Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(5): e1012058, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38768227

RESUMO

Viral disruption of innate immune signaling is a critical determinant of productive infection. The Human Cytomegalovirus (HCMV) UL26 protein prevents anti-viral gene expression during infection, yet the mechanisms involved are unclear. We used TurboID-driven proximity proteomics to identify putative UL26 interacting proteins during infection to address this issue. We find that UL26 forms a complex with several immuno-regulatory proteins, including several STAT family members and various PIAS proteins, a family of E3 SUMO ligases. Our results indicate that UL26 prevents STAT phosphorylation during infection and antagonizes transcriptional activation induced by either interferon α (IFNA) or tumor necrosis factor α (TNFα). Additionally, we find that the inactivation of PIAS1 sensitizes cells to inflammatory stimulation, resulting in an anti-viral transcriptional environment similar to ΔUL26 infection. Further, PIAS1 is important for HCMV cell-to-cell spread, which depends on the presence of UL26, suggesting that the UL26-PIAS1 interaction is vital for modulating intrinsic anti-viral defense.

2.
J Neurooncol ; 163(3): 623-634, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37389756

RESUMO

PURPOSE: Gliomas and their surrounding microenvironment constantly interact to promote tumorigenicity, yet the underlying posttranscriptional regulatory mechanisms that govern this interplay are poorly understood. METHODS: Utilizing our established PAC-seq approach and PolyAMiner bioinformatic analysis pipeline, we deciphered the NUDT21-mediated differential APA dynamics in glioma cells. RESULTS: We identified LAMC1 as a critical NUDT21 alternative polyadenylation (APA) target, common in several core glioma-driving signaling pathways. qRT-PCR analysis confirmed that NUDT21-knockdown in glioma cells results in the preferred usage of the proximal polyA signal (PAS) of LAMC1. Functional studies revealed that NUDT21-knockdown-induced 3'UTR shortening of LAMC1 is sufficient to cause translational gain, as LAMC1 protein is upregulated in these cells compared to their respective controls. We demonstrate that 3'UTR shortening of LAMC1 after NUDT21 knockdown removes binding sites for miR-124/506, thereby relieving potent miRNA-based repression of LAMC1 expression. Remarkably, we report that the knockdown of NUDT21 significantly promoted glioma cell migration and that co-depletion of LAMC1 with NUDT21 abolished this effect. Lastly, we observed that LAMC1 3'UTR shortening predicts poor prognosis of low-grade glioma patients from The Cancer Genome Atlas. CONCLUSION: This study identifies NUDT21 as a core alternative polyadenylation factor that regulates the tumor microenvironment through differential APA and loss of miR-124/506 inhibition of LAMC1. Knockdown of NUDT21 in GBM cells mediates 3'UTR shortening of LAMC1, contributing to an increase in LAMC1, increased glioma cell migration/invasion, and a poor prognosis.


Assuntos
Fator de Especificidade de Clivagem e Poliadenilação , Glioma , MicroRNAs , Humanos , Regiões 3' não Traduzidas , Glioma/genética , MicroRNAs/metabolismo , Poliadenilação , Transdução de Sinais , Microambiente Tumoral , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo
3.
Sci Adv ; 9(7): eade4814, 2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36800428

RESUMO

Alternative polyadenylation (APA) creates distinct transcripts from the same gene by cleaving the pre-mRNA at poly(A) sites that can lie within the 3' untranslated region (3'UTR), introns, or exons. Most studies focus on APA within the 3'UTR; however, here, we show that CPSF6 insufficiency alters protein levels and causes a developmental syndrome by deregulating APA throughout the transcript. In neonatal humans and zebrafish larvae, CPSF6 insufficiency shifts poly(A) site usage between the 3'UTR and internal sites in a pathway-specific manner. Genes associated with neuronal function undergo mostly intronic APA, reducing their expression, while genes associated with heart and skeletal function mostly undergo 3'UTR APA and are up-regulated. This suggests that, under healthy conditions, cells toggle between internal and 3'UTR APA to modulate protein expression.


Assuntos
Poliadenilação , Peixe-Zebra , Animais , Humanos , Recém-Nascido , Regiões 3' não Traduzidas , Éxons , Íntrons/genética , Peixe-Zebra/genética , Embrião não Mamífero
4.
Nat Commun ; 13(1): 6054, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36229431

RESUMO

Oral-facial-digital (OFD) syndromes are a heterogeneous group of congenital disorders characterized by malformations of the face and oral cavity, and digit anomalies. Mutations within 12 cilia-related genes have been identified that cause several types of OFD, suggesting that OFDs constitute a subgroup of developmental ciliopathies. Through homozygosity mapping and exome sequencing of two families with variable OFD type 2, we identified distinct germline variants in INTS13, a subunit of the Integrator complex. This multiprotein complex associates with RNA Polymerase II and cleaves nascent RNA to modulate gene expression. We determined that INTS13 utilizes its C-terminus to bind the Integrator cleavage module, which is disrupted by the identified germline variants p.S652L and p.K668Nfs*9. Depletion of INTS13 disrupts ciliogenesis in human cultured cells and causes dysregulation of a broad collection of ciliary genes. Accordingly, its knockdown in Xenopus embryos leads to motile cilia anomalies. Altogether, we show that mutations in INTS13 cause an autosomal recessive ciliopathy, which reveals key interactions between components of the Integrator complex.


Assuntos
Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Ciliopatias , Síndromes Orofaciodigitais , Cílios/genética , Ciliopatias/genética , Homozigoto , Humanos , Mutação , Síndromes Orofaciodigitais/genética , RNA , RNA Polimerase II/genética
5.
Mol Cell ; 82(22): 4232-4245.e11, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36309014

RESUMO

RNA polymerase II (RNAPII) pausing in early elongation is critical for gene regulation. Paused RNAPII can be released into productive elongation by the kinase P-TEFb or targeted for premature termination by the Integrator complex. Integrator comprises endonuclease and phosphatase activities, driving termination by cleavage of nascent RNA and removal of stimulatory phosphorylation. We generated a degron system for rapid Integrator endonuclease (INTS11) depletion to probe the direct consequences of Integrator-mediated RNA cleavage. Degradation of INTS11 elicits nearly universal increases in active early elongation complexes. However, these RNAPII complexes fail to achieve optimal elongation rates and exhibit persistent Integrator phosphatase activity. Thus, only short transcripts are significantly upregulated following INTS11 loss, including transcription factors, signaling regulators, and non-coding RNAs. We propose a uniform molecular function for INTS11 across all RNAPII-transcribed loci, with differential effects on particular genes, pathways, or RNA biotypes reflective of transcript lengths rather than specificity of Integrator activity.


Assuntos
Endonucleases , RNA Polimerase II , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Endonucleases/genética , Regiões Promotoras Genéticas , RNA , Monoéster Fosfórico Hidrolases/metabolismo , Transcrição Gênica
6.
Nat Commun ; 13(1): 5742, 2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36180473

RESUMO

Integrator is a multi-subunit protein complex associated with RNA polymerase II (Pol II), with critical roles in noncoding RNA 3'-end processing and transcription attenuation of a broad collection of mRNAs. IntS11 is the endonuclease for RNA cleavage, as a part of the IntS4-IntS9-IntS11 Integrator cleavage module (ICM). Here we report a cryo-EM structure of the Drosophila ICM, at 2.74 Å resolution, revealing stable association of an inositol hexakisphosphate (IP6) molecule. The IP6 binding site is located in a highly electropositive pocket at an interface among all three subunits of ICM, 55 Å away from the IntS11 active site and generally conserved in other ICMs. We also confirmed IP6 association with the same site in human ICM. IP6 binding is not detected in ICM samples harboring mutations in this binding site. Such mutations or disruption of IP6 biosynthesis significantly reduced Integrator function in snRNA 3'-end processing and mRNA transcription attenuation. Our structural and functional studies reveal that IP6 is required for Integrator function in Drosophila, humans, and likely other organisms.


Assuntos
Ácido Fítico , RNA Polimerase II , Animais , Drosophila/metabolismo , Endonucleases , Humanos , Ácido Fítico/metabolismo , RNA Polimerase II/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Nuclear Pequeno/metabolismo , RNA não Traduzido
7.
Nat Genet ; 53(7): 994-1005, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33986536

RESUMO

Genome-wide association studies have identified thousands of noncoding variants associated with human traits and diseases. However, the functional interpretation of these variants is a major challenge. Here, we constructed a multi-tissue atlas of human 3'UTR alternative polyadenylation (APA) quantitative trait loci (3'aQTLs), containing approximately 0.4 million common genetic variants associated with the APA of target genes, identified in 46 tissues isolated from 467 individuals (Genotype-Tissue Expression Project). Mechanistically, 3'aQTLs can alter poly(A) motifs, RNA secondary structure and RNA-binding protein-binding sites, leading to thousands of APA changes. Our CRISPR-based experiments indicate that such 3'aQTLs can alter APA regulation. Furthermore, we demonstrate that mapping 3'aQTLs can identify APA regulators, such as La-related protein 4. Finally, 3'aQTLs are colocalized with approximately 16.1% of trait-associated variants and are largely distinct from other QTLs, such as expression QTLs. Together, our findings show that 3'aQTLs contribute substantially to the molecular mechanisms underlying human complex traits and diseases.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Herança Multifatorial , Poliadenilação , Locos de Características Quantitativas , RNA Mensageiro/genética , Regiões 3' não Traduzidas , Estudos de Associação Genética/métodos , Humanos , Poli A
8.
Mol Cell ; 80(2): 345-358.e9, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32966759

RESUMO

Efficient release of promoter-proximally paused RNA Pol II into productive elongation is essential for gene expression. Recently, we reported that the Integrator complex can bind paused RNA Pol II and drive premature transcription termination, potently attenuating the activity of target genes. Premature termination requires RNA cleavage by the endonuclease subunit of Integrator, but the roles of other Integrator subunits in gene regulation have yet to be elucidated. Here we report that Integrator subunit 8 (IntS8) is critical for transcription repression and required for association with protein phosphatase 2A (PP2A). We find that Integrator-bound PP2A dephosphorylates the RNA Pol II C-terminal domain and Spt5, preventing the transition to productive elongation. Thus, blocking PP2A association with Integrator stimulates pause release and gene activity. These results reveal a second catalytic function associated with Integrator-mediated transcription termination and indicate that control of productive elongation involves active competition between transcriptional kinases and phosphatases.


Assuntos
Proteínas de Drosophila/metabolismo , Proteína Fosfatase 2/metabolismo , Subunidades Proteicas/metabolismo , Fatores de Transcrição/metabolismo , Terminação da Transcrição Genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência Conservada , Proteínas de Drosophila/química , Drosophila melanogaster , Regulação da Expressão Gênica , Loci Gênicos , Humanos , Fosforilação , Regiões Promotoras Genéticas , Subunidades Proteicas/química , RNA Polimerase II/química , RNA Polimerase II/metabolismo , Transdução de Sinais , Especificidade por Substrato , Fatores de Transcrição/química
9.
RNA ; 26(9): 1143-1159, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32404348

RESUMO

Tob2, an anti-proliferative protein, promotes deadenylation through recruiting Caf1 deadenylase to the mRNA poly(A) tail by simultaneously interacting with both Caf1 and poly(A)-binding protein (PABP). Previously, we found that changes in Tob2 phosphorylation can alter its PABP-binding ability and deadenylation-promoting function. However, it remained unknown regarding the relevant kinase(s). Moreover, it was unclear whether Tob2 phosphorylation modulates the transcriptome and whether the phosphorylation is linked to Tob2's anti-proliferative function. In this study, we found that c-Jun amino-terminal kinase (JNK) increases phosphorylation of Tob2 at many Ser/Thr sites in the intrinsically disordered region (IDR) that contains two separate PABP-interacting PAM2 motifs. JNK-induced phosphorylation or phosphomimetic mutations at these sites weaken the Tob2-PABP interaction. In contrast, JNK-independent phosphorylation of Tob2 at serine 254 (S254) greatly enhances Tob2 interaction with PABP and its ability to promote deadenylation. We discovered that both PAM2 motifs are required for Tob2 to display these features. Combining mass spectrometry analysis, poly(A) size-distribution profiling, transcriptome-wide mRNA turnover analyses, and cell proliferation assays, we found that the phosphomimetic mutation at S254 (S254D) enhances Tob2's association with PABP, leading to accelerated deadenylation and decay of mRNAs globally. Moreover, the Tob2-S254D mutant accelerates the decay of many transcripts coding for cell cycle related proteins and enhances anti-proliferation function. Our findings reveal a novel mechanism by which Ccr4-Not complex is recruited by Tob2 to the mRNA 3' poly(A)-PABP complex in a phosphorylation dependent manner to promote rapid deadenylation and decay across the transcriptome, eliciting transcriptome reprogramming and suppressed cell proliferation.


Assuntos
Proteínas de Ciclo Celular/genética , Proliferação de Células/genética , Fosforilação/genética , RNA Mensageiro/genética , Transcriptoma/genética , Linhagem Celular , Células HEK293 , Humanos , Poli A/genética , Proteínas de Ligação a Poli(A)/genética , Poliadenilação/genética , Estabilidade de RNA/genética
10.
Mol Cell ; 76(5): 738-752.e7, 2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31809743

RESUMO

The transition of RNA polymerase II (Pol II) from initiation to productive elongation is a central, regulated step in metazoan gene expression. At many genes, Pol II pauses stably in early elongation, remaining engaged with the 25- to 60-nt-long nascent RNA for many minutes while awaiting signals for release into the gene body. However, 15%-20% of genes display highly unstable promoter Pol II, suggesting that paused polymerase might dissociate from template DNA at these promoters and release a short, non-productive mRNA. Here, we report that paused Pol II can be actively destabilized by the Integrator complex. Specifically, we present evidence that Integrator utilizes its RNA endonuclease activity to cleave nascent RNA and drive termination of paused Pol II. These findings uncover a previously unappreciated mechanism of metazoan gene repression, akin to bacterial transcription attenuation, wherein promoter-proximal Pol II is prevented from entering productive elongation through factor-regulated termination.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , RNA Mensageiro/biossíntese , Elongação da Transcrição Genética , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , RNA Polimerase II/genética , RNA Mensageiro/genética
11.
Genes Dev ; 33(21-22): 1525-1538, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31530651

RESUMO

Cellular homeostasis requires transcriptional outputs to be coordinated, and many events post-transcription initiation can dictate the levels and functions of mature transcripts. To systematically identify regulators of inducible gene expression, we performed high-throughput RNAi screening of the Drosophila Metallothionein A (MtnA) promoter. This revealed that the Integrator complex, which has a well-established role in 3' end processing of small nuclear RNAs (snRNAs), attenuates MtnA transcription during copper stress. Integrator complex subunit 11 (IntS11) endonucleolytically cleaves MtnA transcripts, resulting in premature transcription termination and degradation of the nascent RNAs by the RNA exosome, a complex also identified in the screen. Using RNA-seq, we then identified >400 additional Drosophila protein-coding genes whose expression increases upon Integrator depletion. We focused on a subset of these genes and confirmed that Integrator is bound to their 5' ends and negatively regulates their transcription via IntS11 endonuclease activity. Many noncatalytic Integrator subunits, which are largely dispensable for snRNA processing, also have regulatory roles at these protein-coding genes, possibly by controlling Integrator recruitment or RNA polymerase II dynamics. Altogether, our results suggest that attenuation via Integrator cleavage limits production of many full-length mRNAs, allowing precise control of transcription outputs.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , Regulação da Expressão Gênica , Metalotioneína/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/metabolismo , Animais , Linhagem Celular , Cobre/toxicidade , Endorribonucleases/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Ligação Proteica , Clivagem do RNA , Estresse Fisiológico/efeitos dos fármacos
12.
Nucleic Acids Res ; 46(8): 4241-4255, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29471365

RESUMO

Integrator (INT) is a transcriptional regulatory complex associated with RNA polymerase II that is required for the 3'-end processing of both UsnRNAs and enhancer RNAs. Integrator subunits 9 (INTS9) and INTS11 constitute the catalytic core of INT and are paralogues of the cleavage and polyadenylation specificity factors CPSF100 and CPSF73. While CPSF73/100 are known to associate with a third protein called Symplekin, there is no paralog of Symplekin within INT raising the question of how INTS9/11 associate with the other INT subunits. Here, we have identified that INTS4 is a specific and conserved interaction partner of INTS9/11 that does not interact with either subunit individually. Although INTS4 has no significant homology with Symplekin, it possesses N-terminal HEAT repeats similar to Symplekin but also contains a ß-sheet rich C-terminal region, both of which are important to bind INTS9/11. We assess three functions of INT including UsnRNA 3'-end processing, maintenance of Cajal body structural integrity, and formation of histone locus bodies to conclude that INTS4/9/11 are the most critical of the INT subunits for UsnRNA biogenesis. Altogether, these results indicate that INTS4/9/11 compose a heterotrimeric complex that likely represents the Integrator 'cleavage module' responsible for its endonucleolytic activity.


Assuntos
Endorribonucleases/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Drosophila/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/química , RNA Nuclear Pequeno/metabolismo , Técnicas do Sistema de Duplo-Híbrido
13.
Cancer Biol Ther ; 17(2): 129-38, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26650448

RESUMO

Poly (ADP) ribose polymerase (PARP) inhibitors, first evaluated nearly a decade ago, are primarily used in malignancies with known defects in DNA repair genes, such as alterations in breast cancer, early onset 1/2 (BRCA1/2). While no specific mutations in BRCA1/2 have been reported in malignant peripheral nerve sheath tumors (MPNSTs), MPNST cells could be effectively targeted with a PARP inhibitor to drive cells to synthetic lethality due to their complex karyotype and high level of inherent genomic instability. In this study, we assessed the expression levels of PARP1 and PARP2 in MPNST patient tumor samples and correlated these findings with overall survival. We also determined the level of PARP activity in MPNST cell lines. In addition, we evaluated the efficacy of the PARP inhibitor AZD2281 (Olaparib) in MPNST cell lines. We observed decreased MPNST cell proliferation and enhanced apoptosis in vitro at doses similar to, or less than, the doses used in cell lines with established defective DNA repair genes. Furthermore, AZD2281 significantly reduced local growth of MPNST xenografts, decreased the development of macroscopic lung metastases, and increased survival of mice with metastatic disease. Our results suggest that AZD2281 could be an effective therapeutic option in MPNST and should be further investigated for its potential clinical use in this malignancy.


Assuntos
Proliferação de Células/efeitos dos fármacos , Neurilemoma/tratamento farmacológico , Ftalazinas/administração & dosagem , Piperazinas/administração & dosagem , Inibidores de Poli(ADP-Ribose) Polimerases/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Reparo do DNA/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Neurilemoma/genética , Neurilemoma/patologia , Poli(ADP-Ribose) Polimerase-1/biossíntese , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerases/biossíntese , Poli(ADP-Ribose) Polimerases/genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
RNA ; 19(3): 295-305, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23340509

RESUMO

Cytoplasmic poly(A)-binding protein (PABP) C1 recruits different interacting partners to regulate mRNA fate. The majority of PABP-interacting proteins contain a PAM2 motif to mediate their interactions with PABPC1. However, little is known about the regulation of these interactions or the corresponding functional consequences. Through in silico analysis, we found that PAM2 motifs are generally embedded within an extended intrinsic disorder region (IDR) and are located next to cluster(s) of potential serine (Ser) or threonine (Thr) phosphorylation sites within the IDR. We hypothesized that phosphorylation at these Ser/Thr sites regulates the interactions between PAM2-containing proteins and PABPC1. In the present study, we have tested this hypothesis using complementary approaches to increase or decrease phosphorylation. The results indicate that changing the extent of phosphorylation of three PAM2-containing proteins (Tob2, Pan3, and Tnrc6c) alters their ability to interact with PABPC1. Results from experiments using phospho-blocking or phosphomimetic mutants in PAM2-containing proteins further support our hypothesis. Moreover, the phosphomimetic mutations appreciably affected the functions of these proteins in mRNA turnover and gene silencing. Taken together, these results provide a new framework for understanding the roles of intrinsically disordered proteins in the dynamic and signal-dependent control of cytoplasmic mRNA functions.


Assuntos
Motivos de Aminoácidos , Proteína I de Ligação a Poli(A)/química , Proteína I de Ligação a Poli(A)/metabolismo , RNA Mensageiro/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células Cultivadas , Citoplasma/metabolismo , Humanos , Camundongos , Células NIH 3T3 , Fosforilação , Poli A/metabolismo , RNA Mensageiro/genética , Serina/genética , Treonina/genética , Transfecção
15.
Clin Cancer Res ; 18(17): 4633-45, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22821997

RESUMO

PURPOSE: The significance of mTOR activation in uterine leiomyosarcoma (ULMS) and its potential as a therapeutic target were investigated. Furthermore, given that effective therapies likely require combination mTOR blockade with inhibition of other targets, coupled with recent observations suggesting that Aurora-A kinase (Aurk-A) deregulations commonly occur in ULMS, the preclinical impact of dually targeting both pathways was evaluated. EXPERIMENTAL DESIGN: Immunohistochemical staining was used to evaluate expression of activated mTOR components in a large (>200 samples) ULMS tissue microarray. Effects of mTOR blockade (using rapamycin) and Aurk-A inhibition (using MLN8237) alone and in combination on human ULMS cell growth, cell-cycle progression, and apoptosis were assessed in cellular assays. Drug interactions were determined via combination index analyses. The antitumor effects of inhibitors alone or in combination were evaluated in vivo. RESULTS: Enhanced mTOR activation was seen in human ULMS samples. Increased pS6RP and p4EBP1 expression correlated with disease progression; p4EBP1 was found to be an independent prognosticator of patient outcome. Rapamycin inhibited growth and cell-cycle progression of ULMS cell strains/lines in culture. However, only a cytostatic effect on tumor growth was found in vivo. Combining rapamycin with MLN8237 profoundly (and synergistically) abrogated ULMS cells' growth in culture; interestingly, these effects were seen only when MLN8237 was preadministered. This novel therapeutic combination and scheduling regimen resulted in marked tumor growth inhibition in vivo. CONCLUSIONS: mTOR and Aurk-A pathways are commonly deregulated in ULMS. Preclinical data support further exploration of dual mTOR and Aurk-A therapeutic blockade for human ULMS.


Assuntos
Leiomiossarcoma , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Neoplasias Uterinas , Animais , Apoptose/efeitos dos fármacos , Aurora Quinase A , Aurora Quinases , Azepinas/administração & dosagem , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Leiomiossarcoma/tratamento farmacológico , Leiomiossarcoma/metabolismo , Leiomiossarcoma/patologia , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/administração & dosagem , Transdução de Sinais , Sirolimo/administração & dosagem , Serina-Treonina Quinases TOR/antagonistas & inibidores , Transplante Heterólogo , Neoplasias Uterinas/tratamento farmacológico , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia
17.
PLoS One ; 5(4): e10105, 2010 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-20419128

RESUMO

BACKGROUND: Vimentin is a ubiquitous mesenchymal intermediate filament supporting mechano-structural integrity of quiescent cells while participating in adhesion, migration, survival, and cell signaling processes via dynamic assembly/disassembly in activated cells. Soft tissue sarcomas and some epithelial cancers exhibiting "epithelial to mesenchymal transition" phenotypes express vimentin. Withaferin-A, a naturally derived bioactive compound, may molecularly target vimentin, so we sought to evaluate its effects on tumor growth in vitro and in vivo thereby elucidating the role of vimentin in drug-induced responses. METHODS AND FINDINGS: Withaferin-A elicited marked apoptosis and vimentin cleavage in vimentin-expressing tumor cells but significantly less in normal mesenchymal cells. This proapoptotic response was abrogated after vimentin knockdown or by blockade of caspase-induced vimentin degradation via caspase inhibitors or overexpression of mutated caspase-resistant vimentin. Pronounced anti-angiogenic effects of Withaferin-A were demonstrated, with only minimal effects seen in non-proliferating endothelial cells. Moreover, Withaferin-A significantly blocked soft tissue sarcoma growth, local recurrence, and metastasis in a panel of soft tissue sarcoma xenograft experiments. Apoptosis, decreased angiogenesis, and vimentin degradation were all seen in Withaferin-A treated specimens. CONCLUSIONS: In light of these findings, evaluation of Withaferin-A, its analogs, or other anti-vimentin therapeutic approaches in soft tissue sarcoma and "epithelial to mesenchymal transition" clinical contexts is warranted.


Assuntos
Sarcoma/tratamento farmacológico , Vimentina/antagonistas & inibidores , Vitanolídeos/farmacologia , Inibidores da Angiogênese , Animais , Antineoplásicos , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células-Tronco Mesenquimais , Camundongos , Neovascularização Patológica/tratamento farmacológico , Sarcoma/patologia , Vimentina/metabolismo , Vitanolídeos/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Neoplasia ; 10(12): 1393-401, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19048118

RESUMO

Betel quid (BQ)-chewing oral cancer is a prevalent disease in many countries of Southeast Asia. Yet, the precise disease mechanism remains largely unknown. Here, we show that BQ extract-induced cell motility in three oral cancer cells (Ca9-22, SAS, and SCC9) presumably involves the Src family kinases (SFKs). Besides, BQ extract can markedly induce cell migration of wild type mouse embryonic fibroblasts (MEFs) but not MEFs lacking three SFK members, namely, Src, Yes, and Fyn, indicating the requirement of SFKs for BQ-induced cell motility. Betel quid extract can also elevate cellular SFK activities because phosphorylation of tyrosine 416 at the catalytic domain is increased, which in turn promotes phosphorylation of an in vitro substrate, enolase. Furthermore, we identified that areca nut, a major component of BQ, is the key factor accounting for BQ-induced cell migration and invasion through SFKs-mediated signaling pathways. Immunohistochemistry revealed that, particularly in BQ-chewing cases, the activity of SFKs was significantly higher in tumor-adjacent mucosa than that in solid tumor areas (P < .01). These results suggest a possible role of SFKs in tumor-host interface and thus in early tumor invasion in vivo. Consistent with this is the observation that activation of SFKs is colocalized with invasive tumor fronts in oral squamous cell carcinoma. Together, we conclude that SFKs may represent a potential biomarker of invasion and therapeutic target in BQ-induced oral cancer.


Assuntos
Areca , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Bucais/induzido quimicamente , Neoplasias Bucais/patologia , Quinases da Família src/fisiologia , Animais , Carcinógenos , Linhagem Celular Tumoral , Movimento Celular , Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica , Humanos , Camundongos , Neoplasias Bucais/enzimologia , Invasividade Neoplásica , Quinases da Família src/metabolismo
19.
J Bone Miner Res ; 19(7): 1181-90, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15177002

RESUMO

UNLABELLED: Androgens have important effects on the bone metabolism. However, the effect and mechanism of androgen action on the osteoblasts remains unknown. Here we showed that androgens increase phosphorylation and nuclear translocation of Akt. siRNA-AR prevented androgen-induced Akt activation in MC3T3-E1 cells. This suggests that nongenomic androgen activation of Akt is mediated by androgen receptor in osteoblasts. INTRODUCTION: Androgens have important effects on the human skeleton in both males and females. However, the mechanism of androgen action on bone metabolism remains unknown. The aims of this study were to determine the effect and mechanism of androgen action on the osteoblast cells. MATERIALS AND METHODS: Here we showed that 5alpha-dihydrotestosterone (DHT) accelerates cell growth of the MC3T3-E1 cell line in a time- and dose-dependent manner. The specific phosphatidylinositol 3-kinase (PI 3-kinase) inhibitor LY294002 and kinase-deficient Akt mutant can repress the androgen effect on MC3T3-E1 cells. Western blot analysis showed that DHT, 17beta-estradiol, and testosterone (T) induce a rapid and transient phosphorylation of Akt in MC3T3-E1 cells. This activation reached to a plateau after 15 minutes and gradually diminished after 60 minutes of DHT treatment. RESULTS: Fluorescence microscopy showed a distinct increase in immunostaining intensity in the nuclear interior after androgen treatment but no change in the subcellular distribution of Akt when the cells were pretreated with hydroxyflutamide (HF) or LY294002. In addition, small interfering RNA against androgen receptor (siRNA-AR) prevented DHT-induced Akt phosphorylation and cell growth. CONCLUSION: These findings represents the first physiological finding to indicate how steroid hormones such as androgens can mediate the nuclear localization of Akt/PKB in osteoblasts that has previously mainly been linked to growth factor-induced events occurring at the plasma membrane level.


Assuntos
Androgênios/fisiologia , Di-Hidrotestosterona/farmacologia , Osteoblastos/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Androgênios/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular , Núcleo Celular/química , Proliferação de Células , Cromonas/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Camundongos , Morfolinas/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Fosfatidilinositol 3-Quinases/análise , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt , Interferência de RNA , Receptores Androgênicos/efeitos dos fármacos , Receptores Androgênicos/genética , Receptores Androgênicos/fisiologia , Transdução de Sinais/fisiologia , Fosfolipases Tipo C/metabolismo , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...