismo , Ribossomos/metabolismo , Espectrometria de Fluorescência , Fatores de Elongação da Transcrição/genética
19.
EMBO J ; 28(19): 2959-70, 2009 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-19713935

RESUMO

While probing the role of RNA for the function of SET1C/COMPASS histone methyltransferase, we identified SET1RC (SET1 mRNA-associated complex), a complex that contains SET1 mRNA and Set1, Swd1, Spp1 and Shg1, four of the eight polypeptides that constitute SET1C. Characterization of SET1RC showed that SET1 mRNA binding did not require associated Swd1, Spp1 and Shg1 proteins or RNA recognition motifs present in Set1. RNA binding was not observed when Set1 protein and SET1 mRNA were derived from independent genes or when SET1 transcripts were restricted to the nucleus. Importantly, the protein-RNA interaction was sensitive to EDTA, to the translation elongation inhibitor puromycin and to the inhibition of translation initiation in prt1-1 mutants. Taken together, our results support the idea that SET1 mRNA binding was dependent on translation and that SET1RC assembled on nascent Set1 in a cotranslational manner. Moreover, we show that cellular accumulation of Set1 is limited by the availability of certain SET1C components, such as Swd1 and Swd3, and suggest that cotranslational protein interactions may exert an effect in the protection of nascent Set1 from degradation.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ácido Edético/metabolismo , Regulação Fúngica da Expressão Gênica , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Ligação Proteica , Biossíntese de Proteínas , Estrutura Terciária de Proteína , Inibidores da Síntese de Proteínas/metabolismo , Puromicina/metabolismo , RNA Mensageiro/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
20.
Biochemistry ; 47(17): 4898-906, 2008 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-18393533

RESUMO

The current view of ribosomal peptidyl transfer is that the ribosome is a ribozyme and that ribosomal proteins are not involved in catalysis of the chemical reaction. This view is largely based on the first crystal structures of bacterial large ribosomal subunits that did not show any protein components near the peptidyl transferase center (PTC). Recent crystallographic data on the full 70S ribosome from Thermus thermophilus, however, show that ribosomal protein L27 extends with its N-terminus into the PTC in accordance with independent biochemical data, thus raising the question of whether the ribozyme picture is strictly valid. We have carried out extensive computer simulations of the peptidyl transfer reaction in the T. thermophilus ribosome to address the role of L27. The results show a reaction rate similar to that obtained in earlier simulations of the Haloarcula marismortui reaction. Furthermore, deletion of L27 is predicted to only give a minor rate reduction, in agreement with biochemical data, suggesting that the ribozyme view is indeed valid. The N-terminus of L27 is predicted to interact with the A76 phosphate group of the A-site tRNA, thereby explaining the observed impairment of A-site substrate binding for ribosomes lacking L27. Simulations are also reported for the reaction with puromycin, an A-site tRNA analogue which lacks the A76 phosphate group. The calculated energetics shows that this substrate can cause a downward p K a shift of L27 and that the reaction proceeds faster with the L27 N-terminus deprotonated, in contrast to the situation with aminoacyl-tRNA substrates. These results could explain the observed differences in pH dependence between the puromycin and C-puromycin reactions, where the former reaction has been seen to depend on an additional ionizing group besides the attacking amine, and our model predicts this ionizing group to be the N-terminal amine of L27.


Assuntos
Proteínas de Bactérias/metabolismo , Peptídeos/metabolismo , Proteínas Ribossômicas/metabolismo , Thermus thermophilus/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Simulação por Computador , Cinética , Modelos Biológicos , Modelos Moleculares , Mutação , Peptidil Transferases/metabolismo , Conformação Proteica , Prótons , Puromicina/metabolismo , Aminoacil-RNA de Transferência/metabolismo , Proteínas Ribossômicas/química , Proteínas Ribossômicas/genética , Subunidades Ribossômicas Maiores de Bactérias/genética , Subunidades Ribossômicas Maiores de Bactérias/metabolismo , Termodinâmica , Thermus thermophilus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA