Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
RNA ; 28(4): 478-492, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35110373

RESUMEN

Polymorphism drives survival under stress and provides adaptability. Genetic polymorphism of ribosomal RNA (rRNA) genes derives from internal repeat variation of this multicopy gene, and from interindividual variation. A considerable amount of rRNA sequence heterogeneity has been proposed but has been challenging to estimate given the scarcity of accurate reference sequences. We identified four rDNA copies on chromosome 21 (GRCh38) with 99% similarity to recently introduced reference sequence KY962518.1. We customized a GATK bioinformatics pipeline using the four rDNA loci, spanning a total 145 kb, for variant calling and used high-coverage whole-genome sequencing (WGS) data from the 1000 Genomes Project to analyze variants in 2504 individuals from 26 populations. We identified a total of 3791 variant positions. The variants positioned nonrandomly on the rRNA gene. Invariant regions included the promoter, early 5' ETS, most of 18S, 5.8S, ITS1, and large areas of the intragenic spacer. A total of 470 variant positions were observed on 28S rRNA. The majority of the 28S rRNA variants were located on highly flexible human-expanded rRNA helical folds ES7L and ES27L, suggesting that these represent positions of diversity and are potentially under continuous evolution. Several variants were validated based on RNA-seq analyses. Population analyses showed remarkable ancestry-linked genetic variance and the presence of both high penetrance and frequent variants in the 5' ETS, ITS2, and 28S regions segregating according to the continental populations. These findings provide a genetic view of rRNA gene array heterogeneity and raise the need to functionally assess how the 28S rRNA variants affect ribosome functions.


Asunto(s)
Heterogeneidad Genética , Genoma , ADN Ribosómico/genética , Genes de ARNr/genética , Humanos , ARN Ribosómico/genética , ARN Ribosómico 18S , ARN Ribosómico 28S/genética
2.
J Biol Chem ; 298(1): 101450, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34838819

RESUMEN

Cancer cells are dependent upon an abundance of ribosomes to maintain rapid cell growth and proliferation. The rate-limiting step of ribosome biogenesis is ribosomal RNA (rRNA) synthesis by RNA polymerase I (Pol I). Therefore, a goal of the cancer therapeutic field is to develop and characterize Pol I inhibitors. Here, we elucidate the mechanism of Pol I inhibition by a first-in-class small-molecule BMH-21. To characterize the effects of BMH-21 on Pol I transcription, we leveraged high-resolution in vitro transcription assays and in vivo native elongating transcript sequencing (NET-seq). We find that Pol I transcription initiation, promoter escape, and elongation are all inhibited by BMH-21 in vitro. In particular, the transcription elongation phase is highly sensitive to BMH-21 treatment, as it causes a decrease in transcription elongation rate and an increase in paused Pols on the ribosomal DNA (rDNA) template. In vivo NET-seq experiments complement these findings by revealing a reduction in Pol I occupancy on the template and an increase in sequence-specific pausing upstream of G-rich rDNA sequences after BMH-21 treatment. Collectively, these data reveal the mechanism of action of BMH-21, which is a critical step forward in the development of this compound and its derivatives for clinical use.


Asunto(s)
ADN Ribosómico , Compuestos Heterocíclicos de 4 o más Anillos , ARN Polimerasa I , Transcripción Genética , ADN Ribosómico/genética , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , ARN Polimerasa I/genética , ARN Polimerasa I/metabolismo , ARN Polimerasa II/genética , Transcripción Genética/efectos de los fármacos
3.
J Biol Chem ; 298(12): 102690, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36372232

RESUMEN

RNA Polymerase I (Pol I) synthesizes rRNA, which is the first and rate-limiting step in ribosome biogenesis. Factors governing the stability of the polymerase complex are not known. Previous studies characterizing Pol I inhibitor BMH-21 revealed a transcriptional stress-dependent pathway for degradation of the largest subunit of Pol I, RPA194. To identify the E3 ligase(s) involved, we conducted a cell-based RNAi screen for ubiquitin pathway genes. We establish Skp-Cullin-F-box protein complex F-box protein FBXL14 as an E3 ligase for RPA194. We show that FBXL14 binds to RPA194 and mediates RPA194 ubiquitination and degradation in cancer cells treated with BMH-21. Mutation analysis in yeast identified lysines 1150, 1153, and 1156 on Rpa190 relevant for the protein degradation. These results reveal the regulated turnover of Pol I, showing that the stability of the catalytic subunit is controlled by the F-box protein FBXL14 in response to transcription stress.


Asunto(s)
Proteínas F-Box , Proteínas Ligasas SKP Cullina F-box , Transcripción Genética , Dominio Catalítico , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , ARN Polimerasa I/genética , ARN Polimerasa I/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Ubiquitinación , Humanos , Transcripción Genética/genética
4.
Prostate ; 79(16): 1837-1851, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31524299

RESUMEN

BACKGROUND: Advanced prostate cancers depend on protein synthesis for continued survival and accelerated rates of metabolism for growth. RNA polymerase I (Pol I) is the enzyme responsible for ribosomal RNA (rRNA) transcription and a rate-limiting step for ribosome biogenesis. We have shown using a specific and sensitive RNA probe for the 45S rRNA precursor that rRNA synthesis is increased in prostate adenocarcinoma compared to nonmalignant epithelium. We have introduced a first-in-class Pol I inhibitor, BMH-21, that targets cancer cells of multiple origins, and holds potential for clinical translation. METHODS: The effect of BMH-21 was tested in prostate cancer cell lines and in prostate cancer xenograft and mouse genetic models. RESULTS: We show that BMH-21 inhibits Pol I transcription in metastatic, castration-resistant, and enzalutamide treatment-resistant prostate cancer cell lines. The genetic abrogation of Pol I effectively blocks the growth of prostate cancer cells. Silencing of p53, a pathway activated downstream of Pol I, does not diminish this effect. We find that BMH-21 significantly inhibited tumor growth and reduced the Ki67 proliferation index in an enzalutamide-resistant xenograft tumor model. A decrease in 45S rRNA synthesis demonstrated on-target activity. Furthermore, the Pol I inhibitor significantly inhibited tumor growth and pathology in an aggressive genetically modified Hoxb13-MYC|Hoxb13-Cre|Ptenfl/fl (BMPC) mouse prostate cancer model. CONCLUSION: Taken together, BMH-21 is a novel promising molecule for the treatment of castration-resistant prostate cancer.


Asunto(s)
Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , ARN Polimerasa I/antagonistas & inhibidores , Animales , Benzamidas , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Nitrilos , Células PC-3 , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , ARN Polimerasa I/genética , ARN Polimerasa I/metabolismo , ARN Ribosómico/genética , Distribución Aleatoria , Transcripción Genética/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
PLoS Pathog ; 12(2): e1005424, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26891221

RESUMEN

Kaposi's sarcoma herpesvirus (KSHV) causes Kaposi's sarcoma and certain lymphoproliferative malignancies. Latent infection is established in the majority of tumor cells, whereas lytic replication is reactivated in a small fraction of cells, which is important for both virus spread and disease progression. A siRNA screen for novel regulators of KSHV reactivation identified the E3 ubiquitin ligase MDM2 as a negative regulator of viral reactivation. Depletion of MDM2, a repressor of p53, favored efficient activation of the viral lytic transcription program and viral reactivation. During lytic replication cells activated a p53 response, accumulated DNA damage and arrested at G2-phase. Depletion of p21, a p53 target gene, restored cell cycle progression and thereby impaired the virus reactivation cascade delaying the onset of virus replication induced cytopathic effect. Herpesviruses are known to reactivate in response to different kinds of stress, and our study now highlights the molecular events in the stressed host cell that KSHV has evolved to utilize to ensure efficient viral lytic replication.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 8/genética , Estrés Fisiológico/genética , Replicación Viral , Línea Celular Tumoral , Replicación del ADN , Humanos , ARN Interferente Pequeño/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virología , Activación Viral/fisiología , Latencia del Virus/genética , Replicación Viral/genética
6.
J Urol ; 191(3): 842-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24035881

RESUMEN

PURPOSE: DNA damage responses are relevant to prostate cancer initiation, progression and treatment. Few models of the normal and malignant human prostate that maintain stromal-epithelial interactions in vivo exist in which to study DNA damage responses. We evaluated the feasibility of maintaining tissue slice grafts at subcutaneous vs subrenal capsular sites in RAG2(-/-)γC(-/-) mice to study the DNA damage responses of normal and malignant glands. MATERIALS AND METHODS: We compared the take rate and histology of tissue slice grafts from fresh, precision cut surgical specimens that were maintained for 1 to 4 weeks in subcutaneous vs subrenal capsular sites. Induction of γH2AX, p53, ATM and apoptosis was evaluated as a measure of the DNA damage response after irradiation. RESULTS: The take rate of subcutaneous tissue slice grafts was higher than typically reported but lower than at the subrenal capsular site. Subcutaneous tissue slice grafts frequently showed basal cell hyperplasia, squamous metaplasia and cystic atrophy, and cancer did not survive. In contrast, normal and malignant histology was well maintained in subrenal capsular tissue slice grafts. Regardless of implantation site the induction of γH2AX and ATM occurred in tissue slice graft epithelium 1 hour after irradiation and decreased to basal level by 24 hours, indicating DNA damage recognition and repair. As observed previously in prostatic ex vivo models, p53 was not activated. Notably, tumor but not normal cells responded to irradiation by undergoing apoptosis. CONCLUSIONS: To our knowledge this is the first study of DNA damage responses in a patient derived prostate tissue graft model. The subrenal capsular site of RAG2(-/-)γC(-/-) mice optimally maintains normal and malignant histology and function, permitting novel studies of DNA damage responses in a physiological context.


Asunto(s)
Daño del ADN , Neoplasias de la Próstata/genética , Trasplante de Tejidos/métodos , Animales , Apoptosis , Reparación del ADN , Humanos , Inmunohistoquímica , Masculino , Ratones , Microscopía Fluorescente , Clasificación del Tumor , Próstata/patología , Neoplasias de la Próstata/patología
7.
Blood Adv ; 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38739710

RESUMEN

Provirus integration site for Moloney murine leukemia virus (PIM) family serine/threonine kinases perform pro-tumorigenic functions in hematologic malignancies and solid tumors by phosphorylating substrates involved in tumor metabolism, cell survival, metastasis, inflammation, and immune cell invasion. However, a comprehensive understanding of PIM kinase functions is currently lacking. Multiple small molecule PIM kinase inhibitors are currently being evaluated as co-therapeutics in cancer patients. To further illuminate PIM kinase functions in cancer, we deeply profiled PIM1 substrates using the reverse in-gel kinase assay to identify downstream cellular processes targetable with small molecules. Pathway analyses of putative PIM substrates nominated RNA splicing and rRNA processing as PIM-regulated cellular processes. PIM inhibition elicited reproducible splicing changes in PIM-inhibitor-responsive acute myeloid leukemia (AML) cell lines. PIM inhibitors synergized with splicing modulators targeting splicing factor 3b subunit 1 (SF3B1) and serine-arginine protein kinase 1 (SRPK1) to kill AML cells. PIM inhibition also altered rRNA processing, and PIM inhibitors synergized with an RNA polymerase I inhibitor to kill AML cells and block AML tumor growth. These data demonstrate that deep kinase substrate knowledge can illuminate unappreciated kinase functions, nominating synergistic co-therapeutic strategies. This approach may expand the co-therapeutic armamentarium to overcome kinase-inhibitor resistant disease that limits durable responses in malignant disease.

8.
Prostate ; 73(2): 219-26, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22806587

RESUMEN

BACKGROUND: PSA is the most useful prostate cancer marker. However, its levels are increased also in some non-malignant conditions. In circulation, the majority of PSA is complexed with protease inhibitors, including α(1) -antichymotrypsin (ACT). The proportion of the PSA-ACT complex is higher in patients with prostate cancer than in controls without cancer. The expression of ACT has been shown to be higher in prostate cancer than in benign prostatic hyperplasia. However, results regarding the extent which PSA forms complexes within the prostate and whether there are differences in complex formation between normal and malignant prostatic tissue are inconsistent and limited. METHODS: We studied complex formation of PSA secreted by cultured human prostate tissues and in the tissue by in situ proximity ligation assay (PLA). Free, total and active PSA, and the PSA-ACT complex were determined in tissue culture media by immunoassays, immunoblotting, and chromatographic methods. RESULTS: The majority of PSA in tissue culture medium was free and enzymatically active. However, a significant proportion (1.6 ± 0.5%) of immunoreactive PSA was found to be complexed with ACT. Complex formation was confirmed by in situ PLA, which showed more intense staining of PSA-ACT in cancers with Gleason grade 3 than in adjacent benign tissues from the same patients. CONCLUSIONS: These results show that PSA forms complexes already within the prostate and that PSA-ACT levels are increased in moderately differentiated prostate cancer tissue. This may explain, at least partially, why the ratio of serum PSA-ACT to total PSA is increased in prostate cancer.


Asunto(s)
Antígeno Prostático Específico/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , alfa 1-Antiquimotripsina/metabolismo , Adulto , Anciano , Diferenciación Celular/fisiología , Humanos , Masculino , Persona de Mediana Edad , Complejos Multiproteicos/metabolismo , Próstata/patología , Neoplasias de la Próstata/diagnóstico , Unión Proteica/fisiología
9.
Mol Cell Proteomics ; 10(10): M111.009241, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21778410

RESUMEN

The nucleolus is a nuclear organelle that coordinates rRNA transcription and ribosome subunit biogenesis. Recent proteomic analyses have shown that the nucleolus contains proteins involved in cell cycle control, DNA processing and DNA damage response and repair, in addition to the many proteins connected with ribosome subunit production. Here we study the dynamics of nucleolar protein responses in cells exposed to stress and DNA damage caused by ionizing and ultraviolet (UV) radiation in diploid human fibroblasts. We show using a combination of imaging and quantitative proteomics methods that nucleolar substructure and the nucleolar proteome undergo selective reorganization in response to UV damage. The proteomic responses to UV include alterations of functional protein complexes such as the SSU processome and exosome, and paraspeckle proteins, involving both decreases and increases in steady state protein ratios, respectively. Several nonhomologous end-joining proteins (NHEJ), such as Ku70/80, display similar fast responses to UV. In contrast, nucleolar proteomic responses to IR are both temporally and spatially distinct from those caused by UV, and more limited in terms of magnitude. With the exception of the NHEJ and paraspeckle proteins, where IR induces rapid and transient changes within 15 min of the damage, IR does not alter the ratios of most other functional nucleolar protein complexes. The rapid transient decrease of NHEJ proteins in the nucleolus indicates that it may reflect a response to DNA damage. Our results underline that the nucleolus is a specific stress response organelle that responds to different damage and stress agents in a unique, damage-specific manner.


Asunto(s)
Nucléolo Celular/metabolismo , Daño del ADN , Proteínas Nucleares/análisis , Proteínas Nucleares/metabolismo , Proteoma/análisis , Antígenos Nucleares/análisis , Antígenos Nucleares/metabolismo , Nucléolo Celular/efectos de la radiación , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Exosomas/metabolismo , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Humanos , Marcaje Isotópico , Autoantígeno Ku , Microscopía Electrónica de Transmisión , Proteínas Nucleares/genética , Proteoma/genética , Proteoma/metabolismo , Proteínas de Unión al ARN/análisis , Proteínas de Unión al ARN/metabolismo , Radiación Ionizante , Estrés Fisiológico , Transcripción Genética , Rayos Ultravioleta
10.
PLoS One ; 18(5): e0285660, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37167337

RESUMEN

RNA Polymerase I (Pol I) has recently been recognized as a cancer therapeutic target. The activity of this enzyme is essential for ribosome biogenesis and is universally activated in cancers. The enzymatic activity of this multi-subunit complex resides in its catalytic core composed of RPA194, RPA135, and RPA12, a subunit with functions in RNA cleavage, transcription initiation and elongation. Here we explore whether RPA12 influences the regulation of RPA194 in human cancer cells. We use a specific small-molecule Pol I inhibitor BMH-21 that inhibits transcription initiation, elongation and ultimately activates the degradation of Pol I catalytic subunit RPA194. We show that silencing RPA12 causes alterations in the expression and localization of Pol I subunits RPA194 and RPA135. Furthermore, we find that despite these alterations not only does the Pol I core complex between RPA194 and RPA135 remain intact upon RPA12 knockdown, but the transcription of Pol I and its engagement with chromatin remain unaffected. The BMH-21-mediated degradation of RPA194 was independent of RPA12 suggesting that RPA12 affects the basal expression, but not the drug-inducible turnover of RPA194. These studies add to knowledge defining regulatory factors for the expression of this Pol I catalytic subunit.


Asunto(s)
Cromatina , ARN Polimerasa I , Humanos , Dominio Catalítico , ARN Polimerasa I/genética , ARN Polimerasa I/metabolismo , ARN Polimerasa II/metabolismo , Transcripción Genética , Neoplasias/genética , Neoplasias/metabolismo
11.
Proteomics ; 12(19-20): 3044-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22890538

RESUMEN

Efficient extraction and accurate quantification of nucleolar macromolecules are critical for in vitro analysis, especially for studying RNA, DNA, and protein dynamics under identical conditions. There is presently no single method that efficiently and simultaneously isolates these three macromolecular constituents from purified nucleoli. We have developed an optimized method, which without evident loss, extracts, and solubilizes protein recovered from a single sample following TRIzol isolation of RNA and DNA. The solubilized protein can be accurately quantified by protein bicinchoninic acid assay and assessed by polyacrylamide gel electrophoresis. We have successfully applied this approach to extract and quantify all three nucleolar components, and to study nucleolar protein responses after actinomycin D treatment.


Asunto(s)
Nucléolo Celular/química , ADN/aislamiento & purificación , Proteínas Nucleares/aislamiento & purificación , Proteómica/métodos , ARN Nuclear/aislamiento & purificación , Nucléolo Celular/metabolismo , ADN/química , Guanidinas/química , Células HeLa , Humanos , Proteínas Nucleares/análisis , Proteínas Nucleares/química , Fenoles/química , Quinolinas/química , ARN Nuclear/química , Fracciones Subcelulares/química
12.
J Cell Physiol ; 227(6): 2605-12, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21882188

RESUMEN

Transforming growth factor-ß (TGF-ß) is a diverse cytokine regulating growth, apoptosis, differentiation, adhesion, invasion, and extracellular matrix production. Dysregulation of TGF-ß is associated with fibrotic disorders and epithelial-mesenchymal transition, and has been linked with idiopathic pulmonary fibrosis (IPF). Cysteine-rich protein 1 (CRP1) is a small LIM-domain containing protein involved in smooth muscle differentiation. Here, we show that TGF-ß1 increases the expression of CRP1 protein and that CRP1 levels increase in a biphasic fashion. A rapid transient (15-45 min) increase in CRP1 is followed by a subsequent, sustained increase in CRP1 a few hours afterwards that lasts several days. We find that TGF-ß1 regulates the expression of CRP1 through Smad and non-conventional p38 MAPK signaling pathways in a transcription-independent manner and that the induction occurs concomitant with an increase in myofibroblast differentiation. Using CRP1 silencing by shRNA, we identify CRP1 as a novel factor mediating cell contractility. Furthermore, we localize CRP1 to fibroblastic foci in IPF lungs and find that CRP1 is significantly more expressed in IPF as compared to control lung tissue. The results show that CRP1 is a novel TGF-ß1 regulated protein that is expressed in fibrotic lesions and may be relevant in the IPF disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Proteínas con Dominio LIM/metabolismo , Pulmón/metabolismo , Miofibroblastos/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Proteínas Portadoras/genética , Estudios de Casos y Controles , Diferenciación Celular , Línea Celular Tumoral , Forma de la Célula , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Proteínas con Dominio LIM/genética , Pulmón/patología , Ratones , Miofibroblastos/patología , Células 3T3 NIH , Interferencia de ARN , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factores de Tiempo , Transfección , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Prostate ; 72(10): 1060-70, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22072329

RESUMEN

BACKGROUND: Prostate and seminal vesicle are two similar hormone responsive human organs that differ dramatically in their cancer incidence. DNA damage response (DDR) is required for maintenance of genomic integrity. METHODS: In this study we investigated the DDR and cell cycle checkpoint activation of these organs using orthotopic cultures of human surgery-derived tissues and primary cultures of isolated prostate and seminal vesicle cells. RESULTS: We find that the activation of ATM signaling pathway by ionizing radiation (IR) was comparable in both tissues. Previously, we have shown that the prostate secretory cells express low levels of histone variant H2AX and phosphorylated H2AX (γH2AX) after IR. Here we demonstrate that H2AX levels are low also in the secretory seminal vesicle cells suggesting that this is a common phenotype of postmitotic cells. We consequently established primary epithelial cell cultures from both organs to compare their DDR. Interestingly, contrary to human prostate epithelial cells (HPEC), primary seminal vesicle epithelial cells (HSVEC) displayed effective cell cycle checkpoints after IR and expressed higher levels of Wee1A checkpoint kinase. Furthermore, HSVEC but not HPEC cells were able to activate p53 and to induce p21 cell cycle inhibitor. DISCUSSION: Our results show that during replication, the checkpoint enforcement is more proficient in the seminal vesicle than in the prostate epithelium cells. This indicates a more stringent enforcement of DDR in replicating seminal vesicle epithelial cells, and suggests that epithelial regeneration combined with sub-optimal checkpoint responses may contribute to high frequency of genetic lesions in the prostate epithelium.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Daño del ADN/genética , Células Epiteliales/fisiología , Próstata/fisiología , Vesículas Seminales/fisiología , Células Cultivadas , Células Epiteliales/patología , Epitelio/patología , Epitelio/fisiología , Humanos , Masculino , Próstata/patología , Vesículas Seminales/patología
14.
PLoS Pathog ; 6(3): e1000818, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20333249

RESUMEN

Nucleophosmin (NPM) is a multifunctional nuclear phosphoprotein and a histone chaperone implicated in chromatin organization and transcription control. Oncogenic Kaposi's sarcoma herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma, primary effusion lymphoma (PEL) and multicentric Castleman disease (MCD). In the infected host cell KSHV displays two modes of infection, the latency and productive viral replication phases, involving extensive viral DNA replication and gene expression. A sustained balance between latency and reactivation to the productive infection state is essential for viral persistence and KSHV pathogenesis. Our study demonstrates that the KSHV v-cyclin and cellular CDK6 kinase phosphorylate NPM on threonine 199 (Thr199) in de novo and naturally KSHV-infected cells and that NPM is phosphorylated to the same site in primary KS tumors. Furthermore, v-cyclin-mediated phosphorylation of NPM engages the interaction between NPM and the latency-associated nuclear antigen LANA, a KSHV-encoded repressor of viral lytic replication. Strikingly, depletion of NPM in PEL cells leads to viral reactivation, and production of new infectious virus particles. Moreover, the phosphorylation of NPM negatively correlates with the level of spontaneous viral reactivation in PEL cells. This work demonstrates that NPM is a critical regulator of KSHV latency via functional interactions with v-cyclin and LANA.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/metabolismo , Herpesvirus Humano 8/crecimiento & desarrollo , Proteínas Nucleares/metabolismo , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virología , Latencia del Virus/fisiología , Acetilación , Antígenos Virales/genética , Antígenos Virales/metabolismo , Línea Celular Tumoral , Herpesvirus Humano 8/genética , Humanos , Proteínas Nucleares/genética , Nucleofosmina , Fosforilación/fisiología , ARN Interferente Pequeño , Treonina/metabolismo , Replicación Viral/fisiología
15.
Cancer Cell ; 5(5): 465-75, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15144954

RESUMEN

Nucleophosmin (NPM, B23) is an abundant nucleolar phosphoprotein involved in ribosome biogenesis, and interacts with tumor suppressor proteins p53 and Rb. Here we show that NPM is a UV damage response protein that undergoes nucleoplasmic redistribution and regulates p53 and HDM2 levels and their interaction. By utilizing RNAi approaches and analyses of endogenous and ectopically expressed proteins, we demonstrate that NPM binds HDM2 and acts as a negative regulator of p53-HDM2 interaction. Viral stress, enforced by expression of Kaposi's sarcoma virus K cyclin, causes NPM redistribution, K cyclin-NPM association, and p53 stabilization by dissociation of HDM2-p53 complexes. The results demonstrate novel associations of HDM2 and K cyclin with NPM and implicate NPM as a crucial controller of p53 through inhibition of HDM2.


Asunto(s)
Nucléolo Celular/metabolismo , Ciclinas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Nucléolo Celular/efectos de la radiación , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/efectos de la radiación , Glutatión Transferasa/metabolismo , Humanos , Ratones , Nucleofosmina , Osteosarcoma/metabolismo , Osteosarcoma/patología , Pruebas de Precipitina , Biosíntesis de Proteínas , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-mdm2 , ARN Interferente Pequeño/farmacología , Proteína SUMO-1/metabolismo , Proteína p53 Supresora de Tumor/química , Rayos Ultravioleta/efectos adversos , Dedos de Zinc
16.
J Mammary Gland Biol Neoplasia ; 16(2): 109-15, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21390570

RESUMEN

Estrogen receptor-α (ERα) and transforming growth factor (TGF)-ß signaling pathways are major regulators during mammary gland development, function and tumorigenesis. Predominantly, they have opposing roles in proliferation and apoptosis. While ERα signaling supports growth and differentiation and is antiapoptotic, mammary gland epithelia cells are very sensitive to TGF-ß-induced cell cycle arrest and apoptosis. Their regulatory pathways intersect, and ERα blocks TGF-ß pathway by multiple means, including direct interactions of its signaling components, Smads. However, relatively little is known of the dysfunction of their interactions in cancer. A better understanding would help to develop new strategies for breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptor alfa de Estrógeno/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Femenino , Humanos , Transducción de Señal
17.
Cancers (Basel) ; 14(23)2022 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-36497261

RESUMEN

RNA polymerase I is a highly processive enzyme with fast initiation and elongation rates. The structure of Pol I, with its in-built RNA cleavage ability and incorporation of subunits homologous to transcription factors, enables it to quickly and efficiently synthesize the enormous amount of rRNA required for ribosome biogenesis. Each step of Pol I transcription is carefully controlled. However, cancers have highjacked these control points to switch the enzyme, and its transcription, on permanently. While this provides an exceptional benefit to cancer cells, it also creates a potential cancer therapeutic vulnerability. We review the current research on the regulation of Pol I transcription, and we discuss chemical biology efforts to develop new targeted agents against this process. Lastly, we highlight challenges that have arisen from the introduction of agents with promiscuous mechanisms of action and provide examples of agents with specificity and selectivity against Pol I.

18.
Cancers (Basel) ; 14(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35158857

RESUMEN

The discovery of small, "cave-like" invaginations at the plasma membrane, called caveola, has opened up a new and exciting research area in health and diseases revolving around this cellular ultrastructure. Caveolae are rich in cholesterol and orchestrate cellular signaling events. Within caveola, the caveola-associated proteins, caveolins and cavins, are critical components for the formation of these lipid rafts, their dynamics, and cellular pathophysiology. Their alterations underlie human diseases such as lipodystrophy, muscular dystrophy, cardiovascular disease, and diabetes. The expression of caveolins and cavins is modulated in tumors and in tumor stroma, and their alterations are connected with cancer progression and treatment resistance. To date, although substantial breakthroughs in cancer drug development have been made, drug resistance remains a problem leading to treatment failures and challenging translation and bench-to-bedside research. Here, we summarize the current progress in understanding cancer drug resistance in the context of caveola-associated molecules and tumor stroma and discuss how we can potentially design therapeutic avenues to target these molecules in order to overcome treatment resistance.

19.
Cancers (Basel) ; 14(22)2022 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-36428638

RESUMEN

Cancer cells require robust ribosome biogenesis to maintain rapid cell growth during tumorigenesis. Because RNA polymerase I (Pol I) transcription of the ribosomal DNA (rDNA) is the first and rate-limiting step of ribosome biogenesis, it has emerged as a promising anti-cancer target. Over the last decade, novel cancer therapeutics targeting Pol I have progressed to clinical trials. BMH-21 is a first-in-class small molecule that inhibits Pol I transcription and represses cancer cell growth. Several recent studies have uncovered key mechanisms by which BMH-21 inhibits ribosome biosynthesis but the selectivity of BMH-21 for Pol I has not been directly measured. Here, we quantify the effects of BMH-21 on Pol I, RNA polymerase II (Pol II), and RNA polymerase III (Pol III) in vitro using purified components. We found that BMH-21 directly impairs nucleotide addition by Pol I, with no or modest effect on Pols II and III, respectively. Additionally, we found that BMH-21 does not affect the stability of any of the Pols' elongation complexes. These data demonstrate that BMH-21 directly exploits unique vulnerabilities of Pol I.

20.
Sci Immunol ; 7(71): eabh4271, 2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35622902

RESUMEN

Memory CD8+ T cells are characterized by their ability to persist long after the initial antigen encounter and their capacity to generate a rapid recall response. Recent studies have identified a role for metabolic reprogramming and mitochondrial function in promoting the longevity of memory T cells. However, detailed mechanisms involved in promoting their rapid recall response are incompletely understood. Here, we identify a role for the initial and continued activation of the trifunctional rate-limiting enzyme of the de novo pyrimidine synthesis pathway CAD (carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase) as critical in promoting the rapid recall response of previously activated CD8+ T cells. We found that CAD was rapidly phosphorylated upon naïve T cell activation in an mTORC1-dependent manner, yet remained phosphorylated long after initial activation. Previously activated CD8+ T cells displayed continued de novo pyrimidine synthesis in the absence of mitogenic signals, and interfering with this pathway diminished the speed and magnitude of cytokine production upon rechallenge. Inhibition of CAD did not affect cytokine transcript levels but diminished available pre-rRNA (ribosomal RNA), the polycistronic rRNA precursor whose synthesis is the rate-limiting step in ribosomal biogenesis. CAD inhibition additionally decreased levels of detectable ribosomal proteins in previously activated CD8+ T cells. Conversely, overexpression of CAD improved both the cytokine response and proliferation of memory T cells. Overall, our studies reveal a critical role for CAD-induced pyrimidine synthesis and ribosomal biogenesis in promoting the rapid recall response characteristic of memory T cells.


Asunto(s)
Aspartato Carbamoiltransferasa , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante) , Aspartato Carbamoiltransferasa/genética , Aspartato Carbamoiltransferasa/metabolismo , Linfocitos T CD8-positivos/metabolismo , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/genética , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/metabolismo , Citocinas , Pirimidinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA