Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.592
Filtrar
1.
Methods Cell Biol ; 189: 1-21, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39393878

RESUMEN

Despite being tightly regulated, messenger RNA (mRNA) translation, a manner in which cells control expression of genes and rapidly respond to stimuli, is highly dysfunctional and plastic in pathologies including cancer. Conversely, the investigation of molecular mechanisms whereby mRNA translation becomes aberrant in cancer, as well as inhibition thereof, become critical in developing novel therapeutic approaches. More specifically, in malignancies such as chronic lymphocytic leukemia in which aberrant global and transcript specific translation has been linked with poorer patient outcomes, targeting translation is a relevant approach, with various translation inhibitors under development. Here we elaborate on a protein synthesis assay by flow cytometry, O-propargyl-puromycin, demonstrating global mRNA translation rate with a variety of different applications including cell lines, primary cells or co-culture systems in vitro. This method provides a comprehensive tool in quantifying the rate of global mRNA translation in cancer cells, as well as that of the tumor microenvironment cells, or in response to inhibitory therapeutic agents while offering the possibility to simultaneously assess other cellular markers.


Asunto(s)
Biosíntesis de Proteínas , Puromicina , ARN Mensajero , Microambiente Tumoral , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Puromicina/farmacología , Leucemia/genética , Leucemia/patología , Citometría de Flujo/métodos , Línea Celular Tumoral , Técnicas de Cocultivo/métodos
2.
Biomolecules ; 14(9)2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39334897

RESUMEN

Introducing genetic material into hard-to-transfect mammalian cell lines and primary cells is often best achieved through retroviral infection. An ideal retroviral vector should offer a compact, selectable, and screenable marker while maximizing transgene delivery capacity. However, a previously published retroviral vector featuring an EGFP/Puromycin fusion protein failed to meet these criteria in our experiments. We encountered issues such as low infection efficiency, weak EGFP fluorescence, and selection against infected cells. To address these shortcomings, we developed a novel retroviral vector based on the Moloney murine leukemia virus. This vector includes a compact bifunctional EGFP and Puromycin resistance cassette connected by a 2A peptide. Our extensively tested vector demonstrated superior EGFP expression, efficient Puromycin selection, and no growth penalty in infected cells compared with the earlier design. These benefits were consistent across multiple mammalian cell types, underscoring the versatility of our vector. In summary, our enhanced retroviral vector offers a robust solution for efficient infection, reliable detection, and effective selection in mammalian cells. Its improved performance and compact design make it an ideal choice for a wide range of applications involving precise genetic manipulation and characterization in cell-based studies.


Asunto(s)
Vectores Genéticos , Proteínas Fluorescentes Verdes , Virus de la Leucemia Murina de Moloney , Retroviridae , Vectores Genéticos/genética , Humanos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Virus de la Leucemia Murina de Moloney/genética , Retroviridae/genética , Animales , Puromicina/farmacología , Células HEK293 , Transgenes , Ratones
3.
Antimicrob Agents Chemother ; 68(10): e0079324, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39254294

RESUMEN

Plasmodium parasite resistance to antimalarial drugs is a serious threat to public health in malaria-endemic areas. Compounds that target core cellular processes like translation are highly desirable, as they should be capable of killing parasites in their liver and blood stage forms, regardless of molecular target or mechanism. Assays that can identify these compounds are thus needed. Recently, specific quantification of native Plasmodium berghei liver stage protein synthesis, as well as that of the hepatoma cells supporting parasite growth, was achieved via automated confocal feedback microscopy of the o-propargyl puromycin (OPP)-labeled nascent proteome, but this imaging modality is limited in throughput. Here, we developed and validated a miniaturized high content imaging (HCI) version of the OPP assay that increases throughput, before deploying this approach to screen the Pathogen Box. We identified only two hits; both of which are parasite-specific quinoline-4-carboxamides, and analogs of the clinical candidate and known inhibitor of blood and liver stage protein synthesis, DDD107498/cabamiquine. We further show that these compounds have strikingly distinct relationships between their antiplasmodial and translation inhibition efficacies. These results demonstrate the utility and reliability of the P. berghei liver stage OPP HCI assay for the specific, single-well quantification of Plasmodium and human protein synthesis in the native cellular context, allowing the identification of selective Plasmodium translation inhibitors with the highest potential for multistage activity.


Asunto(s)
Antimaláricos , Hígado , Plasmodium berghei , Antimaláricos/farmacología , Plasmodium berghei/efectos de los fármacos , Hígado/parasitología , Animales , Humanos , Ratones , Malaria/parasitología , Malaria/tratamiento farmacológico , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/antagonistas & inhibidores , Puromicina/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos
4.
Sci Rep ; 14(1): 18772, 2024 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-39138277

RESUMEN

Computational pharmacogenomics can potentially identify new indications for already approved drugs and pinpoint compounds with similar mechanism-of-action. Here, we used an integrated drug repositioning approach based on transcriptomics data and structure-based virtual screening to identify compounds with gene signatures similar to three known proteasome inhibitors (PIs; bortezomib, MG-132, and MLN-2238). In vitro validation of candidate compounds was then performed to assess proteasomal proteolytic activity, accumulation of ubiquitinated proteins, cell viability, and drug-induced expression in A375 melanoma and MCF7 breast cancer cells. Using this approach, we identified six compounds with PI properties ((-)-kinetin-riboside, manumycin-A, puromycin dihydrochloride, resistomycin, tegaserod maleate, and thapsigargin). Although the docking scores pinpointed their ability to bind to the ß5 subunit, our in vitro study revealed that these compounds inhibited the ß1, ß2, and ß5 catalytic sites to some extent. As shown with bortezomib, only manumycin-A, puromycin dihydrochloride, and tegaserod maleate resulted in excessive accumulation of ubiquitinated proteins and elevated HMOX1 expression. Taken together, our integrated drug repositioning approach and subsequent in vitro validation studies identified six compounds demonstrating properties similar to proteasome inhibitors.


Asunto(s)
Bortezomib , Reposicionamiento de Medicamentos , Inhibidores de Proteasoma , Humanos , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/química , Reposicionamiento de Medicamentos/métodos , Bortezomib/farmacología , Transcriptoma , Complejo de la Endopetidasa Proteasomal/metabolismo , Línea Celular Tumoral , Células MCF-7 , Simulación del Acoplamiento Molecular , Antineoplásicos/farmacología , Antineoplásicos/química , Puromicina/farmacología , Perfilación de la Expresión Génica , Supervivencia Celular/efectos de los fármacos
5.
Biochem Biophys Res Commun ; 730: 150369, 2024 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-39013264

RESUMEN

Gemcitabine is a nucleoside analog widely used as an anticancer agent against several types of cancer. Although gemcitabine sometimes shows excellent effectiveness, cancer cells are often poorly responsive to or resistant to the drug. Recently, specific strains or dysbiosis of the human microbiome were correlated with drug reactivity and resistance acquisition. Therefore, we aimed to identify antibiotic compounds that can modulate the microbiome to enhance the responsiveness to gemcitabine. To achieve this, we confirmed the gemcitabine responsiveness based on public data and conducted drug screening on a set of 250 antibiotics compounds. Subsequently, we performed experiments to investigate whether the selected compounds could enhance the responsiveness to gemcitabine. First, we grouped a total of seven tumor cell lines into resistant and sensitive group based on the IC50 value (1 µM) of gemcitabine obtained from the public data. Second, we performed high-throughput screening with compound treatments, identifying seven compounds from the resistant group and five from the sensitive group based on dose dependency. Finally, the combination of the selected compound, puromycin dihydrochloride, with gemcitabine in gemcitabine-resistant cell lines resulted in extensive cell death and a significant increase in cytotoxic efficacy. Additionally, mRNA levels associated with cell viability and stemness were reduced. Through this study, we screened antibiotics to further improve the efficacy of existing anticancer drugs and overcome resistance. By combining existing anticancer agents and antibiotic substances, we hope to establish various drug combination therapies and ultimately improve cancer treatment efficacy.


Asunto(s)
Antibacterianos , Desoxicitidina , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Gemcitabina , Ensayos Analíticos de Alto Rendimiento , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Humanos , Ensayos Analíticos de Alto Rendimiento/métodos , Resistencia a Antineoplásicos/efectos de los fármacos , Línea Celular Tumoral , Antibacterianos/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Puromicina/farmacología , Antimetabolitos Antineoplásicos/farmacología , Sinergismo Farmacológico , Antineoplásicos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología
6.
Eur J Med Chem ; 275: 116604, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-38917665

RESUMEN

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.


Asunto(s)
Transducción de Señal , Animales , Relación Estructura-Actividad , Transducción de Señal/efectos de los fármacos , Masculino , Ratones , Estructura Molecular , Relación Dosis-Respuesta a Droga , Humanos , Antígenos CD13/antagonistas & inhibidores , Antígenos CD13/metabolismo , Encefalinas/química , Encefalinas/metabolismo , Encefalinas/farmacología , Puromicina/farmacología , Puromicina/metabolismo , Puromicina/química , Analgésicos/farmacología , Analgésicos/química , Aminopeptidasas/antagonistas & inhibidores , Aminopeptidasas/metabolismo , Ratas
7.
Proc Natl Acad Sci U S A ; 121(7): e2306993121, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38315848

RESUMEN

Puromycin is covalently added to the nascent chain of proteins by the peptidyl transferase activity of the ribosome and the dissociation of the puromycylated peptide typically follows this event. It was postulated that blocking the translocation of the ribosome with emetine could retain the puromycylated peptide on the ribosome, but evidence against this has recently been published [Hobson et al., Elife 9, e60048 (2020); and Enam et al., Elife 9, e60303 (2020)]. In neurons, puromycylated nascent chains remain in the ribosome even in the absence of emetine, yet direct evidence for this has been lacking. Using biochemistry and cryoelectron microscopy, we show that the puromycylated peptides remain in the ribosome exit channel in the large subunit in a subset of neuronal ribosomes stalled in the hybrid state. These results validate previous experiments to localize stalled polysomes in neurons and provide insight into how neuronal ribosomes are stalled. Moreover, in these hybrid-state neuronal ribosomes, anisomycin, which usually blocks puromycylation, competes poorly with puromycin in the puromycylation reaction, allowing a simple assay to determine the proportion of nascent chains that are stalled in this state. In early hippocampal neuronal cultures, over 50% of all nascent peptides are found in these stalled polysomes. These results provide insights into the stalling mechanisms of neuronal ribosomes and suggest that puromycylated peptides can be used to reveal subcellular sites of hybrid-state stalled ribosomes in neurons.


Asunto(s)
Emetina , Ribosomas , Puromicina/farmacología , Microscopía por Crioelectrón , Emetina/análisis , Emetina/metabolismo , Ribosomas/metabolismo , Biosíntesis de Proteínas , Péptidos/metabolismo , Neuronas/metabolismo
8.
STAR Protoc ; 4(4): 102653, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37862174

RESUMEN

Translation is a fundamental process of cellular behavior. Here, we present a protocol for measuring translation in Drosophila epithelial tissues using O-propargyl-puromycin (OPP), a puromycin derivative. We detail steps for larval dissection, OPP incorporation, fixation, OPP labeling, immunostaining, and imaging. We also provide details of quantification analysis. Significantly, OPP addition to methionine-containing media enables polypeptide labeling in living cells. Here, we study wing imaginal discs, an excellent model system for investigating growth, proliferation, pattern formation, differentiation, and cell death. For complete details on the use and execution of this protocol, please refer to Lee et al. (2018), Ji et al. (2019), and Kiparaki et al. (2022).1,2,3.


Asunto(s)
Drosophila , Discos Imaginales , Puromicina/análogos & derivados , Animales , Larva/metabolismo , Puromicina/farmacología
9.
Nucleic Acids Res ; 51(14): 7465-7479, 2023 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-37395404

RESUMEN

Extremely diverse libraries are essential for effectively selecting functional peptides or proteins, and mRNA display technology is a powerful tool for generating such libraries with over 1012-1013 diversity. Particularly, the protein-puromycin linker (PuL)/mRNA complex formation yield is determining for preparing the libraries. However, how mRNA sequences affect the complex formation yield remains unclear. To study the effects of N-terminal and C-terminal coding sequences on the complex formation yield, puromycin-attached mRNAs containing three random codons after the start codon (32768 sequences) or seven random bases next to the amber codon (6480 sequences) were translated. Enrichment scores were calculated by dividing the appearance rate of every sequence in protein-PuL/mRNA complexes by that in total mRNAs. The wide range of enrichment scores (0.09-2.10 for N-terminal and 0.30-4.23 for C-terminal coding sequences) indicated that the N-terminal and C-terminal coding sequences strongly affected the complex formation yield. Using C-terminal GGC-CGA-UAG-U sequences, which resulted in the highest enrichment scores, we constructed highly diverse libraries of monobodies and macrocyclic peptides. The present study provides insights into how mRNA sequences affect the protein/mRNA complex formation yield and will accelerate the identification of functional peptides and proteins involved in various biological processes and having therapeutic applications.


Asunto(s)
Codón de Terminación , Biblioteca de Péptidos , Péptidos/metabolismo , Proteínas/genética , Puromicina/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo
10.
J Med Chem ; 66(5): 3250-3261, 2023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36855911

RESUMEN

Overexpression of the selenoprotein thioredoxin reductase (TrxR) has been documented in malignant tissues and is of pathological significance for many types of tumors. The antibiotic puromycin (Puro) is a protein synthesis inhibitor causing premature polypeptide chain termination during translation. The well-defined action mechanism of Puro makes it a useful tool in biomedical studies. However, the nonselective cytotoxicity of Puro limits its therapeutic applications. We report herein the construction and evaluation of two Puro prodrugs, that is, S1-Puro with a five-membered cyclic disulfide trigger and S2-Puro with a linear disulfide trigger. S1-Puro is selectively activated by TrxR and shows the TrxR-dependent cytotoxicity to cancer cells, while S2-Puro is readily activated by thiols. Furthermore, S1-Puro displays higher stability in plasma than S2-Puro. We expect that this prodrug strategy may promote the further development of Puro as a therapeutic agent.


Asunto(s)
Profármacos , Reductasa de Tiorredoxina-Disulfuro , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Profármacos/farmacología , Puromicina/farmacología
11.
Angew Chem Int Ed Engl ; 62(23): e202216784, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-36973168

RESUMEN

Puromycin derivatives containing an emissive thieno[3,4-d]-pyrimidine core, modified with azetidine and 3,3-difluoroazetidine as Me2 N surrogates, exhibit translation inhibition and bactericidal activity similar to the natural antibiotic. The analogues are capable of cellular puromycylation of nascent peptides, generating emissive products without any follow-up chemistry. The 3,3-difluoroazetidine-containing analogue is shown to fluorescently label newly translated peptides and be visualized in both live and fixed HEK293T cells and rat hippocampal neurons.


Asunto(s)
Péptidos , Ratas , Animales , Humanos , Puromicina/farmacología , Células HEK293
12.
Appl Biochem Biotechnol ; 195(12): 7379-7396, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37000351

RESUMEN

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.


Asunto(s)
Enfermedades Renales , Síndrome Nefrótico , Podocitos , Humanos , Niño , Ratones , Masculino , Femenino , Animales , Doxorrubicina/toxicidad , Síndrome Nefrótico/inducido químicamente , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/metabolismo , Podocitos/metabolismo , Podocitos/patología , Enfermedades Renales/inducido químicamente , Proteinuria/tratamiento farmacológico , Proteinuria/metabolismo , Proteinuria/patología , Puromicina/metabolismo , Puromicina/farmacología , Puromicina/uso terapéutico
13.
Asian Pac J Cancer Prev ; 23(5): 1803-1812, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-35633567

RESUMEN

Cancer is life-threatening disease and being global health problems. Chemotherapy is one of the most used therapy for cancer since many years ago. Chemotherapy is also toxic for normal cell, not specific to the target cells. Consequently, chemotherapy has various side effects. Monoclonal antibody (MAb) has been developed for specific therapy which only has killing effect in cancer cells, but the survival rate of most MAbs around 20%. Therefore, in clinical practice, MAbs administration should combine with chemotherapeutic agents. For effectiveness of therapy and to minimalize adverse effects, anticancer agent with selective cytotoxic effect on target cells is needed, the immunotoxin. OBJECTIVE: This study introduces a novel approach to conjugate monoclonal antibody (Cetuximab) and toxin (Puromycin), in order to selectively inhibit proliferation of triple negative breast cancer (TNBC) and to enhance the efficacy of MAb in target cells killing. METHODS: Cetuximab was conjugated with Puromycin using a linker, i.e SATP (Succinimidyl-acetylthiopropionate) and tested on triple negative breast cancer cell lines (MDA-MB-231) which expressed EGFR (epidermal growth factor receptor). Cetuximab is MAb which targets EGFR. MCF-7 was used as control cells since it has low or no EGFR expression. Cell counting were conducted as viability assay at 24 hours, 48 hours, and 72 hours after treatment. RESULTS: The results showed significant reduction of live cells number in Conjugate 20 µg/mL cultured in MDA-MB-231 compared to MCF-7 after 24 hours, 48 hours, and 72 hours incubation. In all time period of incubation, significant reduction of MDA-MB-231 live cells number was also observed in Conjugate 20 µg/mL compared to Cetuximab 20 µg/mL. CONCLUSION: Synthesized conjugate showed its target-specific effect in TNBC and improved the efficacy of Cetuximab on TNBC. In the future, this conjugate can be a potential anticancer therapy in treating triple-negative breast cancer.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Cetuximab/farmacología , Receptores ErbB/metabolismo , Humanos , Puromicina/farmacología , Puromicina/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo
14.
Int J Mol Sci ; 22(24)2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34948207

RESUMEN

In minimal change nephrotic syndrome, podocyte vesicle transport is enhanced. Adenomatous polyposis coli (APC) anchors microtubules to cell membranes and plays an important role in vesicle transport. To clarify the role of APC in vesicle transport in podocytes, nephrotic syndrome was induced by puromycin amino nucleoside (PAN) injection in mice expressing APC1638T lacking the C-terminal of microtubule-binding site (APC1638T mouse); this was examined in renal tissue changes. The kidney size and glomerular area of APC1638T mice were reduced (p = 0.014); however, the number of podocytes was same between wild-type (WT) mice and APC1638T mice. The ultrastructure of podocyte foot process was normal by electron microscopy. When nephrotic syndrome was induced, the kidneys of WT+PAN mice became swollen with many hyaline casts, whereas these changes were inhibited in the kidneys of APC1638T+PAN mice. Electron microscopy showed foot process effacement in both groups; however, APC1638T+PAN mice had fewer vesicles in the basal area of podocytes than WT+PAN mice. Cytoplasmic dynein-1, a motor protein for vesicle transport, and α-tubulin were significantly reduced in APC1638T+PAN mice associated with suppressed urinary albumin excretion compared to WT+PAN mice. In conclusion, APC1638T mice showed reduced albuminuria associated with suppressed podocyte vesicle transport when minimal change nephrotic syndrome was induced.


Asunto(s)
Poliposis Adenomatosa del Colon/patología , Albuminuria/patología , Síndrome Nefrótico/patología , Podocitos/patología , Transcitosis/fisiología , Poliposis Adenomatosa del Colon/metabolismo , Albuminuria/metabolismo , Animales , Modelos Animales de Enfermedad , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Síndrome Nefrótico/inducido químicamente , Síndrome Nefrótico/metabolismo , Podocitos/metabolismo , Puromicina/farmacología , Puromicina Aminonucleósido/farmacología
15.
Biochem Biophys Res Commun ; 582: 93-99, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34695756

RESUMEN

The genetic manipulation of cells followed by their selection is indispensable for cell biological research. Although antibiotics-resistant genes are commonly used as selection markers, optimization of the condition for each selective agent is required. Here we utilized split-inteins and the drug-selectable marker puromycin N-acetyltransferase (PAC) to develop a system that enables the selection of cells simultaneously or sequentially transfected with multiple genetic constructs, using only puromycin. The active PAC enzyme was reconstituted by intein-mediated trans-splicing at several inherent or engineered serine/cysteine residues. Multiple splitting and reconstitution of active PAC was readily achieved by selecting optimum division sites based on the cellular tolerance to various puromycin concentrations. To achieve the stepwise selection method, PAC-intein fragments were transduced into cells using a virus-like particle (VLP) composed of HIV-1 gag-pol and VSV-G. The PAC-intein-VLP successfully conferred sufficient PAC activity for puromycin selection, which was quickly diminished in the absence of the VLP. Our findings demonstrate a versatile strategy for establishing markers for all-at-once or stepwise selection of multiple genetic manipulations, which will be useful in many fields of biology.


Asunto(s)
Acetiltransferasas/genética , Ingeniería Celular/métodos , Proteínas de Fusión gag-pol/genética , Inteínas/genética , Glicoproteínas de Membrana/genética , Selección Genética , Proteínas del Envoltorio Viral/genética , Acetiltransferasas/metabolismo , Partículas Similares a Virus Artificiales/química , Partículas Similares a Virus Artificiales/metabolismo , Línea Celular Tumoral , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Proteínas de Fusión gag-pol/metabolismo , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Glicoproteínas de Membrana/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Puromicina/farmacología , Transfección/métodos , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteína Fluorescente Roja
16.
Elife ; 92020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33252038

RESUMEN

Wnt signaling is downregulated in embryonal rhabdomyosarcoma (ERMS) and contributes to the block of differentiation. Epigenetic mechanisms leading to its suppression are unknown and could pave the way toward novel therapeutic modalities. We demonstrate that EHMT2 suppresses canonical Wnt signaling by activating expression of the Wnt antagonist DKK1. Inhibition of EHMT2 expression or activity in human ERMS cell lines reduced DKK1 expression and elevated canonical Wnt signaling resulting in myogenic differentiation in vitro and in mouse xenograft models in vivo. Mechanistically, EHMT2 impacted Sp1 and p300 enrichment at the DKK1 promoter. The reduced tumor growth upon EHMT2 deficiency was reversed by recombinant DKK1 or LGK974, which also inhibits Wnt signaling. Consistently, among 13 drugs targeting chromatin modifiers, EHMT2 inhibitors were highly effective in reducing ERMS cell viability. Our study demonstrates that ERMS cells are vulnerable to EHMT2 inhibitors and suggest that targeting the EHMT2-DKK1-ß-catenin node holds promise for differentiation therapy.


Asunto(s)
Epigénesis Genética , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Rabdomiosarcoma Embrionario/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Dimetilsulfóxido/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Predisposición Genética a la Enfermedad , Antígenos de Histocompatibilidad/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Ratones , Ratones Desnudos , Puromicina/farmacología , Pirazinas/farmacología , Piridinas/farmacología , Quinazolinas/farmacología , Interferencia de ARN , Rabdomiosarcoma Embrionario/genética
17.
Int J Biol Macromol ; 165(Pt A): 1373-1381, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-33045297

RESUMEN

Puromycin sensitive aminopeptidase (PSA or NPEPPS) is a M1 class aminopeptidase is selectively inhibited by the natural product puromycin, an aminonucleoside antibiotic produced by the bacterium Streptomyces alboniger. The molecular basis for this selective inhibition has not been understood well. Here, we report the basis for selectivity of puromycin using biochemical, structural and molecular modeling tools on four different M1 family enzymes including human PSA. Except for PSA, the other three enzymes were not inhibited. Instead, the peptide bond in the puromycin is hydrolyzed to O-methyl-L-tyrosine (OMT) and puromycin aminonucleoside (PAN). Neither of the hydrolyzed products, individually or together inhibit any of the four enzymes. Crystal structure of ePepN using crystals that are incubated with puromycin contained the hydrolyzed products instead of intact puromycin. On the other hand, intact puromycin molecule was observed in the crystal structure of the inactive mutant ePepN (E298A)-puromycin complex. Surprisingly, puromycin does not enter the active site of the mutant enzyme but binds near the entrance. Comparison of puromycin binding region in ePepN mutant enzyme and molecular modeling studies suggest that PSA might be inhibited by similar mode of binding there by blocking the entrance of the active site.


Asunto(s)
Modelos Moleculares , Antígeno Prostático Específico/antagonistas & inhibidores , Conformación Proteica , Puromicina/química , Secuencia de Aminoácidos/genética , Escherichia coli/genética , Humanos , Cinética , Masculino , Antígeno Prostático Específico/química , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/ultraestructura , Puromicina/farmacología , Especificidad por Sustrato/genética
18.
Biochem Biophys Res Commun ; 531(2): 112-117, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32782144

RESUMEN

Iron deficiency anemia indicates poor nutrition and is a public health problem. Iron deficiency is also associated with muscle weakness. However, the intracellular mechanisms by which iron deficiency induces muscle weakness are obscure. The purpose of the present study was to evaluate the effect of iron deficiency on protein synthesis in basal and branched-amino acids (BCAA)- and insulin-stimulated state in muscle cells. Differentiated C2C12 myotubes were incubated with an iron chelator, deferoxamine mesylate, and then stimulated with BCAA or insulin to activate protein synthesis. This iron deprivation resulted in a significant reduction in the abundance of iron-containing proteins, such as the mitochondrial complex 1 subunit protein, compared to control cells, but not of protein that does not contain iron, such as citrate synthase. Proteins involved in glucose utilization, such as glucose transpoter-1, hexokinase and AMP-activated protein kinase (AMPK), were upregulated under iron deficiency. Additionally, rates of BCAA- and insulin-stimulated protein synthesis, measured by puromycin incorporation, were lower in iron-deficient myotubes than in control cells. We suggest that low iron availability attenuates BCAA- and insulin-stimulated protein synthesis, possibly via activation of AMPK in myotubes. The present findings advance the understanding of the importance of iron to skeletal muscle protein synthesis and, thus, may contribute to the prevention of sarcopenia and frailty.


Asunto(s)
Aminoácidos de Cadena Ramificada/farmacología , Insulina/farmacología , Deficiencias de Hierro , Fibras Musculares Esqueléticas/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Animales , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Lipasa/genética , Lipasa/metabolismo , Ratones , Proteínas Mitocondriales/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Puromicina/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitina
19.
Elife ; 92020 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-32844746

RESUMEN

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.


Asunto(s)
Extensión de la Cadena Peptídica de Translación/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína , Puromicina , Ribosomas , Animales , Línea Celular Tumoral , Ratones , Inhibidores de la Síntesis de la Proteína/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas/química , Proteínas/metabolismo , Puromicina/metabolismo , Puromicina/farmacología , ARN Mensajero/química , ARN Mensajero/metabolismo , Aminoacil-ARN de Transferencia/química , Aminoacil-ARN de Transferencia/metabolismo , Ribosomas/efectos de los fármacos , Ribosomas/metabolismo
20.
Elife ; 92020 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-32844748

RESUMEN

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.


Asunto(s)
Núcleo Celular , Biosíntesis de Proteínas , Inhibidores de la Síntesis de la Proteína , Puromicina , Animales , Caenorhabditis elegans , Núcleo Celular/química , Núcleo Celular/metabolismo , Emetina/metabolismo , Emetina/farmacología , Péptidos/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/fisiología , Inhibidores de la Síntesis de la Proteína/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Puromicina/metabolismo , Puromicina/farmacología , ARN de Transferencia/metabolismo , Conejos , Ribosomas/metabolismo , Análisis de la Célula Individual
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA