Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1457629, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39281684

RESUMEN

Induced pluripotent stem cells (iPSCs) have emerged as a revolutionary tool in cell therapies due to their ability to differentiate into various cell types, unlimited supply, and potential as off-the-shelf cell products. New advances in iPSC-derived immune cells have generated potent iNK and iT cells which showed robust killing of cancer cells in animal models and clinical trials. With the advent of advanced genome editing technologies that enable the development of highly engineered cells, here we outline 12 strategies to engineer iPSCs to overcome limitations and challenges of current cell-based immunotherapies, including safety switches, stealth edits, avoiding graft-versus-host disease (GvHD), targeting, reduced lymphodepletion, efficient differentiation, increased in vivo persistence, stemness, metabolic fitness, homing/trafficking, and overcoming suppressive tumor microenvironment and stromal cell barrier. With the development of advanced genome editing techniques, it is now possible to insert large DNA sequences into precise genomic locations without the need for DNA double strand breaks, enabling the potential for multiplexed knock out and insertion. These technological breakthroughs have made it possible to engineer complex cell therapy products at unprecedented speed and efficiency. The combination of iPSC derived iNK, iT and advanced gene editing techniques provides new opportunities and could lead to a new era for next generation of cell immunotherapies.


Asunto(s)
Edición Génica , Inmunoterapia , Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Animales , Inmunoterapia/métodos , Edición Génica/métodos , Diferenciación Celular , Neoplasias/terapia , Neoplasias/inmunología , Ingeniería Celular/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos
2.
Front Neurosci ; 18: 1348478, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38449737

RESUMEN

Fragile X Syndrome (FXS) is a neurological disorder caused by epigenetic silencing of the FMR1 gene. Reactivation of FMR1 is a potential therapeutic approach for FXS that would correct the root cause of the disease. Here, using a candidate-based shRNA screen, we identify nine epigenetic repressors that promote silencing of FMR1 in FXS cells (called FMR1 Silencing Factors, or FMR1- SFs). Inhibition of FMR1-SFs with shRNAs or small molecules reactivates FMR1 in cultured undifferentiated induced pluripotent stem cells, neural progenitor cells (NPCs) and post-mitotic neurons derived from FXS patients. One of the FMR1-SFs is the histone methyltransferase EZH2, for which an FDA-approved small molecule inhibitor, EPZ6438 (also known as tazemetostat), is available. We show that EPZ6438 substantially corrects the characteristic molecular and electrophysiological abnormalities of cultured FXS neurons. Unfortunately, EZH2 inhibitors do not efficiently cross the blood-brain barrier, limiting their therapeutic use for FXS. Recently, antisense oligonucleotide (ASO)-based approaches have been developed as effective treatment options for certain central nervous system disorders. We therefore derived efficacious ASOs targeting EZH2 and demonstrate that they reactivate FMR1 expression and correct molecular and electrophysiological abnormalities in cultured FXS neurons, and reactivate FMR1 expression in human FXS NPCs engrafted within the brains of mice. Collectively, our results establish EZH2 inhibition in general, and EZH2 ASOs in particular, as a therapeutic approach for FXS.

3.
Front Neurosci ; 17: 1251228, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37849894

RESUMEN

A common pathological hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is the cytoplasmic mislocalization and aggregation of the DNA/RNA-binding protein TDP-43, but how loss of nuclear TDP-43 function contributes to ALS and FTD pathogenesis remains largely unknown. Here, using large-scale RNAi screening, we identify TARDBP, which encodes TDP-43, as a gene whose loss-of-function results in elevated DNA mutation rate and genomic instability. Consistent with this finding, we observe increased DNA damage in induced pluripotent stem cells (iPSCs) and iPSC-derived post-mitotic neurons generated from ALS patients harboring TARDBP mutations. We find that the increase in DNA damage in ALS iPSC-derived neurons is due to defects in two major pathways for DNA double-strand break repair: non-homologous end joining and homologous recombination. Cells with defects in DNA repair are sensitive to DNA damaging agents and, accordingly, we find that ALS iPSC-derived neurons show a marked reduction in survival following treatment with a DNA damaging agent. Importantly, we find that increased DNA damage is also observed in neurons with nuclear TDP-43 depletion from ALS/FTD patient brain tissues. Collectively, our results demonstrate that ALS neurons with loss of nuclear TDP-43 function have elevated levels of DNA damage and contribute to the idea that genomic instability is a defining pathological feature of ALS/FTD patients with TDP-43 pathology.

4.
Nucleic Acids Res ; 50(22): 12657-12673, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36511872

RESUMEN

Friedreich's ataxia is an incurable disease caused by frataxin (FXN) protein deficiency, which is mostly induced by GAA repeat expansion in intron 1 of the FXN gene. Here, we identified antisense oligonucleotides (ASOs), complementary to two regions within the first intron of FXN pre-mRNA, which could increase FXN mRNA by ∼2-fold in patient fibroblasts. The increase in FXN mRNA was confirmed by the identification of multiple overlapping FXN-activating ASOs at each region, two independent RNA quantification assays, and normalization by multiple housekeeping genes. Experiments on cells with the ASO-binding sites deleted indicate that the ASO-induced FXN activation was driven by indirect effects. RNA sequencing analyses showed that the two ASOs induced similar transcriptome-wide changes, which did not resemble the transcriptome of wild-type cells. This RNA-seq analysis did not identify directly base-paired off-target genes shared across ASOs. Mismatch studies identified two guanosine-rich motifs (CCGG and G4) within the ASOs that were required for FXN activation. The phosphorodiamidate morpholino oligomer analogs of our ASOs did not activate FXN, pointing to a PS-backbone-mediated effect. Our study demonstrates the importance of multiple, detailed control experiments and target validation in oligonucleotide studies employing novel mechanisms such as gene activation.


Asunto(s)
Ataxia de Friedreich , Regulación de la Expresión Génica , Oligonucleótidos Antisentido , Humanos , Ataxia de Friedreich/genética , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/metabolismo , ARN Mensajero/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Frataxina
5.
Dis Markers ; 2022: 3071665, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36277975

RESUMEN

Objective: To investigate the clinical application of preoperative adductor canal block combined with general anaesthesia in elderly patients with total knee arthroplasty. Methods: Seventy-four patients scheduled for elective TKA in Shaanxi Nuclear Industry Hospital No. 215 were selected and were assigned into group A (continuous ACB prior to the induction of anaesthesia) and group B (continuous ACB after extraction of the tracheal catheter post-operatively) according to the random number table method. Pre and postoperative plasma adrenaline and noradrenaline levels were measured; mean arterial pressure (MAP) and heart rate (HR) were recorded at the admission and the surgical skin incision; intraoperative sufentanil dosage, number of analgesic pump presses at 48 h postoperatively; postoperative adverse effects and length of stay were recorded; resting and active VAS pain scores were assessed at 4, 8, 12, 24, and 48 h postoperatively. Results: Group B experienced a substantial increase in MAP and HR at the time of surgical skin incision, while group A registered a smaller change and a stable haemodynamic profile (P < 0.05). The plasma adrenaline and norepinephrine concentrations in group B were elevated compared to the preoperative period, differentially with group A. Group A received less intraoperative sufentanil than Group B (P < 0.05). Conclusion: Collectively, postoperative resting VAS scores and active VAS scores remained lower in TKA patients who were subjected to preoperative and postoperative ACB, while preoperative ACB in conjunction with general anaesthesia decreased intraoperative sufentanil dosage, contained the surgical stress response, and maintained a stable intraoperative haemodynamic state, in what is probably a preferable option for elderly patients undergoing TKA. This study has served as a reference for postoperative patients to reduce their medication and for clinicians in the treatment going forward.


Asunto(s)
Artroplastia de Reemplazo de Rodilla , Bloqueo Nervioso , Humanos , Anciano , Artroplastia de Reemplazo de Rodilla/efectos adversos , Artroplastia de Reemplazo de Rodilla/métodos , Bloqueo Nervioso/métodos , Dolor Postoperatorio/etiología , Sufentanilo/uso terapéutico , Anestesia General , Epinefrina/uso terapéutico , Norepinefrina/uso terapéutico
6.
J Virol ; 92(3)2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29142135

RESUMEN

During the infection cycle of Autographa californica multiple nucleopolyhedrovirus (AcMNPV), two forms of virions are produced, budded virus (BV) and occlusion-derived virus (ODV). Nucleocapsids that form BV have to egress from the nucleus, whereas nucleocapsids that form ODV remain inside the nucleus. The molecular mechanism that determines whether nucleocapsids remain inside or egress from the nucleus is unknown. AC141 (a predicted E3 ubiquitin ligase) and viral ubiquitin (vUbi) have both been shown to be required for efficient BV production. In this study, it was hypothesized that vUbi interacts with AC141, and in addition, that this interaction was required for BV production. Deletion of both ac141 and vubi restricted viral infection to a single cell, and BV production was completely eliminated. AC141 was ubiquitinated by either vUbi or cellular Ubi, and this interaction was required for optimal BV production. Nucleocapsids in BV, but not ODV, were shown to be specifically ubiquitinated by vUbi, including a 100-kDa protein, as well as high-molecular-weight conjugates. The viral ubiquitinated 100-kDa BV-specific nucleocapsid protein was identified as AC66, which is known to be required for BV production and was shown by coimmunoprecipitation and mass spectrometry to interact with AC141. Confocal microscopy also showed that AC141, AC66, and vUbi interact at the nuclear periphery. These results suggest that ubiquitination of nucleocapsid proteins by vUbi functions as a signal to determine if a nucleocapsid will egress from the nucleus and form BV or remain in the nucleus to form ODV.IMPORTANCE Baculoviruses produce two types of virions called occlusion-derived virus (ODV) and budded virus (BV). ODVs are required for oral infection, whereas BV enables the systemic spread of virus to all host tissues, which is critical for killing insects. One of the important steps for BV production is the export of nucleocapsids out of the nucleus. This study investigated the molecular mechanisms that enable the selection of nucleocapsids for nuclear export instead of being retained within the nucleus, where they would become ODV. Our data show that ubiquitination, a universal cellular process, specifically tags nucleocapsids of BV, but not those found in ODV, using a virus-encoded ubiquitin (vUbi). Therefore, ubiquitination may be the molecular signal that determines if a nucleocapsid is destined to form a BV, thus ensuring lethal infection of the host.


Asunto(s)
Proteínas de la Nucleocápside/metabolismo , Nucleocápside/metabolismo , Nucleopoliedrovirus/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Transporte Activo de Núcleo Celular , Animales , Espectrometría de Masas , Nucleopoliedrovirus/genética , Células Sf9 , Spodoptera/virología , Ensamble de Virus , Liberación del Virus
7.
Proc Natl Acad Sci U S A ; 113(5): 1250-5, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26787892

RESUMEN

During cancer development, it is well established that many genes, including tumor suppressor genes, are hypermethylated and transcriptionally repressed, a phenomenon referred to as epigenetic silencing. In general, the factors involved in, and the mechanistic basis of, epigenetic silencing during cancer development are not well understood. We have recently described an epigenetic silencing pathway, directed by the oncogenic B-Raf proto-oncogene (BRAF) variant BRAF(V600E), that mediates widespread epigenetic silencing in colorectal cancer (CRC). Notably, the BRAF(V600E) mutation is also present in 50-70% of melanomas. Here, we show that the same pathway we identified in CRC also directs epigenetic silencing of a similar set of genes in BRAF-positive melanoma. In both CRC and melanoma, BRAF(V600E) promotes epigenetic silencing through up-regulation of v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog G (MAFG), a transcriptional repressor with sequence-specific DNA-binding activity. The elevated concentration of MAFG drives DNA binding on the promoter. Promoter-bound MAFG recruits a set of corepressors that includes its heterodimeric partner BTB and CNC homology 1, basic leucine zipper transcription factor 1 (BACH1), the chromatin remodeling factor chromodomain helicase DNA-binding protein 8 (CHD8), and the DNA methyltransferase DNMT3B, resulting in hypermethylation and transcriptional silencing. Our results reveal a common BRAF(V600E)-directed transcriptional regulatory pathway that mediates epigenetic silencing in unrelated solid tumors and provide strong support for an instructive model of oncoprotein-directed epigenetic silencing.


Asunto(s)
Neoplasias Colorrectales/genética , Epigénesis Genética/fisiología , Silenciador del Gen , Factor de Transcripción MafG/fisiología , Melanoma/genética , Proteínas Proto-Oncogénicas B-raf/fisiología , Proteínas Represoras/fisiología , Línea Celular Tumoral , Metilación de ADN , Humanos , Proto-Oncogenes Mas , Regulación hacia Arriba
8.
Cell Rep ; 11(9): 1350-7, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26004186

RESUMEN

The CREB-regulated transcription coactivator CRTC2 stimulates CREB target gene expression and has a well-established role in modulating glucose and lipid metabolism. Here, we find, unexpectedly, that loss of CRTC2, as well as CREB1 and its coactivator CREB-binding protein (CBP), results in a deficiency in DNA mismatch repair (MMR) and a resultant increased mutation frequency. We show that CRTC2, CREB1, and CBP are transcriptional activators of well-established MMR genes, including EXO1, MSH6, PMS1, and POLD2. Mining of expression profiling databases and analysis of patient samples reveal that CRTC2 and its target MMR genes are downregulated in specific T cell lymphoma subtypes, which are microsatellite unstable. The levels of acetylated histone H3 on the CRTC2 promoter are significantly reduced in lymphoma in comparison to normal tissue, explaining the decreased CRTC2 expression. Our results establish a role for CRTC2 as a lymphoma tumor suppressor gene that preserves genome integrity by stimulating transcription of MMR genes.


Asunto(s)
Reparación de la Incompatibilidad de ADN/genética , Regulación Neoplásica de la Expresión Génica/genética , Genes Supresores de Tumor/fisiología , Genoma Humano , Linfoma de Células T/genética , Factores de Transcripción/genética , Transcripción Genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Humanos , Immunoblotting , Linfoma de Células T/metabolismo , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/metabolismo , Transfección
9.
Mol Cell ; 55(6): 904-915, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25219500

RESUMEN

Most colorectal cancers (CRCs) containing activated BRAF (BRAF[V600E]) have a CpG island methylator phenotype (CIMP) characterized by aberrant hypermethylation of many genes, including the mismatch repair gene MLH1. MLH1 silencing results in microsatellite instability and a hypermutable phenotype. Through an RNAi screen, here we identify the transcriptional repressor MAFG as the pivotal factor required for MLH1 silencing and CIMP in CRCs containing BRAF(V600E). In BRAF-positive human CRC cell lines and tumors, MAFG is bound at the promoters of MLH1 and other CIMP genes, and recruits a corepressor complex that includes its heterodimeric partner BACH1, the chromatin remodeling factor CHD8, and the DNA methyltransferase DNMT3B, resulting in hypermethylation and transcriptional silencing. BRAF(V600E) increases BRAF/MEK/ERK signaling resulting in phosphorylation and elevated levels of MAFG, which drives DNA binding. Analysis of transcriptionally silenced CIMP genes in KRAS-positive CRCs indicates that different oncoproteins direct the assembly of distinct repressor complexes on common promoters.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias Colorrectales/genética , Islas de CpG/genética , Factor de Transcripción MafG/metabolismo , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Represoras/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/patología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Ratones Endogámicos BALB C , Homólogo 1 de la Proteína MutL , Mutación , Neoplasias Experimentales , Fenotipo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas B-raf/genética , Transducción de Señal , Factores de Transcripción/metabolismo , ADN Metiltransferasa 3B
10.
Elife ; 3: e02313, 2014 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-24623306

RESUMEN

Approximately 70% of KRAS-positive colorectal cancers (CRCs) have a CpG island methylator phenotype (CIMP) characterized by aberrant DNA hypermethylation and transcriptional silencing of many genes. The factors involved in, and the mechanistic basis of, CIMP is not understood. Among the CIMP genes are the tumor suppressors p14(ARF), p15(INK4B), and p16(INK4A), encoded by the INK4-ARF locus. In this study, we perform an RNA interference screen and identify ZNF304, a zinc-finger DNA-binding protein, as the pivotal factor required for INK4-ARF silencing and CIMP in CRCs containing activated KRAS. In KRAS-positive human CRC cell lines and tumors, ZNF304 is bound at the promoters of INK4-ARF and other CIMP genes. Promoter-bound ZNF304 recruits a corepressor complex that includes the DNA methyltransferase DNMT1, resulting in DNA hypermethylation and transcriptional silencing. KRAS promotes silencing through upregulation of ZNF304, which drives DNA binding. Finally, we show that ZNF304 also directs transcriptional silencing of INK4-ARF in human embryonic stem cells. DOI: http://dx.doi.org/10.7554/eLife.02313.001.


Asunto(s)
Islas de CpG , Metilación de ADN , Silenciador del Gen , Genes ras , Transcripción Genética , Secuencia de Aminoácidos , Animales , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/genética , Humanos , Datos de Secuencia Molecular , Fenotipo , Interferencia de ARN , Homología de Secuencia de Aminoácido , Factores de Transcripción/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Regulación hacia Arriba
11.
Mol Cell Biol ; 33(13): 2635-47, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23648481

RESUMEN

Multiple endocrine neoplasia type 1 is a familial cancer syndrome resulting from loss-of-function mutations in the MEN1 gene. We previously identified the tumor suppressor MEN1 as a gene required for oncogene-induced senescence in melanocytes, raising the possibility that MEN1 is a melanoma tumor suppressor. Here we show that MEN1 expression is lost in a high percentage of human melanomas and melanoma cell lines. We find that melanocytes depleted of MEN1 are deficient in homologous recombination (HR)-directed DNA repair, which is accompanied by increased nonhomologous end-joining activity. Following DNA damage, MEN1 levels increase as a result of phosphorylation by the DNA damage kinase ATM/ATR. Most importantly, we show that MEN1 functions by directly stimulating the transcription of several genes, including BRCA1, RAD51, and RAD51AP1, that encode proteins involved in HR. MEN1 and its coactivator, the mixed-lineage leukemia histone methyltransferase, are recruited to the BRCA1, RAD51, and RAD51AP1 promoters by estrogen receptor 1, resulting in increased histone H3-lysine 4 trimethylation and transcription. Collectively, our results indicate that MEN1 is a melanoma tumor suppressor that functions by stimulating the transcription of genes involved in HR-directed DNA repair.


Asunto(s)
Melanoma/genética , Proteínas Proto-Oncogénicas/metabolismo , Reparación del ADN por Recombinación/genética , Proteínas de la Ataxia Telangiectasia Mutada , Proteína BRCA1/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Reparación del ADN por Unión de Extremidades/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Regulación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina , Humanos , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas de Unión al ARN , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
12.
Elife ; 1: e00068, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23150797

RESUMEN

The general transcription factor TFIID comprises the TATA-box-binding protein (TBP) and approximately 14 TBP-associated factors (TAFs). Here we find, unexpectedly, that undifferentiated human embryonic stem cells (hESCs) contain only six TAFs (TAFs 2, 3, 5, 6, 7 and 11), whereas following differentiation all TAFs are expressed. Directed and global chromatin immunoprecipitation analyses reveal an unprecedented promoter occupancy pattern: most active genes are bound by only TAFs 3 and 5 along with TBP, whereas the remaining active genes are bound by TBP and all six hESC TAFs. Consistent with these results, hESCs contain a previously undescribed complex comprising TAFs 2, 6, 7, 11 and TBP. Altering the composition of hESC TAFs, either by depleting TAFs that are present or ectopically expressing TAFs that are absent, results in misregulated expression of pluripotency genes and induction of differentiation. Thus, the selective expression and use of TAFs underlies the ability of hESCs to self-renew.DOI:http://dx.doi.org/10.7554/eLife.00068.001.


Asunto(s)
Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores Asociados con la Proteína de Unión a TATA/genética , Proteína de Unión a TATA-Box/genética , Transcripción Genética , Diferenciación Celular , Cromatina/química , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Células Madre Embrionarias/citología , Células HeLa , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Multimerización de Proteína , Transducción de Señal , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Proteína de Unión a TATA-Box/metabolismo
13.
J Virol ; 86(9): 4981-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22379094

RESUMEN

Baculovirus occlusion-derived virus (ODV) infects insect midgut cells under alkaline conditions, a process mediated by highly conserved per os infectivity factors (PIFs), P74 (PIF0), PIF1, PIF2, PIF3, PIF4, and PIF5 (ODV-E56). Previously, a multimolecular complex composed of PIF1, PIF2, PIF3, and P74 was identified which was proposed to play an essential role during ODV entry. Recently, more proteins have been identified that play important roles in ODV oral infectivity, including PIF4, PIF5, and SF58, which might work in concert with previously known PIFs to facilitate ODV infection. In order to understand the ODV entry mechanism, the identification of all components of the PIF complex is crucial. Hence, the aim of this study was to identify additional components of the PIF complex. Coimmunoprecipitation (CoIP) combined with proteomic analysis was used to identify the components of the Autographa californica multiple nucleopolyhedrovirus (AcMNPV) PIF complex. PIF4 and P95 (AC83) were identified as components of the PIF complex while PIF5 was not, and this was confirmed with blue native PAGE and a second CoIP. Deletion of the pif4 gene impaired complex formation, but deletion of pif5 did not. Differentially denaturing SDS-PAGE further revealed that PIF4 forms a stable complex with PIF1, PIF2, and PIF3. P95 and P74 are more loosely associated with this complex. Three other proteins, AC5, AC68, and AC108 (homologue of SF58), were also found by the proteomic analysis to be associated with the PIF complex. Finally the functional significance of the PIF protein interactions is discussed.


Asunto(s)
Nucleopoliedrovirus/metabolismo , Proteínas Virales/metabolismo , Eliminación de Gen , Orden Génico , Espectrometría de Masas , Nucleopoliedrovirus/genética , Unión Proteica , Estabilidad Proteica , Proteínas Virales/química , Proteínas Virales/genética
14.
J Virol ; 86(7): 3985-94, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22278232

RESUMEN

Autographa californica multiple nucleopolyhedrovirus ac68 is a core gene that overlaps lef3 which encodes the single-stranded DNA binding protein. A knockout (KO) virus lacking both lef3 and ac68 was generated (lef3-ac68 2×KO) to enable the functional study of ac68. To produce an ac68KO virus that did not impact lef3 expression, the lef3-ac68 2×KO virus was repaired with a DNA fragment containing lef3 and ac68, in which ac68 contained point mutations so that only LEF3 was expressed. Repair of lef3-ac68 2×KO with just ac68 generated an lef3KO virus. Analysis of the ac68KO virus showed that viral DNA replication and budded virus (BV) levels were unaffected compared to levels in the double-repair or wild-type (WT) control virus. Bioassay analyses of Trichoplusia ni larvae injected with BV directly into the hemolymph, bypassing the gut, showed no difference in mortality rates between the ac68KO and the WT viruses. However, in oral bioassays the ac68KO occlusion bodies failed to kill larvae. These results show that the core gene ac68 encodes a per os infectivity factor (pif6). The lef3KO virus was also analyzed, and virus replication was drastically reduced compared to WT virus, but very low levels of lef3KO virus DNA replication and BV production could be detected. In addition, in transfected cells P143 was transported to the nucleus in the absence of LEF3. This study therefore shows for the first time that even though the loss of LEF3 severely impairs virus replication, it is not absolutely essential for P143 nuclear import or viral replication.


Asunto(s)
Genes Sobrepuestos , Nucleopoliedrovirus/fisiología , Proteínas Virales/metabolismo , Replicación Viral , Secuencia de Aminoácidos , Animales , Línea Celular , Núcleo Celular/metabolismo , Núcleo Celular/virología , Datos de Secuencia Molecular , Mariposas Nocturnas/metabolismo , Mariposas Nocturnas/virología , Nucleopoliedrovirus/genética , Nucleopoliedrovirus/patogenicidad , Transporte de Proteínas , Proteínas Virales/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
15.
Virology ; 409(1): 38-45, 2011 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-20965540

RESUMEN

Autographa californica multiple nucleopolyhedrovirus (AcMNPV) ac92 is a core gene encoding a protein associated with occlusion derived virus (ODV), binds human P53 and also has flavin adenine dinucleotide linked sulfhydryl oxidase activity but its role in the virus life cycle is not known. To determine ac92 function a deletion virus (vAc(92KO)) was generated and transfected Sf9 cells revealed that vAc(92KO) infection was restricted primarily to single cells and budded virus (BV) titer was reduced over 99.99%. However, viral DNA replication was unaffected and development of occlusion bodies in vAc(92KO)-transfected cells evidenced progression to very late phases of viral infection. AC92 localized to both the cytoplasm and nucleus, and was also associated with BV as well as ODV. In BV AC92 was detected in BV envelope and nucleocapsid fractions. Finally it was shown that the ac92 homologue from the Group II alphabaculovirus Mamestra configurata NPV maco96 could only partially rescue vAc(92KO).


Asunto(s)
Genes Virales , Mariposas Nocturnas/virología , Nucleopoliedrovirus/fisiología , Proteínas Virales/genética , Liberación del Virus/fisiología , Replicación Viral/fisiología , Animales , Núcleo Celular/metabolismo , Núcleo Celular/virología , Células Cultivadas , Citoplasma/metabolismo , Citoplasma/virología , Replicación del ADN , Genes Esenciales , Cuerpos de Inclusión Viral/virología , Nucleopoliedrovirus/genética , Nucleopoliedrovirus/metabolismo , Eliminación de Secuencia , Spodoptera/virología , Proteínas Virales/metabolismo
16.
J Virol ; 83(23): 12569-78, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19759145

RESUMEN

Autographa californica multiple nucleopolyhedrovirus (AcMNPV) ac96 is a core gene, but its role in virus replication is still unknown. To determine its role in the baculovirus life cycle, we used the AcMNPV bacmid system to generate an ac96-null virus (vAc(96)(null)). Our analyses showed that the absence of ac96 does not affect budded virus (BV) production or viral DNA replication in infected Sf9 cells. Western blotting and confocal immunofluorescence analysis showed that AC96 is expressed in both the cytoplasm and the nucleus throughout infection. In addition, AC96 was detected in the envelope fractions of both BV and occlusion-derived virus. Injection of vAc(96)(null) BV into the hemocoel killed Trichoplusia ni larvae as efficiently as repaired and control viruses; however, vAc(96)(null) was unable to infect the midgut tissue of Trichoplusia ni larvae when inoculated per os. Therefore, the results of this study show that ac96 encodes a new per os infectivity factor (PIF-4).


Asunto(s)
Infecciones por Virus ADN/veterinaria , Lepidópteros/virología , Nucleopoliedrovirus/patogenicidad , Proteínas Estructurales Virales/fisiología , Replicación Viral , Animales , Línea Celular , Tracto Gastrointestinal/virología , Eliminación de Gen , Spodoptera , Análisis de Supervivencia , Proteínas Estructurales Virales/genética , Virulencia
17.
Virology ; 389(1-2): 66-74, 2009 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-19411088

RESUMEN

Autographa californica multiple nucleopolyhedrovirus (AcMNPV) ac109 is a core gene and its function in the virus life cycle is unknown. To determine its role in the baculovirus life cycle, we used the AcMNPV bacmid system to generate an ac109 deletion virus (vAc(109KO)). Fluorescence and light microscopy showed that transfection of vAc(109KO) results in a single-cell infection phenotype. Viral DNA replication is unaffected and the development of occlusion bodies in vAc(109KO)-transfected cells evidenced progression to the very late phases of viral infection. Western blot and confocal immunofluorescence analysis showed that AC109 is expressed in the cytoplasm and nucleus throughout infection. In addition, AC109 is a structural protein as it was detected in both budded virus (BV) and occlusion derived virus in both the envelope and nucleocapsid fractions. Titration assays by qPCR and TCID(50) showed that vAc(109KO) produced BV but the virions are non-infectious. The vAc(109KO) BV were indistinguishable from the BV of repaired and wild type control viruses as determined by negative staining and electron microscopy.


Asunto(s)
Eliminación de Gen , Nucleopoliedrovirus/genética , Spodoptera/virología , Proteínas del Núcleo Viral/genética , Virión/genética , Secuencia de Aminoácidos , Animales , Línea Celular , ADN Viral/biosíntesis , Datos de Secuencia Molecular , Nucleopoliedrovirus/fisiología , Replicación Viral
18.
Virology ; 385(2): 496-504, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19155039

RESUMEN

The Autographa californica multiple nucleopolyhedrovirus (AcMNPV) encoded protein, EXON0 (AC141), is required for the efficient transport of nucleocapsids out of the nucleus for the production of budded virus (BV). To further elucidate the molecular mechanisms by which EXON0 regulates BV production, EXON0 was tagged at the N-terminus with 3x FLAG-6x His. Protein complexes were isolated by tandem affinity purification and potential EXON0 specific interacting protein partners were gel purified and identified by LC-MS/MS. This analysis showed that the cellular protein, beta-tubulin, co-purified with EXON0 which was confirmed by co-immunoprecipitation. In addition, immunofluorescence showed that EXON0 and beta-tubulin co-localized during virus infection. The microtubule inhibitors colchicine and nocodazole were used to treat AcMNPV infected Sf9 cells and results showed that BV production was reduced by over 85%. These data suggest that the egress of AcMNPV budded virus may be facilitated by the interaction of EXON0 with beta-tubulin and microtubules.


Asunto(s)
Nucleocápside/metabolismo , Nucleopoliedrovirus/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas Virales/metabolismo , Animales , Colchicina/farmacología , Inmunoprecipitación , Microtúbulos/metabolismo , Nocodazol/farmacología , Nucleopoliedrovirus/efectos de los fármacos , Nucleopoliedrovirus/genética , Sistemas de Lectura Abierta/genética , Spodoptera/citología , Spodoptera/virología , Tubulina (Proteína)/genética , Moduladores de Tubulina/farmacología , Proteínas Virales/genética
19.
Virology ; 385(2): 484-95, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19150105

RESUMEN

IE0 and IE1 are the primary viral regulatory proteins of Autographa californica multiple nucleopolyhedrovirus (AcMNPV) involved in the transactivation of early genes, stimulation of late gene expression, and viral DNA replication. The protein interactions required for IE0 or IE1 to achieve these varied roles are not well defined, so to identify proteins that interact with IE0 and IE1, tandem affinity purification (TAP) and LC-MS/MS was used. Analysis of purified proteins identified AC16 (DA26, BV/ODV-E26) from TAP tagged IE0 virus infected Sf9 cells. Co-immunoprecipitation confirmed that AC16 interacts with both IE0 and IE1 and yeast 2-hybrid analysis mapped the domain required for interaction with AC16. Mutation of the AC16 binding domain enhanced BV production by viruses expressing only IE0 but had no effect if only IE1 is expressed. An ac16 deletion virus was constructed and was shown not to affect the temporal expression of IE0 and IE1; however the relative level of IE0 to IE1 was significantly increased.


Asunto(s)
Regulación Viral de la Expresión Génica/genética , Proteínas Inmediatas-Precoces/genética , Nucleopoliedrovirus , Transactivadores/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Eliminación de Gen , Vectores Genéticos , Proteínas Inmediatas-Precoces/química , Proteínas Inmediatas-Precoces/metabolismo , Inmunoprecipitación , Datos de Secuencia Molecular , Nucleopoliedrovirus/genética , Nucleopoliedrovirus/metabolismo , Alineación de Secuencia , Spodoptera/virología , Transactivadores/química , Técnicas del Sistema de Dos Híbridos , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/genética , Replicación Viral/genética
20.
Virology ; 375(1): 265-76, 2008 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-18313716

RESUMEN

The Autographa californica multiple nucleopolyhedrovirus (AcMNPV) late gene exon0 (ac141) is required for the efficient production of budded virus (BV). EXON0 interacts with nucleopcapsid protein BV/ODV-C42 and FP25 and enables egress of nucleocapsids from the nucleus to the cytoplasm. This study examines the functional domains of EXON0 that play a role in BV production. Six putative domains of the 261 amino acid EXON0 were deleted and examined for functionality by determining their ability to rescue an AcMNPV exon0 knockout bacmid. Domain mapping results showed that all the six domains were required but deletion of the N-terminal acidic region and the leucine zipper domains had the greatest impact on BV production. Yeast 2-hybrid and co-immunoprecipitation demonstrated that EXON0 formed dimers. Point mutation analysis demonstrated that the leucine zipper was required for dimer formation and interaction with BV/ODV-C42 and FP25. The charged domain was also required for BV/ODV-C42 interaction.


Asunto(s)
Nucleopoliedrovirus/fisiología , Dominios y Motivos de Interacción de Proteínas/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral , Secuencia de Aminoácidos , Animales , Western Blotting , Línea Celular , Dimerización , Prueba de Complementación Genética , Inmunoprecipitación , Datos de Secuencia Molecular , Proteínas de la Nucleocápside/metabolismo , Mutación Puntual , Alineación de Secuencia , Eliminación de Secuencia , Spodoptera , Técnicas del Sistema de Dos Híbridos , Ensayo de Placa Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...