Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Acta Pharmacol Sin ; 41(11): 1427-1432, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32555510

RESUMEN

Induced pluripotent stem cells (iPSCs) have become an essential research platform to study different human diseases once being discovered by Dr. Shinya Yamanaka in 2006. Another breakthrough in biomedical research is the application of CRISPR/Cas9 system for genome editing in mammalian cells. Although numerous studies have been done to develop methods for gene editing in iPSCs, the current approaches suffer from several limitations, including time and labor consuming, low editing efficiency, and potential off-target effects. In the current study, we report an electroporation-mediated plasmid CRISPR/Cas9 delivery approach for genome editing in iPSCs. With this approach, an edited iPSC cell line could be obtained within 2 weeks. In addition, the transit introducing of CRISPR/Cas9 machinery could minimize genomic integration of Cas9 gene, which avoided potential long-term side effects of Cas9 enzyme. We showed that CRISPR/Cas9-mediated genomic editing did not affect pluripotency and differentiation ability of iPSCs. With the quickly evolving of both iPSC and CRISPR/Cas9-mediated genome editing research fields, we believe that our method can significantly facilitate the application of genome editing in iPSCs research.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas , Edición Génica/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Secuencia de Bases , Diferenciación Celular/genética , Técnicas de Inactivación de Genes/métodos , Humanos , Plásmidos , Ubiquitina Tiolesterasa/genética
2.
Cell Death Differ ; 30(5): 1366-1381, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36899106

RESUMEN

Gasdermin (GSDM)-mediated pyroptosis is functionally involved in multiple diseases, but Gasdermin-B (GSDMB) exhibit cell death-dependent and independent activities in several pathologies including cancer. When the GSDMB pore-forming N-terminal domain is released by Granzyme-A cleavage, it provokes cancer cell death, but uncleaved GSDMB promotes multiple pro-tumoral effects (invasion, metastasis, and drug resistance). To uncover the mechanisms of GSDMB pyroptosis, here we determined the GSDMB regions essential for cell death and described for the first time a differential role of the four translated GSDMB isoforms (GSDMB1-4, that differ in the alternative usage of exons 6-7) in this process. Accordingly, we here prove that exon 6 translation is essential for GSDMB mediated pyroptosis, and therefore, GSDMB isoforms lacking this exon (GSDMB1-2) cannot provoke cancer cell death. Consistently, in breast carcinomas the expression of GSDMB2, and not exon 6-containing variants (GSDMB3-4), associates with unfavourable clinical-pathological parameters. Mechanistically, we show that GSDMB N-terminal constructs containing exon-6 provoke cell membrane lysis and a concomitant mitochondrial damage. Moreover, we have identified specific residues within exon 6 and other regions of the N-terminal domain that are important for GSDMB-triggered cell death as well as for mitochondrial impairment. Additionally, we demonstrated that GSDMB cleavage by specific proteases (Granzyme-A, Neutrophil Elastase and caspases) have different effects on pyroptosis regulation. Thus, immunocyte-derived Granzyme-A can cleave all GSDMB isoforms, but in only those containing exon 6, this processing results in pyroptosis induction. By contrast, the cleavage of GSDMB isoforms by Neutrophil Elastase or caspases produces short N-terminal fragments with no cytotoxic activity, thus suggesting that these proteases act as inhibitory mechanisms of pyroptosis. Summarizing, our results have important implications for understanding the complex roles of GSDMB isoforms in cancer or other pathologies and for the future design of GSDMB-targeted therapies.


Asunto(s)
Neoplasias de la Mama , Piroptosis , Humanos , Femenino , Granzimas/genética , Granzimas/metabolismo , Péptido Hidrolasas/metabolismo , Elastasa de Leucocito/metabolismo , Gasderminas , Proteínas de Neoplasias/metabolismo , Caspasas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Neoplasias de la Mama/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo
3.
J Biol Chem ; 286(15): 12820-4, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21343302

RESUMEN

Plasma membrane repair is an essential process for maintenance of homeostasis at the cellular and tissue levels, whereas compromised repair capacity contributes to degenerative human diseases. Our recent studies show that MG53 is essential for muscle membrane repair, and defects in MG53 function are linked to muscular dystrophy and cardiac dysfunction. Here we report that polymerase I and transcript release factor (PTRF), a gene known to regulate caveolae membrane structure, is an indispensable component of the membrane repair machinery. PTRF acts as a docking protein for MG53 during membrane repair potentially by binding exposed membrane cholesterol at the injury site. Cells lacking expression of endogenous PTRF show defective trafficking of MG53 to membrane injury sites. A mutation in PTRF associated with human disease results in aberrant nuclear localization of PTRF and disrupts MG53 function in membrane resealing. Although RNAi silencing of PTRF leads to defective muscle membrane repair, overexpression of PTRF can rescue membrane repair defects in dystrophic muscle. Our data suggest that membrane-delimited interaction between MG53 and PTRF contributes to initiation of cell membrane repair, which can be an attractive target for treatment or prevention of tissue injury in human diseases.


Asunto(s)
Proteínas Portadoras/metabolismo , Caveolas/metabolismo , Músculo Esquelético/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Proteínas Portadoras/genética , Caveolas/patología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Células HeLa , Cardiopatías/genética , Cardiopatías/metabolismo , Cardiopatías/patología , Células Hep G2 , Humanos , Proteínas de la Membrana , Ratones , Músculo Esquelético/patología , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Distrofias Musculares/terapia , Mutación , Transporte de Proteínas/genética , Proteínas de Unión al ARN/genética , Proteínas de Motivos Tripartitos
4.
J Biol Chem ; 286(11): 9038-48, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21189256

RESUMEN

Bcl-2 homology domain-3 (BH3) peptides are potent cancer therapeutic reagents that target regulators of apoptotic cell death in cancer cells. However, their cytotoxic effects are affected by different expression levels of Bcl-2 family proteins. We recently found that the amphipathic tail-anchoring peptide (ATAP) from Bfl-1, a bifunctional Bcl-2 family member, produced strong pro-apoptotic activity by permeabilizing the mitochondrial outer membrane. Here, we test whether the activity of ATAP requires other cellular factors and whether ATAP has an advantage over the BH3 peptides in targeting cancer cells. Confocal microscopic imaging illustrates specific targeting of ATAP to mitochondria, whereas BH3 peptides show diffuse patterns of cytosolic distribution. Although the pro-apoptotic activities of BH3 peptides are largely inhibited by either overexpression of anti-apoptotic Bcl-2 or Bcl-xL or nullification of pro-apoptotic Bax and Bak in cells, the pro-apoptotic function of ATAP is not affected by these cellular factors. Reconstitution of synthetic ATAP into liposomal membranes results in release of fluorescent molecules of the size of cytochrome c from the liposomes, suggesting that the membrane permeabilizing activity of ATAP does not require additional protein factors. Because ATAP can target to the mitochondrial membrane and its pro-apoptotic activity does not depend on the content of Bcl-2 family proteins, it represents a promising candidate for anti-cancer drugs that can potentially overcome the intrinsic apoptosis-resistant nature of cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Neoplasias/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/farmacología , Proteínas Proto-Oncogénicas/farmacología , Proteína bcl-X/metabolismo , Citocromos c/metabolismo , Células HeLa , Humanos , Liposomas/química , Antígenos de Histocompatibilidad Menor , Neoplasias/metabolismo , Permeabilidad/efectos de los fármacos
5.
FASEB J ; 25(8): 2638-49, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21518849

RESUMEN

We describe a novel transgenic system for tissue-specific and inducible control of gene expression in mice. The system employs a tetracycline-responsive CMV promoter that controls transcription of a short-hairpin RNA (shRNA) that remains nonfunctional until an interrupting reporter cassette is excised by Cre recombinase. Insertion of Dicer and Drosha RNase processing sites within the shRNA allows generation of siRNA to knock down a target gene efficiently. Tissue-specific shRNA expression is achieved through the use of appropriate inducer mice with tissue-specific expression of Cre. We applied this system to regulate expression of junctophilins (JPs), genes essential for maintenance of membrane ultrastructure and Ca(2+) signaling in muscle. Transgenic mice with skeletal muscle-specific expression of shRNA against JP mRNAs displayed no basal change of JP expression before treatment with doxycycline (Dox), while inducible and reversible knockdown of JPs was achieved by feeding mice with Dox-containing water. Dox-induced knockdown of JPs led to abnormal junctional membrane structure and Ca(2+) signaling in adult muscle fibers, consistent with essential roles of JPs in muscle development and function. This transgenic approach can be applied for inducible and reversible gene knockdown or gene overexpression in many different tissues, thus providing a versatile system for elucidating the physiological gene function in viable animal models.


Asunto(s)
Técnicas Genéticas , Ratones Transgénicos/genética , Plásmidos/genética , Animales , Secuencia de Bases , Células CHO , Cricetinae , Cricetulus , Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Células HEK293 , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Interferencia de ARN , ARN Interferente Pequeño/genética , Distribución Tisular
6.
FEBS Lett ; 551(1-3): 29-36, 2003 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-12965200

RESUMEN

Human Bfl-1 is an anti-apoptotic Bcl-2 family member. Here, we found that Bfl-1 was converted into a potent death-promoting protein by green fluorescent protein (GFP) fusion with its N-terminus. The transient expression of GFP-Bfl-1 induced cytochrome c release and triggered apoptosis in 293T cells, which depended on the mitochondrial localization of GFP-Bfl-1. Apoptosis induced by GFP-Bfl-1 was significantly blocked by the pan-caspase inhibitor carbobenzoxy-Val-Ala-Asp-fluoromethyl ketone, but was not blocked by either Bcl-xL or Bfl-1. Our findings provide a useful model for understanding the structural basis of Bcl-2 family proteins that act in an opposite way despite sharing structural similarity between anti-apoptotic and pro-apoptotic proteins.


Asunto(s)
Apoptosis , Proteínas Proto-Oncogénicas c-bcl-2/química , Caspasas/fisiología , Línea Celular , Grupo Citocromo c/metabolismo , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Antígenos de Histocompatibilidad Menor , Mitocondrias/química , Mitocondrias/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/metabolismo , Proteína bcl-X
7.
J Cell Sci ; 120(Pt 16): 2912-23, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17666431

RESUMEN

Many Bcl2 family proteins target intracellular membranes by their C-terminal tail-anchor domain. Bfl1 is a bi-functional Bcl2 family protein with both anti- and pro-apoptotic activities and contains an amphipathic tail-anchoring peptide (ATAP; residues 147-175) with unique properties. Here we show that ATAP targets specifically to mitochondria, and induces caspase-dependent apoptosis that does not require Bax or Bak. Mutagenesis studies revealed that lysine residues flanking the ATAP sequence are involved in targeting of the peptide to the mitochondrial membrane, and charged residues that contribute to the amphipathic nature of ATAP are critical for its pro-apoptotic function. The ATAP sequence is present in another tumor suppressor gene, HCCS1, which contains an additional mitochondria-targeting signal (MTS) close to the ATAP. We propose that both ATAP and MTS could be used as therapeutic peptides to induce cell death in the treatment of cancer cells.


Asunto(s)
Apoptosis , Membranas Mitocondriales/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/química , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular Tumoral , Secuencia Conservada , Humanos , Membrana Dobles de Lípidos/metabolismo , Potencial de la Membrana Mitocondrial , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , Péptidos/química , Permeabilidad , Señales de Clasificación de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/genética , Relación Estructura-Actividad , Proteínas Supresoras de Tumor/genética , Proteínas de Transporte Vesicular , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
8.
J Biol Chem ; 281(21): 14691-9, 2006 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-16574658

RESUMEN

The TRAP/Mediator coactivator complex serves as a functional interface between DNA-bound transactivators and the RNA polymerase II-associated basal transcription apparatus. TRAP220/MED1 is a variably associated subunit of the complex that plays a specialized role in selectively targeting TRAP/Mediator to specific genes. Ablation of the Trap220/Med1 gene in mice impairs embryonic cell growth, yet the underlying mechanism is unknown. In this report, we identified distinct cell growth regulatory genes whose expression is affected by the loss of TRAP220/MED1 by RNA interference. Among the down-regulated genes revealed by cDNA microarray analyses, we identified Aurora-A, a centrosome kinase that plays a critical role in regulating M phase events and is frequently amplified in several types of cancer. In general, we found that TRAP220/MED1 expression is required for high basal levels of Aurora-A gene expression and that ectopic overexpression of TRAP220/MED1 coactivates transcription from the Aurora-A gene promoter. Furthermore, chromatin immunoprecipitation assays show that TRAP220/MED1-containing TRAP/Mediator complexes directly bind to the Aurora-A promoter in vivo. Finally, we present evidence suggesting that TRAP/Mediator is recruited to the Aurora-A gene via direct interactions between TRAP220/MED1 and the Ets-related transcription factor GABP. Taken together, these findings suggest that TRAP220/MED1 plays a novel coregulatory role in facilitating the recruitment of TRAP/Mediator to specific target genes involved in growth and cell cycle progression.


Asunto(s)
Endodesoxirribonucleasas/metabolismo , Regulación Enzimológica de la Expresión Génica , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción/metabolismo , Aurora Quinasa A , Aurora Quinasas , Proliferación Celular , ADN/química , Células HeLa , Humanos , Subunidad 1 del Complejo Mediador , Modelos Genéticos , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Hormonas Tiroideas/metabolismo , Activación Transcripcional
9.
J Cell Biochem ; 91(2): 410-22, 2004 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-14743399

RESUMEN

Various chemotherapeutic agents have been shown to sensitize cancer cells to members of the tumor necrosis factor (TNF) family. However, it is unclear whether sensitization by chemotherapeutic agents involves the transcriptional regulation of apoptosis-related genes. In this study, we investigated mRNA regulation of TNF family receptors and Bcl-2 family members after treating the murine colon cancer cell line, CT26, with various apoptosis inducers. We found that treatment with cycloheximide, a protein synthesis inhibitor, remarkably increased CD40 mRNA levels by semi-quantitative RT-PCR. Other protein synthesis inhibitors, such as anisomycin and emetine, also enhanced CD40 mRNA expression, which was significantly blocked by a NF-kappaB antagonist and a p38 MAP kinase antagonist. After treatment with cycloheximide, and further cultivation in fresh medium, CD40 protein levels were found to increase by flow cytometry. Additionally, we found that cycloheximide treatment appeared to downregulate the Bcl-xL mRNA level but not the Bax mRNA level by RNase protection assay. Because the upregulation of CD40 mRNA and the downregulation of Bcl-xL correlated with CT26 cell death, our results suggest that chemotherapeutic agents, including cycloheximide, may exert their synergistic effects on the TNF family treatment of cancer cells by regulating the mRNA levels of apoptosis-related genes.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transcripción Genética , Animales , Anisomicina/farmacología , Apoptosis/efectos de los fármacos , Antígenos CD40/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Cicloheximida/farmacología , Regulación hacia Abajo , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA