Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(1): 113651, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38175751

RESUMEN

Dynamic chromosome remodeling and nuclear compartmentalization take place during mammalian meiotic prophase I. We report here that the crucial roles of male pachynema-specific protein (MAPS) in pachynema progression might be mediated by its liquid-liquid phase separation in vitro and in cellulo. MAPS forms distinguishable liquid phases, and deletion or mutations of its N-terminal amino acids (aa) 2-9 disrupt its secondary structure and charge properties, impeding phase separation. Maps-/- pachytene spermatocytes exhibit defects in nucleus compartmentalization, including defects in forming sex bodies, altered nucleosome composition, and disordered chromatin accessibility. MapsΔ2-9/Δ2-9 male mice expressing MAPS protein lacking aa 2-9 phenocopy Maps-/- mice. Moreover, a frameshift mutation in C3orf62, the human counterpart of Maps, is correlated with nonobstructive azoospermia in a patient exhibiting pachynema arrest in spermatocyte development. Hence, the phase separation property of MAPS seems essential for pachynema progression in mouse and human spermatocytes.


Asunto(s)
Cromatina , Meiosis , Humanos , Masculino , Ratones , Animales , Cromatina/metabolismo , Fase Paquiteno , Separación de Fases , Profase Meiótica I , Espermatocitos/metabolismo , Mamíferos/genética
2.
Mol Cell ; 84(2): 202-220.e15, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38103559

RESUMEN

Compounds binding to the bromodomains of bromodomain and extra-terminal (BET) family proteins, particularly BRD4, are promising anticancer agents. Nevertheless, side effects and drug resistance pose significant obstacles in BET-based therapeutics development. Using high-throughput screening of a 200,000-compound library, we identified small molecules targeting a phosphorylated intrinsically disordered region (IDR) of BRD4 that inhibit phospho-BRD4 (pBRD4)-dependent human papillomavirus (HPV) genome replication in HPV-containing keratinocytes. Proteomic profiling identified two DNA damage response factors-53BP1 and BARD1-crucial for differentiation-associated HPV genome amplification. pBRD4-mediated recruitment of 53BP1 and BARD1 to the HPV origin of replication occurs in a spatiotemporal and BRD4 long (BRD4-L) and short (BRD4-S) isoform-specific manner. This recruitment is disrupted by phospho-IDR-targeting compounds with little perturbation of the global transcriptome and BRD4 chromatin landscape. The discovery of these protein-protein interaction inhibitors (PPIi) not only demonstrates the feasibility of developing PPIi against phospho-IDRs but also uncovers antiviral agents targeting an epigenetic regulator essential for virus-host interaction and cancer development.


Asunto(s)
Infecciones por Papillomavirus , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Virus del Papiloma Humano , Infecciones por Papillomavirus/tratamiento farmacológico , Infecciones por Papillomavirus/genética , Proteómica , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Papillomaviridae/genética , Papillomaviridae/metabolismo , Proteínas Virales/genética , Replicación Viral/fisiología , Reparación del ADN , Proteínas que Contienen Bromodominio
3.
Methods Mol Biol ; 2466: 61-82, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35585311

RESUMEN

We have developed the CL7/Im7 protein purification system to achieve high-yield, high-purity and high-activity (HHH) products in one step. The system is based on the natural ultrahigh-affinity complex between the two small proteins encoded by colicinogenic plasmids carried by certain E. coli strains, the DNAse domain of colicin E7 (CE7; MW ~ 15 kDa) and its natural endogenous inhibitor, the immunity protein 7 (Im7; MW ~ 10 kDa). CL7 is an engineered variant of CE7, in which the toxic DNA-binding and catalytic activities have been eliminated while retaining the high affinity to Im7. CL7 is used as a protein tag, while Im7 is covalently attached to agarose beads. To make the CL7/Im7 technique easy to use, we have designed a set of the E. coli expression vectors for fusion of a target protein to the protease-cleavable CL7-tag either at the N- or the C-terminus, and also have the options of the dual (CL7/His8) tag. A subset of vectors is dedicated for cloning membrane and multisubunit proteins. The CL7/Im7 system has several notable advatantages over other available affinity purification techniques. First, high concentrations of the small Im7 protein are coupled to the beads resulting in the high column capacities (up to 60 mg/mL). Second, an exceptional stability of Im7 allows for multiple (100+) regeneration cycles with no loss of binding capacities. Third, the CL7-tag improves protein expression levels, solubility and, in some cases, assists folding of the target proteins. Fourth, the on-column proteolytic elution produces purified proteins with few or no extra amino acid residues. Finally, the CL7/Im7 affinity is largely insensitive to high salt concentrations. For many target proteins, loading the bacterial lysates on the Im7 column in high salt is a key to high purity. Altogether, these properties of the CL7/Im7 system allow for a one-step HHH purification of most challenging, biologically and clinically significant proteins.


Asunto(s)
Colicinas , Proteínas de Escherichia coli , Cromatografía de Afinidad/métodos , Colicinas/química , Colicinas/genética , Colicinas/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Péptido Hidrolasas/metabolismo , Preparaciones Farmacéuticas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33602822

RESUMEN

Meiosis is a specialized cell division that creates haploid germ cells from diploid progenitors. Through differential RNA expression analyses, we previously identified a number of mouse genes that were dramatically elevated in spermatocytes, relative to their very low expression in spermatogonia and somatic organs. Here, we investigated in detail 1700102P08Rik, one of these genes, and independently conclude that it encodes a male germline-specific protein, in agreement with a recent report. We demonstrated that it is essential for pachynema progression in spermatocytes and named it male pachynema-specific (MAPS) protein. Mice lacking Maps (Maps-/- ) suffered from pachytene arrest and spermatocyte death, leading to male infertility, whereas female fertility was not affected. Interestingly, pubertal Maps-/- spermatocytes were arrested at early pachytene stage, accompanied by defects in DNA double-strand break (DSB) repair, crossover formation, and XY body formation. In contrast, adult Maps-/- spermatocytes only exhibited partially defective crossover but nonetheless were delayed or failed in progression from early to mid- and late pachytene stage, resulting in cell death. Furthermore, we report a significant transcriptional dysregulation in autosomes and XY chromosomes in both pubertal and adult Maps-/- pachytene spermatocytes, including failed meiotic sex chromosome inactivation (MSCI). Further experiments revealed that MAPS overexpression in vitro dramatically decreased the ubiquitination levels of cellular proteins. Conversely, in Maps-/- pachytene cells, protein ubiquitination was dramatically increased, likely contributing to the large-scale disruption in gene expression in pachytene cells. Thus, MAPS is a protein essential for pachynema progression in male mice, possibly in mammals in general.


Asunto(s)
Infertilidad Masculina/patología , Meiosis , Proteínas Nucleares/fisiología , Fase Paquiteno , Espermatocitos/patología , Espermatogénesis , Animales , Emparejamiento Cromosómico , Reparación del ADN , Femenino , Infertilidad Masculina/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cromosomas Sexuales , Espermatocitos/metabolismo
5.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33436409

RESUMEN

Long noncoding RNAs (lncRNAs) play diverse roles in biological processes, but their expression profiles and functions in cervical carcinogenesis remain unknown. By RNA-sequencing (RNA-seq) analyses of 18 clinical specimens and selective validation by RT-qPCR analyses of 72 clinical samples, we provide evidence that, relative to normal cervical tissues, 194 lncRNAs are differentially regulated in high-risk (HR)-HPV infection along with cervical lesion progression. One such lncRNA, lnc-FANCI-2, is extensively characterized because it is expressed from a genomic locus adjacent to the FANCI gene encoding an important DNA repair factor. Both genes are up-regulated in HPV lesions and in in vitro model systems of HR-HPV18 infection. We observe a moderate reciprocal regulation of lnc-FANCI-2 and FANCI in cervical cancer CaSki cells. In these cells, lnc-FANCI-2 is transcribed from two alternative promoters, alternatively spliced, and polyadenylated at one of two alternative poly(A) sites. About 10 copies of lnc-FANCI-2 per cell are detected preferentially in the cytoplasm. Mechanistically, HR-HPVs, but not low-risk (LR)-HPV oncogenes induce lnc-FANCI-2 in primary and immortalized human keratinocytes. The induction is mediated primarily by E7, and to a lesser extent by E6, mostly independent of p53/E6AP and pRb/E2F. We show that YY1 interacts with an E7 CR3 core motif and transactivates the promoter of lnc-FANCI-2 by binding to two critical YY1-binding motifs. Moreover, HPV18 increases YY1 expression by reducing miR-29a, which targets the 3' untranslated region of YY1 mRNA. These data have provided insights into the mechanisms of how HR-HPV infections contribute to cervical carcinogenesis.


Asunto(s)
Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Papillomavirus Humano 16/genética , MicroARNs/genética , Infecciones por Papillomavirus/genética , ARN Largo no Codificante/genética , Neoplasias del Cuello Uterino/genética , Factor de Transcripción YY1/genética , Empalme Alternativo , Secuencia de Bases , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Cuello del Útero/metabolismo , Cuello del Útero/patología , Cuello del Útero/virología , Factores de Transcripción E2F/genética , Factores de Transcripción E2F/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Femenino , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 16/patogenicidad , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/metabolismo , Papillomavirus Humano 18/patogenicidad , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Queratinocitos/virología , MicroARNs/metabolismo , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Regiones Promotoras Genéticas , ARN Largo no Codificante/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/virología , Factor de Transcripción YY1/metabolismo
6.
Proc Natl Acad Sci U S A ; 116(49): 24583-24592, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31732667

RESUMEN

The mammalian target of rapamycin (mTOR) pathway, which plays a critical role in regulating cellular growth and metabolism, is aberrantly regulated in the pathogenesis of a variety of neoplasms. Here we demonstrate that dual mTORC1/mTORC2 inhibitors OSI-027 and PP242 cause catastrophic macropinocytosis in rhabdomyosarcoma (RMS) cells and cancers of the skin, breast, lung, and cervix, whereas the effects are much less pronounced in immortalized human keratinocytes. Using RMS as a model, we characterize in detail the mechanism of macropinocytosis induction. Macropinosomes are distinct from endocytic vesicles and autophagosomes in that they are single-membrane bound vacuoles formed by projection, ruffling, and contraction of plasma membranes. They are positive for EEA-1 and LAMP-1 and contain watery fluid but not organelles. The vacuoles then merge and rupture, killing the cells. We confirmed the inhibition of mTORC1/mTORC2 as the underpinning mechanism for macropinocytosis. Exposure to rapamycin, an mTORC1 inhibitor, or mTORC2 knockdown alone had little or reduced effect relative to the combination. We further demonstrate that macropinocytosis depends on MKK4 activated by elevated reactive oxygen species. In a murine xenograft model, OSI-027 reduced RMS tumor growth. Molecular characterization of the residual tumors was consistent with the induction of macropinocytosis. Furthermore, relative to the control xenograft tumors, the residual tumors manifested reduced expression of cell proliferation markers and proteins that drive the epithelial mesenchymal transition. These data indicate a role of mTORC2 in regulating tumor growth by macropinocytosis and suggest that dual inhibitors could help block refractory or recurrent RMS and perhaps other neoplasms and other cancer as well.


Asunto(s)
Antineoplásicos/farmacología , Imidazoles/farmacología , Indoles/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Diana Mecanicista del Complejo 2 de la Rapamicina/antagonistas & inhibidores , Pinocitosis/efectos de los fármacos , Purinas/farmacología , Triazinas/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Ciclofosfamida/administración & dosificación , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Imidazoles/administración & dosificación , MAP Quinasa Quinasa 4/metabolismo , Ratones Desnudos , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Triazinas/administración & dosificación , Vacuolas/efectos de los fármacos , Vacuolas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Int J Mol Sci ; 20(21)2019 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-31683862

RESUMEN

Mucosotropic human papillomaviruses (HPVs) cause prevalent anogenital infections, some of which can progress to cancers. It is imperative to identify efficacious drug candidates, as there are few therapeutic options. We have recapitulated a robust productive program of HPV-18 in organotypic raft cultures of primary human keratinocytes. The HPV E7 protein induces S phase reentry, along with DNA damage response (DDR) in differentiated cells to support viral DNA amplification. A number of small molecule inhibitors of DDR regulators are in clinical use or clinical trials to treat cancers. Here, we used our raft culture system to examine effects of inhibitors of ATR/Chk1 and ATM/Chk2 on HPV infection. The inhibitors impaired S-phase reentry and progression as well as HPV DNA amplification. The Chk1 inhibitor MK-8776 was most effective, reducing viral DNA amplification by 90-99% and caused DNA damage and apoptosis, preferentially in HPV infected cells. We found that this sensitivity was imparted by the E7 protein and report that MK-8776 also caused extensive cell death of cervical cancer cell lines. Furthermore, it sensitized the cells to cisplatin, commonly used to treat advanced cervical cancer. Based on these observations, the Chk1 inhibitors could be potential effective agents to be re-purposed to treat the spectrum of HPV infections in single or combination therapy.


Asunto(s)
ADN Viral/antagonistas & inhibidores , Papillomavirus Humano 18/metabolismo , Queratinocitos/efectos de los fármacos , Proteínas E7 de Papillomavirus/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Replicación Viral/efectos de los fármacos , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Células Cultivadas , Cisplatino/farmacología , ADN Viral/genética , ADN Viral/metabolismo , Femenino , Células HeLa , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/fisiología , Humanos , Queratinocitos/metabolismo , Queratinocitos/virología , Masculino , Proteínas E7 de Papillomavirus/genética , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/virología , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/virología , Replicación Viral/genética
9.
mBio ; 10(1)2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696738

RESUMEN

RNA-binding proteins (RBPs) control mRNA processing, stability, transport, editing, and translation. We recently conducted transcriptome analyses comparing normal (i.e., healthy) cervical tissue samples with human papillomavirus (HPV)-positive cervical cancer tissue samples and identified 614 differentially expressed protein-coding transcripts which are enriched in cancer-related pathways and consist of 95 known RBPs. We verified the altered expression of 26 genes with a cohort of 72 cervical samples, including 24 normal cervical samples, 25 cervical intraepithelial neoplasia grade 2 (CIN2) and CIN3 samples, and 23 cervical cancer tissue samples. LY6K (lymphocyte antigen 6 complex locus K), FAM83A (family member with sequence similarity 83), CELSR3, ASF1B, IQGAP3, SEMA3F, CLDN10, MSX1, CXCL5, ASRGL1, ELAVL2, GRB7, KHSRP, NOVA1, PTBP1, and RNASEH2A were identified as novel candidate genes associated with cervical lesion progression and carcinogenesis. HPV16 or HPV18 infection was found to alter the expression of 8 RBP genes (CDKN2A, ELAVL2, GRB7, HSPB1, KHSRP, NOVA1, PTBP1, and RNASEH2A) in human vaginal and foreskin keratinocytes. Both viral E6 and E7 decreased NOVA1 expression, but only E7 increased the expression of RNASEH2A in an E2F1-dependent manner. Proliferating cell nuclear antigen (PCNA) directs RNASEH2 activity with respect to DNA replication by removing the RNA primers to promote Okazaki fragment maturation, and two factors are closely associated with neoplasia progression. Therefore, we predict that the induction of expression of RNASEH2A via viral E7 and E2F1 may promote DNA replication and cancer cell proliferation.IMPORTANCE High-risk HPV infections lead to development of cervical cancer. This study identified the differential expression of 16 novel genes (LY6K, FAM83A, CELSR3, ASF1B, IQGAP3, SEMA3F, CLDN10, MSX1, CXCL5, ASRGL1, ELAVL2, GRB7, KHSRP, NOVA1, PTBP1, and RNASEH2A) in HPV-infected cervical tissue samples and keratinocytes. Eight of these genes (CDKN2A, ELAVL2, GRB7, HSPB1, KHSRP, NOVA1, PTBP1, and RNASEH2A) encode RNA-binding proteins. Further studies indicated that both HPV16 and HPV18 infections lead to the aberrant expression of selected RBP-encoding genes. We found that viral E6 and E7 decrease NOVA1 expression but that E7 increases RNASEH2A expression via E2F1. The altered expression of these genes may be utilized as biomarkers for high-risk (HR)-HPV carcinogenesis and progression.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , Interacciones Huésped-Patógeno , Papillomavirus Humano 16/fisiología , Papillomavirus Humano 18/fisiología , Proteínas E7 de Papillomavirus/metabolismo , Ribonucleasa H/metabolismo , Neoplasias del Cuello Uterino/patología , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Queratinocitos/patología , Queratinocitos/virología , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología
10.
Proc Natl Acad Sci U S A ; 115(47): E11138-E11147, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30385631

RESUMEN

Human papillomaviruses (HPVs) cause epithelial proliferative diseases. Persistent infection of the mucosal epithelia by the high-risk genotypes can progress to high-grade dysplasia and cancers. Viral transcription and protein activities are intimately linked to regulation by histone acetyltransferases and histone deacetylases (HDACs) that remodel chromatin and regulate gene expression. HDACs are also essential to remodel and repair replicating chromatin to enable the progression of replication forks. As such, Vorinostat (suberoylanilide hydroximic acid), and other pan-HDAC inhibitors, are used to treat lymphomas. Here, we investigated the effects of Vorinostat on productive infection of the high-risk HPV-18 in organotypic cultures of primary human keratinocytes. HPV DNA amplifies in the postmitotic, differentiated cells of squamous epithelia, in which the viral oncoproteins E7 and E6 establish a permissive milieu by destabilizing major tumor suppressors, the pRB family proteins and p53, respectively. We showed that Vorinostat significantly reduced these E6 and E7 activities, abrogated viral DNA amplification, and inhibited host DNA replication. The E7-induced DNA damage response, which is critical for both events, was also compromised. Consequently, Vorinostat exposure led to DNA damage and triggered apoptosis in HPV-infected, differentiated cells, whereas uninfected tissues were spared. Apoptosis was attributed to highly elevated proapoptotic Bim isoforms that are known to be repressed by EZH2 in a repressor complex containing HDACs. Two other HDAC inhibitors, Belinostat and Panobinostat, also inhibited viral DNA amplification and cause apoptosis. We suggest that HDAC inhibitors are promising therapeutic agents to treat benign HPV infections, abrogate progeny virus production, and hence interrupt transmission.


Asunto(s)
Replicación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Papillomavirus Humano 18/genética , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/tratamiento farmacológico , Vorinostat/farmacología , Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2/metabolismo , Células Cultivadas , Reparación del ADN/efectos de los fármacos , ADN Viral/genética , Histonas/metabolismo , Papillomavirus Humano 18/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/farmacología , Queratinocitos/virología , Membrana Mucosa/virología , Panobinostat/farmacología , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/transmisión , Sulfonamidas/farmacología , Proteína p53 Supresora de Tumor/metabolismo
12.
Proc Natl Acad Sci U S A ; 114(38): E7949-E7958, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28855339

RESUMEN

Posttranslational histone modifications play important roles in regulating chromatin-based nuclear processes. Histone H2AK119 ubiquitination (H2Aub) is a prevalent modification and has been primarily linked to gene silencing. However, the underlying mechanism remains largely obscure. Here we report the identification of RSF1 (remodeling and spacing factor 1), a subunit of the RSF complex, as a H2Aub binding protein, which mediates the gene-silencing function of this histone modification. RSF1 associates specifically with H2Aub, but not H2Bub nucleosomes, through a previously uncharacterized and obligatory region designated as ubiquitinated H2A binding domain. In human and mouse cells, genes regulated by RSF1 overlap significantly with those controlled by RNF2/Ring1B, the subunit of Polycomb repressive complex 1 (PRC1) which catalyzes the ubiquitination of H2AK119. About 82% of H2Aub-enriched genes, including the classic PRC1 target Hox genes, are bound by RSF1 around their transcription start sites. Depletion of H2Aub levels by Ring1B knockout results in a significant reduction of RSF1 binding. In contrast, RSF1 knockout does not affect RNF2/Ring1B or H2Aub levels but leads to derepression of H2Aub target genes, accompanied by changes in H2Aub chromatin organization and release of linker histone H1. The action of RSF1 in H2Aub-mediated gene silencing is further demonstrated by chromatin-based in vitro transcription. Finally, RSF1 and Ring1 act cooperatively to regulate mesodermal cell specification and gastrulation during Xenopus early embryonic development. Taken together, these data identify RSF1 as a H2Aub reader that contributes to H2Aub-mediated gene silencing by maintaining a stable nucleosome pattern at promoter regions.


Asunto(s)
Silenciador del Gen/fisiología , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Nucleosomas/metabolismo , Transactivadores/metabolismo , Ubiquitinación/fisiología , Animales , Células HeLa , Histonas/genética , Humanos , Ratones , Proteínas Nucleares/genética , Nucleosomas/genética , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Regiones Promotoras Genéticas/fisiología , Transactivadores/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
13.
Proc Natl Acad Sci U S A ; 114(26): E5138-E5147, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28607052

RESUMEN

Protein purification is an essential primary step in numerous biological studies. It is particularly significant for the rapidly emerging high-throughput fields, such as proteomics, interactomics, and drug discovery. Moreover, purifications for structural and industrial applications should meet the requirement of high yield, high purity, and high activity (HHH). It is, therefore, highly desirable to have an efficient purification system with a potential to meet the HHH benchmark in a single step. Here, we report a chromatographic technology based on the ultra-high-affinity (Kd ∼ 10-14-10-17 M) complex between the Colicin E7 DNase (CE7) and its inhibitor, Immunity protein 7 (Im7). For this application, we mutated CE7 to create a CL7 tag, which retained the full binding affinity to Im7 but was inactivated as a DNase. To achieve high capacity, we developed a protocol for a large-scale production and highly specific immobilization of Im7 to a solid support. We demonstrated its utility with one-step HHH purification of a wide range of traditionally challenging biological molecules, including eukaryotic, membrane, toxic, and multisubunit DNA/RNA-binding proteins. The system is simple, reusable, and also applicable to pulldown and kinetic activity/binding assays.


Asunto(s)
Proteínas Portadoras/química , Cromatografía de Afinidad/métodos , Colicinas/química , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/aislamiento & purificación , Proteínas de Escherichia coli/química , Escherichia coli/química
14.
J Med Chem ; 59(23): 10470-10478, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27933957

RESUMEN

Human papillomavirus (HPV) high-risk genotypes such as HPV-16 and HPV-18 cause the majority of anogenital tract carcinomas, including cervical cancer, the second most common malignancy in women worldwide. Currently there are no approved antiviral agents that reduce or eliminate HPV and reverse virus-associated pathology. We synthesized and evaluated several alkoxyalkyl acyclic nucleoside phosphonate diesters and identified octadecyloxyethyl benzyl 9-[(2-phosphonomethoxy)ethyl]guanine (ODE-Bn-PMEG) as an active compound which strongly inhibited transient amplification of HPV-11, -16, and -18 origin-containing plasmid DNA in transfected cells at concentrations well below its cytotoxic concentrations. ODE-Bn-PMEG demonstrated increased uptake in human foreskin fibroblast cells and was readily converted in vitro to the active antiviral metabolite, PMEG diphosphate. The P-chiral enantiomers of ODE-Bn-PMEG were obtained and appeared to have equivalent antiviral activities against HPV. ODE-Bn-PMEG is a promising candidate for the local treatment of HPV-16 and HPV-18 and other high-risk types, an important unmet medical need.


Asunto(s)
Antivirales/farmacología , ADN Viral/efectos de los fármacos , Guanina/análogos & derivados , Técnicas de Amplificación de Ácido Nucleico , Organofosfonatos/farmacología , Papillomaviridae/efectos de los fármacos , Antivirales/síntesis química , Antivirales/química , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Guanina/síntesis química , Guanina/química , Guanina/farmacología , Células HEK293 , VIH/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/virología , Estructura Molecular , Organofosfonatos/síntesis química , Organofosfonatos/química , Papillomaviridae/genética , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
15.
Proc Natl Acad Sci U S A ; 113(33): 9333-8, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27482104

RESUMEN

High-risk human papillomaviruses (HPVs) are causative agents of anogenital cancers and a fraction of head and neck cancers. The mechanisms involved in the progression of HPV neoplasias to cancers remain largely unknown. Here, we report that O-linked GlcNAcylation (O-GlcNAc) and O-GlcNAc transferase (OGT) were markedly increased in HPV-caused cervical neoplasms relative to normal cervix, whereas O-GlcNAcase (OGA) levels were not altered. Transduction of HPV16 oncogene E6 or E6/E7 into mouse embryonic fibroblasts (MEFs) up-regulated OGT mRNA and protein, elevated the level of O-GlcNAc, and promoted cell proliferation while reducing cellular senescence. Conversely, in HPV-18-transformed HeLa cervical carcinoma cells, inhibition of O-GlcNAc with a low concentration of a chemical inhibitor impaired the transformed phenotypes in vitro. We showed that E6 elevated c-MYC via increased protein stability attributable to O-GlcNAcylation on Thr58. Reduction of HPV-mediated cell viability by a high concentration of O-GlcNAc inhibitor was partially rescued by elevated c-MYC. Finally, knockdown of OGT or O-GlcNAc inhibition in HeLa cells or in TC-1 cells, a mouse cell line transformed by HPV16 E6/E7 and activated K-RAS, reduced c-MYC and suppressed tumorigenesis and metastasis. Thus, we have uncovered a mechanism for HPV oncoprotein-mediated transformation. These findings may eventually aid in the development of effective therapeutics for HPV-associated malignancies by targeting aberrant O-GlcNAc.


Asunto(s)
Carcinogénesis , N-Acetilglucosaminiltransferasas/fisiología , Proteínas Oncogénicas Virales/fisiología , Proteínas Represoras/fisiología , Neoplasias del Cuello Uterino/etiología , Animales , Línea Celular Tumoral , Femenino , Genes myc , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas E7 de Papillomavirus/fisiología , Neoplasias del Cuello Uterino/virología
16.
Vaccine ; 34(27): 3171-3177, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27113165

RESUMEN

The prevalent human papillomaviruses (HPVs) infect human epithelial tissues. Infections by the mucosotropic HPV genotypes cause hyperproliferative ano-genital lesions. Persistent infections by high-risk (HR) HPVs such as HPV-16, HPV-18 and related types can progress to high grade intraepithelial neoplasias and cancers. Prophylactic HPV vaccines are based on DNA-free virus-like particles (VLPs) composed of the major capsid protein L1 of HPV-16, -18, -6 and -11 (Gardasil) or HPV-16 and -18 (Cervarix). Sera from vaccinated animals effectively prevent HPV pseudovirions to infect cell lines and mouse cervical epithelia. Both vaccines have proven to be highly protective in people. HPV pseudovirions are assembled in HEK293TT cells from matched L1 and L2 capsid proteins to encapsidate a reporter gene. Pseudovirions and genuine virions have structural differences and they infect cell lines or primary human keratinocytes (PHKs) with different efficiencies. In this study, we show that sera and isolated IgG from women immunized with Gardasil prevent authentic HPV-18 virions from infecting PHKs, whereas non-immune sera and purified IgG thereof are uniformly ineffective. Using early passage PHKs, neutralization is achieved only if immune sera are added within 2-4h of infection. We attribute the timing effect to a conformational change in HPV virions, thought to occur upon initial binding to heparan sulfate proteoglycans (HSPG) on the cell surface. This interpretation is consistent with the inability of immune IgG bound to or taken up by PHKs to neutralize the virus. Interestingly, the window of neutralization increases to 12-16h in slow growing, late passage PHKs, suggestive of altered cell surface molecules. In vivo, this window might be further lengthened by the time required to activate the normally quiescent basal cells to become susceptible to infection. Our observations help explain the high efficacy of HPV vaccines.


Asunto(s)
Anticuerpos Antivirales/inmunología , Vacuna Tetravalente Recombinante contra el Virus del Papiloma Humano Tipos 6, 11 , 16, 18/uso terapéutico , Sueros Inmunes/inmunología , Queratinocitos/virología , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Células Cultivadas , Femenino , Proteoglicanos de Heparán Sulfato/metabolismo , Papillomavirus Humano 18 , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Pruebas de Neutralización , Infecciones por Papillomavirus/prevención & control
17.
Ann Plast Surg ; 76(4): 446-52, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26954733

RESUMEN

Wound healing involves a number of factors that results in the production of a "closed" wound. Studies have shown, in animal models, acceleration of wound healing with the addition of adipose-derived stromal cells (ADSC). The cause for the positive effect which these cells have on wound healing has not been elucidated. We have previously shown that addition of ADSC to the dermal equivalent in 3-dimensional skin cultures accelerates reepithelialization. We now demonstrate that conditioned media (CM) from cultured ADSC produced a similar rate of healing. This result suggests that a feedback from the 3-dimensional epithelial cultures to ADSC was not necessary to effect the accelerated reepithelialization. Mass spectrometry of CM from ADSC and primary human fibroblasts revealed differences in secretomes, some of which might have roles in the accelerating wound healing. Thus, the use of CM has provided some preliminary information on a possible mode of action.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Repitelización/fisiología , Grasa Subcutánea Abdominal/citología , Técnicas de Cultivo de Tejidos/métodos , Medios de Cultivo Condicionados , Fibroblastos/fisiología , Humanos , Piel , Cicatrización de Heridas/fisiología
18.
Stem Cells Dev ; 25(7): 556-68, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26886504

RESUMEN

Podocalyxin-like protein (PODXL) is a member of CD34 family proteins. It is the protein that carries many post-translational epitopes responsible for various pluripotent surface markers including TRA-1-60, TRA-1-81, GCTM2, GP200, and mAb84. However, PODXL has not attracted the attention of stem cell biologists. Here, we report several features of PODXL mRNA and protein in pluripotent stem cells. Similar to the modification-dependent pluripotent epitopes, PODXL transcripts and carrier protein are also features of pluripotency. PODXL is highly expressed in early human embryos from oocytes up to four-cell stages. During reprogramming of human cells to pluripotency, in contrast to TRA-1-60 and TRA-1-81, PODXL is activated by KLF4 at a very early time of reprogramming. Although TRA-1-60 and TRA-1-81 are completely lost upon differentiation, a residual PODXL(+) population exists even after extended differentiation and they were identified by the universal human PODXL epitope 3D3. Unlike TRA-1-60 and TRA-1-81 epitopes that are unique to primate pluripotent stem cells (PSCs), PODXL carrier protein can be used as a murine surface marker. Most importantly, antibody to 3D3 epitope causes massive necrosis and apoptosis of human PSCs (hPSCs). We suggest that 3D3 antibody could be employed to eliminate the tumorigenic pluripotent cells in hPSC-derived cells for cell transplantation.


Asunto(s)
Anticuerpos/toxicidad , Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Sialoglicoproteínas/metabolismo , Animales , Apoptosis , Células Cultivadas , Epítopos/inmunología , Células HEK293 , Células HeLa , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Sialoglicoproteínas/genética , Sialoglicoproteínas/inmunología
19.
Virol Sin ; 30(2): 92-100, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25924993

RESUMEN

The prevalent human papillomaviruses (HPVs) infect either cutaneous or mucosal epithelium. Active Infections lead to epithelial hyperprolifeation and are usually cleared in healthy individuals within a year. Persistent infections in the anogenital tracts by certain high-risk genotypes such as HPV-16, HPV-18 and closely related types, can progress to high grade dysplasias and carcinomas in women and men, including cervical, vulva, penile and anal cancers. A significant fraction of the head and neck cancers are also caused by HPV-16. The viral oncogenes responsible for neoplastic conversion are E6 and E7 that disrupt the pathways controlled by the two major tumor suppressor genes, p53 and members of pRB family. Because HPV cannot be propagated in conventional submerged monolayer cell cultures, organotypic epithelial raft cultures that generate a stratified and differentiated epithelium have been used to study the viral life cycle. This article describes several systems to examine aspects of the viral productive phase, along with the advantages and limitations. Animal model systems of HPV carcinogenesis are also briefly described.


Asunto(s)
Interacciones Huésped-Patógeno , Neoplasias/virología , Papillomaviridae/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Técnicas de Cultivo de Órganos
20.
Methods Mol Biol ; 1249: 93-109, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25348300

RESUMEN

The productive program of the human papillomaviruses takes place in terminally differentiating squamous epithelia. In this chapter, we provide the protocols for robust production of HPV-18 in organotypic cultures of early passages of primary human keratinocytes. A critical step is the generation of genomic HPV plasmids in vivo by using Cre-loxP-mediated excisional recombination from a vector plasmid. We discuss the rationale for this approach. This system produces high yields of infectious virus and facilitates genetic analyses of HPV protein functions and their regulation in the context of recapitulated host tissue environment.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Papillomavirus Humano 18/crecimiento & desarrollo , Queratinocitos/virología , Animales , Sitios de Ligazón Microbiológica/genética , Proteínas de la Cápside/metabolismo , Separación Celular , Células Cultivadas , ADN Viral/genética , ADN Viral/aislamiento & purificación , Genoma Viral , Humanos , Ratones , Plásmidos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Virión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...