Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 301
Filtrar
1.
PLoS Genet ; 17(11): e1009868, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34752469

RESUMO

While comprehensive molecular profiling of histone H3.3 mutant pediatric high-grade glioma has revealed extensive dysregulation of the chromatin landscape, the exact mechanisms driving tumor formation remain poorly understood. Since H3.3 mutant gliomas also exhibit high levels of copy number alterations, we set out to address if the H3.3K27M oncohistone leads to destabilization of the genome. Hereto, we established a cell culture model allowing inducible H3.3K27M expression and observed an increase in mitotic abnormalities. We also found enhanced interaction of DNA replication factors with H3.3K27M during mitosis, indicating replication defects. Further functional analyses revealed increased genomic instability upon replication stress, as represented by mitotic bulky and ultrafine DNA bridges. This co-occurred with suboptimal 53BP1 nuclear body formation after mitosis in vitro, and in human glioma. Finally, we observed a decrease in ultrafine DNA bridges following deletion of the K27M mutant H3F3A allele in primary high-grade glioma cells. Together, our data uncover a role for H3.3 in DNA replication under stress conditions that is altered by the K27M mutation, promoting genomic instability and potentially glioma development.


Assuntos
Neoplasias Encefálicas/genética , Replicação do DNA/genética , Instabilidade Genômica , Glioma/genética , Histonas/fisiologia , Neoplasias Encefálicas/patologia , Criança , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Humanos , Mitose/genética
3.
Bull Cancer ; 108(4): 385-398, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33685627

RESUMO

Numerous epigenetic alterations are observed in cancer cells, and dysregulation of mono-ubiquitination of histone H2B (H2Bub1) has often been linked to tumorigenesis. H2Bub1 is a dynamic post-translational histone modification associated with transcriptional elongation and DNA damage response. Histone H2B monoubiquitination occurs in the site of lysine 120, written predominantly by E3 ubiquitin ligases RNF20/RNF40 and deubiquitinated by ubiquitin specific peptidase 22 (USP22). RNF20/40 is often altered in the primary tumors including colorectal cancer, breast cancer, ovarian cancer, prostate cancer, and lung cancer, and the loss of H2Bub1 is usually associated with poor prognosis in tumor patients. The purpose of this review is to summarize the current knowledge of H2Bub1 in transcription, DNA damage response and primary tumors. This review also provides novel options for exploiting the potential therapeutic target H2Bub1 in personalized cancer therapy.


Assuntos
Regulação Neoplásica da Expressão Gênica , Histonas/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Ubiquitinadas/fisiologia , Carcinoma/etiologia , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/terapia , Dano ao DNA , Reparo do DNA , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Progressão da Doença , Humanos , Proteínas de Neoplasias/genética , Neoplasias/etiologia , Neoplasias/genética , Neoplasias/terapia , Medicina de Precisão , Elongação da Transcrição Genética , Ubiquitina Tiolesterase/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
4.
Mol Biol Cell ; 32(8): 712-721, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33596090

RESUMO

Similar to other core biological processes, the vast majority of cell division components are essential for viability across human cell lines. However, recent genome-wide screens have identified a number of proteins that exhibit cell line-specific essentiality. Defining the behaviors of these proteins is critical to our understanding of complex biological processes. Here, we harness differential essentiality to reveal the contributions of the four-subunit centromere-localized CENP-O complex, whose precise function has been difficult to define. Our results support a model in which the CENP-O complex and BUB1 act in parallel pathways to recruit a threshold level of PLK1 to mitotic kinetochores, ensuring accurate chromosome segregation. We demonstrate that targeted changes to either pathway sensitizes cells to the loss of the other component, resulting in cell-state dependent requirements. This approach also highlights the advantage of comparing phenotypes across diverse cell lines to define critical functional contributions and behaviors that could be exploited for the targeted treatment of disease.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Histonas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Centrômero/metabolismo , Centrômero/fisiologia , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/fisiologia , Segregação de Cromossomos , Histonas/genética , Histonas/fisiologia , Humanos , Cinetocoros/fisiologia , Mitose/fisiologia , Ligação Proteica , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Quinase 1 Polo-Like
5.
J Mol Med (Berl) ; 99(3): 425-438, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33484281

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumorigenic virus and the etiological agent of an endothelial tumor (Kaposi's sarcoma) and two B cell proliferative diseases (primary effusion lymphoma and multicentric Castleman's disease). While in patients with late stage of Kaposi's sarcoma the majority of spindle cells are KSHV-infected, viral copies are rapidly lost in vitro, both upon culture of tumor-derived cells or from newly infected endothelial cells. We addressed this discrepancy by investigating a KSHV-infected endothelial cell line in various culture conditions and in tumors of xenografted mice. We show that, in contrast to two-dimensional endothelial cell cultures, KSHV genomes are maintained under 3D cell culture conditions and in vivo. Additionally, an increased rate of newly infected cells was detected in 3D cell culture. Furthermore, we show that the PI3K/Akt/mTOR and ATM/γH2AX pathways are modulated and support an improved KSHV persistence in 3D cell culture. These mechanisms may contribute to the persistence of KSHV in tumor tissue in vivo and provide a novel target for KS specific therapeutic interventions. KEY MESSAGES: In vivo maintenance of episomal KSHV can be mimicked in 3D spheroid cultures 3D maintenance of KSHV is associated with an increased de novo infection frequency PI3K/Akt/mTOR and ATM/ γH2AX pathways contribute to viral maintenance.


Assuntos
Técnicas de Cultura de Células em Três Dimensões , Células Endoteliais/virologia , Herpesvirus Humano 8/fisiologia , Cultura de Vírus/métodos , Animais , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Transformada , Doxiciclina/farmacologia , Células Endoteliais/citologia , Genoma Viral , Xenoenxertos , Histonas/fisiologia , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , Plasmídeos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Sarcoma de Kaposi/virologia , Transdução de Sinais/fisiologia , Esferoides Celulares/transplante , Esferoides Celulares/virologia , Serina-Treonina Quinases TOR/fisiologia , Latência Viral , Liberação de Vírus , Replicação Viral
6.
Ecotoxicol Environ Saf ; 209: 111832, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33383341

RESUMO

Cobalt has been known for its neurotoxicity in numerous studies. However, the molecular mechanism underlying cobalt-induced neurotoxicity remains largely unknown. In this study, two neuroblastoma (SHSY5Y and N2a) cell lines and a phaeochromocytoma (PC12) line were used as in vitro models. Cells were treated for 24 h with 50, 100, 200, 300, 400 µM cobalt chloride (CoCl2) or cultured with 300 µM CoCl2 for 4, 8, 12 and 24 h to investigate the effects of histone acetylation on CoCl2-induced neurodegenerative damages. Our findings demonstrate that CoCl2 suppresses the acetylation of histone H3 and H4 in a time-dependent and dosage-dependent manner. Furthermore, CoCl2 selectively decreases the expression and activity of histone acetyltransferase (HAT) but has no effects on histone deacetylase (HDAC) in SHSY5Y cells. More importantly, we show that 100 ng/mL HDAC inhibitor trichostatin (TSA) pre-treatment partly attenuates 300 µM CoCl2-induced neurodegenerative damages in SHSY5Y cells. Mechanistic analyses show that CoCl2-induced neurodegenerative damages are associated with the dysfunction of APP, BACE1, PSEN1, NEP and HIF-1α genes, whose expression are partly mediated by histone modification. In summary, we demonstrate that histone acetylation is involved in CoCl2-induced neurodegenerative damages. Our study indicates an important connection between histone modification and the pathological process of neurodegenerative damages and provides a mechanism for cobalt-mediated epigenetic regulation.


Assuntos
Cobalto/toxicidade , Histonas/fisiologia , Sistema Nervoso/efeitos dos fármacos , Acetilação/efeitos dos fármacos , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Linhagem Celular Tumoral , Cobalto/metabolismo , Epigênese Genética/efeitos dos fármacos , Inibidores de Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Testes de Toxicidade
7.
Curr Gene Ther ; 21(1): 60-71, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33183201

RESUMO

BACKGROUND: Epigenetic modification pattern is considered as a characteristic feature in blood malignancies. Modifications in the DNA methylation modulators are recurrent in lymphoma and leukemia, so that the distinct methylation pattern defines different types of leukemia. Generally, the role of epigenetics is less understood, and most investigations are focused on genetic abnormalities and cytogenic studies to develop novel treatments for patients with hematologic disorders. Recently, understanding the underlying mechanism of acute lymphoblastic leukemia (ALL), especially epigenetic alterations as a driving force in the development of ALL opens a new era of investigation for developing promising strategy, beyond available conventional therapy. OBJECTIVE: This review will focus on a better understanding of the epigenetic mechanisms in cancer development and progression, with an emphasis on epigenetic alterations in ALL including, DNA methylation, histone modification, and microRNA alterations. Other topics that will be discussed include the use of epigenetic alterations as a promising therapeutic target in order to develop novel, well-suited approaches against ALL. CONCLUSION: According to the literature review, leukemogenesis of ALL is extensively influenced by epigenetic modifications, particularly DNA hyper-methylation, histone modification, and miRNA alteration.


Assuntos
Carcinogênese/genética , Metilação de DNA , Epigênese Genética , Histonas/fisiologia , MicroRNAs , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Humanos , Terapêutica
8.
Mol Cell ; 79(5): 836-845.e7, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32649884

RESUMO

The inactive X chromosome (Xi) is inherently susceptible to genomic aberrations. Replication stress (RS) has been proposed as an underlying cause, but the mechanisms that protect from Xi instability remain unknown. Here, we show that macroH2A1.2, an RS-protective histone variant enriched on the Xi, is required for Xi integrity and female survival. Mechanistically, macroH2A1.2 counteracts its structurally distinct and equally Xi-enriched alternative splice variant, macroH2A1.1. Comparative proteomics identified a role for macroH2A1.1 in alternative end joining (alt-EJ), which accounts for Xi anaphase defects in the absence of macroH2A1.2. Genomic instability was rescued by simultaneous depletion of macroH2A1.1 or alt-EJ factors, and mice deficient for both macroH2A1 variants harbor no overt female defects. Notably, macroH2A1 splice variant imbalance affected alt-EJ capacity also in tumor cells. Together, these findings identify macroH2A1 splicing as a modulator of genome maintenance that ensures Xi integrity and may, more broadly, predict DNA repair outcome in malignant cells.


Assuntos
Processamento Alternativo , Reparo do DNA , Epigênese Genética , Instabilidade Genômica , Histonas/fisiologia , Anáfase , Animais , Linhagem Celular , Instabilidade Cromossômica , Cromossomos Humanos X , Feminino , Histonas/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
Elife ; 92020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32391789

RESUMO

Neutrophils are important innate immune cells that tackle invading pathogens with different effector mechanisms. They acquire this antimicrobial potential during their maturation in the bone marrow, where they differentiate from hematopoietic stem cells in a process called granulopoiesis. Mature neutrophils are terminally differentiated and short-lived with a high turnover rate. Here, we show a critical role for linker histone H1 on the differentiation and function of neutrophils using a genome-wide CRISPR/Cas9 screen in the human cell line PLB-985. We systematically disrupted expression of somatic H1 subtypes to show that individual H1 subtypes affect PLB-985 maturation in opposite ways. Loss of H1.2 and H1.4 induced an eosinophil-like transcriptional program, thereby negatively regulating the differentiation into the neutrophil lineage. Importantly, H1 subtypes also affect neutrophil differentiation and the eosinophil-directed bias of murine bone marrow stem cells, demonstrating an unexpected subtype-specific role for H1 in granulopoiesis.


Assuntos
Células-Tronco Hematopoéticas/citologia , Histonas/fisiologia , Neutrófilos/citologia , Animais , Medula Óssea/fisiologia , Sistemas CRISPR-Cas , Diferenciação Celular , Linhagem Celular , Eosinófilos/citologia , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Hematopoese , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Fatores de Transcrição/fisiologia
10.
Trends Cancer ; 6(3): 247-260, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32101727

RESUMO

Alternative lengthening of telomeres (ALT) is a mechanism of telomere maintenance that is observed in many of the most recalcitrant cancer subtypes. Telomeres in ALT cancer cells exhibit a distinctive nucleoprotein architecture shaped by the mismanagement of chromatin that fosters cycles of DNA damage and replicative stress that activate homology-directed repair (HDR). Mutations in specific chromatin-remodeling factors appear to be key determinants of the emergence and survival of ALT cancer cells. However, these may represent vulnerabilities for the targeted elimination of ALT cancer cells that infiltrate tissues and organs to become devastating tumors. In this review we examine recent findings that provide new insights into the factors and mechanisms that mediate telomere length maintenance and survival of ALT cancer cells.


Assuntos
Neoplasias/genética , Homeostase do Telômero , Cromatina/ultraestrutura , Evolução Clonal , Proteínas Correpressoras/antagonistas & inibidores , Proteínas Correpressoras/fisiologia , Dano ao DNA , Reparo do DNA , Replicação do DNA , DNA de Neoplasias/metabolismo , DNA de Neoplasias/ultraestrutura , Histonas/fisiologia , Recombinação Homóloga , Humanos , Modelos Genéticos , Chaperonas Moleculares/antagonistas & inibidores , Chaperonas Moleculares/fisiologia , Mutação , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Neoplasias/ultraestrutura , Conformação de Ácido Nucleico , Telomerase/genética , Telomerase/fisiologia , Proteína Nuclear Ligada ao X/antagonistas & inibidores , Proteína Nuclear Ligada ao X/fisiologia
11.
Proc Natl Acad Sci U S A ; 116(44): 22152-22157, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31611394

RESUMO

A lysine-to-methionine mutation at lysine 27 of histone 3 (H3K27M) has been shown to promote oncogenesis in a subset of pediatric gliomas. While there is evidence that this "oncohistone" mutation acts by inhibiting the histone methyltransferase PRC2, the details of this proposed mechanism nevertheless continue to be debated. Recent evidence suggests that PRC2 must simultaneously bind both H3K27M and H3K27me3 to experience competitive inhibition of its methyltransferase activity. In this work, we used PRC2 inhibitor treatments in a transgenic H3K27M cell line to validate this dependence in a cellular context. We further used designer chromatin inhibitors to probe the geometric constraints of PRC2 engagement of H3K27M and H3K27me3 in a biochemical setting. We found that PRC2 binds to a bivalent inhibitor unit consisting of an H3K27M and an H3K27me3 nucleosome and exhibits a distance dependence in its affinity for such an inhibitor, which favors closer proximity of the 2 nucleosomes within a chromatin array. Together, our data precisely delineate fundamental aspects of the H3K27M inhibitor and support a model wherein PRC2 becomes trapped at H3K27M-H3K27me3 boundaries.


Assuntos
Histonas/genética , Complexo Repressor Polycomb 2/fisiologia , Substituição de Aminoácidos , Sítios de Ligação , Linhagem Celular , Histona Metiltransferases/antagonistas & inibidores , Histona Metiltransferases/química , Histonas/química , Histonas/fisiologia , Humanos , Modelos Moleculares , Complexo Repressor Polycomb 2/química , Complexo Repressor Polycomb 2/metabolismo
12.
Restor Neurol Neurosci ; 37(4): 291-313, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31227672

RESUMO

BACKGROUND: We have shown that histone H1 is a binding partner for polysialic acid (PSA) and that it improves functional recovery, axon regrowth/sprouting, and target reinnervation after mouse femoral nerve injury. OBJECTIVE: Here, we analyzed whether histone H1 affects functional recovery, axon regrowth/sprouting, and target reinnervation after spinal cord injury of adult mice. Furthermore, we tested in vitro histone H1's effect on astrocytic gene expression, cell shape and migration as well as on cell survival of cultured motoneurons. METHODS: We applied histone H1 to compressed spinal cord and determined functional recovery and number of fibrillary acidic protein (GFAP)- and neuron-glial antigen 2 (NG2)- positive glial cells, which contribute to glial scarring. Histone H1's effect on migration of astrocytes, astrocytic gene expression and motoneuronal survival was determined using scratch-wounded astroglial monolayer cultures, astrocyte cultures for microarray analysis, and motoneuron cell culture under oxidative stress conditions, respectively. RESULTS: Histone H1 application improves locomotor functions and enhances monoaminergic and cholinergic reinnervation of the spinal cord. Expression levels of GFAP and NG2 around the lesion site were decreased in histone H1-treated mice relative to vehicle-treated mice six weeks after injury. Histone H1 reduced astrocytic migration, changed the shape of GFAP- and NG2-positive glial cells and altered gene expression. Gene ontology enrichment analysis indicated that in particular genes coding for proteins involved in proliferation, differentiation, migration and apoptosis are dysregulated. The up- and down-regulation of distinct genes was confirmed by qPCR and Western blot analysis. Moreover, histone H1 reduced hydrogen peroxide-induced cell death of cultured motoneurons. CONCLUSIONS: The combined observations indicate that histone H1 locally applied to the lesion site, improves regeneration after spinal cord injury. Some of these beneficial functions of histone H1 in vivo and in vitro can be attributed to its interaction with PSA-carrying neural cell adhesion molecule.


Assuntos
Astrócitos/fisiologia , Axônios/fisiologia , Movimento Celular/fisiologia , Expressão Gênica/fisiologia , Histonas/fisiologia , Locomoção/fisiologia , Neurônios Motores/fisiologia , Regeneração Nervosa/fisiologia , Neuroglia/fisiologia , Recuperação de Função Fisiológica/fisiologia , Ácidos Siálicos/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Animais , Astrócitos/efeitos dos fármacos , Axônios/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Histonas/farmacologia , Locomoção/efeitos dos fármacos , Camundongos , Neurônios Motores/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico
13.
PLoS Biol ; 17(6): e3000331, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31226107

RESUMO

Eukaryotes have evolved elaborate mechanisms to ensure that chromosomes segregate with high fidelity during mitosis and meiosis, and yet specific aneuploidies can be adaptive during environmental stress. Here, we identify a chromatin-based system required for inducible aneuploidy in a human pathogen. Candida albicans utilizes chromosome missegregation to acquire tolerance to antifungal drugs and for nonmeiotic ploidy reduction after mating. We discovered that the ancestor of C. albicans and 2 related pathogens evolved a variant of histone 2A (H2A) that lacks the conserved phosphorylation site for kinetochore-associated Bub1 kinase, a key regulator of chromosome segregation. Using engineered strains, we show that the relative gene dosage of this variant versus canonical H2A controls the fidelity of chromosome segregation and the rate of acquisition of tolerance to antifungal drugs via aneuploidy. Furthermore, whole-genome chromatin precipitation analysis reveals that Centromere Protein A/ Centromeric Histone H3-like Protein (CENP-A/Cse4), a centromeric histone H3 variant that forms the platform of the eukaryotic kinetochore, is depleted from tetraploid-mating products relative to diploid parents and is virtually eliminated from cells exposed to aneuploidy-promoting cues. We conclude that genetically programmed and environmentally induced changes in chromatin can confer the capacity for enhanced evolvability via chromosome missegregation.


Assuntos
Proteína Centromérica A/metabolismo , Segregação de Cromossomos/fisiologia , Histonas/metabolismo , Aneugênicos/metabolismo , Aneuploidia , Candida albicans/genética , Candida albicans/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrômero/metabolismo , Proteína Centromérica A/fisiologia , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Evolução Molecular , Histonas/fisiologia , Cinetocoros/metabolismo , Meiose , Mitose , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo
14.
Life Sci ; 230: 89-96, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31129138

RESUMO

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief. Given the comments of Dr Elisabeth Bik regarding this article "… the Western blot bands in all 400+ papers are all very regularly spaced and have a smooth appearance in the shape of a dumbbell or tadpole, without any of the usual smudges or stains. All bands are placed on similar looking backgrounds, suggesting they were copy/pasted from other sources, or computer generated", the journal requested the authors to provide the raw data. However, the authors were not able to fulfil this request and therefore the Editor-in-Chief decided to retract the article.


Assuntos
Neoplasias Colorretais/metabolismo , Histonas/metabolismo , Histonas/fisiologia , Acilação , Carcinogênese , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/genética , Sobrevivência Celular , Neoplasias Colorretais/fisiopatologia , Progressão da Doença , Fator 15 de Diferenciação de Crescimento/metabolismo , Histona Desacetilase 2/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais , Proteínas ras/fisiologia
15.
Sci Rep ; 9(1): 3831, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30846735

RESUMO

The mRNA processing and export factor, Iws1, interacts with the histone H3/H4 chaperone, Spt6 (Supt6 in mouse gene ontology) and recruits the lysine methyltransferase, Setd2, to chromatin to regulate H3K36me3. This recruitment is known to be crucial for pre-mRNA splicing and Iws1 has been shown to interact with REF1/Aly to mediate mRNA export. However, the role of this complex has not yet been examined in embryonic development. Here, we show that knockdown of either Iws1 or Supt6 blocked embryo development, primarily at the 8/16-cell stage, indicating that Iws1 and Supt6 are crucial for mouse preimplantation development. In the knockdown embryos, we observed decreases in pre-mRNA splicing, mRNA export and the expression of the lineage-specific transcription factor, Nanog. We found that either Iws1 or Supt6 are required for H3K36 trimethylation and that concurrent knockdown of both Iws1 and Supt6 blocks embryonic development at the 2-cell stage. We show that H3K36me3 is modulated by the Pi3k/Akt pathway, as inhibition of this pathway reduced the global level of H3K36me3 while activation of the pathway increased the level of this modification in 2-cell embryos. We observed that Iws1 interacts with nuclear Akt in early embryos, and herein propose that Akt modulates H3K36me3 through interaction with Iws1. Together, our results indicate that the Iws1 and Supt6 play crucial roles in embryonic genome activation, lineage specification, and histone modification during mouse early development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Código das Histonas , Histonas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Ligação a RNA/fisiologia , Transdução de Sinais , Fatores de Transcrição/fisiologia , Animais , Metilação de DNA , Feminino , Técnicas de Silenciamento de Genes , Histonas/fisiologia , Lisina , Masculino , Camundongos , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição/metabolismo
16.
Cancer Lett ; 448: 61-69, 2019 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-30742944

RESUMO

Giant cell tumors of bone (GCTB) are semi-malignant tumors associated with extensive osteolytic defects and massive bone destructions. They display a locally aggressive behavior and a very high recurrence rate. Recently, a single mutation has been identified in GCTB affecting the H3F3A gene coding for the histone variant H3.3 (H3.3-G34W). The aim of this study was to investigate whether H3.3-G34W is sufficient to drive tumorigenesis in GCTB. Initially, we confirmed the high frequency of this mutation in 94% of 84 analyzed tissue samples. Using a siRNA based approach we could selectively knockdown H3.3-G34W in primary neoplastic stromal cells isolated from tumor tissue (GCTSC). H3.3-G34W knockdown caused a significant inhibition of cell proliferation, migration and colony formation capacity in vitro. Xenotransplantation of GCTSCs onto the chorioallantoic membrane of fertilized chicken eggs further demonstrated a significant impact of H3.3-G34W knockdown on tumor engraftment and growth in vivo. Our data indicate that H3.3-G34W is sufficient to drive tumorigenesis in GCTB. Apart from the application of H3.3-G34W screening as diagnostic tool, our data suggest that H3.3-G4W represents a promising target for the development of new GCTB therapies.


Assuntos
Neoplasias Ósseas/patologia , Tumor de Células Gigantes do Osso/patologia , Histonas/fisiologia , Células Estromais/patologia , Neoplasias Ósseas/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Técnicas de Silenciamento de Genes , Tumor de Células Gigantes do Osso/genética , Histonas/genética , Humanos , Fenótipo
17.
Cancer Res ; 79(7): 1305-1317, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30718359

RESUMO

Long noncoding RNAs (lncRNA) play critical roles in the development of cancer, including hepatocellular carcinoma (HCC). However, the mechanisms underlying their deregulation remain largely unexplored. In this study, we report that two lncRNAs frequently downregulated in HCC function as tumor suppressors and are epigenetically silenced by histone methyltransferase EZH2. lncRNAs TCAM1P-004 and RP11-598D14.1 were inhibited by EZH-mediated trimethylation of H3K27me3 at their promoters. Downregulation of TCAM1P-004 and RP11-598D14.1 was frequently observed in HCC tumors compared with adjacent normal tissues. Both lncRNAs inhibited cell growth, cell survival, and transformation in HCC cells in vitro as well as tumor formation in vivo. Using RNA pull-down and mass spectrometry, we demonstrated that TCAM1P-004 bound IGF2BP1 and HIST1H1C, whereas RP11-598D14.1 bound IGF2BP1 and STAU1. These lncRNA-protein interactions were critical in regulating p53, MAPK, and HIF1α pathways that promoted cell proliferation in HCC. Overexpression of EZH2 was critical in repressing TCAM1P-004 and RP11-598D14.1, and EZH2-TCAM1P-004/RP11-598D14.1-regulated pathways were prevalent in human HCC. Aberrant suppression of TCAM1P-004 and RP11-598D14.1 led to loss of their tumor-suppressive effects by disrupting the interaction with IGF2BP1, HIST1H1C, and STAU1, which in turn promoted HCC development and progression. Collectively, these findings demonstrate the role of TCAMP1P-004 and RP11-598D14.1 in suppressing tumor growth and suggest that EZH2 may serve as a therapeutic target in HCC. SIGNIFICANCE: EZH2-mediated loss of lncRNAs TCAM1P-004 and RP11-598D14.1 hinders the formation of tumor suppressor lncRNA-protein complexes and subsequently promotes HCC growth.


Assuntos
Carcinoma Hepatocelular/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigênese Genética , Inativação Gênica , Genoma Humano , Neoplasias Hepáticas/genética , RNA Longo não Codificante/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Genes Supressores de Tumor , Histonas/fisiologia , Humanos , Neoplasias Hepáticas/patologia , Metilação , Proteínas de Ligação a RNA/fisiologia
18.
Urol J ; 16(3): 289-294, 2019 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-30206922

RESUMO

PURPOSE: To investigate the effects of heroin on sperm parameters, histone-to-protamine transition ratios in mature sperm, and serum reproductive hormone levels in active heroin users. MATERIALS AND METHODS: Semen and blood samples were collected from 25 men who used only heroin for at least 12 months and the same number healthy men who did not use any drugs and did not suffer from infertility problems. Computer-based analysis, Aniline blue staining, and hormonal assessment were performed to provide valuable new information on the relationship between addiction and semen profile and serum reproductive hor-mone levels. RESULTS: Our finding showed that semen pH (7.8 vs. 7.75), sperm motility (42.93 ± 3.89% vs. 68.9 ± 2.68%), and viability (73.27 ± 3.85% vs. 86.48 ± 1.05%), and sperm histone replacement abnormalities (32.33 ± 10.89% vs. 5.56 ± 0.85%) were significant differences in addicted group vs. non-exposed ones (P ? .05). In addition, serum sex hormone levels were not significantly differed between groups. There was a correlation between the amount of daily heroin consumption and LH level. We also observed that duration of drug dependence is correlated with sperm abnormalities. CONCLUSION: We concluded that heroin consumption affect sperm maturities such as histone-to-protamine ratio and impair semen profile in general and particularly sperm morphology and motility. Heroin may be considered as one of the idiopathic male infertility reason.


Assuntos
Hormônios Esteroides Gonadais/sangue , Dependência de Heroína , Análise do Sêmen , Espermatogênese , Adulto , Proteínas Cromossômicas não Histona/fisiologia , Dependência de Heroína/sangue , Dependência de Heroína/complicações , Histonas/fisiologia , Humanos , Masculino , Protaminas
19.
Cells ; 8(1)2018 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-30577532

RESUMO

In acute pancreatitis, histones are released by infiltrating neutrophils, but how histones modulate pancreatic acinar cell function has not been investigated. We have examined histone modulation of rat pancreatic acini and pancreatic acinar tumor cell AR4-2J by calcium imaging. Histones were found to have no effect on calcium in pancreatic acini but blocked calcium oscillations induced by cholecystokinin or acetylcholine. Both mixed (Hx) and individual (H1, H2A, H2B, H3, H4) histones induced calcium oscillations in AR4-2J. RT-PCR and Western blot verified the expression of histone-targeted Toll-like receptor (TLR) 2, 4 and 9. Immunocytochemistry identified TLR2/TLR4 on apical plasma membrane and TLR9 in zymogen granule regions in pancreatic acini. TLR2 was found on neighboring and TLR9 on peripheral plasma membranes, but TLR4 was in the nucleus in AR4-2J clusters. Neither TLR2 agonist zymosan-A nor TLR4 agonist lipopolysaccharide had any effect on calcium, but TLR9 agonist ODN1826 induced calcium oscillations; TLR9 antagonist ODN2088 blocked H4-induced calcium oscillations in AR4-2J, which also disappeared after treatment of AR4-2J with glucocorticoid dexamethasone, with concurrent TLR9 migration from plasma membrane to cell interiors. TLR9 down regulation with siRNA suppressed H4-induced calcium oscillations. These data together suggest that extracellular histones activate plasma membrane TLR9 to trigger calcium oscillations in AR4-2J cells.


Assuntos
Células Acinares/metabolismo , Sinalização do Cálcio , Carcinoma de Células Acinares/metabolismo , Histonas/fisiologia , Pâncreas Exócrino , Neoplasias Pancreáticas/metabolismo , Receptor Toll-Like 9/metabolismo , Células Acinares/citologia , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Pâncreas Exócrino/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/metabolismo , Receptor Toll-Like 9/antagonistas & inibidores
20.
Cell ; 175(1): 186-199.e19, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30220457

RESUMO

Mutations or aberrant upregulation of EZH2 occur frequently in human cancers, yet clinical benefits of EZH2 inhibitor (EZH2i) remain unsatisfactory and limited to certain hematological malignancies. We profile global posttranslational histone modification changes across a large panel of cancer cell lines with various sensitivities to EZH2i. We report here oncogenic transcriptional reprogramming mediated by MLL1's interaction with the p300/CBP complex, which directs H3K27me loss to reciprocal H3K27ac gain and restricts EZH2i response. Concurrent inhibition of H3K27me and H3K27ac results in transcriptional repression and MAPK pathway dependency in cancer subsets. In preclinical models encompassing a broad spectrum of EZH2-aberrant solid tumors, a combination of EZH2 and BRD4 inhibitors, or a triple-combination including MAPK inhibition display robust efficacy with very tolerable toxicity. Our results suggest an attractive precision treatment strategy for EZH2-aberrant tumors on the basis of tumor-intrinsic MLL1 expression and concurrent inhibition of epigenetic crosstalk and feedback MAPK activation.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Histona-Lisina N-Metiltransferase/fisiologia , Proteína de Leucina Linfoide-Mieloide/fisiologia , Animais , Carcinogênese/genética , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Epigênese Genética/genética , Epigenômica/métodos , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Código das Histonas/efeitos dos fármacos , Código das Histonas/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Histonas/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Mutação , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Complexo Repressor Polycomb 2/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Ativação Transcricional , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Fatores de Transcrição de p300-CBP/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA